1
|
Gao Z. New insights into Smad3 in cardiac fibrosis. Gene 2025; 952:149418. [PMID: 40089084 DOI: 10.1016/j.gene.2025.149418] [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: 01/26/2025] [Revised: 03/04/2025] [Accepted: 03/13/2025] [Indexed: 03/17/2025]
Abstract
Damage to myocardial tissues, leading to myocardial fibrosis, is a significant pathological hallmark across various heart diseases. SMAD3, a central transcriptional regulator within the transforming growth factor-beta (TGF-β) signaling pathway, plays a pivotal role in the pathological progression of myocardial fibrosis and cardiac remodeling. It intricately regulates physiological and pathological processes encompassing cell proliferation, differentiation, tissue repair, and fibrosis. Notably, SMAD3 exerts crucial influences in myocardial fibrosis subsequent to myocardial infarction, pressure overload-induced myocardial fibrosis, diabetic cardiomyopathy (DCM), aging-associated cardiac fibrosis and myocarditis-related myocardial fibrosis. The targeted modulation of genes or the utilization of compounds, including traditional Chinese medicine (paeoniflorin, baicalin, and genistein et al.) and other pharmaceutical agents that modulate SMAD3, may offer avenues for restraining the pathological cascade of myocardial fibrosis. Consequently, targeted regulation of SMAD3 associated with myocardial fibrosis may herald novel therapeutic paradigms for ameliorating myocardial diseases.
Collapse
Affiliation(s)
- Zhen Gao
- Liaocheng Vocational and Technical College, Shandong, China.
| |
Collapse
|
2
|
Goumtsa AF, Nguelefack-Mbuyo EP, Nokam F, Koho CW, Dial CMM, Nguelefack TB. Antihypertrophic effects of the seed ethanolic extract of Aframomum pruinosum Gagnep. (Zingiberaceae) against isoproterenol-induced cardiac hypertrophy in male Wistar rat. Toxicol Rep 2025; 14:101855. [PMID: 39802597 PMCID: PMC11719414 DOI: 10.1016/j.toxrep.2024.101855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025] Open
Abstract
The seeds of Aframomum pruinosum are popularly used in the management of cardiovascular conditions. This study was undertaken to evaluate the capacity of the seed ethanolic extract of A. pruinosum (EE) to prevent the development of cardiac hypertrophy in rats. Isoproterenol (0.3 mg/kg/day, sc) was injected to male rats alone or concomitantly with EE (37.5, 75, or 150 mg/kg, per os) or propranolol (20 mg/kg/day, per os) for 7 consecutive days and systolic blood pressure (SBP), diastolic blood pressure (DBP), and heart rate measurements were performed. Cardiac homogenates were used to assay myeloperoxidase (MPO), superoxide dismutase (SOD), catalase, nitric oxide (NO) and reduced glutathione (GSH). Also, sections of heart tissue were stained with Hematoxylin-Eosin, Masson trichrome, or for immunohistological labelling of atrial natriuretic peptide (ANP). Isoproterenol administration caused a decline in SBP and DBP (p < 0.001). Heart rate, cardiac mass, cardiomyocyte surface, and MPO levels were significantly (p < 0.001) increased. All these alterations were significantly prevented (p < 0.01 and p < 0.001) by EE. EE inhibited immune cell infiltration and cardiac fibrosis elicited by isoproterenol injection. The overexpression of ANP in the atrium and ventricle induced by the isoproterenol was significantly (p < 0.001) prevented by EE. EE possesses antihypertrophic effect against isoproterenol-induced cardiac hypertrophy that may result from its antifibrotic, anti-inflammatory properties, as well as its capacity to down regulate the expression of ANP.
Collapse
Affiliation(s)
| | | | - Florence Nokam
- University of Dschang, Department of Animal Biology, Dschang, Dschang, Cameroon
| | - Cédric Wamba Koho
- University of Dschang, Department of Animal Biology, Dschang, Dschang, Cameroon
| | | | | |
Collapse
|
3
|
Stankovic S, Mutavdzin Krneta S, Djuric D, Milosevic V, Milenkovic D. Plant Polyphenols as Heart's Best Friends: From Health Properties, to Cellular Effects, to Molecular Mechanisms of Action. Int J Mol Sci 2025; 26:915. [PMID: 39940685 PMCID: PMC11816429 DOI: 10.3390/ijms26030915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 12/22/2024] [Accepted: 01/18/2025] [Indexed: 02/16/2025] Open
Abstract
Polyphenols are micronutrients found in fruits, vegetables, tea, coffee, cocoa, medicinal herbs, fish, crustaceans, and algae. They can also be synthesized using recombinant microorganisms. Interest in plant-derived natural compounds has grown due to their potential therapeutic effects with minimal side effects. This is particularly important as the aging population faces increasing rates of chronic diseases such as cancer, diabetes, arthritis, cardiovascular, and neurological disorders. Studies have highlighted polyphenols' capacity to reduce risk factors linked to the onset of chronic illnesses. This narrative review discusses polyphenol families and their metabolism, and the cardioprotective effects of polyphenols evidenced from in vitro studies, as well as from in vivo studies, on different animal models of cardiac disease. This study also explores the molecular mechanisms underlying these benefits. Current research suggests that polyphenols may protect against ischemia, hypertension, cardiac hypertrophy, heart failure, and myocardial injury through complex mechanisms, including epigenetic and genomic modulation. However, further studies under nutritionally and physiologically relevant conditions, using untargeted multigenomic approaches, are needed to more comprehensively elucidate these mechanisms and firmly prove the cardioprotective effects of polyphenols.
Collapse
Affiliation(s)
- Sanja Stankovic
- Center for Medical Biochemistry, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Slavica Mutavdzin Krneta
- Institute of Medical Physiology “Richard Burian”, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (S.M.K.); (D.D.)
| | - Dragan Djuric
- Institute of Medical Physiology “Richard Burian”, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (S.M.K.); (D.D.)
| | - Verica Milosevic
- Department of Anatomy, Faculty of Medicine, University of Niš, 18000 Nis, Serbia;
| | - Dragan Milenkovic
- Department of Food, Bioprocessing and Nutrition Sciences, Plants for Human Health Institute, North Carolina State University, Kannapolis, NC 28081, USA
- Department of Nutrition, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
4
|
Bader Eddin L, Nagoor Meeran MF, Kumar Jha N, Goyal SN, Ojha S. Isoproterenol mechanisms in inducing myocardial fibrosis and its application as an experimental model for the evaluation of therapeutic potential of phytochemicals and pharmaceuticals. Animal Model Exp Med 2025; 8:67-91. [PMID: 39690876 PMCID: PMC11798751 DOI: 10.1002/ame2.12496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 08/14/2024] [Indexed: 12/19/2024] Open
Abstract
Cardiac injury initiates repair mechanisms and results in cardiac remodeling and fibrosis, which appears to be a leading cause of cardiovascular diseases. Cardiac fibrosis is characterized by the accumulation of extracellular matrix proteins, mainly collagen in the cardiac interstitium. Many experimental studies have demonstrated that fibrotic injury in the heart is reversible; therefore, it is vital to understand different molecular mechanisms that are involved in the initiation, progression, and resolution of cardiac fibrosis to enable the development of antifibrotic agents. Of the many experimental models, one of the recent models that has gained renewed interest is isoproterenol (ISP)-induced cardiac fibrosis. ISP is a synthetic catecholamine, sympathomimetic, and nonselective β-adrenergic receptor agonist. The overstimulated and sustained activation of β-adrenergic receptors has been reported to induce biochemical and physiological alterations and ultimately result in cardiac remodeling. ISP has been used for decades to induce acute myocardial infarction. However, the use of low doses and chronic administration of ISP have been shown to induce cardiac fibrosis; this practice has increased in recent years. Intraperitoneal or subcutaneous ISP has been widely used in preclinical studies to induce cardiac remodeling manifested by fibrosis and hypertrophy. The induced oxidative stress with subsequent perturbations in cellular signaling cascades through triggering the release of free radicals is considered the initiating mechanism of myocardial fibrosis. ISP is consistently used to induce fibrosis in laboratory animals and in cardiomyocytes isolated from animals. In recent years, numerous phytochemicals and synthetic molecules have been evaluated in ISP-induced cardiac fibrosis. The present review exclusively provides a comprehensive summary of the pathological biochemical, histological, and molecular mechanisms of ISP in inducing cardiac fibrosis and hypertrophy. It also summarizes the application of this experimental model in the therapeutic evaluation of natural as well as synthetic compounds to demonstrate their potential in mitigating myocardial fibrosis and hypertrophy.
Collapse
Affiliation(s)
- Lujain Bader Eddin
- Department of Pharmacology and Therapeutics, College of Medicine and Health SciencesUAE UniversityAl AinUnited Arab Emirates
| | - Mohamed Fizur Nagoor Meeran
- Department of Pharmacology and Therapeutics, College of Medicine and Health SciencesUAE UniversityAl AinUnited Arab Emirates
| | - Niraj Kumar Jha
- School of Bioengineering & BiosciencesLovely Professional UniversityPhagwaraIndia
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha UniversityChennaiIndia
| | - Samer N. Goyal
- Shri Vile Parle Kelvani Mandal's Institute of PharmacyDhuleMaharashtraIndia
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health SciencesUAE UniversityAl AinUnited Arab Emirates
- Zayed Bin Sultan Center for Health SciencesUnited Arab Emirates UniversityAl AinUnited Arab Emirates
| |
Collapse
|
5
|
Chakraborty P, Dewanjee S. Unrevealing the mechanisms behind the cardioprotective effect of wheat polyphenolics. Arch Toxicol 2024; 98:3543-3567. [PMID: 39215839 DOI: 10.1007/s00204-024-03850-y] [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: 06/05/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Cardiovascular diseases pose a major threat to both life expectancy and quality of life worldwide, and a concerning level of disease burden has been attained, particularly in middle- and low-income nations. Several drugs presently in use lead to multiple adverse events. Thus, it is urgently needed to develop safe, affordable, and effective management of cardiovascular diseases. Emerging evidence reveals a positive association between polyphenol consumption and cardioprotection. Whole wheat grain and allied products are good sources of polyphenolic compounds bearing enormous cardioprotective potential. Polyphenolic extract of the entire wheat grain contains different phenolic compounds viz. ferulic acid, caffeic acid, chlorogenic acid, p-coumaric acid, sinapic acid, syringic acid, vanillic acid, apigenin, quercetin, luteolin, etc. which exert cardioprotection by reducing oxidative stress and interfering with different toxicological processes. The antioxidant capacity has been thought to exert the cardioprotective mechanism of wheat grain polyphenolics, which predominantly suppresses oxidative stress, inflammation and fibrosis by downregulating several pathogenic signaling events. However, the combined effect of polyphenolics appears to be more prominent than that of a single molecule, which might be attained due to the synergy resulting in multimodal cardioprotective benefits from multiple phenolics. The current article covers the bioaccessibility and possible effects of wheat-derived polyphenolics in protecting against several cardiovascular disorders. This review discusses the mechanistic pharmacology of individual wheat polyphenols on the cardiovascular system. It also highlights the comparative superiority of polyphenolic extracts over a single phenolic.
Collapse
Affiliation(s)
- Pratik Chakraborty
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India.
| |
Collapse
|
6
|
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
|
7
|
Shokri F, Ramezani-Aliakbari K, Zarei M, Komaki A, Raoufi S, Naddaf H, Ramezani-Aliakbari F. Cardioprotective effect of Vitamin D on cardiac hypertrophy through improvement of mitophagy and apoptosis in an experimental rat model of levothyroxine -induced hyperthyroidism. Mol Biol Rep 2024; 51:969. [PMID: 39249564 DOI: 10.1007/s11033-024-09897-5] [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: 06/28/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Mitochondria are known to be involved in mediating the calorigenic effects of thyroid hormones. With an abundance of these hormones, alterations in energy metabolism and cellular respiration take place, leading to the development of cardiac hypertrophy. Vitamin D has recently gained attention due to its involvement in the regulation of mitochondrial function, demonstrating promising potential in preserving the integrity and functionality of the mitochondrial network. The present study aimed to investigate the therapeutic potential of Vitamin D on cardiac hypertrophy induced by hyperthyroidism, with a focus on the contributions of mitophagy and apoptosis as possible underlying molecular mechanisms. METHODS AND RESULTS The rats were divided into three groups: control; hyperthyroid; hyperthyroid + Vitamin D. Hyperthyroidism was induced by Levothyroxine administration for four weeks. Serum thyroid hormones levels, myocardial damage markers, cardiac hypertrophy indices, and histological examination were assessed. The assessment of Malondialdehyde (MDA) levels and the expression of the related genes were conducted using heart tissue samples. Vitamin D pretreatment exhibited a significant improvement in the hyperthyroidism-induced decline in markers indicative of myocardial damage, oxidative stress, and indices of cardiac hypertrophy. Vitamin D pretreatment also improved the downregulation observed in myocardial expression levels of genes involved in the regulation of mitophagy and apoptosis, including PTEN putative kinase 1 (PINK1), Mitofusin-2 (MFN2), Dynamin-related Protein 1 (DRP1), and B cell lymphoma-2 (Bcl-2), induced by hyperthyroidism. CONCLUSIONS These results suggest that supplementation with Vitamin D could be advantageous in preventing the progression of cardiac hypertrophy and myocardial damage.
Collapse
Affiliation(s)
- Farid Shokri
- Department of Physiology, School of medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Mohammad Zarei
- Department of Physiology, School of medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Safoura Raoufi
- Department of Physiology, School of medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hanieh Naddaf
- Core facility lab, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fatemeh Ramezani-Aliakbari
- Department of Physiology, School of medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
8
|
Ott C. Mapping the interplay of immunoproteasome and autophagy in different heart failure phenotypes. Free Radic Biol Med 2024; 218:149-165. [PMID: 38570171 DOI: 10.1016/j.freeradbiomed.2024.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/25/2024] [Accepted: 03/30/2024] [Indexed: 04/05/2024]
Abstract
Proper protein degradation is required for cellular protein homeostasis and organ function. Particularly, in post-mitotic cells, such as cardiomyocytes, unbalanced proteolysis due to inflammatory stimuli and oxidative stress contributes to organ dysfunction. To ensure appropriate protein turnover, eukaryotic cells exert two main degradation systems, the ubiquitin-proteasome-system and the autophagy-lysosome-pathway. It has been shown that proteasome activity affects the development of cardiac dysfunction differently, depending on the type of heart failure. Studies analyzing the inducible subtype of the proteasome, the immunoproteasome (i20S), demonstrated that the i20S plays a double role in diseased hearts. While i20S subunits are increased in cardiac hypertrophy, atrial fibrillation and partly in myocarditis, the opposite applies to diabetic cardiomyopathy and ischemia/reperfusion injury. In addition, the i20S appears to play a role in autophagy modulation depending on heart failure phenotype. This review summarizes the current literature on the i20S in different heart failure phenotypes, emphasizing the two faces of i20S in injured hearts. A selection of established i20S inhibitors is introduced and signaling pathways linking the i20S to autophagy are highlighted. Mapping the interplay of the i20S and autophagy in different types of heart failure offers potential approaches for developing treatment strategies against heart failure.
Collapse
Affiliation(s)
- Christiane Ott
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Molecular Toxicology, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
| |
Collapse
|
9
|
Mu X, Yu H, Li H, Feng L, Ta N, Ling L, Bai L, A R, Borjigidai A, Pan Y, Fu M. Metabolomics analysis reveals the effects of Salvia Miltiorrhiza Bunge extract on ameliorating acute myocardial ischemia in rats induced by isoproterenol. Heliyon 2024; 10:e30488. [PMID: 38737264 PMCID: PMC11088323 DOI: 10.1016/j.heliyon.2024.e30488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 04/10/2024] [Accepted: 04/28/2024] [Indexed: 05/14/2024] Open
Abstract
Salvia miltiorrhiza Bunge (SM) is a widespread herbal therapy for myocardial ischemia (MI). Nevertheless, the therapeutic signaling networks of SM extract on MI is yet unknown. Emerging evidences suggested that alterations in cardiac metabolite influences host metabolism and accelerates MI progression. Herein, we employed an isoproterenol (ISO)-induced acute myocardial ischemia (AMI) rat model to confirm the pharmacological effects of SM extract (0.8, 0.9, 1.8 g/kg/day) via assessment of the histopathological alterations that occur within the heart tissue and associated cytokines; we also examined the underlying SM extract-mediated signaling networks using untargeted metabolomics. The results indicated that 25 compounds with a relative content higher than 1 % in SM aqueous extract were identified using LC-MS/MS analysis, which included salvianolic acid B, lithospermic acid, salvianolic acid A, and caffeic acid as main components. An in vivo experiment showed that pretreatment with SM extract attenuated ISO-induced myocardial injury, shown as decreased myocardial ischemic size, transformed electrocardiographic, histopathological, and serum biochemical aberrations, reduced levels of proinflammatory cytokines, inhibited oxidative stress (OS), and reversed the trepidations of the cardiac tissue metabolic profiles. Metabolomics analysis shows that the levels of 24 differential metabolites (DMs) approached the same value as controls after SM extract therapy, which were primarily involved in histidine; alanine, aspartate, and glutamate; glycerophospholipid; and glycine, serine, and threonine metabolisms through metabolic pathway analysis. Correlation analysis demonstrated that the levels of modulatory effects of SM extract on the inflammation and OS were related to alterations in endogenous metabolites. Overall, SM extract demonstrated significant cardioprotective effects in an ISO-induced AMI rat model, alleviating myocardial injury, inflammation and oxidative stress, with metabolomics analysis indicating potential therapeutic pathways for myocardial ischemia.
Collapse
Affiliation(s)
- Xiyele Mu
- Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou 571199, China
- NMPA Key Laboratory of Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Hongzhen Yu
- Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou 571199, China
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, 100081, China
| | - Huifang Li
- NMPA Key Laboratory of Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Lan Feng
- NMPA Key Laboratory of Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Na Ta
- NMPA Key Laboratory of Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Ling Ling
- NMPA Key Laboratory of Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Li Bai
- Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou 571199, China
- NMPA Key Laboratory of Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Rure A
- Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou 571199, China
- NMPA Key Laboratory of Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Almaz Borjigidai
- Key Laboratory of Ethnomedicine of Ministry of Education, Center on Translational Neuroscience, School of Pharmacy, Minzu University of China, Beijing, 100081, China
| | - Yipeng Pan
- Department of Transplantation, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570100, China
| | - Minghai Fu
- Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou 571199, China
- NMPA Key Laboratory of Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao 028000, China
| |
Collapse
|
10
|
Sahu R, Rawal RK. Modulation of the c-JNK/p38-MAPK signaling pathway: Investigating the therapeutic potential of natural products in hypertension. PHYTOMEDICINE PLUS 2024; 4:100564. [DOI: 10.1016/j.phyplu.2024.100564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
11
|
Das A, Nikhil A, Shiekh PA, Yadav B, Jagavelu K, Kumar A. Ameliorating impaired cardiac function in myocardial infarction using exosome-loaded gallic-acid-containing polyurethane scaffolds. Bioact Mater 2024; 33:324-340. [PMID: 38076649 PMCID: PMC10701288 DOI: 10.1016/j.bioactmat.2023.11.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 11/06/2023] [Accepted: 11/16/2023] [Indexed: 06/21/2024] Open
Abstract
Myocardial infarction (MI) can be tackled by implanting cardiac patches which provide mechanical support to the heart. However, most tissue-engineered scaffolds face difficulty in attenuating oxidative stress, maintaining mechanical stability, and regenerating damaged cardiomyocytes. Here, we fabricated elastic cryogels using polyurethane modified with antioxidant gallic acid in its backbone (PUGA) and further coated them with decellularized extracellular matrix (dECM) to improve adhesiveness, biocompatibility and hemocompatibility. The scaffold was functionalized with exosomes (EXO) isolated from adipose-derived stem cells having regenerative potential. PUGA-dECM + EXO was tested in a rat model with induced MI where echocardiography after 8 weeks of implantation showed significant recovery in treatment group. Histological analysis revealed a decrease in fibrosis after application of patch and promotion of angiogenesis with reduced oxidative stress was shown by immunostaining. Expression of cardiac tissue contractile function marker was also observed in treatment groups. Thus, the proposed biomaterial has a promising application to be utilized as a patch for cardiac regeneration. More detailed studies with larger animal species are needed for using these observations for specific applications.
Collapse
Affiliation(s)
- Ankita Das
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, U.P., India
| | - Aman Nikhil
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, U.P., India
| | - Parvaiz Ahmad Shiekh
- SMART Lab, Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Babita Yadav
- Department of Pharmacology, Council of Scientific and Industrial Research (CSIR)-Central Drug Research Institute, Lucknow, 226031, U.P., India
| | - Kumaravelu Jagavelu
- Department of Pharmacology, Council of Scientific and Industrial Research (CSIR)-Central Drug Research Institute, Lucknow, 226031, U.P., India
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, U.P., India
- Centre for Environmental Sciences and Engineering, Indian Institute of Technology Kanpur, Kanpur, 208016, U.P., India
- Centre of Excellence for Orthopaedics and Prosthetics, Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur, 208016, U.P., India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, 208016, U.P., India
| |
Collapse
|
12
|
Wahid M, Saqib F, Abbas G, Shah S, Alshammari A, Albekairi TH, Ali A, Khurm M, Mubarak MS. Cardioprotective and hypotensive mechanistic insights of hydroethanolic extract of Cucumis melo L. kernels in isoprenaline-induced cardiotoxicity based on metabolomics and in silico electrophysiological models. Front Pharmacol 2024; 14:1277594. [PMID: 38348351 PMCID: PMC10859416 DOI: 10.3389/fphar.2023.1277594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 12/26/2023] [Indexed: 02/15/2024] Open
Abstract
Background: Cardiovascular diseases (CVD) continue to threaten health worldwide, and account for a significant portion of deaths and illnesses. In both developing and industrialized nations, they challenge their health systems. There are several traditional uses of Cucurbitaceae seeds in Pakistan, India, Iran, and China, including treating cardiovascular, neurological, and urogenital diseases. Methods: In the present work, integrated techniques of metabolomics profiling and computational cardiomyocyte stimulation were used to investigate possible mechanisms of C. melo in isoprenaline (ISO)-induced myocardial infarction. In vitro, vasoconstrictions, paired atria, and in vivo invasive blood pressure measurement models were performed to explore the mechanism of action of C. melo hydroethanolic seed extract (Cm-EtOH). Results: Results showed that Cm-EtOH demonstrates NO-based endothelium-derived relaxing factor (EDRF) vasorelaxant response, negative chronotropic and inotropic response in the atrium, and hypotensive effects in normotensive rats. Results also revealed that Cm-EtOH decreases cardiomyocyte hypertrophy and reverts the altered gene expressions, biochemical, and metabolites in ISO-induced myocardial infarction (MI) rats. The extract additionally reversed ISO-induced MI-induced oxidative stress, energy consumption, and amino acid metabolism. Moreover, C. melo seeds increased EDRF function, energy production, and antioxidant capacity to treat myocardial and vascular disorders. In computational cardiomyocyte simulation, gallic acid reduced action potential duration, upstroke velocity (dV/dtmax), and effective refractory period. Conclusion: This study highlights the therapeutic potential of C. melo seeds to treat cardiovascular diseases and provides mechanistic insight into its antihypertensive and cardioprotective activities.
Collapse
Affiliation(s)
- Muqeet Wahid
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Fatima Saqib
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Ghulam Abbas
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Shahid Shah
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Thamer H. Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Anam Ali
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Muhammad Khurm
- School of Pharmacy, Xi’an Jiaotong University, Xi’an, China
| | | |
Collapse
|
13
|
Kim HB, Hong YJ, Lee SH, Kee HJ, Kim M, Ahn Y, Jeong MH. Gallic Acid Inhibits Proliferation and Migration of Smooth Muscle Cells in a Pig In-Stent Restenosis Model. Chonnam Med J 2024; 60:32-39. [PMID: 38304132 PMCID: PMC10828086 DOI: 10.4068/cmj.2024.60.1.32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 02/03/2024] Open
Abstract
In-stent restenosis (ISR) develops primarily due to neointimal hyperplasia. Gallic acid (GA) has anti-inflammatory, antioxidant, and cardioprotective effects. This study sought to investigate the effects of GA on neointimal hyperplasia and proliferation and migration of vascular smooth muscle cells (VSMCs) in a pig ISR model. In vitro proliferation and migration experiments were confirmed, after VSMCs were treated with platelet-derived growth factor (PDGF-BB) and GA (100 µM) using a 3-(4,5-dimethylthiazol)-2,5-diphenyltetrazolium bromide (MTT) assay and a scratch wound assay for 24 hours and 48 hours. A bare metal stent (BMS) was implanted in the pig coronary artery to induce ISR with overdilation (1.1-1.2:1), and GA (10 mg/kg/day) was administered for 4 weeks. At the 4-week follow-up, optical coherence tomography (OCT) and histopathological analyses were performed. GA decreased the proliferation of VSMCs by PDGF-BB for 24 hours (89.24±24.56% vs. 170.04±19.98%, p<0.001) and 48 hours (124.87±7.35% vs. 187.64±4.83%, p<0.001). GA inhibited the migration of VSMCs induced by PDGF-BB for 24 hours (26.73±2.38% vs. 65.38±9.73%, p<0.001) and 48 hours (32.96±3.04% vs. 77.04±10.07%, p<0.001). Using OCT, % neointimal hyperplasia was shown to have significantly decreased in the GA group compared with control vehicle group (28.25±10.07% vs. 37.60±10.84%, p<0.001). GA effectively reduced neointimal hyperplasia by inhibiting the proliferation and migration of VSMCs in a pig ISR model. GA could be a potential treatment strategy for reducing ISR after stent implantation.
Collapse
Affiliation(s)
- Han Byul Kim
- Division of Cardiology, Chonnam National University Hospital, Gwangju, Korea
| | - Young Joon Hong
- Division of Cardiology, Chonnam National University Hospital, Gwangju, Korea
- Division of Cardiology, Chonnam National University Medical School, Gwangju, Korea
| | - Seung Hun Lee
- Division of Cardiology, Chonnam National University Hospital, Gwangju, Korea
| | - Hae Jin Kee
- Division of Cardiology, Chonnam National University Hospital, Gwangju, Korea
| | - Munki Kim
- Division of Cardiology, Chonnam National University Hospital, Gwangju, Korea
| | - Youngkeun Ahn
- Division of Cardiology, Chonnam National University Hospital, Gwangju, Korea
- Division of Cardiology, Chonnam National University Medical School, Gwangju, Korea
| | - Myung Ho Jeong
- Division of Cardiology, Chonnam National University Hospital, Gwangju, Korea
- Division of Cardiology, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
14
|
Momeni Z, Danesh S, Ahmadpour M, Eshraghi R, Farkhondeh T, Pourhanifeh MH, Samarghandian S. Protective Roles and Therapeutic Effects of Gallic Acid in the Treatment of Cardiovascular Diseases: Current Trends and Future Directions. Curr Med Chem 2024; 31:3733-3751. [PMID: 37815180 DOI: 10.2174/0109298673259299230921150030] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/19/2023] [Accepted: 08/18/2023] [Indexed: 10/11/2023]
Abstract
Cardiovascular diseases (CVDs) are serious life-threatening illnesses and significant problematic issues for public health having a heavy economic burden on all society worldwide. The high incidence of these diseases as well as high mortality rates make them the leading causes of death and disability. Therefore, finding novel and more effective therapeutic methods is urgently required. Gallic acid, an herbal medicine with numerous biological properties, has been utilized in the treatment of various diseases for thousands of years. It has been demonstrated that gallic acid possesses pharmacological potential in regulating several molecular and cellular processes such as apoptosis and autophagy. Moreover, gallic acid has been investigated in the treatment of CVDs both in vivo and in vitro. Herein, we aimed to review the available evidence on the therapeutic application of gallic acid for CVDs including myocardial ischemia-reperfusion injury and infarction, drug-induced cardiotoxicity, hypertension, cardiac fibrosis, and heart failure, with a focus on underlying mechanisms.
Collapse
Affiliation(s)
- Zahra Momeni
- Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Sepideh Danesh
- Research Hub Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Ahmadpour
- Research Hub Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Eshraghi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Tahereh Farkhondeh
- Department of Toxicology and Pharmacology, School of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Hub Institute, Tehran University of Medical Sciences, Tehran, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Saeed Samarghandian
- University of Neyshabur Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
15
|
Shi Y, Liu C, Xiong S, Yang L, Yang C, Qiao W, Liu Y, Liu S, Liu J, Dong G. Ling-Gui-Qi-Hua formula alleviates left ventricular myocardial fibrosis in rats with heart failure with preserved ejection fraction by blocking the transforming growth factor-β1 /Smads signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 317:116849. [PMID: 37385575 DOI: 10.1016/j.jep.2023.116849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/06/2023] [Accepted: 06/25/2023] [Indexed: 07/01/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ling-Qui-Qi-Hua (LGQH) decoction, composed of Poria cocos (Schw.) Wolf, Cinnamomum cassia (L.) J. Presl, Paeonia veitchii Lynch, and Atractylodes macrocephala Koidz., is a compound formula derived from Ling-Gui-Zhu-Gan decoction recorded in the Treatise on Febrile and Miscellaneous. It has shown cardioprotective effects on patients or rats with heart failure with preserved ejection fraction (HFpEF). Nevertheless, the active ingredients of LGQH and its anti-fibrotic mechanism remain unknown. AIM OF THE STUDY To determine the active ingredients in LGQH decoction and verify that LGQH decoction may inhibit left ventricular (LV) myocardial fibrosis in HFpEF rats by blocking the transforming growth factor-β1 (TGF-β1)/Smads signaling pathway from the perspective of animal experiments. MATERIALS AND METHODS First, liquid chromatography-mass spectrometry (LC-MS) technology was used to identify active components in the LGQH decoction. Secondly, a rat model of the metabolic syndrome-associated HFpEF phenotype was established and subsequently received LGQH intervention. The mRNA and protein expression of targets in the TGF-β1/Smads pathway were detected by quantitative real-time polymerase chain reaction and western blot analysis. Finally, molecular docking was conducted to examine the interactions between the active ingredients in the LGQH decoction and key proteins of the TGF-β1/Smads pathways. RESULTS According to LC-MS analysis, the LGQH decoction contained 13 active ingredients. In animal experiments, LGQH attenuated LV hypertrophy, enlargement, and diastolic function in HEpEF rats. Mechanically, LGQH not only down-regulated TGF-β1, Smad2, Smad3, Smad4, α-SMA, Coll I, and Coll III mRNA expressions and TGF-β1, Smad2, Smad3, P-Smad2/Smad3, Smad4, α-SMA, and Coll I protein expressions, but also up-regulated Smad7 mRNA and protein expressions, which ultimately led to myocardial fibrosis. Furthermore, molecular docking confirmed that 13 active ingredients in the LGQH decoction have excellent binding activities to the critical targets of the TGF-β1/Smads pathway. CONCLUSION LGQH is a modified herbal formulation with multiple active ingredients. It might alleviate LV remodeling and diastolic dysfunction and inhibit LV myocardial fibrosis by blocking TGF-β1/Smads pathways in HFpEF rats.
Collapse
Affiliation(s)
- Yujiao Shi
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100091, China
| | - Chunqiu Liu
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100091, China
| | - Shuang Xiong
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100091, China
| | - Ling Yang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100091, China
| | - Chenguang Yang
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100091, China
| | - Wenbo Qiao
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100091, China
| | - Yongcheng Liu
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100091, China
| | - Siyu Liu
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100091, China
| | - Jiangang Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100091, China.
| | - Guoju Dong
- Department of Cardiovascular Internal Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100091, China; National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100091, China.
| |
Collapse
|
16
|
Bai L, Han X, Kee HJ, He X, Kim SH, Jeon MJ, Zhou H, Jeong SM, Kee SJ, Jeong MH. Protocatechuic acid prevents isoproterenol-induced heart failure in mice by downregulating kynurenine-3-monooxygenase. J Cell Mol Med 2023; 27:2290-2307. [PMID: 37482908 PMCID: PMC10424289 DOI: 10.1111/jcmm.17869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 06/12/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023] Open
Abstract
Protocatechuic acid (3,4-dihydroxybenzoic acid) prevents oxidative stress, inflammation and cardiac hypertrophy. This study aimed to investigate the therapeutic effects of protocatechuic acid in an isoproterenol-induced heart failure mouse model and to identify the underlying mechanisms. To establish the heart failure model, C57BL/6NTac mice were given high-dose isoproterenol (80 mg/kg body weight) for 14 days. Echocardiography revealed that protocatechuic acid reversed the isoproterenol-induced downregulation of fractional shortening and ejection fraction. Protocatechuic acid attenuated cardiac hypertrophy as evidenced by the decreased heart-weight-to-body-weight ratio and the expression of Nppb. RNA sequencing analysis identified kynurenine-3-monooxygenase (Kmo) as a potential target of protocatechuic acid. Protocatechuic acid treatment or transfection with short-interfering RNA against Kmo ameliorated transforming growth factor β1-induced upregulation of Kmo, Col1a1, Col1a2 and Fn1 in vivo or in neonatal rat cardiac fibroblasts. Kmo knockdown attenuated the isoproterenol-induced increase in cardiomyocyte size, as well as Nppb and Col1a1 expression in H9c2 cells or primary neonatal rat cardiomyocytes. Moreover, protocatechuic acid attenuated Kmo overexpression-induced increases in Nppb mRNA levels. Protocatechuic acid or Kmo knockdown decreased isoproterenol-induced ROS generation in vivo and in vitro. Thus, protocatechuic acid prevents heart failure by downregulating Kmo. Therefore, protocatechuic acid and Kmo constitute a potential novel therapeutic agent and target, respectively, against heart failure.
Collapse
Affiliation(s)
- Liyan Bai
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
- Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
- Emergency Critical Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xiongyi Han
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
- Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
- Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, People's Republic of China
| | - Hae Jin Kee
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
- Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Xiaonan He
- Emergency Critical Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Seong Hoon Kim
- Department of Parasitology and Tropical Medicine, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Mi Jin Jeon
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
- Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Hongyan Zhou
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
- Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Seong Min Jeong
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
- Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Seung-Jung Kee
- Department of Laboratory Medicine, Chonnam National University Medical School and Hospital, Gwangju, Republic of Korea
| | - Myung Ho Jeong
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
- Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
- Department of Cardiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| |
Collapse
|
17
|
Cui H, Han S, Dai Y, Xie W, Zheng R, Sun Y, Xia X, Deng X, Cao Y, Zhang M, Shang H. Gut microbiota and integrative traditional Chinese and western medicine in prevention and treatment of heart failure. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 117:154885. [PMID: 37302262 DOI: 10.1016/j.phymed.2023.154885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/27/2023] [Accepted: 05/15/2023] [Indexed: 06/13/2023]
Abstract
BACKGROUND Heart failure (HF) is the terminal stage of multiple cardiovascular diseases, with high mortality and morbidity. More and more studies have proved that gut microbiota may play a role in the process of HF, which is expected to become a new therapeutic target. The combination of traditional Chinese and Western medicine has vast therapeutic potential of complementation against HF. PURPOSE This manuscript expounds on the research progress of mechanisms of gut microbiota participating in the occurrence and prognosis of HF and the role of integrative traditional Chinese and Western medicine from 1987 to 2022. The combination of traditional Chinese and Western medicine in the prevention and treatment of HF from the perspective of gut microbiota has been discussed. METHODS Studies focusing on the effects and their mechanisms of gut microbiota in HF and the role of integrative traditional Chinese and Western medicine were identified and summarized, including contributions from February 1987 until August 2022. The investigation was carried out in accordance with the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) guidelines. We searched PubMed, Embase, Cochrane Library, CNKI, Wanfang, and VIP databases up to April 2023 by using the relevant keywords and operators. RESULTS A total of 34 articles were finally included in this review.16 RCTs and 13 basic researches, and 3 clinical research studies involving 7 relevant outcome indicators(cardiac function evaluation index, changes in gut microbiota, inflammatory factors, metabolites of gut microbiota, serum nutritional index protein, quality of life score, intestinal permeability and all-cause mortality). Compared with healthy controls, serum TNF-α and TMAO levels were significantly higher in patients with heart failure [MD = 5.77, 95%CI(4.97, 6.56), p < 0.0001; SMD = 1.92, 95%CI(1.70, 2.14), p < 0.0001]. Escherichia coli and Thick-walled bacteria increased significantly [SMD = -0.99, 95%CI(-1.38, -0.61), p < 0.0001, SMD = 2.58, 95%CI(2.23, 2.93), p < 0.0001];The number of bacteroides and lactobacillus decreased [SMD = -2.29, 95%CI(-2.54, -2.04), p < 0.0001; SMD = -1.55, 95%CI(-1.8, -1.3), p < 0.0001]. There was no difference in bifidobacterium [SMD = 0.16, 95%CI(-0.22, 0.54), p = 0.42]. In the published literature, it is not difficult to see that most of the results are studied and proved based on animal experiments or clinical trials, involving the cellular level, while the mechanism and mode of action of the molecular biology of traditional Chinese medicine are less elaborated, which is related to the characteristics of multi-components and multi-targets of traditional Chinese medicine. The above are the shortcomings of published literature, which can also be the direction of future research. CONCLUSION Heart failure patients have decreased beneficial bacteria such as Bacillus mimics and Lactobacillus in the intestinal flora and increased harmful flora like thick-walled flora. And increase the inflammatory response of the body and the expression of trimethylamine oxide (TMAO) in the serum. And The prevention and treatment of integrative traditional Chinese and Western medicine against heart failure based on gut microbiota and its metabolites is a promising research direction.
Collapse
Affiliation(s)
- Herong Cui
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China; School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Songjie Han
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yanan Dai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Wei Xie
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Rui Zheng
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yang Sun
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Xiaofeng Xia
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiaopeng Deng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yaru Cao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Mei Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China.
| |
Collapse
|
18
|
Liu X, Tian R, Tao H, Wu J, Yang L, Zhang Y, Meng X. The cardioprotective potentials and the involved mechanisms of phenolic acids in drug-induced cardiotoxicity. Eur J Pharmacol 2022; 936:175362. [DOI: 10.1016/j.ejphar.2022.175362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/22/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
|
19
|
Long K, Zhao Z, Chen J, Zhi L, Wang C, Liao D, Wang M, Gao P. Yang-xin-xue keli exerts therapeutic effects via regulating mitochondrial homeostasis and function in doxorubicin-induced rat heart failure. Front Pharmacol 2022; 13:931453. [PMID: 36110548 PMCID: PMC9468485 DOI: 10.3389/fphar.2022.931453] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Heart failure, especially chronic heart failure, is generally induced by the accumulation of reactive oxygen species (ROS), as well as the subsequent loss of mitochondrial permeability transition pore (mPTP) openings and pathological mitochondrial dysfunction. Herein, we explored the therapeutic effects of the Chinese medicine Yangxin Keli (YXXKL) on chronic heart failure and its underlying working mechanism. Methods: To mimic oxidative stress-induced chronic heart failure, a rat heart failure model was induced by the administration of DOX. Transthoracic echocardiography was performed to confirm the successful establishment of the heart failure model by observing significantly decreased cardiac function in the rats. Mitochondrial membrane potential, function, and ATP synthesis activity were measured after YXXKL was employed. Results The administration of YXXKL not only significantly improved cardiac function but also reversed the myocardium loss and fibrosis induced via DOX. Moreover, the administration of YXXKL also increased ATP synthesis and mitochondrial DNA mass in left ventricular tissues, which indicated that mitochondria may be a key target of YXXKL. Thus, we employed rat cardiomyocyte H9c2 and primary rat cardiac myocytes (RCMs) to induce oxidative stress-induced myocardial injury via DOX treatment. YXXKL-medicated serum promoted cell proliferation, which was inhibited by the addition of IC30 DOX, and the serum also inhibited cell apoptosis, which was promoted by the addition of IC50 DOX. YXKL-medicated serum was able to scavenge ROS and maintain the mitochondrial membrane potential as well as promote mitochondrial function, including the promotion of ATP synthesis, mitochondrial DNA mass, and transcriptional activity. Furthermore, we also observed that YXXKL-medicated serum inhibited DOX-induced autophagy/mitophagy by scavenging ROS. Conclusion: Taken together, we conclude that YXXKLI may exert therapeutic effects on oxidative stress-related heart failure via the regulation of mitochondria.
Collapse
Affiliation(s)
- Kunlan Long
- Intensive Care Unit, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ziyi Zhao
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Chen
- Intensive Care Unit, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lijia Zhi
- Intensive Care Unit, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chunxia Wang
- Intensive Care Unit, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dan Liao
- Intensive Care Unit, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Meng Wang
- Intensive Care Unit, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peiyang Gao
- Intensive Care Unit, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Peiyang Gao,
| |
Collapse
|
20
|
Han X, Bai L, Kee HJ, Jeong MH. Syringic acid mitigates isoproterenol-induced cardiac hypertrophy and fibrosis by downregulating Ereg. J Cell Mol Med 2022; 26:4076-4086. [PMID: 35719043 PMCID: PMC9279583 DOI: 10.1111/jcmm.17449] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/22/2022] [Accepted: 05/11/2022] [Indexed: 12/01/2022] Open
Abstract
Gallic acid has been reported to mitigate cardiac hypertrophy, fibrosis and arterial hypertension. The effects of syringic acid, a derivative of gallic acid, on cardiac hypertrophy and fibrosis have not been previously investigated. This study aimed to examine the effects of syringic acid on isoproterenol‐treated mice and cells. Syringic acid mitigated the isoproterenol‐induced upregulation of heart weight to bodyweight ratio, pathological cardiac remodelling and fibrosis in mice. Picrosirius red staining, quantitative real‐time polymerase chain reaction (qRT‐PCR) and Western blotting analyses revealed that syringic acid markedly downregulated collagen accumulation and fibrosis‐related factors, including Fn1. The results of RNA sequencing analysis of Ereg expression were verified using qRT‐PCR. Syringic acid or transfection with si‐Ereg mitigated the isoproterenol‐induced upregulation of Ereg, Myc and Ngfr. Ereg knockdown mitigated the isoproterenol‐induced upregulation of Nppb and Fn1 and enhancement of cell size. Mechanistically, syringic acid alleviated cardiac hypertrophy and fibrosis by downregulating Ereg. These results suggest that syringic acid is a potential therapeutic agent for cardiac hypertrophy and fibrosis.
Collapse
Affiliation(s)
- Xiongyi Han
- Heart Research Center of Chonnam National University Hospital, Gwangju, Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Liyan Bai
- Heart Research Center of Chonnam National University Hospital, Gwangju, Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Hae Jin Kee
- Heart Research Center of Chonnam National University Hospital, Gwangju, Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Myung Ho Jeong
- Heart Research Center of Chonnam National University Hospital, Gwangju, Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Korea.,Department of Cardiology, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
21
|
Saqib F, Wahid M, Al-Huqail AA, Ahmedah HT, Bigiu N, Irimie M, Moga M, Marc Vlaic RA, Pop OL, Chicea LM. Metabolomics based mechanistic insights to vasorelaxant and cardioprotective effect of ethanolic extract of Citrullus lanatus (Thunb.) Matsum. & Nakai. seeds in isoproterenol induced myocardial infraction. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 100:154069. [PMID: 35364560 DOI: 10.1016/j.phymed.2022.154069] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 02/14/2022] [Accepted: 03/19/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Cardiovascular diseases (CVDs) are a significant cause of morbidity and death in the current world, posing a challenge to both developing and industrialized nation's health systems. Citrullus lanatus (Thunb.) Matsum. & Nakai. seeds have long been utilized to supplement and enhance health and treat cardiovascular illnesses. However, its treatments for CVDs are still unknown. More research is required to fully comprehend the impact of C. lanatus seeds on vasorelaxation and myocardial infractions. PURPOSE Therefore, an integrated metabolomics profiling technique was used to investigate possible pathways of C. lanatus in isoproterenol (ISO)-induced myocardial infarction (MI). Isoproterenol causes long-term cardiac hypertrophy by causing cardiomyocyte compensatory loss, eventually leading to heart failure. METHODS In vitro models of vasoconstriction, atrium, and in vivo models of invasive blood pressure measurement and isoproterenol (ISO) induced cardiac hypertrophy in rats were used to understand underlying mechanistic by LC-MS/MS based dynamic metabolomics analysis of the serum and heart samples to be investigated the effect of ethanolic extract of C. lanatus (Cl.EtOH). RESULTS Cl.EtOH exhibited vasorelaxant, negative chronotropic, and inotropic effects in in-vitro models whereas, a potent hypotensive effect was observed in normotensive rats. The Cl.EtOH protected the animals from ISO-induced myocardial infarction (MI) with therapeutic interventions in left ventricular thickness, cardiomyocyte hypertrophy, mRNA gene expression, biochemical assays, and metabolomic profiling of serum and heart tissues. CONCLUSIONS For the first time, our study confirmed that C. lanatus seeds (Cl.EtOH) possess significant antihypertensive and prevent ISO-induced myocardial infarction. These findings comprehensively demonstrated mechanistic insights of Cl.EtOH in vasorelaxation and myocardial infarction. The current study provides evidence for further mechanistic studies and the development of C. lanatus seeds as a potential therapeutic intervention for patients with cardiovascular disorders.
Collapse
Affiliation(s)
- Fatima Saqib
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Muqeet Wahid
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Arwa Abdulkreem Al-Huqail
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia.
| | - Hanadi Talal Ahmedah
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Rabigh 25732, Saudi Arabia
| | - Nicusor Bigiu
- Faculty of Medicine, Transilvania University of Brasov, 500019 Brasov.
| | - Marius Irimie
- Faculty of Medicine, Transilvania University of Brasov, 500019 Brasov
| | - Marius Moga
- Faculty of Medicine, Transilvania University of Brasov, 500019 Brasov
| | - Romina Alina Marc Vlaic
- Food Engineering Department, Faculty of Food Science and Technology, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania.
| | - Oana Lelia Pop
- Department of Food Science, University of Agricultural Science and Veterinary Medicine, 400372 Cluj-Napoca, Romania
| | | |
Collapse
|
22
|
Bai J, Lin QY, An X, Liu S, Wang Y, Xie Y, Liao J. Low-Dose Gallic Acid Administration Does Not Improve Diet-Induced Metabolic Disorders and Atherosclerosis in Apoe Knockout Mice. J Immunol Res 2022; 2022:7909971. [PMID: 35652108 PMCID: PMC9150997 DOI: 10.1155/2022/7909971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/21/2022] [Accepted: 05/04/2022] [Indexed: 01/17/2023] Open
Abstract
Diets rich in polyphenols are known to be beneficial for cardiovascular health. Gallic acid (GA) is a plant-derived triphenolic chemical with multiple cardio-protective properties, such as antiobesity, anti-inflammation, and antioxidation. However, whether GA could protect against atherosclerotic cardiovascular diseases is still not defined. Here, we investigated the effects of low-dose GA administration on diet-induced metabolic disorders and atherosclerosis in the atherosclerosis-prone apolipoprotein E (Apoe) knockout mice fed on a high-fat Western-type diet (WTD) for 8 weeks. Our data showed that GA administration by oral gavage at a daily dosage of 20 mg/kg body weight did not significantly ameliorate WTD-induced hyperlipidemia, hepatosteatosis, adipogenesis, or insulin resistance; furthermore, GA administration did not significantly ameliorate WTD-induced atherosclerosis. In conclusion, our data demonstrate that low-dose GA administration does not elicit significant health effect on diet-induced metabolic disorders or atherosclerosis in the Apoe knockout mice. Whether GA could be beneficial for atherosclerotic cardiovascular diseases therefore needs further exploration.
Collapse
Affiliation(s)
- Jie Bai
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Qiu-Yue Lin
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiangbo An
- Department of Interventional Therapy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Shuang Liu
- College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Yao Wang
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yunpeng Xie
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Jiawei Liao
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| |
Collapse
|
23
|
Harlev I, Holmes JW, Cohen N. The influence of boundary conditions and protein availability on the remodeling of cardiomyocytes. Biomech Model Mechanobiol 2022; 21:189-201. [PMID: 34661804 DOI: 10.1007/s10237-021-01526-5] [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: 06/07/2021] [Accepted: 10/03/2021] [Indexed: 11/27/2022]
Abstract
The heart muscle is capable of growing and remodeling in response to changes in its mechanical and hormonal environment. While this capability is essential to the healthy function of the heart, under extreme conditions it may also lead to heart failure. In this work, we derive a thermodynamically based and microscopically motivated model that highlights the influence of mechanical boundary conditions and hormonal changes on the remodeling process in cardiomyocytes. We begin with a description of the kinematics associated with the remodeling process. Specifically, we derive relations between the macroscopic deformation, the number of sarcomeres, the sarcomere stretch, and the number of myofibrils in the cell. We follow with the derivation of evolution equations that describe the production and the degradation of protein in the cytosol. Next, we postulate a dissipation-based formulation that characterizes the remodeling process. We show that this process stems from a competition between the internal energy, the entropy, the energy supplied to the system by ATP and other sources, and dissipation mechanisms. To illustrate the merit of this framework, we study four initial and boundary conditions: (1) a myocyte undergoing isometric contractions in the presence of either an infinite or a limited supply of proteins and (2) a myocyte that is free to dilate along the radial direction with an infinite and a limited supply of proteins. This work underscores the importance of boundary conditions on the overall remodeling response of cardiomyocytes, suggesting a plausible mechanism that might play a role in distinguishing eccentric vs. concentric hypertrophy.
Collapse
Affiliation(s)
- Ido Harlev
- Department of Materials Science and Engineering, Technion - Israel Institute of Technology, 3200003, Haifa, Israel
| | - Jeffrey W Holmes
- Division of Cardiovascular Disease, Division of Cardiothoracic Surgery, Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Noy Cohen
- Department of Materials Science and Engineering, Technion - Israel Institute of Technology, 3200003, Haifa, Israel.
| |
Collapse
|
24
|
Hdac8 Inhibitor Alleviates Transverse Aortic Constriction-Induced Heart Failure in Mice by Downregulating Ace1. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6227330. [PMID: 35126818 PMCID: PMC8813277 DOI: 10.1155/2022/6227330] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022]
Abstract
Background Heart failure is characterized by activation of the renin-angiotensin-aldosterone system, which is involved in the regulation of cardiac hypertrophy and hypertension. Recently, we reported that Hdac8 inhibition alleviates isoproterenol-induced and angiotensin II-induced cardiac hypertrophy or hypertension in mice. Here, the effect and regulatory mechanisms of the Hdac8 selective inhibitor PCI34051 on pressure overload-induced heart failure were examined. Methods and Results At week 6 posttransverse aortic constriction (TAC), mice were administered with PCI34051 (3, 10, or 30 mg/kg bodyweight/day) for 2 weeks. The therapeutic effects of PCI34051 on TAC-induced cardiac and lung hypertrophy were determined by examining the heart weight-to-bodyweight and lung weight-to-bodyweight ratios and the cross-sectional cardiomyocyte area. Echocardiography analysis revealed that PCI34051 mitigated TAC-induced decreased ejection fraction and fractional shortening. Additionally, the expression of Hdac8 was upregulated in the cardiac and pulmonary tissues of TAC mice. The expression levels of Ace1 and Agtr1 were upregulated, whereas those of Ace2 and Agtr2 were downregulated in TAC mice. PCI34051 treatment or Hdac8 knockdown alleviated inflammation as evidenced by Rela downregulation and Nfkbia upregulation in mice, as well as in cardiomyocytes, but not in cardiac fibroblasts. Hdac8 overexpression-induced Rela pathway activation was downregulated in Ace1 knockdown cells. Picrosirius red staining, real-time polymerase chain reaction, and western blotting analyses revealed that PCI34051 alleviated fibrosis and downregulated fibrosis-related genes. Moreover, PCI34051 or Hdac8 knockdown in rat cardiac fibroblasts alleviated cardiac fibrosis through the Tgfb1-Smad2/3 pathway. The results of overexpression and knockdown experiments revealed that Hdac8 and Ace1 promote inflammation and fibrosis. Conclusions Treatment with PCI34051 enhanced cardiac and lung functions in the TAC-induced heart failure mouse model. These data suggest that HDAC8 is a potential novel therapeutic target for heart failure accompanied by pathological lung diseases.
Collapse
|
25
|
Shackebaei D, Hesari M, Ramezani-Aliakbari S, Hoseinkhani Z, Ramezani-Aliakbari F. Gallic acid protects against isoproterenol-induced cardiotoxicity in rats. Hum Exp Toxicol 2022; 41:9603271211064532. [PMID: 35193428 DOI: 10.1177/09603271211064532] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Gallic acid (GA) is a polyphenolic agent with interesting pharmacological impacts on the cardiovascular system. OBJECTIVE The present study purposed to study the protective effects of GA at 25 and 50 mg/kg against isoproterenol (ISO)-induced cardiac damage in ischemia/reperfusion (I/R) in rats. METHODS Male Wistar rats were randomly assigned into six groups: Control, Control treated with GA at 25 mg/kg (GA25), Control treated with GA at 50 mg/kg (GA50), Hypertrophic rats induced by ISO (ISO), Hypertrophic rats treated with GA at 25 mg/kg (ISO+GA25), and Hypertrophic rats treated with GA at 50 mg/kg (ISO+GA50). Heart isolation was performed to induce a cardiac I/R injury model. Cardiac hemodynamic parameters were recorded. Serum Lactate Dehydrogenase (LDH) and Creatine Kinase-MB (CK-MB) and cardiac Superoxide dismutases (SOD) levels were evaluated. The gene expression of Sarcoplasmic reticulum Ca2+-ATPase (SERCA2a) was assessed. RESULTS We found that GA at 50 mg/kg was significantly increased cardiac function at post I/R period in ISO-induced hypertrophic hearts. Moreover, it suppressed cardiac hypertrophy, the serum LDH and CK-MB levels in ISO injected rats. Administration of GA at 50 mg/kg was significantly increased SOD level and SERCA2a gene expression in the hypertrophic hearts. CONCLUSION GA at 50 mg/kg could improve cardiac performance possibly by increasing antioxidant defense enzymes, reducing cell damage, and enhancing SERCA2a gene expression in hypertrophic heart induced by ISO in I/R injury conditions.
Collapse
Affiliation(s)
- Dareuosh Shackebaei
- Medical Biology Research Center, Health Technology Institute, 48464Kermanshah University of Medical Sciences, Kermanshah, Iran.,Cardiovascular Research Center, 48464Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mahvash Hesari
- Medical Biology Research Center, Health Technology Institute, 48464Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Soudabeh Ramezani-Aliakbari
- Medical Biology Research Center, Health Technology Institute, 48464Kermanshah University of Medical Sciences, Kermanshah, Iran.,Medical School, 48464Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zohreh Hoseinkhani
- Medical Biology Research Center, Health Technology Institute, 48464Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Ramezani-Aliakbari
- Medical Biology Research Center, Health Technology Institute, 48464Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Physiology, School of Medicine, 48430Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
26
|
JNK signaling-dependent regulation of histone acetylation are involved in anacardic acid alleviates cardiomyocyte hypertrophy induced by phenylephrine. PLoS One 2021; 16:e0261388. [PMID: 34914791 PMCID: PMC8675748 DOI: 10.1371/journal.pone.0261388] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/30/2021] [Indexed: 12/30/2022] Open
Abstract
Cardiac hypertrophy is a complex process induced by the activation of multiple signaling pathways. We previously reported that anacardic acid (AA), a histone acetyltransferase (HAT) inhibitor, attenuates phenylephrine (PE)-induced cardiac hypertrophy by downregulating histone H3 acetylation at lysine 9 (H3K9ac). Unfortunately, the related upstream signaling events remained unknown. The mitogen-activated protein kinase (MAPK) pathway is an important regulator of cardiac hypertrophy. In this study, we explored the role of JNK/MAPK signaling pathway in cardiac hypertrophy induced by PE. The mice cardiomyocyte hypertrophy model was successfully established by treating cells with PE in vitro. This study showed that p-JNK directly interacts with HATs (P300 and P300/CBP-associated factor, PCAF) and alters H3K9ac. In addition, both the JNK inhibitor SP600125 and the HAT inhibitor AA attenuated p-JNK overexpression and H3K9ac hyperacetylation by inhibiting P300 and PCAF during PE-induced cardiomyocyte hypertrophy. Moreover, we demonstrated that both SP600125 and AA attenuate the overexpression of cardiac hypertrophy-related genes (MEF2A, ANP, BNP, and β-MHC), preventing cardiomyocyte hypertrophy and dysfunction. These results revealed a novel mechanism through which AA might protect mice from PE-induced cardiomyocyte hypertrophy. In particular, AA inhibits the effects of JNK signaling on HATs-mediated histone acetylation, and could therefore be used to prevent and treat pathological cardiac hypertrophy.
Collapse
|
27
|
Gu H, Gwon MH, Kim SM, Yun JM. Dietary glucosinolates inhibit splenic inflammation in high fat/cholesterol diet-fed C57BL/6 mice. Nutr Res Pract 2021; 15:798-806. [PMID: 34858556 PMCID: PMC8601941 DOI: 10.4162/nrp.2021.15.6.798] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/23/2020] [Accepted: 05/04/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND/OBJECTIVES Obesity is associated with chronic inflammation. The spleen is the largest organ of the lymphatic system and has an important role in immunity. Obesity-induced inflammatory responses are triggered by Toll-like receptor (TLR)-myeloid differentiation primary response 88 (MyD88) pathway signaling. Phenethyl isothiocyanate (PEITC) and 3,3′-diindolylmethane (DIM), major dietary glucosinolates present in cruciferous vegetables, have been reported to produce anti-inflammatory effects on various diseases. However, the effects of PEITC and DIM on the obesity-induced inflammatory response in the spleen are unclear. The purpose of this study was to examine the anti-inflammatory effects of PEITC and DIM on the spleen and their mechanism in high fat/cholesterol diet (HFCD)-fed C57BL/6 mice. MATERIALS/METHODS We established an animal model of HFCD-induced obesity using C57BL/6 mice. The mice were divided into six groups: normal diet with AIN-93G diet (CON), high fat diet (60% calories from fat) with 1% cholesterol (HFCD), HFCD with PEITC 30 mg/kg/day or 75 mg/kg/day (HFCD+P30, HFCD+P75), and HFCD with DIM 1.5 mg/kg/day or 7.5 mg/kg/day (HFCD+D1.5, HFCD+D7.5). Enzyme-linked immunosorbent assay was used to evaluate pro-inflammatory cytokine secretion. Western blot and quantitative polymerase chain reaction were used to analyze protein and mRNA levels of nuclear factor kappa B (NF-κB) p65, interleukin 6 (IL-6), cyclooxygenase 2 (COX-2), TLR2, TLR4, and MyD88 in spleen tissue. RESULTS Serum IL-6 levels were significantly higher in the HFCD group than in groups fed a HFCD with PEITC or DIM. Levels of NF-κB p65 protein and TLR2/4, MyD88, NF-κB p65, IL-6, and COX-2 mRNA were significantly higher in the HFCD group than in the CON group and were reduced by the PEITC and DIM supplements. CONCLUSIONS PEITC- and DIM-supplemented diets improved splenic inflammation by modulating the TLR2/4-MyD88 pathway in HFCD-fed mice. We suggest that dietary glucosinolates may at least partially improve obesity-induced inflammation of the spleen.
Collapse
Affiliation(s)
- HyunJi Gu
- Department of Food and Nutrition, Chonnam National University, Gwangju 61186, Korea
| | - Min-Hee Gwon
- Department of Education, Graduate School of Education, Chonnam National University, Gwangju 61186, Korea
| | - Sang-Min Kim
- Department of Food and Nutrition, Chonnam National University, Gwangju 61186, Korea
| | - Jung-Mi Yun
- Department of Food and Nutrition, Chonnam National University, Gwangju 61186, Korea
| |
Collapse
|
28
|
Hu G, Zhou X. Gallic Acid Ameliorates Atopic Dermatitis-Like Skin Inflammation Through Immune Regulation in a Mouse Model. Clin Cosmet Investig Dermatol 2021; 14:1675-1683. [PMID: 34815684 PMCID: PMC8605796 DOI: 10.2147/ccid.s327825] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/12/2021] [Indexed: 12/17/2022]
Abstract
Background Gallic acid (GA) has an anti-inflammatory effect by regulating inflammatory molecules. This study aimed to investigate the effect of GA on atopic dermatitis (AD)-like skin inflammation. Methods 4-dinitrochlorobenzene (DNCB) was used to induce an AD-like skin inflammation model. The effect of GA on DNCB-induced inflammation was assessed by measuring the thickness and histopathological examination of the ear. Serum IgE and TNF-α levels were detected. The effect of GA on lymph nodes was determined by measuring the weights and mRNA/protein expression levels of TNF-α, IL-4, IFN-γ and IL-17. Ratio of Treg cells and Th17 cells was also analyzed. Results It was found that the thickness and pathology of the ear were significantly improved by GA in the DNCB-induced mice. Serum IgE and TNF-α levels were significantly reduced in GA-treated model mice compared to the model group. GA treatment lowered the weight of lymph node and the expression of mRNAs of TNF-α, IL-4, IFN-γ, and IL-17 of lymph node. In the ear, inflammatory factors (IL-4, IL-5, IL-17, or IL-23) showed a significant decrease in GA-treated model mice versus model mice, while the expression levels of IL-10 and TGF-β showed a great increase in GA-treated model mice. ROR-γt showed a decrease in GA-treated model group, along with an increase expression of SOCS3. Conclusion GA could ameliorate AD-like skin inflammation possibly through Th17 mediated immune regulation in a DNCB-induced mouse model.
Collapse
Affiliation(s)
- Guohong Hu
- Dermatology Hospital of Jiangxi Province, Nanchang, 330001, Jiangxi, People's Republic of China
| | - Xiansheng Zhou
- Dermatology Hospital of Jiangxi Province, Nanchang, 330001, Jiangxi, People's Republic of China
| |
Collapse
|
29
|
Du Y, Demillard LJ, Ren J. Catecholamine-induced cardiotoxicity: A critical element in the pathophysiology of stroke-induced heart injury. Life Sci 2021; 287:120106. [PMID: 34756930 DOI: 10.1016/j.lfs.2021.120106] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/24/2021] [Accepted: 10/26/2021] [Indexed: 01/20/2023]
Abstract
Cerebrovascular diseases such as ischemic stroke, brain hemorrhage, and subarachnoid hemorrhage provoke cardiac complications such as heart failure, neurogenic stress-related cardiomyopathy and Takotsubo cardiomyopathy. With regards to the pathophysiology of stroke-induced heart injury, several mechanisms have been postulated to contribute to this complex interaction between brain and heart, including damage from gut dysbiosis, immune and systematic inflammatory responses, microvesicle- and microRNA-mediated vascular injury and damage from a surge of catecholamines. All these cerebrovascular diseases may trigger pronounced catecholamine surges through diverse ways, including stimulation of hypothalamic-pituitary adrenal axis, dysregulation of autonomic system, and secretion of adrenocorticotropic hormone. Primary catecholamines involved in this pathophysiological response include norepinephrine (NE) and epinephrine. Both are important neurotransmitters that connect the nervous system with the heart, leading to cardiac damage via myocardial ischemia, calcium (Ca2+) overload, oxidative stress, and mitochondrial dysfunction. In this review, we will aim to summarize the molecular mechanisms behind catecholamine-induced cardiotoxicity including Ca2+ overload, oxidative stress, apoptosis, cardiac hypertrophy, interstitial fibrosis, and inflammation. In addition, we will focus on how synchronization among these pathways evokes cardiotoxicity.
Collapse
Affiliation(s)
- Yuxin Du
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Laurie J Demillard
- School of Pharmacy, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
30
|
Sundaresan S, John S, Paneerselvam G, Andiapppan R, Christopher G, Selvam GS. Gallic acid attenuates cadmium mediated cardiac hypertrophic remodelling through upregulation of Nrf2 and PECAM-1signalling in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 87:103701. [PMID: 34237468 DOI: 10.1016/j.etap.2021.103701] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 07/01/2021] [Accepted: 07/03/2021] [Indexed: 06/13/2023]
Abstract
Gallic acid (GA) is an abundant natural polyphenolic compound found in vegetable and fruits that reduces the cardiac disease risk factor. This study aims to evaluate GA's role on cadmium (Cd) induced cardiac remodelling in experimental rats. Male Wistar rats were exposed to Cd (15 ppm) in drinking water and administered with GA orally (15 mg/kg/d) for 60 days. The results showed that GA regulated the lipid profile and reduced the LDL to 57 % compared with Cd treated rats. GA inhibited cardiac marker enzymes activity of CK-NAC (to 72.7 %) and CK-MB (to 100.3 %). Moreover, GA attenuated lipid peroxidation and enhanced the cardiac glutathione S transferase (GST) activity (89.2 %), glutathione peroxidase (GPx) (87 %), superoxide dismutase (SOD) (88.4 %) and catalase (CAT) activity (86.5 %). Histopathological examination showed that GA impaired the ventricular hypertrophy and fibrotic proliferation induced by Cd in rats. The combination of GA + Cd, decreased the gene expression of ANP (1-fold), BNP (0.5-fold) and β- MHC (0.9-fold). Furthermore, GA significantly reduced the expression of profibrotic (TGF-β) and proinflammatory (MCP-1) gene in Cd intoxicated rats. GA upregulated the expression of Nrf2 (2-fold), HO-1 (3-fold), and PECAM-1 (0.6-fold), which augments the detoxifying enzyme activity and cellular immunity in Cd intoxicated rats. The increased protein expression of Nrf2, PECAM-1 and decreased AKT-1 levels confirmed the mechanical action of GA during the hypertrophic condition. Thus, our results suggest that GA could act as a potential therapeutic agent regulating Nrf2 and PECAM-1 signalling pathways, thereby ameliorating Cd-induced pathological cardiac remodelling.
Collapse
Affiliation(s)
- Sasikumar Sundaresan
- Department of Biochemistry, Molecular Cardiology Unit, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu, India
| | - Samu John
- Rajiv Gandhi Center for Biotechnology, Thycaud, Poojapura, P.O, Thiruvananthapuram, Kerala, India
| | - Gomathi Paneerselvam
- Department of Biochemistry, Molecular Cardiology Unit, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu, India
| | | | | | - Govindan Sadasivam Selvam
- Department of Biochemistry, Molecular Cardiology Unit, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu, India.
| |
Collapse
|
31
|
Bai L, Kee HJ, Han X, Zhao T, Kee SJ, Jeong MH. Protocatechuic acid attenuates isoproterenol-induced cardiac hypertrophy via downregulation of ROCK1-Sp1-PKCγ axis. Sci Rep 2021; 11:17343. [PMID: 34462460 PMCID: PMC8405624 DOI: 10.1038/s41598-021-96761-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/12/2021] [Indexed: 12/25/2022] Open
Abstract
Cardiac hypertrophy is an adaptive response of the myocardium to pressure overload or adrenergic agonists. Here, we investigated the protective effects and the regulatory mechanism of protocatechuic acid, a phenolic compound, using a mouse model of isoproterenol-induced cardiac hypertrophy. Our results demonstrated that protocatechuic acid treatment significantly downregulated the expression of cardiac hypertrophic markers (Nppa, Nppb, and Myh7), cardiomyocyte size, heart weight to body weight ratio, cross-sectional area, and thickness of left ventricular septum and posterior wall. This treatment also reduced the expression of isoproterenol-induced ROCK1, Sp1, and PKCγ both in vivo and in vitro. To investigate the mechanism, we performed knockdown and overexpression experiments. The knockdown of ROCK1, Sp1, or PKCγ decreased the isoproterenol-induced cell area and the expression of hypertrophic markers, while the overexpression of Sp1 or PKCγ increased the levels of hypertrophic markers. Protocatechuic acid treatment reversed these effects. Interestingly, the overexpression of Sp1 increased cell area and induced PKCγ expression. Furthermore, experiments using transcription inhibitor actinomycin D showed that ROCK1 and Sp1 suppression by protocatechuic acid was not regulated at the transcriptional level. Our results indicate that protocatechuic acid acts via the ROCK1/Sp1/PKCγ axis and therefore has promising therapeutic potential as a treatment for cardiac hypertrophy.
Collapse
Affiliation(s)
- Liyan Bai
- Heart Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju, 61469, Republic of Korea
- Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea
| | - Hae Jin Kee
- Heart Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju, 61469, Republic of Korea.
- Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea.
| | - Xiongyi Han
- Heart Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju, 61469, Republic of Korea
- Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea
| | - Tingwei Zhao
- Heart Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju, 61469, Republic of Korea
- Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea
| | - Seung-Jung Kee
- Department of Laboratory Medicine, Chonnam National University, Medical School and Hospital, Gwangju, 61469, Republic of Korea
| | - Myung Ho Jeong
- Heart Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju, 61469, Republic of Korea.
- Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, 61469, Republic of Korea.
- Department of Cardiology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea.
| |
Collapse
|
32
|
Saqib F, Ali A, Ahmedah HT, Irimie CA, Toma SI, Popovici BE, Moga M, Irimie M. Cardioprotective, hypotensive and toxicological studies of Populus ciliata (Wall. ex Royle). Biomed Pharmacother 2021; 142:112065. [PMID: 34449312 DOI: 10.1016/j.biopha.2021.112065] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/11/2021] [Accepted: 08/17/2021] [Indexed: 01/10/2023] Open
Abstract
Populus ciliata Wall ex. Royle has folkloric repute to treat various cardiovascular ailments and related disorders. The current study was designed to evaluate the toxic profile, cardioprotective and hypotensive effects of Populus ciliata (Wall. ex Royle). Populus ciliata crude ethanolic extract (Pc. Cr) and its aqueous (Pc. Aq) & organic (Pc. Dcm) fractions were tested on isolated aorta of rat and rabbit having intact and non-intact endothelium respectively. Pc. Cr & Pc. Aq relaxed the contractions induced by PE (1 µM)-induced and K+ (80 mM)-induced on aorta, possibly by mediating endothelium derived relaxing factor (EDRF) in intact endothelium and voltage dependent L-type calcium channels blocking (CCB) mechanism in non-intact endothelium. Pc. Cr showed anti-hypertensive & cardioprotective activity by decreasing force of contraction & heart rate on isolated rabbit paired atria and reduced blood pressure in anesthetized rat. Cardioprotective effect of Pc. Cr was assessed in isoproterenol induced acute myocardial infarction (AMI) and left ventricular hypertrophy (LVH) in Sprague Dawley rats. In LVH, Pc. Cr exerted positive effects by decreasing angiotensin II & renin and increasing cGMP & nitric oxide (NO) with reduced cardiac fibrosis, necrosis and cardiac cell size. In AMI, Pc. Cr responded effectively by decreasing cardiac markers creatinine kinase (CK), creatinine kinase myocardial band (CK-MB) and lactate dehydrogenase (LD) in blood associated with less edema and necrosis. Presence of catechin, vinallic acid, P-coumeric acid and quercitin identified through HPLC support the effectiveness of Pc. Cr in hypertension, AMI and LVH. Pc. Cr showed no significant adverse effects in Sprague Dawley albino rats after acute & sub-acute treatment in histopathological investigation. Extract of Populus ciliata showed vasorelaxant, hypotensive and cardioprotective effect in Sprague Dawley albino rats and white albino rabbit by mediating EDRF and voltage dependent L-type CCB mechanism respectively.
Collapse
Affiliation(s)
- Fatima Saqib
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan.
| | - Asad Ali
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan.
| | - Hanadi Talal Ahmedah
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Rabigh 25732, Saudi Arabia.
| | | | | | | | - Marius Moga
- Faculty of Medicine, Transilvania University of Brasov, Romania.
| | - Marius Irimie
- Faculty of Medicine, Transilvania University of Brasov, Romania.
| |
Collapse
|
33
|
Krueger ES, Lloyd TS, Tessem JS. The Accumulation and Molecular Effects of Trimethylamine N-Oxide on Metabolic Tissues: It's Not All Bad. Nutrients 2021; 13:nu13082873. [PMID: 34445033 PMCID: PMC8400152 DOI: 10.3390/nu13082873] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/15/2021] [Accepted: 08/19/2021] [Indexed: 02/07/2023] Open
Abstract
Since elevated serum levels of trimethylamine N-oxide (TMAO) were first associated with increased risk of cardiovascular disease (CVD), TMAO research among chronic diseases has grown exponentially. We now know that serum TMAO accumulation begins with dietary choline metabolism across the microbiome-liver-kidney axis, which is typically dysregulated during pathogenesis. While CVD research links TMAO to atherosclerotic mechanisms in vascular tissue, its molecular effects on metabolic tissues are unclear. Here we report the current standing of TMAO research in metabolic disease contexts across relevant tissues including the liver, kidney, brain, adipose, and muscle. Since poor blood glucose management is a hallmark of metabolic diseases, we also explore the variable TMAO effects on insulin resistance and insulin production. Among metabolic tissues, hepatic TMAO research is the most common, whereas its effects on other tissues including the insulin producing pancreatic β-cells are largely unexplored. Studies on diseases including obesity, diabetes, liver diseases, chronic kidney disease, and cognitive diseases reveal that TMAO effects are unique under pathologic conditions compared to healthy controls. We conclude that molecular TMAO effects are highly context-dependent and call for further research to clarify the deleterious and beneficial molecular effects observed in metabolic disease research.
Collapse
Affiliation(s)
- Emily S. Krueger
- Department of Nutrition, Dietetics and Food Science, Brigham Young University, Provo, UT 84602, USA; (E.S.K.); (T.S.L.)
| | - Trevor S. Lloyd
- Department of Nutrition, Dietetics and Food Science, Brigham Young University, Provo, UT 84602, USA; (E.S.K.); (T.S.L.)
- Medical Education Program, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Jeffery S. Tessem
- Department of Nutrition, Dietetics and Food Science, Brigham Young University, Provo, UT 84602, USA; (E.S.K.); (T.S.L.)
- Correspondence: ; Tel.: +1-801-422-9082
| |
Collapse
|
34
|
Zhu J, Yi X, Ding H, Zhong L, Fang L. Integrated Transcriptomics and Reverse Pharmacophore Mapping-based Network Pharmacology to Explore the Mechanisms of Natural Compounds against Doxorubicin-induced Cardiotoxicity. Comb Chem High Throughput Screen 2021; 25:1707-1721. [PMID: 34397328 DOI: 10.2174/1386207324666210816122629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 06/08/2021] [Accepted: 06/27/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Doxorubicin-induced cardiotoxicity (DIC) has greatly limited the clinical benefits of this frontline drug in oncotherapy. Drug combination with natural compounds (NCs) that possess potency against DIC is considered as a promising intervention strategy. However, the mechanisms of action (MoAs) underlying such drug interactions remain poorly understood. The aim of this study was to systematically pursuit of the molecular mechanisms of NCs against DIC. METHODS First, the gene expression signatures of DIC were characterized from transcriptomics datasets with doxorubicin-treated and untreated cardiomyocytes using differentially expressed gene identification, functional enrichment analysis, and protein-protein interaction network analysis. Secondly, reverse pharmacophore mapping-based network pharmacology was employed to illustrate the MoAs of 82 publicly reported NCs with anti-DIC potency. Cluster analysis based on their enriched pathways was performed to gain systematic insights into the anti-DIC mechanisms of the NCs. Finally, the typical compounds were validated using gene set enrichment analysis (GSEA) of the relevant gene expression profiles from a public gene expression database. RESULTS Based on their anti-DIC MoAs, the 82 NCs could be divided into four groups, which corresponded to ten MoA clusters. GSEA and literature evidence on these compounds were provided to validate the MoAs identified through this bioinformatics analysis. The results suggested that NCs exerted potency against DIC through both common and different MoAs. CONCLUSION This strategy integrating different types of bioinformatics approaches is expected to create new insights for elucidating the MoAs of NCs against DIC.
Collapse
Affiliation(s)
- Junfeng Zhu
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Xiaojiao Yi
- Department of Pharmacy, Xixi Hospital of Hangzhou, Hangzhou 310023, China
| | - Haiying Ding
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Like Zhong
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Luo Fang
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| |
Collapse
|
35
|
Yamasan BE, Mercan T, Erkan O, Ozdemir S. Ellagic Acid Prevents Ca 2+ Dysregulation and Improves Functional Abnormalities of Ventricular Myocytes via Attenuation of Oxidative Stress in Pathological Cardiac Hypertrophy. Cardiovasc Toxicol 2021; 21:630-641. [PMID: 33909254 DOI: 10.1007/s12012-021-09654-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 04/20/2021] [Indexed: 01/25/2023]
Abstract
The aim of this study was to investigate whether ellagic acid (EA) treatment can prevent changes in contractile function and Ca2+ regulation of cardiomyocytes in pathologic cardiac hypertrophy. Groups were assigned as Con group; an ISO group in which the rats received isoproterenol alone (5 mg/kg/day); and an ISO + EA group in which the rats received isoproterenol and EA (20 mg/kg/day) for 4 weeks. Subsequently, fractional shortening, intracellular Ca2+ signals, and L-type Ca2+ currents of isolated ventricular myocytes were recorded. Protein expression levels were also determined by the Western blotting method. The survival rate was increased, and the upregulated cardiac hypertrophy markers were significantly attenuated with the EA treatment. The fractional shortening and relaxation rate of myocytes was decreased in the ISO group, whereas EA significantly improved these changes. Ventricular myocytes of the ISO + EA rats displayed lower diastolic Ca2+ levels, higher Ca2+ transients, shorter Ca2+ decay, and higher L-type Ca2+ currents than those of ISO rats. Protein expression analyses indicated that the upregulated p-PLB and p-CaMKII expressions were restored by EA treatment, suggesting improved calcium handling in the ISO + EA rat heart. Moreover, ISO rats displayed significantly increased expression of p-22phox and p47phox subunits of NOX2 protein. Expression of the p22phox subunit was reduced with EA administration, while the decrease in p47phox did not reach a significant level. The increased ROS impairs Ca2+ homeostasis and contractile activity of cardiac myocytes, whereas chronic EA administration prevents Ca2+ dysregulation and functional abnormalities associated with pathological cardiac hypertrophy via the diminution of oxidative stress.
Collapse
Affiliation(s)
- Bilge E Yamasan
- Department of Biophysics, Akdeniz University Faculty of Medicine, Antalya, Turkey
| | - Tanju Mercan
- Department of Biophysics, Akdeniz University Faculty of Medicine, Antalya, Turkey
| | - Orhan Erkan
- Department of Biophysics, Akdeniz University Faculty of Medicine, Antalya, Turkey
| | - Semir Ozdemir
- Department of Biophysics, Akdeniz University Faculty of Medicine, Antalya, Turkey.
| |
Collapse
|
36
|
Zhang X, Zhang Z, Wang P, Han Y, Liu L, Li J, Chen Y, Liu D, Wang J, Tian X, Zhao Q, Yan F. Bawei Chenxiang Wan Ameliorates Cardiac Hypertrophy by Activating AMPK/PPAR-α Signaling Pathway Improving Energy Metabolism. Front Pharmacol 2021; 12:653901. [PMID: 34149410 PMCID: PMC8209424 DOI: 10.3389/fphar.2021.653901] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 05/13/2021] [Indexed: 12/31/2022] Open
Abstract
Bawei Chenxiang Wan (BCW), a well-known traditional Chinese Tibetan medicine formula, is effective for the treatment of acute and chronic cardiovascular diseases. In the present study, we investigated the effect of BCW in cardiac hypertrophy and underlying mechanisms. The dose of 0.2, 0.4, and 0.8 g/kg BCW treated cardiac hypertrophy in SD rat model induced by isoprenaline (ISO). Our results showed that BCW (0.4 g/kg) could repress cardiac hypertrophy, indicated by macro morphology, heart weight to body weight ratio (HW/BW), left ventricle heart weight to body weight ratio (LVW/BW), hypertrophy markers, heart function, pathological structure, cross-sectional area (CSA) of myocardial cells, and the myocardial enzymes. Furthermore, we declared the mechanism of BCW anti-hypertrophy effect was associated with activating adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK)/peroxisome proliferator-activated receptor-α (PPAR-α) signals, which regulate carnitine palmitoyltransferase1β (CPT-1β) and glucose transport-4 (GLUT-4) to ameliorate glycolipid metabolism. Moreover, BCW also elevated mitochondrial DNA-encoded genes of NADH dehydrogenase subunit 1(ND1), cytochrome b (Cytb), and mitochondrially encoded cytochrome coxidase I (mt-co1) expression, which was associated with mitochondria function and oxidative phosphorylation. Subsequently, knocking down AMPK by siRNA significantly can reverse the anti-hypertrophy effect of BCW indicated by hypertrophy markers and cell surface of cardiomyocytes. In conclusion, BCW prevents ISO-induced cardiomyocyte hypertrophy by activating AMPK/PPAR-α to alleviate the disturbance in energy metabolism. Therefore, BCW can be used as an alternative drug for the treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Xiaoying Zhang
- Department of Pharmacology, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Zhiying Zhang
- Department of Pharmacology, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Pengxiang Wang
- Department of Pharmacology, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Yiwei Han
- Department of Pharmacology, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Lijun Liu
- Department of Pharmacology, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Jie Li
- Department of Pharmacology, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Yichun Chen
- Department of Pharmacology, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Duxia Liu
- Department of Pharmacology, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Jinying Wang
- School of Medical Science, Jinan University, Guangzhou, China
| | - Xiaoying Tian
- School of Medical Science, Jinan University, Guangzhou, China
| | - Qin Zhao
- Department of Pharmacology, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Fengxia Yan
- School of Medical Science, Jinan University, Guangzhou, China
| |
Collapse
|
37
|
Chen Y, Pan R, Zhang J, Liang T, Guo J, Sun T, Fu X, Wang L, Zhang L. Pinoresinol diglucoside (PDG) attenuates cardiac hypertrophy via AKT/mTOR/NF-κB signaling in pressure overload-induced rats. JOURNAL OF ETHNOPHARMACOLOGY 2021; 272:113920. [PMID: 33607200 DOI: 10.1016/j.jep.2021.113920] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/01/2021] [Accepted: 02/08/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pinoresinol diglucoside (PDG), the active compound extracted from Eucommia ulmoides, Styrax sp. and Forsythia suspensa, plays the roles in regulating hypertension, inflammation and oxidative stress. AIMS Considering that hypertension and inflammation has been proved to contribute to cardiac remodeling, we tested the effects of PDG on cardiac hypertrophy (CM). METHODS Male Sprague Dawley (SD) rats were used to construct hypertrophic rats by partial abdominal aortic constriction (AAC)-surgery. PDG solution (2 mg/ml) was used to treat AAC-induced rats by intraperitoneal injection at low dose (L-PDG, 2.5 mg/kg per day), medium dose (M-PDG, 5 mg/kg per day), and high dose (H-PDG, 7.5 mg/kg per day) for 3 weeks post AAC-surgery. CM was evaluated by the ratio of left ventricular weight to body weight ratio (LVW/BW), left ventricular wall thickness by H&E staining, and collagen content deposit by Masson's staining. Further, isoproterenol (ISO) and phenylephrine (PE) were used to produce cellular models of CM in neonatal rat ventricular cardiomyocytes (NRVMs). PDG pre-treated NRVMs 2 h at low dose (L-PDG, 2.5 μg/ml), medium dose (M-PDG, 5 μg/ml), and high dose (H-PDG, 7.5 μg/ml) for 24 h with or without PE- and ISO-stimulation. CM was evaluated by the expressions of hypertrophic biomarkers. Next, the hypertrophic biomarkers and pro-inflammatory cytokines were measured using quantitative real-time PCR (qRT-PCR), the expressions of protein kinase B (AKT)/mammalian target of rapamycin (mTOR)/transcription factor nuclear factor-kappa B (NF-kB) signaling pathway were determined by Western blotting. RESULTS PDG treatment prevented cardiac histomorphology damages, decreased upregulations of hypertrophic biomarkers, and prevented fibrosis and inflammation after pressure overload resulting from AAC-surgery. Consistently, PDG remarkably inhibited the changes of cardiomyocyte hypertrophic biomarkers and inflammatory responses in cellular models of CM. Interestingly, PDG administration inhibited the activation of AKT/mTOR/NF-kB signaling pathway both in vivo and in vitro. CONCLUSIONS PDG prevents AAC-induced CM in vivo, PE- and ISO-induced CM in vitro. The AKT/mTOR/NF-kB signaling pathway could be the potential therapeutic target involved in the protection of PDG. These findings provide novel evidence that PDG might be a promising therapeutic strategy for CM.
Collapse
Affiliation(s)
| | | | | | | | | | - Tai Sun
- School of Basic Medicine, PR China
| | | | - Ling Wang
- Medical Experiment and Training Center, Weifang Medical University, Weifang 261053, PR China
| | | |
Collapse
|
38
|
Zhao T, Kee HJ, Bai L, Kim MK, Kee SJ, Jeong MH. Selective HDAC8 Inhibition Attenuates Isoproterenol-Induced Cardiac Hypertrophy and Fibrosis via p38 MAPK Pathway. Front Pharmacol 2021; 12:677757. [PMID: 33959033 PMCID: PMC8093872 DOI: 10.3389/fphar.2021.677757] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 03/29/2021] [Indexed: 12/21/2022] Open
Abstract
Histone deacetylase (HDAC) expression and enzymatic activity are dysregulated in cardiovascular diseases. Among Class I HDACs, HDAC2 has been reported to play a key role in cardiac hypertrophy; however, the exact function of HDAC8 remains unknown. Here we investigated the role of HDAC8 in cardiac hypertrophy and fibrosis using the isoproterenol-induced cardiac hypertrophy model system.Isoproterenol-infused mice were injected with the HDAC8 selective inhibitor PCI34051 (30 mg kg−1 body weight). Enlarged hearts were assessed by HW/BW ratio, cross-sectional area, and echocardiography. RT-PCR, western blotting, histological analysis, and cell size measurements were performed. To elucidate the role of HDAC8 in cardiac hypertrophy, HDAC8 knockdown and HDAC8 overexpression were also used. Isoproterenol induced HDAC8 mRNA and protein expression in mice and H9c2 cells, while PCI34051 treatment decreased cardiac hypertrophy in isoproterenol-treated mice and H9c2 cells. PCI34051 treatment also reduced the expression of cardiac hypertrophic markers (Nppa, Nppb, and Myh7), transcription factors (Sp1, Gata4, and Gata6), and fibrosis markers (collagen type I, fibronectin, and Ctgf) in isoproterenol-treated mice. HDAC8 overexpression stimulated cardiac hypertrophy in cells, whereas HDAC8 knockdown reversed those effects. HDAC8 selective inhibitor and HDAC8 knockdown reduced the isoproterenol-induced activation of p38 MAPK, whereas HDAC8 overexpression promoted p38 MAPK phosphorylation. Furthermore, p38 MAPK inhibitor SB203580 significantly decreased the levels of p38 MAPK phosphorylation, as well as ANP and BNP protein expression, induced by HDAC8 overexpression.Here we show that inhibition of HDAC8 activity or expression suppresses cardiac hypertrophy and fibrosis. These findings suggest that HDAC8 could be a promising target to treat cardiac hypertrophy and fibrosis by regulating p38 MAPK.
Collapse
Affiliation(s)
- Tingwei Zhao
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Hae Jin Kee
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Liyan Bai
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Moon-Ki Kim
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Seung-Jung Kee
- Department of Laboratory Medicine, Chonnam National University, Medical School and Hospital, Gwangju, Republic of Korea
| | - Myung Ho Jeong
- Heart Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Republic of Korea.,Department of Cardiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| |
Collapse
|
39
|
Lee JH, Park M, Jung K, Hong G, Lee HL, Kim DW, Kim CE, Kang KS. Identification of gallic acid as a active ingredient of Syzygium aromaticum against tacrolimus-induced damage in renal epithelial LLC-PK1 cells and rat kidney. Bioorg Med Chem Lett 2021; 41:128012. [PMID: 33838305 DOI: 10.1016/j.bmcl.2021.128012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/19/2021] [Accepted: 03/28/2021] [Indexed: 11/29/2022]
Abstract
Tacrolimus (FK506), a calcineurin inhibitor, is an effective immunosuppressive agent mainly used to lower the risk of organ rejection after allogeneic organ transplant. However, FK506-associated adverse effects, such as nephrotoxicity, may limit its therapeutic use. In this study, we confirmed that epigallocatechin-3-gallate (EGCG), sanguiin H-6, and gallic acid increased cell survival following FK506-induced cytotoxicity in renal epithelial LLC-PK1. Among these compounds, gallic acid exerted the strongest protective effect, further confirmed in the FK506-induced nephrotoxicity rat model. Additionally, we identified supporting evidence for the nephroprotective function of gallic acid using molecular docking and bioavailability investigations.
Collapse
Affiliation(s)
- Ji Hwan Lee
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
| | - Musun Park
- Department of Physiology, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea; Intellectual Information Team, Future Medicine Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Kiwon Jung
- Institute of Pharmaceutical Sciences, College of Pharmacy, CHA University, Sungnam 13844, Republic of Korea
| | - Gyeongmin Hong
- Institute of Pharmaceutical Sciences, College of Pharmacy, CHA University, Sungnam 13844, Republic of Korea
| | - Hye Lim Lee
- Department of Pediatrics, College of Korean Medicine, Daejeon University, Daejeon, Republic of Korea
| | - Dong-Wook Kim
- Department of Pharmaceutical Engineering, Cheongju University, Cheongju 28530, Republic of Korea
| | - Chang-Eop Kim
- Department of Physiology, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea.
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea.
| |
Collapse
|
40
|
Lin YM, Badrealam KF, Kuo CH, Daddam J, Asokan Shibu M, Lin KH, Ho TJ, Viswanadha VP, Kuo WW, Huang CY. Small Molecule Compound Nerolidol attenuates Hypertension induced hypertrophy in spontaneously hypertensive rats through modulation of Mel-18-IGF-IIR signalling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 84:153450. [PMID: 33611212 DOI: 10.1016/j.phymed.2020.153450] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 12/16/2020] [Accepted: 12/22/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Cardiovascular diseases are caused by multitudes of stress factors like hypertension and their outcomes are associated with high mortality and morbidity worldwide. Nerolidol, a naturally occurring sesquiterpene found in several plant species, embodies various pharmacological benefits against numerous health disorders. However, their effects on hypertension induced cardiac complications are not completely understood. PURPOSE The present study is to elucidate the efficacy of nerolidol against hypertension related cardiac hypertrophy in spontaneously hypertensive rats (SHRs). STUDY DESIGN For preliminary in vitro studies, H9c2 cardiomyoblasts cells were challenged with 200 nM Angiotensin-II (AngII) for 12 h and were then treated with nerolidol for 24 h. The hypertrophic effect in H9c2 cells were analyzed by actin staining and the modulations in hypertrophic protein markers and mediators were determined by Western blotting analysis. For in vivo experiments, sixteen week-old male Wistar Kyoto (WKY) and SHRs were segregated into five groups (n = 9): Control WKY, hypertensive SHRs, SHRs with low dose (75 mg/kg b.w/day) nerolidol, SHRs with high dose (150 mg/kg b.w/day) nerolidol and SHR rats treated with an anti-hypertensive drug captopril (50 mg/kg b.w/day). Nerolidol treatment was given orally for 8 weeks and were analysed through Echocardiography. After euthanasia, hematoxylin and eosin staining, Immunohistochemical analysis and Western blotting was performed on left ventricle tissue. RESULTS Western blotting analysis revealed that nerolidol significantly attenuates AngII induced expression of hypertrophic markers ANP and BNP in H9c2 cardiomyoblasts. In addition, actin staining further ascertained the potential of nerolidol to ameliorate AngII induced cardiac hypertrophy. Moreover, nerolidol administration suppressed the hypertrophic signalling mediators like calcineurin, GATA4, Mel-18, HSF-2 and IGFIIR in a dose-dependent fashion. In silico studies also ascertained the role of Mel-18 in the ameliorative effects of nerolidol. Further, these intriguing in vitro results were further confirmed in in vivo SHR model. Oral neraolidol in SHRs efficiently reduced blood pressure and ameliorated hypertension induced cardiac hypertrophic effects by effectively reducing the levels of proteins involved in cardiac MeL-18-HSF2-IGF-IIR signalling. CONCLUSION Collectively, the data reveals that the cardioprotective effect of nerolidol against hypertension induced hypertrophy involves reduction in blood pressure and regulation of the cardiac Mel-18-IGFIIR signalling cascade.
Collapse
Affiliation(s)
- Yueh-Min Lin
- Department of Pathology, Changhua Christian Hospital, Changhua 500, Taiwan; Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Taipei 11260, Taiwan
| | - Khan Farheen Badrealam
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taiwan
| | - Jayasimharayalu Daddam
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Marthandam Asokan Shibu
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Kuan-Ho Lin
- College of Medicine, China Medical University, Taichung, Taiwan; Department of Emergency Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Tsung-Jung Ho
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Hualien 97002, Taiwan; Department of Chinese Medicine, Hualien Tzu Chi Hospital, Hualien 97002, Taiwan; School of Post-Baccalaureate Chinese Medicine, College of Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | | | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung; Ph.D. Program for Biotechnology Industry, China Medical University, Taichuang 406, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan; Department of Biological Science and Technology, Asia University, Taichung, Taiwan; Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien 970, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan.
| |
Collapse
|
41
|
Najjar RS, Schwartz AM, Wong BJ, Mehta PK, Feresin RG. Berries and Their Polyphenols as a Potential Therapy for Coronary Microvascular Dysfunction: A Mini-Review. Int J Mol Sci 2021; 22:3373. [PMID: 33806050 PMCID: PMC8036956 DOI: 10.3390/ijms22073373] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 12/16/2022] Open
Abstract
Ischemia with no obstructive coronary artery disease (INOCA) is a common diagnosis with a higher prevalence in women compared to men. Despite the absence of obstructive coronary artery disease and no structural heart disease, INOCA is associated with major adverse cardiovascular outcomes as well a significant contributor to angina and related disability. A major feature of INOCA is coronary microvascular dysfunction (CMD), which can be detected by non-invasive imaging and invasive coronary physiology assessments in humans. CMD is associated with epicardial endothelial-dependent and -independent dysfunction, diffuse atherosclerosis, and left-ventricular hypertrophy, all of which lead to insufficient blood flow to the myocardium. Inflammatory and oxidative stress signaling, upregulation of the renin-angiotensin-aldosterone system and adrenergic receptor signaling are major drivers of CMD. Treatment of CMD centers around addressing cardiovascular risk factors; however, there are limited treatment options for those who do not respond to traditional anti-anginal therapies. In this review, we highlight the ability of berry-derived polyphenols to modulate those pathways. The evidence supports the need for future clinical trials to investigate the effectiveness of berries and their polyphenols in the treatment of CMD in INOCA patients.
Collapse
Affiliation(s)
- Rami S. Najjar
- Department of Nutrition, Georgia State University, Atlanta, GA 30302, USA;
| | - Arielle M. Schwartz
- J. Willis Hurst Internal Medicine Residency Program, Emory University, Atlanta, GA 30322, USA;
| | - Brett J. Wong
- Department of Kinesiology & Health, Georgia State University, Atlanta, GA 30302, USA;
| | - Puja K. Mehta
- Division of Cardiology, Emory Women’s Heart Center, Emory University School of Medicine, Atlanta, GA 30322, USA
- Division of Cardiology, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Rafaela G. Feresin
- Department of Nutrition, Georgia State University, Atlanta, GA 30302, USA;
| |
Collapse
|
42
|
Estrada AC, Yoshida K, Saucerman JJ, Holmes JW. A multiscale model of cardiac concentric hypertrophy incorporating both mechanical and hormonal drivers of growth. Biomech Model Mechanobiol 2021; 20:293-307. [PMID: 32970240 PMCID: PMC7897221 DOI: 10.1007/s10237-020-01385-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 09/08/2020] [Indexed: 01/19/2023]
Abstract
Growth and remodeling in the heart is driven by a combination of mechanical and hormonal signals that produce different patterns of growth in response to exercise, pregnancy, and various pathologies. In particular, increases in afterload lead to concentric hypertrophy, a thickening of the walls that increases the contractile ability of the heart while reducing wall stress. In the current study, we constructed a multiscale model of cardiac hypertrophy that connects a finite-element model representing the mechanics of the growing left ventricle to a cell-level network model of hypertrophic signaling pathways that accounts for changes in both mechanics and hormones. We first tuned our model to capture published in vivo growth trends for isoproterenol infusion, which stimulates β-adrenergic signaling pathways without altering mechanics, and for transverse aortic constriction (TAC), which involves both elevated mechanics and altered hormone levels. We then predicted the attenuation of TAC-induced hypertrophy by two distinct genetic interventions (transgenic Gq-coupled receptor inhibitor overexpression and norepinephrine knock-out) and by two pharmacologic interventions (angiotensin receptor blocker losartan and β-blocker propranolol) and compared our predictions to published in vivo data for each intervention. Our multiscale model captured the experimental data trends reasonably well for all conditions simulated. We also found that when prescribing realistic changes in mechanics and hormones associated with TAC, the hormonal inputs were responsible for the majority of the growth predicted by the multiscale model and were necessary in order to capture the effect of the interventions for TAC.
Collapse
|
43
|
A coaxially structured trilayered gallic acid-based antioxidant vascular graft for treating coronary artery disease. Eur Polym J 2021. [DOI: 10.1016/j.eurpolymj.2020.110203] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
44
|
Han D, Zhang QY, Zhang YL, Han X, Guo SB, Teng F, Yan X, Li HH. Gallic Acid Ameliorates Angiotensin II-Induced Atrial Fibrillation by Inhibiting Immunoproteasome- Mediated PTEN Degradation in Mice. Front Cell Dev Biol 2020; 8:594683. [PMID: 33251220 PMCID: PMC7673442 DOI: 10.3389/fcell.2020.594683] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/13/2020] [Indexed: 11/13/2022] Open
Abstract
Atrial fibrillation (AF) is the most prevalent cardiac arrhythmia and is a major cause of stroke and heart failure. We and others have found that gallic acid (GA) plays a beneficial role in cardiac hypertrophic remodeling and hypertension. However, the effect of GA on angiotensin II (Ang II)-induced AF and atrial remodeling as well as the underlying mechanisms remain unknown. AF was induced in mice by Ang II infusion (2000 ng/kg/min) for 3 weeks. Blood pressure was measured using the tail-cuff method. Atrial volume was evaluated by echocardiography. Atrial remodeling was studied using hematoxylin and eosin, Masson's trichrome, and immunohistochemical staining. Atrial oxidative stress was assessed by dihydroethidium staining. The gene expression of fibrotic and inflammatory markers and protein levels of signaling mediators were measured by quantitative real-time PCR and western blot analysis. In mice, GA administration significantly attenuated Ang II-induced elevation of blood pressure, AF incidence and duration, atrial dilation, fibrosis, inflammation, and oxidative stress compared with the vehicle control. Furthermore, GA downregulated Ang II-induced activity and expression of immunoproteasome subunits (β2i and β5i), which reduced PTEN degradation and led to the inactivation of AKT1 and downstream signaling mediators. Importantly, blocking PTEN activity by VO-Ohpic markedly reversed the GA-mediated protective effects on Ang II-induced AF and atrial remodeling. Therefore, our results provide novel evidence that GA exerts a cardioprotective role by inhibiting immunoproteasome activity, which attenuates PTEN degradation and activation of downstream signaling, and may represent a promising candidate for treating hypertensive AF.
Collapse
Affiliation(s)
- Dan Han
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qi-Yu Zhang
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yun-Long Zhang
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Capital Medical University, Beijing, China
| | - Xiao Han
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Capital Medical University, Beijing, China
| | - Shu-Bin Guo
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Capital Medical University, Beijing, China
| | - Fei Teng
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Capital Medical University, Beijing, China
| | - Xiao Yan
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Capital Medical University, Beijing, China
| | - Hui-Hua Li
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China.,Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Capital Medical University, Beijing, China
| |
Collapse
|
45
|
Abstract
Arbutin is a glycoside reported for its anti-oxidant, anti-inflammatory and anti-tumor properties. However, the cardioprotective effect of Arbutin is not well established. The study aims to understand the effect of arbutin on isoproterenol (ISO)-induced cardiac hypertrophy in mice. The animals were pretreated with Arbutin for a week and ISO was administered for 10 days and then sacrificed. Cardiac injury markers such as creatinine kinase and lactate dehydrogenase concentrations were measured in the serum. The mRNA expression of cardiac hypertrophy markers namely atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) were measured using qRT-PCR. The levels of pro-inflammatory cytokines TNF-α and IL-6 were quantified by ELISA in isolated tissues and serum. Other tissue anti-oxidant parameters such as GST, GSH, SOD and TBARS were also measured. TUNEL assay was performed to detect apoptosis. Histology studies were performed using H & E and Masson trichome staining. Immunoblot analysis was used to quantify the protein expression of TLR-4 and NF-κB. ISO-alone-treated group showed significant increase in CK-MB, LDH along with increase in hypertrophic markers ANP and BNP, TNF-α and IL-6 levels in serum and tissues and increased cardiomyocyte apoptosis. Anti-oxidant parameters were significantly decreased and TLR-4 and NF-κB protein expression was found to be upregulated in comparison to the control group. Pretreatment with Arbutin-exhibited significant inhibition of TLR-4/NF-κB pathway with decreased levels of pro-inflammatory cytokines and enhanced myocardial anti-oxidant status. Our study demonstrated that pretreatment with Arbutin exhibits marked protective effects on ISO-induced cardiac hypertrophy in mice. Thus, Arbutin may be used as potential pharmacological interventions in the management of cardiac hypertrophy.
Collapse
|
46
|
Viswanadha VP, Dhivya V, Beeraka NM, Huang CY, Gavryushova LV, Minyaeva NN, Chubarev VN, Mikhaleva LM, Tarasov VV, Aliev G. The protective effect of piperine against isoproterenol-induced inflammation in experimental models of myocardial toxicity. Eur J Pharmacol 2020; 885:173524. [PMID: 32882215 DOI: 10.1016/j.ejphar.2020.173524] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/24/2020] [Accepted: 08/28/2020] [Indexed: 12/19/2022]
Abstract
Myocardial infarction (MI) eventually exacerbates inflammatory response due to the release of inflammatory and pro-inflammatory factors. The aim of this study is to explore the protective efficacy of piperine supplementation against the inflammatory response in isoproterenol (ISO)-induced MI. Masson Trichome staining was executed to determine myocardial tissue architecture. Immunohistochemistry was performed for IL-6, TNF-α. RT-PCR studies were performed to ascertain the gene expression of IL-6, TNF-α, iNOS, eNOS, MMP-2, MMP-9, and collagen-III. Western blotting was performed to determine expression of HIF-1α, VEGF, Nrf-2, NF-ƙB, Cox-2, p-38, phospho-p38, ERK-1/2, phospho-ERK-1/2, and collagen-I. HIF-1α, VEGF, and iNOS expression were significantly upregulated with concomitant decline in eNOS expression in the heart myocardial tissue of rats received ISO alone whereas piperine pretreatment prevented these changes in ISO administered rats. Current results revealed ROS-mediated activation of MAPKs, namely, p-p38, p-ERK1/2 in the heart tissue of ISO administered group. Piperine pretreatment significantly prevented these changes in ISO treated group. NF-κB is involved in the modulation of gene expressions responsible for tissue repair. ISO-induced NF-κB-p65 expression was significantly reduced in the group pretreated with piperine and mitigated extent of myocardial inflammation. A significant increase in cardiac fibrosis upon ISO treatment was reported due to the increased hydroxyproline content, MMP-2 & 9 and upregulation of collagen-I protein compared to control group. All these cardiac hypertrophy markers were decreased in 'piperine pretreated ISO administered group' compared to group received ISO injection. Current findings concluded that piperine as a nutritional intervention could prevent inflammation of myocardium in ISO-induced MI.
Collapse
Affiliation(s)
- Vijaya Padma Viswanadha
- Translational Research Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India; China Medical University, Lifu Teaching Building 12F, 91 Hsueh-Shih Road, Taichung, 40402, Taiwan.
| | - Velumani Dhivya
- Translational Research Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Narasimha Murthy Beeraka
- Translational Research Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Chih-Yang Huang
- China Medical University, Lifu Teaching Building 12F, 91 Hsueh-Shih Road, Taichung, 40402, Taiwan
| | - Liliya V Gavryushova
- Department of Therapeutic Dentistry, Saratov State Medical University named after V.I. Razumovsky, 410012, Saratov, Russia
| | - Nina N Minyaeva
- National Research University Higher School of Economics, 20 Myasnitskaya Street, Moscow, 101000, Russia
| | - Vladimir N Chubarev
- Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
| | - Liudmila M Mikhaleva
- Research Institute of Human Morphology, Russian Academy of Medical Science, Street Tsyurupa 3, Moscow, 117418, Russia
| | - Vadim V Tarasov
- Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
| | - Gjumrakch Aliev
- Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia; Research Institute of Human Morphology, Russian Academy of Medical Science, Street Tsyurupa 3, Moscow, 117418, Russia; Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, Moscow Region, 142432, Russia; GALLY International Research Institute, 7733 Louis Pasteur Drive, #330, San Antonio, TX, 78229, USA.
| |
Collapse
|
47
|
Yan X, Zhang QY, Zhang YL, Han X, Guo SB, Li HH. Gallic Acid Attenuates Angiotensin II-Induced Hypertension and Vascular Dysfunction by Inhibiting the Degradation of Endothelial Nitric Oxide Synthase. Front Pharmacol 2020; 11:1121. [PMID: 32848742 PMCID: PMC7396711 DOI: 10.3389/fphar.2020.01121] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/10/2020] [Indexed: 01/15/2023] Open
Abstract
Hypertension is a major cause of heart attack and stroke. Our recent study revealed that gallic acid (GA) exerts protective effects on pressure overload-induced cardiac hypertrophy and dysfunction. However, the role of GA in angiotensin II (Ang II)-induced hypertension and vascular remodeling remains unknown. C57BL/6J mice were subjected to saline and Ang II infusion. Systolic blood pressure was measured using a tail-cuff system. Vascular remodeling and oxidative stress were examined by histopathological staining. Vasodilatory function was evaluated in the aortic ring. Our findings revealed that GA administration significantly ameliorated Ang II-induced hypertension, vascular inflammation, and fibrosis. GA also abolished vascular endothelial dysfunction and oxidative stress in Ang II-infused aortas. Mechanistically, GA treatment attenuated Ang II-induced upregulation of the immunoproteasome catalytic subunits β2i and β5i leading to reduction of the trypsin-like and chymotrypsin-like activity of the proteasome, which suppressed degradation of endothelial nitric oxide synthase (eNOS) and reduction of nitric oxide (NO) levels. Furthermore, blocking eNOS activity by using a specific inhibitor (L-NG-nitroarginine methyl ester) markedly abolished the GA-mediated beneficial effect. This study identifies GA as a novel immunoproteasome inhibitor that may be a potential therapeutic agent for hypertension and vascular dysfunction.
Collapse
Affiliation(s)
- Xiao Yan
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, and Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| | - Qi-Yu Zhang
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yun-Long Zhang
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, and Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| | - Xiao Han
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, and Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| | - Shu-Bin Guo
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, and Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China
| | - Hui-Hua Li
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, and Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing, China.,Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
48
|
Saqib F, Mujahid K, Aslam MA, Modhi A, Moga MA, Bobescu E, Marceanu L. Ex vivo and in vivo studies of Viola tricolor Linn. as potential cardio protective and hypotensive agent: Inhibition of voltage-gated Ca ++ ion channels. FASEB J 2020; 34:9102-9119. [PMID: 32475023 DOI: 10.1096/fj.202000658r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 04/12/2020] [Accepted: 04/20/2020] [Indexed: 11/11/2022]
Abstract
Viola tricolor Linn. is used as cardio-protective and anti-hypertensive agent in traditional medicine. Current study objective was to evaluate cardio-protective and hypotensive effects of Viola tricolor L. in vitro and in vivo studies. Viola tricolor L. crude extract (Vt.Cr) and its fractions (Aqueous and organic) were tested at rabbit atria and aorta coupled to Power Lab Data Acquisition System for cardio depressant and vasorelaxant effects in vitro whereas in vivo Blood Pressure was checked by invasive method in normotensive ketamine-diazepam anesthetized rats. Isoproterenol was employed for acute myocardial infarction (AMI) and left ventricular hypertrophy (LVH) development and cardioprotective effects of Vt.Cr were evaluated hemodynamically and histopathologically. Vt.Cr and its fractions decreased heart rate and contractile force in paired atria and relaxed Phenylephrine (1 µM) and K+ (80 mM) stimulated contractions in aorta possibly mediated through Voltage dependent L-type calcium channels blockage supported by in vivo hypotensive action. In LVH, Vt.Cr lowered Angiotensin Converting Enzymes and renin, increased cyclic Guanosine Monophosphate and nitric oxide levels, decreased cardiomyocytes size and fibrosis attributed to Gallic acid as detected by High Performance Liquid Chromatography. Partial positive results were seen hemodynamically and histologically in AMI Viola tricolor L. showed vasorelaxant, cardio-relaxant, hypotensive, and cardio protective effect validating traditional practice in cardiovascular disorders.
Collapse
Affiliation(s)
- Fatima Saqib
- Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Khizra Mujahid
- Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | | | - Alotaibi Modhi
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | | | - Elena Bobescu
- Faculty of Medicine, Transilvania University Brasov, Brasov, Romania
| | - Luigi Marceanu
- Faculty of Medicine, Transilvania University Brasov, Brasov, Romania
| |
Collapse
|
49
|
Amniotic membrane mesenchymal stem cells labeled by iron oxide nanoparticles exert cardioprotective effects against isoproterenol (ISO)-induced myocardial damage by targeting inflammatory MAPK/NF-κB pathway. Drug Deliv Transl Res 2020; 11:242-254. [PMID: 32441012 DOI: 10.1007/s13346-020-00788-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The aim of the present study is to investigate the protective effects of human amniotic membrane-derived mesenchymal stem cells (hAMSCs) labeled by superparamagnetic iron oxide nanoparticles (SPIONs) against isoproterenol (ISO)-induced myocardial injury in the presence and absence of a magnetic field. ISO was injected subcutaneously for 4 consecutive days to induce myocardial injury in male Wistar rats. The hAMSCs were incubated with 100 μg/ml SPIONs and injected to rats in magnet-dependent and magnet-independent groups via the tail vein. The size and shape of nanoparticles were determined by dynamic light scattering (DLS) and transmission electron microscopy (TEM). Prussian blue staining was used to determine cell uptake of nanoparticles. Myocardial fibrosis, heart function, characterization of hAMSCs, and histopathological changes were determined using Masson's trichrome, echocardiography, flow cytometry, and H&E staining, respectively. Enzyme-linked immunosorbent assay (ELISA) was used to the expression pro-inflammatory cytokines. Immunohistochemistry assay was used to determine the expression of nuclear factor-κB (NF-κB) and the Ras/mitogen-activated protein kinase (MAPK). SPION-labeled MSCs in the presence of magnetic field significantly improved cardiac function and reduced fibrosis and tissue damage by suppressing inflammation in a NF-κB/MAPK-dependent mechanism (p < 0. 05). Collectively, our findings demonstrate that SPION-labeled MSCs in the presence of magnetic field can be a good treatment option to reduce inflammation following myocardial injury. Graphical abstract.
Collapse
|
50
|
Cymbopogon Proximus Essential Oil Protects Rats against Isoproterenol-Induced Cardiac Hypertrophy and Fibrosis. Molecules 2020; 25:molecules25081786. [PMID: 32295062 PMCID: PMC7221672 DOI: 10.3390/molecules25081786] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/08/2020] [Accepted: 04/11/2020] [Indexed: 01/19/2023] Open
Abstract
Cardiac hypertrophy is an independent risk factor of many cardiovascular diseases. Several cardiovascular protective properties of Cymbopogon proximus have been reported. However, no reports investigating the direct effect of C. proximus essential oil on the heart are available. The goal of this study was to explore the cardioprotective effect of C. proximus on cardiac hypertrophy and fibrosis. Male albino rats were administered C. proximus essential oil in the presence or absence of hypertrophic agonist isoproterenol. Cardiac hypertrophy and fibrosis were assessed using real-time polymerase chain reaction (PCR) and histological examination. Pre- treatment of rats with C. proximus decreased the ratio of heart weight to body weight and gene expression of hypertrophy markers atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), and β-myosin heavy chain (β-MHC), which were induced by isoproterenol. Moreover, C. proximus prevented the increase in gene expression of fibrosis markers procollagen I and procollagen III and alleviated the collagen volume fraction caused by isoproterenol. The pre- treatment with C. proximus essential oil conferred cardio-protection against isoproterenol- induced cardiac hypertrophy and fibrosis.
Collapse
|