1
|
Zhang R, Wang P, Jin Y, Xie Q, Xiao P. Imperatorin's Effect on Myocardial Infarction Based on Network Pharmacology and Molecular Docking. Cardiovasc Ther 2025; 2025:7551459. [PMID: 39834616 PMCID: PMC11745561 DOI: 10.1155/cdr/7551459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 12/31/2024] [Indexed: 01/22/2025] Open
Abstract
Purpose: Myocardial infarction (MI), a severe cardiovascular disease, is the result of insufficient blood supply to the myocardium. Despite the improvements of conventional therapies, new approaches are needed to improve the outcome post-MI. Imperatorin is a natural compound with multiple pharmacological properties and potential cardioprotective effects. Therefore, this work investigated imperatorin's therapeutic effects and its mechanism of action in an MI mouse model. Methods: Network pharmacology, molecular docking, and experimental validation were performed for exploring the pharmacokinetic properties, therapeutic effects, and molecular targets of imperatorin in MI. Permanent ligation of the left anterior descending artery was performed in male C57BL/6 mice to induce MI before treatment with imperatorin once per day for 1 week. Echocardiography, heart histology, RNA sequencing, and quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) as well as western blotting were carried out for evaluating cardiac function and structure, as well as gene and protein expression. Results: Imperatorin significantly improved cardiac function, preserved cardiac structure, attenuated cardiac remodeling and fibrosis, and reduced cardiomyocyte apoptosis in MI mice. Eight differentially expressed genes overlapping with key target genes were found, two upregulated and six downregulated. A key target in signaling pathways associated with imperatorin effect in MI was angiotensin-converting enzyme (ACE). Imperatorin inhibited ACE-angiotensin II (Ang II)-angiotensin II Type 1 receptor (AT1R) axis in MI mice. Conclusion: Imperatorin attenuated MI by inhibiting the ACE-Ang II-AT1R axis. Thus, imperatorin might be considered a potential therapeutic agent to cure MI.
Collapse
MESH Headings
- Animals
- Myocardial Infarction/drug therapy
- Myocardial Infarction/pathology
- Myocardial Infarction/metabolism
- Myocardial Infarction/physiopathology
- Myocardial Infarction/genetics
- Male
- Furocoumarins/pharmacology
- Furocoumarins/chemistry
- Mice, Inbred C57BL
- Disease Models, Animal
- Network Pharmacology
- Molecular Docking Simulation
- Apoptosis/drug effects
- Signal Transduction/drug effects
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/metabolism
- Ventricular Remodeling/drug effects
- Fibrosis
- Ventricular Function, Left/drug effects
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/drug effects
- Angiotensin II
- Gene Expression Regulation
- Gene Regulatory Networks
Collapse
Affiliation(s)
- Ruizhe Zhang
- Department of Cardiology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Peng Wang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yao Jin
- Department of Cardiology, The Fourth Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Qingya Xie
- Department of Cardiology, The Fourth Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Pingxi Xiao
- Department of Cardiology, The Fourth Affiliated Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
2
|
Panthiya L, Tocharus J, Chaichompoo W, Suksamrarn A, Tocharus C. Hexahydrocurcumin mitigates angiotensin II-induced proliferation, migration, and inflammation in vascular smooth muscle cells. EXCLI JOURNAL 2023; 22:466-481. [PMID: 37534221 PMCID: PMC10391613 DOI: 10.17179/excli2023-6124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 05/30/2023] [Indexed: 08/04/2023]
Abstract
The proliferation and migration of vascular smooth muscle cells (VSMCs) play vital roles in the pathogenesis of atherosclerosis and hypertension. It has been proposed and verified that hexahydrocurcumin (HHC), a metabolite form of curcumin, has cardiovascular protective effects. This study examined the effect of HHC on angiotensin II (Ang II)-induced proliferation, migration, and inflammation in rat aortic VSMCs and explored the molecular mechanisms related to the processes. The results showed that HHC significantly suppressed Ang II-induced proliferation, migration, and inflammation in VSMCs. HHC inhibited Ang II-induction of the increase in cyclin D1 and decrease in p21 expression in VSMCs. Moreover, HHC attenuated the generation of reactive oxygen species (ROS), and the expression of nuclear factor kappa B (NF-κB), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and matrix metalloproteinases-9 (MMP9) in Ang II-induced VSMCs. The proliferation, migration, inflammation, and ROS production were also inhibited by GKT137831 (NADPH oxidase, NOX1/4 inhibitor) and the combination of HHC and GKT137831. In addition, HHC restored the Ang-II inhibited expression of peroxisome proliferator-activated receptor-γ (PPAR-γ) and peroxisome proliferator activated receptor-γ coactivator-1α (PGC-1α). These findings indicate that HHC may play a protective role in Ang II-promoted proliferation, migration, and inflammation by suppressing NADPH oxidase mediated ROS generation and elevating PPAR-γ and PGC-1α expression. See also Figure 1(Fig. 1).
Collapse
Affiliation(s)
- Luckika Panthiya
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Graduate School, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Jiraporn Tocharus
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Waraluck Chaichompoo
- Department of Chemistry and Center of Excellence of Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok 10240, Thailand
| | - Apichart Suksamrarn
- Department of Chemistry and Center of Excellence of Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok 10240, Thailand
| | - Chainarong Tocharus
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Functional Food Research Center for Well-Being, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
3
|
Zhao Y, Qian Y, Sun Z, Shen X, Cai Y, Li L, Wang Z. Role of PI3K in the Progression and Regression of Atherosclerosis. Front Pharmacol 2021; 12:632378. [PMID: 33767629 PMCID: PMC7985550 DOI: 10.3389/fphar.2021.632378] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 01/29/2021] [Indexed: 12/11/2022] Open
Abstract
Phosphatidylinositol 3 kinase (PI3K) is a key molecule in the initiation of signal transduction pathways after the binding of extracellular signals to cell surface receptors. An intracellular kinase, PI3K activates multiple intracellular signaling pathways that affect cell growth, proliferation, migration, secretion, differentiation, transcription and translation. Dysregulation of PI3K activity, and as aberrant PI3K signaling, lead to a broad range of human diseases, such as cancer, immune disorders, diabetes, and cardiovascular diseases. A growing number of studies have shown that PI3K and its signaling pathways play key roles in the pathophysiological process of atherosclerosis. Furthermore, drugs targeting PI3K and its related signaling pathways are promising treatments for atherosclerosis. Therefore, we have reviewed how PI3K, an important regulatory factor, mediates the development of atherosclerosis and how targeting PI3K can be used to prevent and treat atherosclerosis.
Collapse
Affiliation(s)
- Yunyun Zhao
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yongjiang Qian
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xinyi Shen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yaoyao Cai
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
4
|
Zhang L, Wei C, Ruan Y, Zhang Y, Zhou Y, Lei D. Serum containing Buyang Huanwu decoction prevents age-associated migration and invasion of human vascular smooth muscle cells by up regulating SIRT1 expression. Biosci Trends 2018; 12:282-290. [PMID: 29952352 DOI: 10.5582/bst.2018.01063] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The migration and invasion of vascular smooth muscle cells (VSMCs) caused by advanced aging play an important role in diffuse intimal thickening, facilitate adverse arterial remodeling and contribute to the initiation and progression of cardiovascular diseases. The inhibitory function of Buyang Huanwu decoction (BYHWD) has been found on aortic intimal hyperplasia and VSMC proliferation, but its effect on age-associated migration and invasion remains unknown. Here, we used an in vitro angiotensin II (Ang II)-induced senescence model to study the effects of serum containing BYHWD (BYHWS) on the migratory and invasive capacities, matrix metalloprotease type 2 (MMP-2) expression and modulation of sirtuin1 (SIRT1) signaling in human aorta VSMCs (HA-VAMCs). Our results showed that BYHWS was able to inhibit Ang II-induced migration and invasion, with down-regulation of MMP-2. In addition, manipulation of SIRT1 by either over-expression or siRNA knockdown ameliorated or promoted cellular migration and invasion, respectively. Moreover, BYHWS reversed senescence-mediated decrease of SIRT1 levels and SIRT1 was required for BYHWS regulation on migration and invasion of senescent HA-VAMCs. In summary, our data demonstrated that BYHWS suppressed the migration and invasion of age-associated VSMC via an increase of the SIRT1 level, which provides novel insights for the therapy of age-associated cardiovascular diseases.
Collapse
MESH Headings
- Aging/drug effects
- Aging/physiology
- Angiotensin II/pharmacology
- Aorta/cytology
- Aorta/physiology
- Cardiovascular Diseases/drug therapy
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Cellular Senescence/drug effects
- Cellular Senescence/physiology
- Down-Regulation
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Gene Knockdown Techniques
- Humans
- Matrix Metalloproteinase 2/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/physiology
- RNA, Small Interfering/metabolism
- Signal Transduction/drug effects
- Sirtuin 1/genetics
- Sirtuin 1/metabolism
- Up-Regulation
Collapse
Affiliation(s)
- Li Zhang
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University
| | - Chunshan Wei
- Department of Liver Disease, Shenzhen Hospital Affiliated to Guangzhou University of Chinese Medicine
| | - Yunjun Ruan
- Department of Cardiology, Guangzhou General Hospital of Guangzhou Military Command
| | - Yanan Zhang
- Veterinary medicine, Northeast Agricultural University
| | - Yuliang Zhou
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University
| | - Da Lei
- Department of Cardiology, The First Affiliated Hospital of Guangdong Pharmaceutical University
| |
Collapse
|
5
|
Salvia fruticosa Induces Vasorelaxation In Rat Isolated Thoracic Aorta: Role of the PI3K/Akt/eNOS/NO/cGMP Signaling Pathway. Sci Rep 2017; 7:686. [PMID: 28386068 PMCID: PMC5429649 DOI: 10.1038/s41598-017-00790-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 03/13/2017] [Indexed: 12/27/2022] Open
Abstract
Salvia fruticosa (SF) Mill. is traditionally used for its antihypertensive actions. However, little is known about its pharmacologic and molecular mechanisms of action. Here we determined the effects of an ethanolic extract of SF leaves on rings of isolated thoracic aorta from Sprague-Dawley rats. Our results show that SF extract increased nitric oxide production and relaxed endothelium-intact rings in a dose-dependent (0.3 µg/ml–1 mg/ml) manner, and the maximum arterial relaxation (Rmax) was significantly reduced with endothelium denudation. Pretreatment of endothelium-intact rings with L-NAME (a non-selective inhibitor of nitric oxide synthase, 100 µM), or ODQ (an inhibitor of soluble guanylyl cyclase, 10 µM) significantly diminished SF-mediated vasorelaxation. Furthermore, SF induced Akt phosphorylation as well as increased cGMP levels in rings treated with increasing doses of SF. Prior exposure to PI3K inhibitors, wortmannin (0.1 µM) or LY294002 (10 µM), decreased cGMP accumulation and attenuated the SF-induced vasorelaxation by approximately 50% (Rmax). SF-evoked relaxation was not affected by indomethacin, verapamil, glibenclamide, tetraethylammonium, pyrilamine or atropine. Taken together, our results indicate that SF induces endothelium-dependent vasorelaxation through the PI3K/Akt/eNOS/NO/sGC/cGMP signaling pathway. Our data illustrate the health-orientated benefits of consuming SF which may act as an antihypertensive agent to reduce the burden of cardiovascular complications.
Collapse
|