1
|
Li S, He P, Liu J, Zang S, Luo J, Luo Y, Zhu S, Zang L. Ferulic acid protects against stress-induced myocardial injury in mice. Toxicol Appl Pharmacol 2025; 498:117309. [PMID: 40120650 DOI: 10.1016/j.taap.2025.117309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Excessive stress is a known contributor to cardiovascular diseases (CVD), and ferulic acid (FA), a natural phenolic compound, has demonstrated significant antioxidant and anti-inflammatory properties. This study investigates the protective effects of FA against stress-induced myocardial injury (SIMI) and elucidates the underlying mechanisms. An acute SIMI model was established in mice using low-temperature water immersion restraint. Cardiac function was assessed via cardiac index and histopathological analysis. Serum levels of corticosterone (CORT), lactate dehydrogenase (LDH), and brain natriuretic peptide (BNP) were quantified using enzyme-linked immunosorbent assay (ELISA), along with inflammatory markers TNF-α and IL-1β. The oxidative stress parameters, including malondialdehyde (MDA), glutathione (GSH), superoxide dismutase (SOD), and reactive oxygen species (ROS), were analyzed using colorimetric methods and fluorescent probes. Immunohistochemistry (IHC) and Western Blot were used to analyze the expression of proteins related to TNF, MAPK, PPAR-α/PGC-1α, and Nrf2 signaling pathways. Results indicated that FA pretreatment improved cardiac index, myocardial structural integrity, and reduced inflammatory cell infiltration. Serum levels of LDH, BNP, CORT, TNF-α, and IL-1β were significantly decreased in FA-treated SIMI mice. Elevated MDA and ROS levels, along with decreased GSH and SOD levels in the SIMI group, were reversed by FA pretreatment, likely through activation of the PPARα/PGC-1α and Nrf2 signaling pathways. Additionally, FA inhibited the TNF-α/TNFR1 and ERK/JNK MAPK pathways, contributing to its protective effects. In conclusion, FA mitigates SIMI by alleviating oxidative stress and inflammatory responses.
Collapse
Affiliation(s)
- Siyong Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Peiyi He
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jiahe Liu
- The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | | | - Jiahao Luo
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yi Luo
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Shuguang Zhu
- The First Affiliated Hospital cardiothoracic surgery department, Guangdong Pharmaceutical University, Guangzhou 510080, China.
| | - Linquan Zang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
2
|
Yang J, Liao D, He L, Fan W. Puerarin raises exosomal miR- 342 - 3p by inhibiting lncRNA NEAT1 in umbilical cord mesenchymal stem cells to alleviate renal tubular epithelial cell pyroptosis in chronic renal failure. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04164-1. [PMID: 40274620 DOI: 10.1007/s00210-025-04164-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 04/09/2025] [Indexed: 04/26/2025]
Abstract
The therapeutic effect of exosomes (Exo) secreted by the Puerarin-treated umbilical cord mesenchymal stem cells (UC-MSC) on chronic renal failure (CRF) was explored. UC-MSC were treated by Puerarin, or transfected by lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1) overexpression vectors, or both. HK-2 cells were treated with LPS/H2O2 to establish the CRF cell model. The CRF rat model was constructed by adenine administration. Exo derived from UC-MSC were used to treat the CRF cell and rat models. The therapeutic effect of the Puerarin-treated UC-MSC-derived Exo on CRF was evaluated by a series of logical tests. NEAT1 had binding sites for miR-342-3p. miR-342-3p in the Puerarin-treated UC-MSC-derived Exo was increased. miR-342-3p in the NEAT1-overexpressed and Puerarin-treated UC-MSC-derived Exo was lower than that in the Puerarin-treated UC-MSC-derived Exo. The CRF cell model treated by the Puerarin-treated UC-MSC-derived Exo expressed higher miR-342-3p, and lower TGF-β1, SMAD2/3, Cleaved-caspase-1, GSDMD-N, IL-1β and IL-18. The relieved renal injury, up-regulated renal miR-342-3p, down-regulated renal TGF-β1, SMAD2/3, Cleaved-caspase-1 and GSDMD-N, and decreased serum creatinine and blood urea nitrogen were found in the CRF rat model treated by the Puerarin-treated UC-MSC-derived Exo. However, compared to the CRF cell and rat models treated by the Puerarin-treated UC-MSC-derived Exo, those treated by the NEAT1-overexpressed and Puerarin-treated UC-MSC-derived Exo showed the opposite results. Puerarin elevates exosomal miR-342-3p by suppressing NEAT1 in UC-MSC, thereby inactivating the TGF-β1/SMAD pathway in renal tubular epithelial cells to alleviate pyroptosis in CRF. The Puerarin-treated UC-MSC-derived Exo may be useful in treating CRF.
Collapse
Affiliation(s)
- Jing Yang
- Department of Nephrology, the First Affiliated Department of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming City, 650032, Yunnan Province, China
| | - Dongxue Liao
- Department of Nephrology, the First Affiliated Department of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming City, 650032, Yunnan Province, China
| | - Lanxin He
- Department of Nephrology, the First Affiliated Department of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming City, 650032, Yunnan Province, China
| | - Wenxing Fan
- Department of Nephrology, the First Affiliated Department of Kunming Medical University, 295 Xichang Road, Wuhua District, Kunming City, 650032, Yunnan Province, China.
| |
Collapse
|
3
|
Patil P, Kumar P. Exploring kudzu: Extraction, quantification, and health impacts of bioactive compounds. Fitoterapia 2025; 182:106453. [PMID: 40020789 DOI: 10.1016/j.fitote.2025.106453] [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: 09/27/2024] [Revised: 02/13/2025] [Accepted: 02/21/2025] [Indexed: 03/03/2025]
Abstract
Kudzu (Pueraria species) is a perennial plant within the Fabaceae family, native to China, Japan, and India. It is known for its therapeutic properties, mainly due to its high content of isoflavones, including puerarin, daidzein, daidzin, genistein, and genistin. These isoflavones are found throughout the plant and are important in developing pharmaceutical drugs. This review comprehensively analyzes naturally occurring isoflavones in Kudzu, focusing on advanced and green techniques for their extraction, purification, and identification. Additionally, it highlights their health benefits and the growing demand in the global food and pharmaceutical industries. Due to their superior efficiency, scalability, and cost-effectiveness, contemporary eco-friendly extraction methods like ultrasound, microwave, enzyme-assisted, and supercritical fluid extraction are gaining prominence in this endeavor. They are crucial in optimizing the extraction process, driving innovation within industries, and harnessing natural sources, ultimately boosting global economies. Scientific studies confirm that Kudzu isoflavones have various anti-diabetic, neuroprotective, anti-cancer, antioxidant, alcohol detoxification, and cardiovascular protective effects. This review encourages further exploration of Kudzu isoflavones as a nutritional food source. It also highlights advancements in extraction methods within pharmaceuticals and natural products, underscoring the superiority of modern techniques over conventional ones. Additionally, critical analysis of the trends, limitations, and scope of Kudzu-extracted isoflavones for novel food applications can further advance scientific understanding.
Collapse
Affiliation(s)
- Poonam Patil
- Department of Food Engineering and Technology, Sant Longowal Institute of Engineering and Technology, Longowal 148106, Punjab, India.
| | - Pradyuman Kumar
- Department of Food Engineering and Technology, Sant Longowal Institute of Engineering and Technology, Longowal 148106, Punjab, India.
| |
Collapse
|
4
|
Qian Q, Wu J, Wang C, Yang Z, Cheng Y, Zheng Y, Wang X, Wang H. 6-PPD triggered lipid metabolism disorder and inflammatory response in larval zebrafish (Danio rerio) by regulating PPARγ/NF-κB pathway. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 368:125785. [PMID: 39900129 DOI: 10.1016/j.envpol.2025.125785] [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: 09/29/2024] [Revised: 01/26/2025] [Accepted: 02/01/2025] [Indexed: 02/05/2025]
Abstract
As a synthetic rubber antioxidant, the environmental monitoring concentrations of N-(1,3-Dimethylbutyl)-N'-phenyl-p-phenylenediamine (6-PPD) have exceeded the risk threshold, attracting widespread attention. Although investigations into the harmful effects on zebrafish have commenced, a comprehensive exploration of its toxicological impacts and underlying molecular mechanisms remains to be conducted. By using zebrafish as a model, this study systematically evaluated 6-PPD-induced lipid metabolism disorders and inflammation response following environmental exposure. Bioinformatics analysis revealed that 6-PPD target genes enriched in the hepatitis B pathway, indicating potential hepatic toxicity via inflammatory pathways. Therefore, we hypothesize that 6-PPD could trigger hepatotoxicity through the crosstalk between lipid metabolism and inflammation. Further experiments substantiated this hypothesis by showing lipid accumulation in the liver following 6-PPD exposure, along with elevated triglyceride (TG) and total cholesterol (TC) levels, and imbalanced expression of lipid metabolism-related marker genes. Additionally, 6-PPD exposure induced the accumulation of reactive oxygen species (ROS) and inhibited the differentiation and maturation of immune cells, resulting in immune evasion. Most of these abnormalities were exacerbated in a dose-dependent manner with increasing concentrations of 6-PPD. The addition of the PPARγ pathway agonist puerarin (PUE) or NF-κB pathway inhibitor quinazoline (QNZ) to 6-PPD exposure group mitigated these toxic effects, validating our conjecture that lipid metabolism disorder and inflammatory responses may result from the regulation of the PPARγ/NF-κB pathway.
Collapse
Affiliation(s)
- Qiuhui Qian
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215009, China; School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Ji Wu
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215009, China
| | - Cuizhen Wang
- Sanquan College of Xinxiang Medical University, Xinxiang, 453513, China
| | - Zheng Yang
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215009, China
| | - Ying Cheng
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215009, China
| | - Yuansi Zheng
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Xuedong Wang
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215009, China
| | - Huili Wang
- National and Local Joint Engineering Laboratory of Municipal Sewage Resource Utilization Technology, School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215009, China; School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
5
|
Li Y, He Q, Yu X, Xiong S, You H, Xuan Y, Liao W, Chen Z, Li H, Wang W, Chen Y, Wang X. Puerarin Reversing Autophagy-Lysosomal Dysfunction via Acid Sphingomyelinase Inhibition in Cardiomyocytes. J Cell Mol Med 2025; 29:e70427. [PMID: 39993967 PMCID: PMC11850092 DOI: 10.1111/jcmm.70427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 09/04/2024] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
Heart failure (HF) is a major cardiovascular disorder characterised by high prevalence and mortality rate. Recent studies have emphasised the role of autophagy in development and progression of HF. Dysfunctions in lysosomes and autophagic processes are closely associated with the aetiology of HF. Puerarin (PUE), a traditional Chinese medicine known for its antioxidant, anti-inflammatory and antiapoptotic properties, is widely used for the treatment of HF. However, the effectiveness of PUE in HF management via the modulation of autophagy requires further investigation. We used a mouse model of transverse aortic constriction to investigate protective effects of PUE on the autophagy-lysosomal pathway (ALP). We assessed heart function using echocardiography and performed histological staining for fibrosis and hypertrophy. RT-qPCR for atrial natriuretic peptide (ANP)/brain natriuretic peptide (BNP) and Western blotting for p62/LC3/LAMP1/Beclin1 were performed. Immunofluorescence was used to identify the autophagosomes, autolysosomes and lysosomes. In addition, immunohistochemistry was performed to detect acid sphingomyelinase (ASM) and ceramides. ASM siRNA was transfected into cardiomyocytes to evaluate autophagy. PUE treatment significantly reduced myocardial fibrosis and hypertrophy in HF-induced mice. PUE also effectively ameliorated ALP impairment in HF-induced mice and H9c2 cells. Mechanistically, PUE restored lysosomal homeostasis by inhibiting ASM expression and lysosomal transport, thereby enhancing lysosomal activity. These results underscore the therapeutic potential of PUE in correcting the ASM-mediated disruption of the HF-linked autophagy-lysosomal pathway.
Collapse
Affiliation(s)
- Yin‐ping Li
- Research Centre of Basic Integrative Medicine, School of Basic Medical SciencesGuangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
| | - Qian He
- Research Centre of Basic Integrative Medicine, School of Basic Medical SciencesGuangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
| | - Xiao‐ying Yu
- Department of Pharmacology, School of PharmaceuticalGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Sheng‐tao Xiong
- Research Centre of Basic Integrative Medicine, School of Basic Medical SciencesGuangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
| | - Hong‐jing You
- Department of Pharmacology, School of PharmaceuticalGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Yue Xuan
- Research Centre of Basic Integrative Medicine, School of Basic Medical SciencesGuangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
| | - Wei‐yan Liao
- Research Centre of Basic Integrative Medicine, School of Basic Medical SciencesGuangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
| | - Ze‐yu Chen
- Research Centre of Basic Integrative Medicine, School of Basic Medical SciencesGuangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
| | - Hai‐yan Li
- Department of Pharmacology, School of PharmaceuticalGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Wei Wang
- Department of Pharmacology, School of PharmaceuticalGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Yang Chen
- Department of Pharmacology, School of PharmaceuticalGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Xiao Wang
- Research Centre of Basic Integrative Medicine, School of Basic Medical SciencesGuangzhou University of Chinese MedicineGuangzhouGuangdongP.R. China
| |
Collapse
|
6
|
Jiang S, Qiu S, Mu Y, Liu C, Han Y, Jiang J, Wang Y. Puerarin reduces susceptibility to ventricular arrhythmias and inhibits ferroptosis via Sirt1/Nrf2 signaling in high-fat-diet rats. Free Radic Biol Med 2025; 227:472-484. [PMID: 39647799 DOI: 10.1016/j.freeradbiomed.2024.12.005] [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: 09/08/2024] [Revised: 11/18/2024] [Accepted: 12/02/2024] [Indexed: 12/10/2024]
Abstract
Obesity is a significant risk factor for cardiac arrhythmias, and the ferroptosis is closely related to cardiac arrhythmias. This study aimed to investigate whether puerarin (Pue), a natural isoflavone, could reduce the susceptibility to ventricular arrhythmias (VAs) associated with obesity and inhibit ferroptosis, with a particular focus on the Sirt1/Nrf2 signaling pathway. Male rats were randomly divided into three groups: normal chow diet (NC), high-fat diet (HFD), and HFD with Pue treatment (100mg/kg, HFD + Pue). After 16 weeks, electrophysiological, structural, and molecular analysis were performed. Compared to the NC group, HFD rats exhibited prolonged QT interval and Tpeak-Tend interval, amplified transmural dispersion of ventricular repolarization, and increased susceptibility to VAs. Pue treatment significantly ameliorated these electrophysiological abnormalities and reduced VAs susceptibility. HFD rats showed cardiac hypertrophy, fibrosis, and inflammation, which were alleviated by Pue application. Cardiac lipid peroxidation, iron deposition, mitochondrial abnormality, and ferroptosis marker induction were observed in HFD rats. Further, treatment with Pue improved these alterations. Additionally, molecular docking analysis confirmed the interaction of Pue with Sirt1 and Nrf2. Furthermore, Pue treatment upregulated Sirt1 and Nrf2 expression in HFD rats, thereby reducing reactive oxygen species (ROS) generation and ferroptosis. Moreover, Pue protected cardiomyocytes against palmitic acid (PA)-induced injury by inhibiting ferroptosis via the Sirt1/Nrf2 pathway in H9c2 cells. Overall, our study shows for the first time that Pue reduces susceptibility to VAs and inhibits ferroptosis in HFD rats by modulating the Sirt1/Nrf2 signaling pathway, offering a potential therapeutic strategy for obesity-related cardiac arrhythmias.
Collapse
MESH Headings
- Animals
- Sirtuin 1/metabolism
- Sirtuin 1/genetics
- NF-E2-Related Factor 2/metabolism
- NF-E2-Related Factor 2/genetics
- Rats
- Male
- Diet, High-Fat/adverse effects
- Ferroptosis/drug effects
- Isoflavones/pharmacology
- Signal Transduction/drug effects
- Arrhythmias, Cardiac/drug therapy
- Arrhythmias, Cardiac/metabolism
- Arrhythmias, Cardiac/etiology
- Arrhythmias, Cardiac/prevention & control
- Arrhythmias, Cardiac/pathology
- Rats, Sprague-Dawley
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Obesity/drug therapy
- Obesity/metabolism
Collapse
Affiliation(s)
- Shuang Jiang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050, Beijing, China
| | - Suhua Qiu
- Department of Pharmacology, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Yu Mu
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050, Beijing, China
| | - Chilu Liu
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050, Beijing, China
| | - Yanxing Han
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050, Beijing, China
| | - Jiandong Jiang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050, Beijing, China.
| | - Yuhong Wang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050, Beijing, China.
| |
Collapse
|
7
|
Wang J, Zheng Q, Chen Z, Liu X, Wan S, Wang L. Puerarin alleviates renal ischemia/reperfusion injury by inhibiting apoptosis and endoplasmic reticulum stress via Nrf2/HO-1 pathway. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2025; 28:187-193. [PMID: 39850122 PMCID: PMC11756738 DOI: 10.22038/ijbms.2024.80438.17412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 09/07/2024] [Indexed: 01/25/2025]
Abstract
Objectives To explore the effects of puerarin on renal ischemia/reperfusion injury and the possible mechanism. Materials and Methods The experimental mice were injected with puerarin (50 or 100 mg/kg) per day or equal sterile saline by intraperitoneal injection for one week, and a renal I/R injury model was constructed. HK-2 cells were incubated with puerarin (1 uM and 10 uM) before the H/R model. Immunohistochemistry, immunocytochemistry, and Western blot analysis were used to detect the protein associated with apoptosis and endoplasmic reticulum stress. Results Puerarin could improve renal function and attenuate tissue structural damage after renal I/R. Meanwhile, puerarin alleviated apoptosis and endoplasmic reticulum stress by decreasing expression levels of specific biomarkers such as caspase-3, GRP78, CHOP, and p-elF2α/ elF2α in animals and HK-2 cells. The up-regulated expression of Nrf2 and HO-1 protein after puerarin treatment indicated that the Nrf2/HO-1 signaling pathway might mediate the protective mechanism of puerarin against renal I/R. Conclusion Our results suggest that puerarin alleviated renal ischemia/reperfusion injury by inhibiting apoptosis and endoplasmic reticulum stress via the Nrf2/HO-1 pathway and offered new insights for preventing and treating renal I/R.
Collapse
Affiliation(s)
- Jingsong Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
- These authors contributed equally to this work
| | - Qingyuan Zheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
- These authors contributed equally to this work
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Shanshan Wan
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| |
Collapse
|
8
|
Wei J, Lv Q, Luan F, Zhang X, Guo D, Zhai B, Chen S, Zou J, Shi Y. Exploration of potential mechanism of Sanhua Jiangzhi granules for the treatment of hyperlipidemia based on network pharmacology and experimental verification. Fitoterapia 2024; 179:106271. [PMID: 39461567 DOI: 10.1016/j.fitote.2024.106271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 10/17/2024] [Accepted: 10/20/2024] [Indexed: 10/29/2024]
Abstract
Sanhua Jiangzhi Granules (SJG) is a traditional Chinese patent medicine known for regulating lipid metabolism. In this study, we utilized UPLC-TOF-MS to analyze the components of SJG and, in conjunction with network pharmacology, identified 125 core chemical constituents. These components were individually queried and intersected with targets related to hyperlipidemia, resulting in the identification of 312 core targets. KEGG and GO analyses suggested that the mechanism of SJG in treating hyperlipidemia may primarily involve the PPAR signaling pathway. To further validate the efficacy and underlying signaling mechanisms of SJG, we conducted experiments using 60 rats. The results indicated that SJG significantly reduced body weight, lowered serum levels of total cholesterol (TC), triglycerides (TG), and low-density lipoprotein cholesterol (LDL-C), while increasing high-density lipoprotein cholesterol (HDLC) levels. Enzyme-linked immunosorbent assay (ELISA) results demonstrated that SJG decreased hepatic TC and TG levels and mitigated lipid accumulation in the liver. Hematoxylin and eosin (HE) staining indicated that SJG improved liver pathological morphology and reduced the risk of fatty liver disease. Western blot analyses showed that treatment with SJG down-regulated the expression of stearoyl-CoA desaturase (SCD), 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), phospholipid transfer protein (PLTP), and fatty acid-binding protein 1 (FABP1), while up-regulating the expression of cholesterol 7α-hydroxylase (CYP7A1), carnitine palmitoyltransferase 1 (CPT-1), and PPARα by activating the PPAR signaling pathway. In conclusion, this study demonstrated that SJG activates the PPAR signaling pathway, leading to decreased body weight, lowered blood lipid levels, reduced hepatic TC and TG, and improved liver pathology in rats.
Collapse
Affiliation(s)
- Junfei Wei
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Qian Lv
- Weinan Testing Institute, 714000, China
| | - Fei Luan
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Xiaofei Zhang
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Dongyan Guo
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Bingtao Zhai
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Shucun Chen
- Shangluo Hospital of Traditional Chinese Medicine, Shangluo 726000, China
| | - Junbo Zou
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang 712046, China.
| | - Yajun Shi
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Pharmacy College, Shaanxi University of Chinese Medicine, Xianyang 712046, China.
| |
Collapse
|
9
|
Rahmani S, Roohbakhsh A, Pourbarkhordar V, Karimi G. The Cardiovascular Protective Function of Natural Compounds Through AMPK/SIRT1/PGC-1α Signaling Pathway. Food Sci Nutr 2024; 12:9998-10009. [PMID: 39723061 PMCID: PMC11666815 DOI: 10.1002/fsn3.4553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/02/2024] [Accepted: 10/06/2024] [Indexed: 12/28/2024] Open
Abstract
Cardiovascular disease (CVD) poses a major risk to human health and exert a heavy burden on individuals, society, and healthcare systems. Therefore, it is critical to identify CVD's underlying mechanism(s) and target them using effective agents. Natural compounds have shown promise as antioxidants with cardioprotective functions against CVD injuries due to their antioxidative solid capacity and high safety profile. Several CVDs, such as heart failure, ischemia/reperfusion, atherosclerosis, and cardiomyopathies, are closely linked to mitochondrial dysfunction. It is well established that activating the AMPK/SIRT1/PGC-1α pathway during CVD promotes mitochondrial function. Therefore, targeting the AMPK/SIRT1/PGC-1α pathway provides a foundation for novel therapeutic strategies to combat CVD. A key goal of our search was to find natural compounds that target this biological pathway and have beneficial effects on CVD.
Collapse
Affiliation(s)
- Sohrab Rahmani
- Student Research CommitteeMashhad University of Medical SciencesMashhadIran
- Department of Pharmacodynamics and Toxicology, School of PharmacyMashhad University of Medical SciencesMashhadIran
| | - Ali Roohbakhsh
- Department of Pharmacodynamics and Toxicology, School of PharmacyMashhad University of Medical SciencesMashhadIran
- Pharmaceutical Research Center, Institute of Pharmaceutical TechnologyMashhad University of Medical SciencesMashhadIran
| | - Vahid Pourbarkhordar
- Student Research CommitteeMashhad University of Medical SciencesMashhadIran
- Department of Pharmacodynamics and Toxicology, School of PharmacyMashhad University of Medical SciencesMashhadIran
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of PharmacyMashhad University of Medical SciencesMashhadIran
- Pharmaceutical Research Center, Institute of Pharmaceutical TechnologyMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
10
|
Jin Z, Lan Y, Li J, Wang P, Xiong X. The role of Chinese herbal medicine in the regulation of oxidative stress in treating hypertension: from therapeutics to mechanisms. Chin Med 2024; 19:150. [PMID: 39468572 PMCID: PMC11520704 DOI: 10.1186/s13020-024-01022-9] [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: 05/07/2024] [Accepted: 10/11/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Although the pathogenesis of essential hypertension is not clear, a large number of studies have shown that oxidative stress plays an important role in the occurrence and development of hypertension and target organ damage. PURPOSE This paper systematically summarizes the relationship between oxidative stress and hypertension, and explores the potential mechanisms of Chinese herbal medicine (CHM) in the regulation of oxidative stress in hypertension, aiming to establish a scientific basis for the treatment of hypertension with CHM. METHODS To review the efficacy and mechanism by which CHM treat hypertension through targeting oxidative stress, data were searched from PubMed, EMBASE, the Cochrane Central Register of Controlled Trials, the Chinese National Knowledge Infrastructure, the VIP Information Database, the Chinese Biomedical Literature Database, and the Wanfang Database from their inception up to January 2024. NPs were classified and summarized by their mechanisms of action. RESULTS In hypertension, the oxidative stress pathway of the body is abnormally activated, and the antioxidant system is inhibited, leading to the imbalance between the oxidative and antioxidative capacity. Meanwhile, excessive production of reactive oxygen species can lead to endothelial damage and vascular dysfunction, resulting in inflammation and immune response, thereby promoting the development of hypertension and damaging the heart, brain, kidneys, blood vessels, and other target organs. Numerous studies suggested that inhibiting oxidative stress may be the potential therapeutic target for hypertension. In recent years, the clinical advantages of traditional Chinese medicine (TCM) in the treatment of hypertension have gradually attracted attention. TCM, including active ingredients of CHM, single Chinese herb, TCM classic formula and traditional Chinese patent medicine, can not only reduce blood pressure, improve clinical symptoms, but also improve oxidative stress, thus extensively affect vascular endothelium, renin-angiotensin-aldosterone system, sympathetic nervous system, target organ damage, as well as insulin resistance, hyperlipidemia, hyperhomocysteinemia and other pathological mechanisms and hypertension related risk factors. CONCLUSIONS CHM display a beneficial multi-target, multi-component, overall and comprehensive regulation characteristics, and have potential value for clinical application in the treatment of hypertension by regulating the level of oxidative stress.
Collapse
Affiliation(s)
- Zixuan Jin
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5 Beixian Ge, Xicheng District, Beijing, 100053, China
| | - Yu Lan
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5 Beixian Ge, Xicheng District, Beijing, 100053, China
| | - Junying Li
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5 Beixian Ge, Xicheng District, Beijing, 100053, China
| | - Pengqian Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Xingjiang Xiong
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5 Beixian Ge, Xicheng District, Beijing, 100053, China.
| |
Collapse
|
11
|
Wan Q, Lu Q, Luo S, Guan C, Zhang H. The beneficial health effects of puerarin in the treatment of cardiovascular diseases: from mechanisms to therapeutics. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7273-7296. [PMID: 38709267 DOI: 10.1007/s00210-024-03142-3] [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: 03/29/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Cardiovascular diseases (CVDs) are the leading causes of death globally that seriously threaten human health. Although novel western medicines have continued to be discovered over the past few decades to inhibit the progression of CVDs, new drug research and development for treating CVDs with less side effects and adverse reactions are continuously being desired. Puerarin is a natural product found in a variety of medicinal plants belonging to the flavonoid family with potent biological and pharmacological activities. Abundant research findings in the literature have suggested that puerarin possesses a promising prospect in treating CVDs. In recent years, numerous new molecular mechanisms of puerarin have been explored in experimental and clinical studies, providing new evidence for this plant metabolite to protect against CVDs. This article systematically introduces the history of use, bioavailability, and various dosage forms of puerarin and further summarizes recently published data on the major research advances and their underlying therapeutic mechanisms in treating CVDs. It may provide references for researchers in the fields of pharmacology, natural products, and internal medicine.
Collapse
Affiliation(s)
- Qiang Wan
- Affiliated Hospital of Jiangxi University of Chinese Medicine, 445 Bayi Avenue, Nanchang, 330006, China.
- Clinical Medical College, Jiangxi University of Chinese Medicine, 445 Bayi Avenue, Nanchang, 330006, China.
| | - Qiwen Lu
- Graduate School, Jiangxi University of Chinese Medicine, 1688 Meiling Avenue, Nanchang, 330004, China
| | - Sang Luo
- Graduate School, Jiangxi University of Chinese Medicine, 1688 Meiling Avenue, Nanchang, 330004, China
| | - Chengyan Guan
- Graduate School, Jiangxi University of Chinese Medicine, 1688 Meiling Avenue, Nanchang, 330004, China
| | - Hao Zhang
- Graduate School, Jiangxi University of Chinese Medicine, 1688 Meiling Avenue, Nanchang, 330004, China
| |
Collapse
|
12
|
Xue J, Ren H, Zhang Q, Gu J, Xu Q, Sun J, Zhang L, Zhou MS. Puerarin attenuates myocardial ischemic injury and endoplasmic reticulum stress by upregulating the Mzb1 signal pathway. Front Pharmacol 2024; 15:1442831. [PMID: 39206261 PMCID: PMC11350615 DOI: 10.3389/fphar.2024.1442831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Objective This study investigated the role of Mzb1 in puerarin protection against heart injury and dysfunction in acute myocardial infarction (AMI) mice. Methods C57BL/6 mice were pretreated with and without puerarin at doses of 50 mg/kg and 100 mg/kg for 14 days before establishing the AMI model. An AMI model was induced by ligating the left descending anterior coronary artery, and AC16 cardiomyocytes were treated with H2O2 in vitro. Echocardiography was performed to measure cardiac function. DHE staining, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase assay, and DCFH-DA oxidative fluorescence staining were used to determine reactive oxygen species (ROS) production in vivo and in vitro. Bioinformatics analysis was used to predict potential upstream transcription factors of Mzb1. Results Puerarin dose-dependently reduced myocardial infarction area and injury, accompanied by the improvement of cardiac function in AMI mice. AMI mice manifested an increase in myocardial oxidative stress, endoplasmic reticulum (ER) stress, apoptosis, and mitochondrial biogenesis dysfunction, which were inhibited by pretreatment with puerarin. Puerarin also prevented Mzb1 downregulation in the hearts of AMI mice or H2O2-treated AC16 cells. Consistent with the in vivo findings, puerarin inhibited H2O2-induced cardiomyocyte apoptosis, ER stress, and mitochondrial dysfunction, which were attenuated by siRNA Mzb1. Furthermore, the JASPAR website predicted that KLF4 may be a transcription factor for Mzb1. The expression of KLF4 was partially reversed by puerarin in the cardiomyocyte injury model, and KLF4 inhibitor (kenpaullone) inhibited Mzb1 expression and affected its function. Conclusion These results suggest that puerarin can protect against cardiac injury by attenuating oxidative stress and endoplasmic reticulum stress through upregulating the KLF4/Mzb1 pathway and that puerarin may expand our armamentarium for the prevention and treatment of ischemic heart diseases.
Collapse
Affiliation(s)
- Jiaojiao Xue
- Science and Experiment Research Center, Shenyang Medical College, Shenyang, China
- School of Basic Medicine, Shenyang Medical College, Shenyang, China
- Department of Pathology, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Haolin Ren
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qi Zhang
- Science and Experiment Research Center, Shenyang Medical College, Shenyang, China
- School of Basic Medicine, Shenyang Medical College, Shenyang, China
| | - Jing Gu
- Science and Experiment Research Center, Shenyang Medical College, Shenyang, China
- School of Basic Medicine, Shenyang Medical College, Shenyang, China
| | - Qian Xu
- Science and Experiment Research Center, Shenyang Medical College, Shenyang, China
| | - Jiaxi Sun
- Science and Experiment Research Center, Shenyang Medical College, Shenyang, China
- School of Basic Medicine, Shenyang Medical College, Shenyang, China
| | - Lu Zhang
- Science and Experiment Research Center, Shenyang Medical College, Shenyang, China
| | - Ming-Sheng Zhou
- School of Basic Medicine, Shenyang Medical College, Shenyang, China
| |
Collapse
|
13
|
Ming Z, Ruishi X, Linyi X, Yonggang Y, Haoming L, Xintian L. The gut-liver axis in fatty liver disease: role played by natural products. Front Pharmacol 2024; 15:1365294. [PMID: 38686320 PMCID: PMC11056694 DOI: 10.3389/fphar.2024.1365294] [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: 01/04/2024] [Accepted: 02/01/2024] [Indexed: 05/02/2024] Open
Abstract
Fatty liver disease, a condition characterized by fatty degeneration of the liver, mainly classified as non-alcoholic fatty liver disease (NAFLD) and alcoholic liver disease (ALD), has become a leading cause of cirrhosis, liver cancer and death. The gut-liver axis is the bidirectional relationship between the gut and its microbiota and its liver. The liver can communicate with the gut through the bile ducts, while the portal vein transports the products of the gut flora to the liver. The intestinal flora and its metabolites directly and indirectly regulate hepatic gene expression, leading to an imbalance in the gut-liver axis and thus contributing to the development of liver disease. Utilizing natural products for the prevention and treatment of various metabolic diseases is a prevalent practice, and it is anticipated to represent the forthcoming trend in the development of drugs for combating NAFLD/ALD. This paper discusses the mechanism of the enterohepatic axis in fatty liver, summarizes the important role of plant metabolites in natural products in fatty liver treatment by regulating the enterohepatic axis, and provides a theoretical basis for the subsequent development of new drugs and clinical research.
Collapse
Affiliation(s)
- Zhu Ming
- Changchun University of Chinese Medicine, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Xie Ruishi
- Changchun University of Chinese Medicine, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Xu Linyi
- Changchun University of Chinese Medicine, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | | | - Luo Haoming
- Changchun University of Chinese Medicine, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Lan Xintian
- Changchun University of Chinese Medicine, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
14
|
Jiang L, Liu J, Yang Z, Wang J, Ke W, Zhang K, Zhang C, Zuo H. Downregulation of the CD151 protects the cardiac function by the crosstalk between the endothelial cells and cardiomyocytes via exosomes. PLoS One 2024; 19:e0297121. [PMID: 38349935 PMCID: PMC10863850 DOI: 10.1371/journal.pone.0297121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 12/27/2023] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Heart failure (HF) is the last stage in the progression of various cardiovascular diseases. Although it is documented that CD151 contributes to regulate the myocardial infarction, the function of CD151 on HF and involved mechanisms are still unclear. METHOD AND RESULTS In the present study, we found that the recombinant adeno-associated virus (rAAV)-mediated endothelial cell-specific knockdown of CD151-transfected mice improved transverse aortic constriction (TAC)-induced cardiac function, attenuated myocardial hypertrophy and fibrosis, and increased coronary perfusion, whereas overexpression of the CD151 protein aggravated cardiac dysfunction and showed the opposite effects. In vitro, the cardiomyocytes hypertrophy induced by PE were significantly improved, while the proliferation and migration of cardiac fibroblasts (CFs) were significantly reduced, when co-cultured with the CD151-silenced endothelial cells (ECs). To further explore the mechanisms, the exosomes from the CD151-silenced ECs were taken by cardiomyocyte (CMs) and CFs, verified the intercellular communication. And the protective effects of CD151-silenced ECs were inhibited when exosome inhibitor (GW4869) was added. Additionally, a quantitative proteomics method was used to identify potential proteins in CD151-silenced EC exosomes. We found that the suppression of CD151 could regulate the PPAR signaling pathway via exosomes. CONCLUSION Our observations suggest that the downregulation of CD151 is an important positive regulator of cardiac function of heart failure, which can regulate exosome-stored proteins to play a role in the cellular interaction on the CMs and CFs. Modulating the exosome levels of ECs by reducing CD151 expression may offer novel therapeutic strategies and targets for HF treatment.
Collapse
Affiliation(s)
- Luying Jiang
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The 3rd Department of Cardiology, The First Affiliated Hospital of the Medical College, Shihezi University, Shihezi, China
| | - Jingbo Liu
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Children Health Care, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Zhenjia Yang
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The 3rd Department of Cardiology, The First Affiliated Hospital of the Medical College, Shihezi University, Shihezi, China
| | - Jianyu Wang
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Wenkai Ke
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Kaiyue Zhang
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunran Zhang
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The 3rd Department of Cardiology, The First Affiliated Hospital of the Medical College, Shihezi University, Shihezi, China
| | - Houjuan Zuo
- Department of Internal Medicine, Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
Du L, Lu H, Wang Z, Liu C, Xiao Y, Guo Z, Li Y. Therapeutic Potential of Ginsenoside Rb1-PLGA Nanoparticles for Heart Failure Treatment via the ROS/PPARα/PGC1α Pathway. Molecules 2023; 28:8118. [PMID: 38138606 PMCID: PMC10745441 DOI: 10.3390/molecules28248118] [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: 10/18/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
(1) Background: Ginsenoside Rb1-PLGA nanoparticles (GRb1@PLGA@NPs) represent a novel nanotherapeutic system, yet their therapeutic efficacy and underlying mechanisms for treating heart failure (HF) remain unexplored. This study aims to investigate the potential mechanisms underlying the therapeutic effects of GRb1@PLGA@NPs in HF treatment; (2) Methods: The left anterior descending coronary artery ligation was employed to establish a HF model in Sprague-Dawley rats, along with an in vitro oxidative stress model using H9c2 myocardial cells. Following treatment with GRb1@PLGA@NPs, cardiac tissue pathological changes and cell proliferation were observed. Additionally, the serum levels of biomarkers such as NT-proBNP, TNF-α, and IL-1β were measured, along with the expression of the ROS/PPARα/PGC1α pathway; (3) Results: GRb1@PLGA@NPs effectively ameliorated the pathological status of cardiac tissues in HF rats, mitigated oxidative stress-induced myocardial cell damage, elevated SOD and MMP levels, and reduced LDH, MDA, ROS, NT-proBNP, TNF-α, and IL-1β levels. Furthermore, the expression of PPARα and PGC1α proteins was upregulated; (4) Conclusions: GRb1@PLGA@NPs may attenuate myocardial cell injury and treat HF through the ROS/PPARα/PGC1α pathway.
Collapse
Affiliation(s)
- Lixin Du
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (L.D.); (H.L.); (Y.X.)
| | - Huiling Lu
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (L.D.); (H.L.); (Y.X.)
| | - Ziyan Wang
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (Z.W.); (C.L.); (Z.G.)
| | - Chengxin Liu
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (Z.W.); (C.L.); (Z.G.)
| | - Yifei Xiao
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (L.D.); (H.L.); (Y.X.)
| | - Zhihua Guo
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (Z.W.); (C.L.); (Z.G.)
| | - Ya Li
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (L.D.); (H.L.); (Y.X.)
| |
Collapse
|
16
|
Tang H, Zhang S, Lu X, Geng T. Effects of puerarin on the intervertebral disc degeneration and biological characteristics of nucleus pulposus cells. PHARMACEUTICAL BIOLOGY 2023; 61:12-22. [PMID: 36524765 PMCID: PMC9762855 DOI: 10.1080/13880209.2022.2147548] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 09/21/2022] [Accepted: 11/05/2022] [Indexed: 06/17/2023]
Abstract
CONTEXT Intervertebral disc degeneration (IDD) is the pathological basis of spinal degenerative diseases. Puerarin (PU) is an isoflavonoid with functions and medicinal properties. OBJECTIVE To explore the effect of PU on IDD and its potential mechanism of action. MATERIALS AND METHODS Sprague-Dawley (SD) rats were divided into sham, IDD, low PU, and high PU groups. Rat nucleus pulposus cells (NPCs) were isolated and divided into control, IL-1β, 100 and 200 μmol/mL PU, TAK-242 (TLR4 inhibitor), or 200 μmol/mL PU + LPS (TLR4 activator) groups. The water content, inflammatory factors, proliferation activity, TLR4/NF-κB pathway activity, apoptosis rate, protein expression of apoptosis, and histology of the extracellular matrix (ECM) were analysed. RESULTS In vivo: Compared with the IDD group, disorganization of intervertebral disc tissue was significantly improved, water content (2.80 ± 0.24 mg, 3.91 ± 0.31 mg vs. 2.02 ± 0.21 mg) and expression levels of collagen II and aggrecan were significantly increased, and the levels of inflammatory factors and the expression levels of TLR4, MyD88, and p-p65 were significantly decreased in IDD rats treated with PU. In vitro: Compared with the IL-1β group, the proliferation activity of IL-1β-treated NPCs and the expression of collagen II and aggrecan were significantly increased, while the apoptosis rate, levels of inflammatory factors, and the expression levels of TLR4, MyD88, and p-p65 were significantly decreased in IL-1β-treated NPCs treated with PU. LPS reversed the biological function changes of IL-1β-treated NPCs induced by PU. CONCLUSIONS PU can delay the progression of IDD by inhibiting activation of the TLR4/NF-κB pathway.
Collapse
Affiliation(s)
- Hengtao Tang
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Song Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinchang Lu
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tongyu Geng
- Department of Orthopaedics, Shangqiu First People’s Hospital, Shangqiu, China
| |
Collapse
|
17
|
Pu Y, Cheng CK, Zhang H, Luo JY, Wang L, Tomlinson B, Huang Y. Molecular mechanisms and therapeutic perspectives of peroxisome proliferator-activated receptor α agonists in cardiovascular health and disease. Med Res Rev 2023; 43:2086-2114. [PMID: 37119045 DOI: 10.1002/med.21970] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 03/10/2023] [Accepted: 04/12/2023] [Indexed: 04/30/2023]
Abstract
The prevalence of cardiovascular disease (CVD) has been rising due to sedentary lifestyles and unhealthy dietary patterns. Peroxisome proliferator-activated receptor α (PPARα) is a nuclear receptor regulating multiple biological processes, such as lipid metabolism and inflammatory response critical to cardiovascular homeostasis. Healthy endothelial cells (ECs) lining the lumen of blood vessels maintains vascular homeostasis, where endothelial dysfunction associated with increased oxidative stress and inflammation triggers the pathogenesis of CVD. PPARα activation decreases endothelial inflammation and senescence, contributing to improved vascular function and reduced risk of atherosclerosis. Phenotypic switch and inflammation of vascular smooth muscle cells (VSMCs) exacerbate vascular dysfunction and atherogenesis, in which PPARα activation improves VSMC homeostasis. Different immune cells participate in the progression of vascular inflammation and atherosclerosis. PPARα in immune cells plays a critical role in immunological events, such as monocyte/macrophage adhesion and infiltration, macrophage polarization, dendritic cell (DC) embedment, T cell activation, and B cell differentiation. Cardiomyocyte dysfunction, a major risk factor for heart failure, can also be alleviated by PPARα activation through maintaining cardiac mitochondrial stability and inhibiting cardiac lipid accumulation, oxidative stress, inflammation, and fibrosis. This review discusses the current understanding and future perspectives on the role of PPARα in the regulation of the cardiovascular system as well as the clinical application of PPARα ligands.
Collapse
Affiliation(s)
- Yujie Pu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Chak Kwong Cheng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Hongsong Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jiang-Yun Luo
- Institute for Cardiovascular Development and Regenerative Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Brian Tomlinson
- Faculty of Medicine, Macau University of Science & Technology, Macau, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
18
|
da Silva FC, Brandão DC, Ferreira EA, Siqueira RP, Ferreira HSV, Da Silva Filho AA, Araújo TG. Tailoring Potential Natural Compounds for the Treatment of Luminal Breast Cancer. Pharmaceuticals (Basel) 2023; 16:1466. [PMID: 37895937 PMCID: PMC10610388 DOI: 10.3390/ph16101466] [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: 08/29/2023] [Revised: 09/24/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Breast cancer (BC) is the most diagnosed cancer worldwide, mainly affecting the epithelial cells from the mammary glands. When it expresses the estrogen receptor (ER), the tumor is called luminal BC, which is eligible for endocrine therapy with hormone signaling blockade. Hormone therapy is essential for the survival of patients, but therapeutic resistance has been shown to be worrying, significantly compromising the prognosis. In this context, the need to explore new compounds emerges, especially compounds of plant origin, since they are biologically active and particularly promising. Natural products are being continuously screened for treating cancer due to their chemical diversity, reduced toxicity, lower side effects, and low price. This review summarizes natural compounds for the treatment of luminal BC, emphasizing the activities of these compounds in ER-positive cells. Moreover, their potential as an alternative to endocrine resistance is explored, opening new opportunities for the design of optimized therapies.
Collapse
Affiliation(s)
- Fernanda Cardoso da Silva
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Universidade Federal de Uberlândia, Patos de Minas 38700-002, MG, Brazil; (F.C.d.S.); (D.C.B.); (R.P.S.); (H.S.V.F.)
| | - Douglas Cardoso Brandão
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Universidade Federal de Uberlândia, Patos de Minas 38700-002, MG, Brazil; (F.C.d.S.); (D.C.B.); (R.P.S.); (H.S.V.F.)
| | - Everton Allan Ferreira
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Juiz de Fora, Juiz de Fora 36036-900, MG, Brazil; (E.A.F.); (A.A.D.S.F.)
| | - Raoni Pais Siqueira
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Universidade Federal de Uberlândia, Patos de Minas 38700-002, MG, Brazil; (F.C.d.S.); (D.C.B.); (R.P.S.); (H.S.V.F.)
| | - Helen Soares Valença Ferreira
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Universidade Federal de Uberlândia, Patos de Minas 38700-002, MG, Brazil; (F.C.d.S.); (D.C.B.); (R.P.S.); (H.S.V.F.)
| | - Ademar Alves Da Silva Filho
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Juiz de Fora, Juiz de Fora 36036-900, MG, Brazil; (E.A.F.); (A.A.D.S.F.)
| | - Thaise Gonçalves Araújo
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Universidade Federal de Uberlândia, Patos de Minas 38700-002, MG, Brazil; (F.C.d.S.); (D.C.B.); (R.P.S.); (H.S.V.F.)
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia 38405-302, MG, Brazil
| |
Collapse
|
19
|
Zhao W, Shan X, Li X, Lu S, Xia L, Chen H, Zhang C, Guo W, Xu M, Lu R, Zhao P. Icariin inhibits hypertrophy by regulation of GPER1 and CaMKII/HDAC4/MEF2C signaling crosstalk in ovariectomized mice. Chem Biol Interact 2023; 384:110728. [PMID: 37739049 DOI: 10.1016/j.cbi.2023.110728] [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: 11/19/2022] [Revised: 08/16/2023] [Accepted: 09/20/2023] [Indexed: 09/24/2023]
Abstract
Icariin (ICA), a flavonoid phytoestrogen, was isolated from traditional Chinese medicine Yin Yang Huo (Epimedium brevicornu Maxim.). Previous studies reporting the cardioprotective effects of ICA are available; however, little is known about the impact of ICA on cardioprotection under conditions of reduced estrogen levels. This study aimed to provide detailed information regarding the antihypertrophic effects of ICA in ovariectomized female mice. Female mice were subjected to ovariectomy (OVX) and transverse aortic constriction and then orally treated with ICA at doses of 30, 60 or 120 mg/kg/day for 4 weeks. Morphological assessments, echocardiographic parameters, histological analyses, and immunofluorescence were performed to evaluate cardiac hypertrophy. Cardiomyocytes from mice or rats were stimulated using phenylephrine, and cell surface and hypertrophy markers were tested using immunofluorescence and qPCR. Western blotting, qPCR, and luciferase reporter gene assays were used to assess the expression of proteins and mRNA and further investigate the proteins related to the G-protein coupled estrogen receptor (GPER1) and CaMKII/HDAC4/MEF2C signaling pathways in vivo and in vitro. ICA blocks cardiac hypertrophy induced by pressure overload in OVX mice. Additionally, we demonstrated that ICA activated GPER1 and inhibited the nuclear export or promoted the nuclear import of histone deacetylase 4 (HDAC4) through regulation of phosphorylation of calmodulin-dependent protein kinase II (CaMKII) and further improved the repression of myocyte enhancer factor-2C (MEF2C). ICA ameliorated cardiac hypertrophy in OVX mice by activating GPER1 and inhibiting the CaMKII/HDAC4/MEF2 signaling pathway.
Collapse
Affiliation(s)
- Wenxia Zhao
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China; School of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaoli Shan
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xueqin Li
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuang Lu
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Xia
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huihua Chen
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chen Zhang
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Guo
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ming Xu
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rong Lu
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Pei Zhao
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
20
|
Pan Y, Lin T, Shao L, Zhang Y, Han Q, Sheng L, Guo R, Sun T, Zhang Y. Lignin/Puerarin Nanoparticle-Incorporated Hydrogel Improves Angiogenesis through Puerarin-Induced Autophagy Activation. Int J Nanomedicine 2023; 18:5095-5117. [PMID: 37705868 PMCID: PMC10496927 DOI: 10.2147/ijn.s412835] [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: 04/13/2023] [Accepted: 08/25/2023] [Indexed: 09/15/2023] Open
Abstract
Purpose Puerarin is the main isoflavone extracted from Radix Puerariae lobata (Willd.) and exerts a strong protective effect on endothelial cells. This isoflavone also exerts proven angiogenic effects; however, the potential underlying mechanism has not been fully explored. Here in this work, we aimed to determine the proangiogenesis effect of a puerarin-attached lignin nanoparticle-incorporated hydrogel and explore the underlying mechanism. Materials and Methods Puerarin-attached lignin nanoparticles were fabricated and mixed with the GelMA hydrogel. After the hydrogel was characterized, the angiogenic effect was evaluated in a mouse hind-limb ischemia model. To further explore the mechanism of angiogenesis, human endothelial cell line EA.hy926 was exposure to different concentrations of puerarin. Wound healing assays and tube formation assays were used to investigate the effects of puerarin on cell migration and angiogenesis. qPCR and Western blotting were performed to determine the changes in the levels of angiogenesis indicators, autophagy indicators and PPARβ/δ. 3-MA was used to assess the role of autophagy in the puerarin-mediated angiogenesis effect in vivo and in vitro. Results The hydrogel significantly improved blood flow restoration in mice with hind-limb ischemia. This effect was mainly due to puerarin-mediated increases in the angiogenic capacity of endothelial cells and the promotion of autophagy activation. A potential underlying mechanism might be that puerarin-mediated activation of autophagy could induce an increase in PPARβ/δ expression. Conclusion The puerarin-attached lignin nanoparticle-incorporated hydrogel effectively alleviated blood perfusion in mice with hind-limb ischemia. Puerarin has a prominent proangiogenic effect. The potential mechanisms might be that puerarin-mediated autophagy activation and increase in PPARβ/δ.
Collapse
Affiliation(s)
- Yingjing Pan
- Foshan Stomatological Hospital, School of Medicine, Foshan University, Foshan, 528225, People’s Republic of China
| | - Tianci Lin
- Foshan Stomatological Hospital, School of Medicine, Foshan University, Foshan, 528225, People’s Republic of China
| | - Longquan Shao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, People’s Republic of China
| | - Yulin Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, People’s Republic of China
| | - Qiao Han
- Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510182, People’s Republic of China
| | - Liyuan Sheng
- Shenzhen Institute, Peking University, Shenzhen, 518057, People’s Republic of China
| | - Rui Guo
- Department of Biomedical Engineering, Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Ting Sun
- Foshan Stomatological Hospital, School of Medicine, Foshan University, Foshan, 528225, People’s Republic of China
| | - Yanli Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, People’s Republic of China
| |
Collapse
|
21
|
Lu Y, Zeng Z, Bao X, Wu M, Jing Z, Feng J. Pristimerin protects against pathological cardiac hypertrophy through improvement of PPARα pathway. Toxicol Appl Pharmacol 2023; 473:116572. [PMID: 37269933 DOI: 10.1016/j.taap.2023.116572] [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: 04/05/2023] [Revised: 05/24/2023] [Accepted: 05/29/2023] [Indexed: 06/05/2023]
Abstract
Pristimerin (PM), serving as a biological component mainly obtained from Celastraceae and Hippocrateaceae families, has been extensively explored for its numerous pharmacological activities, especially anti-cancer activity. However, the function of PM on pathological cardiac hypertrophy is poorly understood. This work was intended to investigate the effects of PM on pressure-overload induced myocardial hypertrophy and its potential pathways. Mouse model of pathological cardiac hypertrophy was generated by transverse aortic constriction (TAC) or minipump administration of the β-adrenergic agonist ISO for 4 weeks, and PM (0.5 mg/Kg/d, i.p.) was treated for 2 weeks. PPARα-/- mice received TAC surgery were used for mechanism exploration. Moreover, neonatal rat cardiomyocytes (NRCMs) were utilized to explore the effect of PM following Angiotensin II (Ang II, 1.0 μM) administration. We found that PM attenuated pressure-overload induced cardiac dysfunction, myocardial hypertrophy and fibrosis in mice. Likewise, PM incubation dramatically reversed Ang II-mediated cardiomyocytes hypertrophy in NRCMs. RNA-Sequence showed that PM selectively contributed to improvement of PPARα/PGC1 signaling, while silencing PPARα abrogated the beneficial effects of PM on Ang II-treated NRCMs. Importantly, PM ameliorated Ang II-induced mitochondrial dysfunction and decrease in metabolic genes, whereas knockdown of PPARα eliminated these alterations in NRCMs. Similarly, PM presented limited protective effects on pressure-overload induced systolic dysfunction and myocardial hypertrophy in PPARα deficient mice. Overall, this study revealed that PM exerted protective activity against pathological cardiac hypertrophy through improvement of PPARα/PGC1 pathway.
Collapse
Affiliation(s)
- Ye Lu
- Department of Interventional Center and Vascular Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; Department of Vascular Surgery, Changhai Hospital, Navy Medical University, Shanghai, PR China
| | - Zhaoxiang Zeng
- Department of Vascular Surgery, Changhai Hospital, Navy Medical University, Shanghai, PR China
| | - Xianhao Bao
- Department of Vascular Surgery, Changhai Hospital, Navy Medical University, Shanghai, PR China
| | - Mingwei Wu
- Department of Vascular Surgery, Changhai Hospital, Navy Medical University, Shanghai, PR China
| | - Zaiping Jing
- Department of Vascular Surgery, Changhai Hospital, Navy Medical University, Shanghai, PR China
| | - Jiaxuan Feng
- Department of Interventional Center and Vascular Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; Department of Vascular Surgery, Changhai Hospital, Navy Medical University, Shanghai, PR China.
| |
Collapse
|
22
|
Zhao Z, Gao W, Ding X, Xu X, Xiao C, Mao G, Xing W. The association between dietary intake of flavonoids and its subclasses and the risk of metabolic syndrome. Front Nutr 2023; 10:1195107. [PMID: 37476404 PMCID: PMC10354435 DOI: 10.3389/fnut.2023.1195107] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/13/2023] [Indexed: 07/22/2023] Open
Abstract
Background The healthiest way to prevent metabolic syndrome (MetS) is through behavioral and nutritional adjustments. We examined the relationship between total flavonoids intake, flavonoid subclasses, and clinically manifest MetS. Methods A cross-sectional analysis was conducted among 28,719 individuals from the National Health and Nutrition Examination Survey (NHANES) and Food and Nutrient Database for Dietary Studies (FNDDS) 2007-2011 and 2017-2018. Two 24-h reviews were conducted to determine flavonoids intake and subclasses. The link between flavonoids intake and MetS was investigated using a multivariate logistic regression model. Results Q2 and Q3 of total flavonoids intake were associated with 20 and 19% lower risk of incident MetS after adjusting age and sex. Anthocyanidins and flavanones intake in Q2 and Q3 substantially reduced the MetS risk compared to Q1. MetS risk decreased steadily as the total intake of flavonoids increased to 237.67 mg/d. Flavanones and anthocyanidins also displayed V-shaped relationship curves (34.37 and 23.13 mg/d). Conclusion MetS was adversely linked with total flavonoids intake, flavanones, and anthocyanidins. Moreover, the most effective doses of total flavonoids, flavanones, and anthocyanidins were 237.67, 34.37, and 23.13 mg/d, respectively, potentially preventing MetS.
Collapse
Affiliation(s)
- Zhenlei Zhao
- Zhejiang Provincial Key Lab of Geriatrics, Zhejiang Hospital, Hangzhou, China
| | - Wenyan Gao
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Xiaoli Ding
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Xiaogang Xu
- Zhejiang Provincial Key Lab of Geriatrics, Zhejiang Hospital, Hangzhou, China
| | - Changqian Xiao
- Zhejiang Provincial Key Lab of Geriatrics, Zhejiang Hospital, Hangzhou, China
| | - Genxiang Mao
- Zhejiang Provincial Key Lab of Geriatrics, Zhejiang Hospital, Hangzhou, China
| | - Wenmin Xing
- Zhejiang Provincial Key Lab of Geriatrics, Zhejiang Hospital, Hangzhou, China
| |
Collapse
|
23
|
Ma Q, Wang M, Li L, Zhang X, Cui L, Mou J, Sun G, Zhang Q. Jiedu Quyu Decoction mitigates monocrotaline-induced right-sided heart failure associated with pulmonary artery hypertension by inhibiting NLRP3 inflammasome in rats. JOURNAL OF ETHNOPHARMACOLOGY 2023; 313:116556. [PMID: 37142147 DOI: 10.1016/j.jep.2023.116556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Right-side heart failure could accelerate mortality in patients of pulmonary hypertension, Jiedu Quyu Decoction (JDQYF) was used to manage pulmonary hypertension, but its right-sided heart protective effect associated with pulmonary artery hypertension is still unclear. AIM OF THE STUDY Here, we evaluated the therapeutic effect of JDQYF on monocrotaline-induced right-sided heart failure associated with pulmonary arterial hypertension in Sprague-Dawley (SD) rats and investigated the potential mechanism of action. MATERIALS AND METHODS The main chemical components of JDQYF were detected and analyzed using ultra-high-performance liquid chromatography quadrupole time-of-flight mass spectrometry. The effects of JDQYF were investigated using a rat model of monocrotaline-induced right-sided heart failure associated with pulmonary arterial hypertension. We assessed the morphology of cardiac tissue using histopathology and the structure and function of the right heart using echocardiography. The biomarkers of heart failure, atrial natriuretic peptide and B-type natriuretic peptide, as well as serum pro-inflammatory markers, interleukin (IL)-1β, and IL-18, were measured by enzyme-linked immunosorbent assay (ELISA). Furthermore, the mRNA and protein expression levels of NLRP3 (NOD-, LRR-, and pyrin domain-containing 3), capase-1, IL-1β, and IL-18 in the right heart tissue were examined by real-time quantitative reverse transcription PCR and western blotting. RESULTS JDQYF improved ventricular function, alleviated pathological lesions in the right cardiac tissue, reduced the expression levels of biomarkers of heart failure and serum pro-inflammatory factors (IL-1β and IL-18), and downregulated the mRNA and protein expression levels of NLRP3, caspase-1, IL-1β, and IL-18 in the right cardiac tissue. CONCLUSIONS JDQYF possesses cardioprotective effect against right heart failure induced by pulmonary arterial hypertension, possibly owing to reduction of cardiac inflammation through the inhibition of NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Qiuxiao Ma
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China; Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China.
| | - Min Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China.
| | - Lanfang Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China.
| | - Xinyu Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Lixin Cui
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Junyu Mou
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China.
| | - Guibo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China.
| | - Qiong Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| |
Collapse
|
24
|
Kang AW, Sun C, Li HT, Zhong K, Zeng XH, Gu ZF, Li BQ, Zhang XN, Gao JL, Chen TX. Puerarin extends the lifespan of Drosophila melanogaster by activating autophagy. Food Funct 2023; 14:2149-2161. [PMID: 36752212 DOI: 10.1039/d2fo02800j] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Lifespan longevity has attracted increasing attention with societal development. To counter the effects of aging on longevity, we focused on the natural chemicals of plants. In this study, we investigated the effects of puerarin supplementation on the lifespan of Drosophila melanogaster. Puerarin supplementation significantly extended the lifespan of D. melanogaster at 60 μM and 120 μM by upregulating proteasome subunit beta 5 (prosbeta5) and sirtuin-1 (Sirt1). However, puerarin-induced longevity of male flies (F0 generation) may not be passed on to descendants. Additionally, a puerarin diet for 10 and 25 days did not influence the body weight and food intake of male Canton-S flies. Puerarin significantly improved the climbing ability, starvation resistance, and oxidation resistance of male flies by upregulating the expression of Shaker, catalase (CAT), superoxide dismutase 1 (SOD1), and Methuselah, and downregulating poly [ADP-ribose] polymerase (PARP-1) and major heat shock 70 kDa protein Aa (HSP70). Moreover, 120 μM puerarin supplementation for 25 days significantly increased adenosine 5' triphosphate (ATP) content by increasing adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) levels. Additionally, the puerarin diet for 25 days suppressed male fecundity in male flies by decreasing the levels of Bam and Punt. Mechanistically, puerarin enhanced lysosome-involved autophagy by promoting the expression of lysosome markers [β-galactosidase and lysosomal associated membrane protein 1 (LAMP1)], and elevating the levels of autophagy-related genes, including autophagy-associated gene 1 (ATG1), ATG5, and ATG8b. However, puerarin decreased the phosphorylation of the target of rapamycin (TOR) protein. In conclusion, puerarin is a promising compound for improving the longevity of D. melanogaster by activating autophagy.
Collapse
Affiliation(s)
- Ai-Wen Kang
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong 226001, Jiangsu, P. R. China.
| | - Chi Sun
- Research Center of Gerontology and Longevity, Affiliated Hospital of Nantong University, Nantong University, Nantong 226001, China. .,Department of Geriatrics, Affiliated Hospital of Nantong University, Nantong University, Nantong 226001, China
| | - Hai-Tao Li
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong 226001, Jiangsu, P. R. China.
| | - Kun Zhong
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong 226001, Jiangsu, P. R. China.
| | - Xu-Hui Zeng
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong 226001, Jiangsu, P. R. China.
| | - Zhi-Feng Gu
- Research Center of Gerontology and Longevity, Affiliated Hospital of Nantong University, Nantong University, Nantong 226001, China.
| | - Bing-Qian Li
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong 226001, Jiangsu, P. R. China.
| | - Xiao-Ning Zhang
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong 226001, Jiangsu, P. R. China.
| | - Jian-Lin Gao
- Research Center of Gerontology and Longevity, Affiliated Hospital of Nantong University, Nantong University, Nantong 226001, China.
| | - Tian-Xing Chen
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong 226001, Jiangsu, P. R. China.
| |
Collapse
|
25
|
Zhao L, Zhou T, Chen J, Cai W, Shi R, Duan Y, Yuan L, Xing C. Colon specific delivery of miR-155 inhibitor alleviates estrogen deficiency related phenotype via microbiota remodeling. Drug Deliv 2022; 29:2610-2620. [PMID: 35938574 PMCID: PMC9364735 DOI: 10.1080/10717544.2022.2108163] [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] [Indexed: 11/27/2022] Open
Abstract
Compelling data have indicated menopause-associated increase in cardiovascular disease in women, while the underlying mechanisms remain largely unknown. It is established that changes of intestinal microbiota affect cardiovascular function in the context of metabolic syndrome. We here aimed to explore the possible link between host intestinal function, microbiota, and cardiac function in the ovariectomy (OVX) mouse model. Mice were ovariectomized to induce estrogen-related metabolic syndrome and cardiovascular defect. Microbiota was analyzed by 16s rRNA sequencing. miRNA and mRNA candidates expression were tested by qPCR. Cardiac function was examined by echocardiography. Colon specific delivery of miRNA candidates was achieved by oral gavage of Eudragit S100 functionalized microspheres. In comparison with the sham-operated group, OVX mice showed compromised cardiac function, together with activated inflammation in the visceral adipose tissue and heart. Lactobacillus abundance was significantly decreased in the gut of OVX mice. Meanwhile, miR-155 was mostly upregulated in the intestinal epithelium and thus the feces over other candidates, which in turn decreased Lactobacillus abundance in the intestine when endocytosed. Oral delivery of miR-155 antagonist restored the protective microbiota and thus protected the cardiac function in the OVX mice. This study has established a possible regulatory axis of intestinal miRNAs-microbiota-estrogen deficiency related phenotype in the OVX model. Colon specific delivery of therapeutic miRNAs would possibly restore the microbiota toward protective phenotype in the context of metabolic syndrome.
Collapse
Affiliation(s)
- Lianbi Zhao
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Tian Zhou
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jianmei Chen
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Wenbin Cai
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Ruijing Shi
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yunyou Duan
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Lijun Yuan
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Changyang Xing
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
26
|
Wang X, He K, Ma L, Wu L, Yang Y, Li Y. Puerarin attenuates isoproterenol‑induced myocardial hypertrophy via inhibition of the Wnt/β‑catenin signaling pathway. Mol Med Rep 2022; 26:306. [PMID: 35946454 PMCID: PMC9437969 DOI: 10.3892/mmr.2022.12822] [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: 03/23/2022] [Accepted: 07/20/2022] [Indexed: 11/06/2022] Open
Abstract
Myocardial hypertrophy (MH) is an independent risk factor for cardiovascular disease, which in turn lead to arrhythmia or heart failure. Therefore, attention must be paid to formulation of therapeutic strategies for MH. Puerarin is a key bioactive ingredient isolated from Pueraria genera of plants that is beneficial for the treatment of MH. However, its molecular mechanism of action has not been fully determined. In the present study, 40 µM puerarin was demonstrated to be a safe dose for human AC16 cells using Cell Counting Kit‑8 assay. The protective effects of puerarin against MH were demonstrated in AC16 cells stimulated with isoproterenol (ISO). These effects were characterized by a significant decrease in surface area of cells (assessed using fluorescence staining) and mRNA and protein expression levels of MH‑associated biomarkers, including atrial and brain natriuretic peptide, assessed using reverse transcription‑quantitative PCR and western blotting, as well as β‑myosin heavy chain mRNA expression levels. Mechanistically, western blotting demonstrated that puerarin inhibited activation of the Wnt signaling pathway. Puerarin also significantly decreased phosphorylation of p65; this was mediated via crosstalk between the Wnt and NF‑κB signaling pathways. An inhibitor (Dickkopf‑1) and activator (IM‑12) of the Wnt signaling pathway were used to demonstrate that puerarin‑mediated effects alleviated ISO‑induced MH via the Wnt signaling pathway. The results of the present study demonstrated that puerarin pre‑treatment may be a potential therapeutic strategy for preventing ISO‑induced MH and managing MH in the future.
Collapse
Affiliation(s)
- Xiaoying Wang
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Kai He
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Linlin Ma
- College of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai 201318, P.R. China
| | - Lan Wu
- College of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai 201318, P.R. China
| | - Yan Yang
- Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, Shanghai 201106, P.R. China
| | - Yanfei Li
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| |
Collapse
|
27
|
Yan J, Honglei Y, Yun W, Sheng D, Yun H, Anhua Z, Na F, Min L, Dandan S, Jing W, Junming T, Wenjun Z, Xiju H. Puerarin ameliorates myocardial remodeling of spontaneously hypertensive rats through inhibiting TRPC6-CaN-NFATc3 pathway. Eur J Pharmacol 2022; 933:175254. [PMID: 36087696 DOI: 10.1016/j.ejphar.2022.175254] [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: 12/20/2021] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/29/2022]
Abstract
Puerarin (Pue) has been widely used in the treatment of hypertension and cardiovascular diseases, but the basic mechanism of Pue on myocardial remodeling (MR) of hypertension is not clear. The purpose of this study was to investigate the effect and mechanism of Pue on MR and provide the basis for the clinical application. Thirty male spontaneously hypertensive rats (SHR) and six male Wistar Kyoto rats (WKY) aged 3 months were used in this study, SHR rats were randomly divided into 5 groups, Pue (40 or 80 mg/kg/d, ip) and telmisartan (TELMI) (30 mg/kg/d, ig) were administrated for 12 weeks. We used Echocardiography to detect the cardiac function. Morphology and structure of myocardium were observed. H9C2 cells were subjected to 1 μM Ang Ⅱ in vitro, 100 μM Pue, 0.5 μM Calmodulin-dependent calcineurin (CaN) inhibitor Cyclosporin A (CsA) and 1 μM specific transient receptor potential channel 6 (TRPC6) inhibitor SAR7334 were used in H9C2 cells. Long-term administration of Pue could significantly improve cardiac function, improve morphology and structure of myocardium in vivo. Pue could reduce MR related proteins expression (ACTC1, TGF-β1, CTGF, β-MHC and BNP), attenuate ROS, restore MMP and decrease Ca2+-overload in vitro. Further study indicated that Pue could decrease TRPC6 expression and inhibit nuclear factor of activated T cells 3 (NFATc3) nuclear translocation in vitro. These results suggested that puerarin could ameliorate myocardial remodeling through inhibiting TRPC6-CaN-NFATc3 in spontaneously hypertensive rats.
Collapse
Affiliation(s)
- Jiang Yan
- Department of Ultrasound, Taihe Hospital, Jinzhou Medicical University Union Training Base, Shiyan, 442000, China
| | - Yu Honglei
- Department of Ultrasound, Taihe Hospital, Jinzhou Medicical University Union Training Base, Shiyan, 442000, China
| | - Wu Yun
- Department of Ultrasound, Wuhan Asia General Hospital, Wuhan, 430000, China
| | - Dong Sheng
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China
| | - He Yun
- Department of Ultrasound, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Zhang Anhua
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Feng Na
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Lu Min
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Shi Dandan
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Wang Jing
- School of Public Health and Health, Hubei University of Medicine, Shiyan, 442000, China
| | - Tang Junming
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Zhang Wenjun
- Department of Ultrasound, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China.
| | - He Xiju
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China; Department of Ultrasound, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China.
| |
Collapse
|
28
|
Tan L, Chen Z, Ruan Y, Xu H. Differential regulatory roles of microRNAs during intramuscular adipogenesis in Chinese Guizhou Congjiang Xiang pigs. Epigenetics 2022; 17:1800-1819. [PMID: 35695092 DOI: 10.1080/15592294.2022.2086675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Intramuscular fat development is regulated by a series of complicated processes, with non-coding RNA (ncRNA) such as microRNA (miRNA) having a critical role during intramuscular preadipocyte proliferation and differentiation in pigs. In the present study, the miRNA expression profiles of intramuscular preadipocytes from the longissimus dorsi muscle of Chinese Guizhou Congjiang Xiang pigs were detected by RNA-seq during various differentiation stages, namely, day 0 (D0), day 4 (D4), and day 8 (D8). A total of 67, 95, and 16 differentially expressed (DE) miRNAs were detected between D4 and D0, D8 and D0, and D8 and D4, respectively. According to gene ontology and Kyoto Encyclopedia of Genes analysis, target genes of DE miRNAs were enriched in categories and pathways related to lipid metabolic process, lipid biosynthetic process, as well as the PI3K-Akt, AMPK, and MAPK signalling pathways. Notably, miR-148a-3p was differentially expressed, with highest expressed abundance in D0, D4, and D8. Overexpression of miR-148a-3p in intramuscular preadipocytes increased cell proliferation and differentiation, and decreased apoptosis, in comparison to the knockdown of miR-148a-3p in intramuscular preadipocytes. Luciferase activity assays, quantitative polymerase-chain reaction, and western blot analysis confirmed that miR-148a-3p regulated adipogenesis by repressing PPARGC1A expression. Accordingly, the effect of miR-148a-3p mimic was attenuated by overexpression of PPARGC1A intramuscular preadipocytes. Furthermore, miR-148a-3p promoted intramuscular preadipocyte differentiation by inhibiting the AMPK/ACC/CPT1C signalling pathway. Taken together, we identified expression profiles of miRNAs in intramuscular preadipocytes and determined that miR-148a-3p acted as a promoter of adipogenesis.
Collapse
Affiliation(s)
- Lulin Tan
- College of Life Science, Guizhou University, Guiyang, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China.,Guizhou Animal Husbandry and Veterinary Research Institute, Guizhou Academy of Agricultural Sciences, Guiyang, China
| | - Zhaojun Chen
- The Potato Institute of Guizhou Province, Guizhou Academy of Agricultural Sciences, Guiyang, China
| | - Yong Ruan
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China
| | - Houqiang Xu
- College of Life Science, Guizhou University, Guiyang, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China
| |
Collapse
|
29
|
Hou M, Lu L, Wu X, Liu H. LCZ696 Ameliorates Isoproterenol-Induced Acute Heart Failure in Rats by Activating the Nrf2 Signaling Pathway. Appl Bionics Biomech 2022; 2022:6077429. [PMID: 35528528 PMCID: PMC9076311 DOI: 10.1155/2022/6077429] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 11/25/2022] Open
Abstract
Objective LCZ696 (sacubitril/valsartan) is an angiotensin II (Ang II) type 1 receptor-neprilysin inhibitor, with effects of immunosuppression, anti-inflammation, antiapoptosis, and antioxidation. The present study was aimed at determining whether LCZ696 has a protective effect against isoproterenol-induced acute heart failure (AHF) in rats. Methods SD rats were randomly divided into four groups: control group, HF group, LCZ696 group, and enalapril group. The cardiac function of rats was evaluated using echocardiographic parameters, heart weight (HW), serum levels of cardiac troponin I (cTnI), and lactate dehydrogenase (LDH). HE is staining, which was used to determine the pathological damage of rat myocardial tissue. Also, we measured oxidative stress markers including reactive oxygen species (ROS), malondialdehyde (MDA), superoxide dismutase (SOD), and catalase (CAT). Finally, the expression of Nrf2 signaling pathway-related proteins was determined using Western blot. Results Compared with the HF group, LCZ696 could significantly improve cardiac function and myocardial injury in rats and reduce AHF-induced oxidative stress. In addition, the results of Western blot confirmed that LCZ696 could upregulate the expression of Nrf2 and HO-1 while decreasing Keap1 expression. Conclusion LCZ696 ameliorates isoproterenol-induced AHF in rats by alleviating oxidative stress injury and activating the Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Min Hou
- Department of Emergency, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Linxin Lu
- Department of Emergency, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaobo Wu
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Lymphoma, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Hongxuan Liu
- Department of Emergency, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
30
|
Xu B, Li J, Chen X, Kou M. Puerarin attenuates cisplatin-induced apoptosis of hair cells through the mitochondrial apoptotic pathway. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119208. [PMID: 35032475 DOI: 10.1016/j.bbamcr.2021.119208] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/14/2021] [Accepted: 12/28/2021] [Indexed: 06/14/2023]
Abstract
Puerarin, one of the main components of Pueraria lobata, has been reported to possess a wide range of pharmacological activities, including anti-inflammatory, antioxidative and anti-apoptotic effects. However, the role of puerarin in ototoxic drug-induced hair cell injury has not been well characterized. This study explored whether puerarin protects against cisplatin-induced hair cell damage and its potential mechanisms. The viability of puerarin-treated HEI-OC1 cells was assessed by CCK8 assay. Reactive oxygen species (ROS) was estimated with flow cytometric analysis using Cellrox Green fluorescent probe. Apoptosis-related protein levels were detected by western blot analysis. Immunostaining of the organ of Corti was performed to determine mice cochlear hair cell survival. Our results showed that puerarin improved cell viability and suppressed apoptosis in the cisplatin-damaged HEI-OC1 cells and cochlear hair cells. Mechanistic studies revealed that puerarin attenuated mitochondrial apoptosis pathway by regulating apoptotic related proteins, such as Bax and cleaved caspase-3, and attenuated ROS accumulation after cisplatin damage. Moreover, puerarin was involved in regulating the Akt pathway in HEI-OC1 cells in response to cisplatin. Our results demonstrated that puerarin administration decreased the sensitivity to apoptosis dependent on the mitochondrial apoptotic pathway by reducing ROS generation, which could be used as a new protective agent against cisplatin-induced ototoxicity.
Collapse
Affiliation(s)
- Bingqiang Xu
- Department of Radiology, Shaanxi Provincial People's Hospital, No.256, You Yi West Street, Xi'an 710068, PR China
| | - Juedan Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, 98 XiWu Road, Xi'an 710004, PR China; Department of General Dentistry and Emergency Room, College of Stomatology, Xi'an Jiaotong University, 98 XiWu Road, Xi'an 710004, PR China
| | - Xiaolong Chen
- Department of Radiology, Shaanxi Provincial People's Hospital, No.256, You Yi West Street, Xi'an 710068, PR China
| | - Mingqing Kou
- Department of Radiology, Shaanxi Provincial People's Hospital, No.256, You Yi West Street, Xi'an 710068, PR China.
| |
Collapse
|
31
|
Epigallocatechin-3-gallate exerts cardioprotective effects related to energy metabolism in pressure overload-induced cardiac dysfunction. Arch Biochem Biophys 2022; 723:109217. [DOI: 10.1016/j.abb.2022.109217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 01/14/2023]
|
32
|
Qin W, Guo J, Gou W, Wu S, Guo N, Zhao Y, Hou W. Molecular mechanisms of isoflavone puerarin against cardiovascular diseases: What we know and where we go. CHINESE HERBAL MEDICINES 2022; 14:234-243. [PMID: 36117660 PMCID: PMC9476793 DOI: 10.1016/j.chmed.2021.12.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/09/2021] [Accepted: 12/29/2021] [Indexed: 12/09/2022] Open
|
33
|
Jiang Z, Cui X, Qu P, Shang C, Xiang M, Wang J. Roles and mechanisms of puerarin on cardiovascular disease:A review. Biomed Pharmacother 2022; 147:112655. [DOI: 10.1016/j.biopha.2022.112655] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/13/2022] [Accepted: 01/16/2022] [Indexed: 12/13/2022] Open
|
34
|
Cui YK, Hong YX, Wu WY, Han WM, Wu Y, Wu C, Li GR, Wang Y. Acacetin ameliorates cardiac hypertrophy by activating Sirt1/AMPK/PGC-1α pathway. Eur J Pharmacol 2022; 920:174858. [PMID: 35219729 DOI: 10.1016/j.ejphar.2022.174858] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/15/2022] [Accepted: 02/22/2022] [Indexed: 12/20/2022]
Abstract
Cardiac hypertrophy is a major risk factor for developing heart failure. This study investigates the effects of the natural flavone acacetin on myocardial hypertrophy in cellular level and whole animals. In cardiomyocytes from neonatal rat with hypertrophy induced by angiotensin II (Ang II), acacetin at 0.3, 1, and 3 μM reduced the increased myocyte surface area, brain natriuretic peptide (BNP), and ROS production by upregulating anti-oxidative molecules (i.e. Nrf2, SOD1, SOD2, HO-1), anti-apoptotic protein Bcl-2, and downregulating the pro-apoptotic protein Bax and the inflammatory cytokine IL-6 in a concentration-dependent manner. In addition, acacetin rescued Ang II-induced impairment of PGC-1α, PPARα and pAMPK. These beneficial effects of acacetin were mediated by activation of Sirt1, which was confirmed in cardiac hypertrophy induced by abdominal aorta constriction (AAC) in SD rats. Acacetin prodrug (10 mg/kg, s.c., b.i.d.) treatment reduced the elevated artery blood pressure, improved the increased heart size and thickness of left ventricular wall and the ventricular fibrosis associated with inhibiting myocardial fibrosis and BNP, and reversed the impaired protective signal molecules including PGC-1α, Nrf2, PPARα, pAMPK and Sirt1 of left ventricular tissue. Our results demonstrate the novel pharmacological effect that acacetin ameliorates cardiac hypertrophy via Sirt1-mediated activation of AMPK/PGC-1α signal molecules followed by reducing oxidation, inflammation and apoptosis.
Collapse
Affiliation(s)
- Yu-Kai Cui
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361009, China
| | - Yi-Xiang Hong
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361009, China
| | - Wei-Yin Wu
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361009, China
| | - Wei-Min Han
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361009, China
| | - Yao Wu
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361009, China
| | - Chan Wu
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361009, China
| | - Gui-Rong Li
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361009, China; Nanjing Amazigh Pharma Limited, Nanjing, Jiangsu, 210032, China.
| | - Yan Wang
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361009, China.
| |
Collapse
|
35
|
Zhang F, Liu L, Xie Y, Wang J, Chen X, Zheng S, Li Y, Dang Y. Cardiac contractility modulation ameliorates myocardial metabolic remodeling in a rabbit model of chronic heart failure through activation of AMPK and PPAR-α pathway. Open Med (Wars) 2022; 17:365-374. [PMID: 35799598 PMCID: PMC8864057 DOI: 10.1515/med-2022-0415] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/03/2021] [Accepted: 11/20/2021] [Indexed: 11/15/2022] Open
Abstract
Abstract
Metabolic remodeling contributes to the pathological process of heart failure (HF). We explored the effects of cardiac contractility modulation (CCM) on myocardial metabolic remodeling in the rabbit model with HF. The HF in rabbit model was established by pressure uploading and then CCM was applied. We evaluated the cardiac structure and function by echocardiography, serum BNP level, and hematoxylin and eosin and Masson’s trichrome staining. We detected the accumulation of glycogen and lipid droplets in myocardial tissues by periodic acid-Schiff and Oil Red O staining. Then, we measured the contents of glucose, free fatty acid (FFA), lactic acid, pyruvate, and adenosine triphosphate (ATP) levels in myocardial tissues by corresponding kits and the expression levels of key factors related to myocardial substrate uptake and utilization by western blotting were analyzed. CCM significantly restored the cardiac structure and function in the rabbit model with HF. CCM therapy further decreased the accumulation of glycogen and lipid droplets. Furthermore, CCM reduced the contents of FFA, glucose, and lactic acid, and increased pyruvate and ATP levels in HF tissues. The protein expression levels related to myocardial substrate uptake and utilization were markedly improved with CCM treatment by further activating adenosine monophosphate-activated protein kinase and peroxisome proliferator-activated receptor-α signaling pathways.
Collapse
Affiliation(s)
- Feifei Zhang
- Department of Cardiology Center, Hebei General Hospital, Xipingxilu 348, Xinhua, Shijiazhuang, 050051, Hebei Province, China
| | - Litian Liu
- Department of Cardiology Center, Hebei General Hospital, Xipingxilu 348, Xinhua, Shijiazhuang, 050051, Hebei Province, China
| | - Yuetao Xie
- Department of Cardiology Center, Hebei General Hospital, Xipingxilu 348, Xinhua, Shijiazhuang, 050051, Hebei Province, China
| | - Jiaqi Wang
- Department of Cardiology Center, Hebei General Hospital, Xipingxilu 348, Xinhua, Shijiazhuang, 050051, Hebei Province, China
| | - Xuefeng Chen
- Department of Cardiology Center, Hebei General Hospital, Xipingxilu 348, Xinhua, Shijiazhuang, 050051, Hebei Province, China
| | - Shihang Zheng
- Department of Cardiology Center, Hebei General Hospital, Xipingxilu 348, Xinhua, Shijiazhuang, 050051, Hebei Province, China
| | - Yingxiao Li
- Department of Cardiology Center, Hebei General Hospital, Xipingxilu 348, Xinhua, Shijiazhuang, 050051, Hebei Province, China
| | - Yi Dang
- Department of Cardiology Center, Hebei General Hospital, Xipingxilu 348, Xinhua, Shijiazhuang, 050051, Hebei Province, China
| |
Collapse
|
36
|
Li KX, Feng Y, Fan XX, Sun X, Li Y, Wu D, Liu L, Cui CP, Xiong X, Li HD, Zhou M, Ma HL, Liu Y, Zhang R, Li BY. Bradykinin-mediated estrogen-dependent depressor response by direct activation of female-specific distribution of myelinated Ah-type baroreceptor neurons in rats. CNS Neurosci Ther 2021; 28:435-447. [PMID: 34964272 PMCID: PMC8841294 DOI: 10.1111/cns.13792] [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: 10/16/2021] [Revised: 12/06/2021] [Accepted: 12/14/2021] [Indexed: 12/03/2022] Open
Abstract
Aim To understand the direct impact of bradykinin in autonomic control of circulation through baroreflex afferent pathway. Methods The mean arterial pressure (MAP) was monitored while bradykinin and its agonists were applied via nodose (NG) microinjection, the expression of bradykinin receptors (BRs) in the NG (1st‐order) and nucleus tractus solitarius (NTS, 2nd‐order) were tested in adult male, age‐matched female, and ovariectomized rats under physiological and hypertensive conditions. Additionally, bradykinin‐induced depolarization was also tested in identified baroreceptor and baroreceptive neurons using whole‐cell patch‐clamp technique. Results Under physiological condition, bradykinin‐induced dose‐ and estrogen‐dependent reductions of MAP with lower estimated EC50 in females. B2R agonist mediated more dramatic MAP reduction with long‐lasting effect compared with B1R activation. These functional observations were consistent with the molecular and immunostaining evidences. However, under hypertensive condition, the MAP reduction was significantly less dramatic in N’‐Nitro‐L‐Arginine‐methyl ester (L‐NAME) induced secondary and spontaneous hypertension rats in males compared with female rats. Electrophysiological data showed that bradykinin‐elicited concentration‐dependent membrane depolarization with discharges during initial phase in identified myelinated Ah‐types baroreceptor neurons, not myelinated A‐types; while, higher concentration of bradykinin was required for depolarization of unmyelinated C‐types without initial discharges. Conclusion These datasets have demonstrated for the first time that bradykinin mediates direct activation of baroreflex afferent function to trigger estrogen‐dependent depressor response, which is due mainly to the direct activation/neuroexcitation of female‐specific myelinated Ah‐type baroreceptor neurons leading to a sexual dimorphism in parasympathetic domination of blood pressure regulation via activation of B2R/B1R expression in baroreflex afferent pathway.
Collapse
Affiliation(s)
- Ke-Xin Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yan Feng
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xiong-Xiong Fan
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xun Sun
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Ying Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Di Wu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Li Liu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Chang-Peng Cui
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xue Xiong
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Hu-Die Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Meng Zhou
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Hai-Lan Ma
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yang Liu
- Department of clinical Laboratory, The 1st Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Rong Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Bai-Yan Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, and Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| |
Collapse
|
37
|
Hou N, Li LR, Shi YY, Yuan WC, Zhao GJ, Liu XW, Cai SA, Huang Y, Zhan HX, Pan WB, Luo CF. Azilsartan ameliorates ventricular hypertrophy in rats suffering from pressure overload-induced cardiac hypertrophy by activating the Keap1-Nrf2 signalling pathway. J Pharm Pharmacol 2021; 73:1715-1725. [PMID: 34343333 DOI: 10.1093/jpp/rgab097] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 06/15/2021] [Indexed: 01/15/2023]
Abstract
OBJECTIVES Investigate if azilsartan protects against myocardial hypertrophy by upregulating nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated pathways. METHODS Abdominal aortic constriction (AAC)-induced cardiac hypertrophy in rats was applied. Azilsartan or vehicle was administered daily for 6 weeks in sham or AAC rats. Cardiac morphology and ventricular function were determined. Azilsartan effects upon neonatal rat cardiomyocyte (NRCM) hypertrophy and molecular mechanisms were studied in angiotensin (Ang) II-stimulated NRCMs in vitro. Nrf2-small interfering RNA (siRNA) was used to knockdown Nrf2 expression. Messenger RNA (mRNA)/protein expression of Kelch-like erythroid cell-derived protein (Keap)1 and Nrf2 and its downstream antioxidant enzymes was determined by real-time reverse transcription-quantitative polymerase chain reaction and western blotting, respectively. KEY FINDINGS Azilsartan treatment ameliorated cardiac hypertrophy/fibrosis significantly in AAC rats. Azilsartan increased expression of Nrf2 protein but decreased expression of Keap1 protein. Upregulation of protein expression of Nrf2's downstream antioxidant enzymes by azilsartan treatment was observed. Azilsartan inhibited Ang II-induced NRCM hypertrophy significantly and similar effects on the Keap1-Nrf2 pathway were observed in vivo. Nrf2 knockdown markedly counteracted the beneficial effects of azilsartan on NRCM hypertrophy and the Keap1-Nrf2 pathway. CONCLUSIONS Azilsartan restrained pressure overload-induced cardiac remodelling by activating the Keap1-Nrf2 pathway and increasing expression of downstream antioxidant enzymes to alleviate oxidative stress.
Collapse
Affiliation(s)
- Ning Hou
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Li-Rong Li
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Tungwah Hospital of Sun Yat-sen University, Dongguan, China
| | - Yong-Ying Shi
- Department of Geriatrics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Wen-Chang Yuan
- Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Clinical Laboratory, General Hospital of Xinjiang Military Region, Urumqi, China
| | - Gan-Jian Zhao
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xia-Wen Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shao-Ai Cai
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yin Huang
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Cardiology, Xiangtan Central Hospital, Xiangtan, China
| | - Hao-Xin Zhan
- Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Wei-Biao Pan
- Tungwah Hospital of Sun Yat-sen University, Dongguan, China
| | - Cheng-Feng Luo
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
38
|
Zhang L, Liu L, Wang M. Effects of puerarin on chronic inflammation: Focus on the heart, brain, and arteries. Aging Med (Milton) 2021; 4:317-324. [PMID: 34964013 PMCID: PMC8711227 DOI: 10.1002/agm2.12189] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 12/05/2021] [Accepted: 12/05/2021] [Indexed: 11/23/2022] Open
Abstract
Age-associated increases in physical and mental stress, known as allostatic load, could lead to a chronic low-grade inflammation in the heart, brain, and arteries. This low-grade inflammation potentially contributes to adverse structural and functional remodeling, such as intimal medial thickening, endothelial dysfunction, arterial stiffening, cardiac hypertrophy and ischemia, and cognitive decline. These cellular and tissue remodeling is the fertile soil for the development of age-associated structural and functional disorders in the cardiovascular and cerebrovascular systems in the pathogenesis of obesity, type II diabetes, hypertension, atherosclerosis, heart dysfunction, and cognitive decline. Growing evidence indicates that puerarin, a polyphenol, extracted from Puerara Labota, efficiently alleviates the initiation and progression of obesity, type II diabetes, hypertension, atherosclerosis, cardiac ischemia, cardiac arrythmia, cardiac hypertrophy, ischemic stroke, and cognition decline via suppression of oxidative stress and inflammation. This mini review focuses on recent advances in the effects of puerarin on the oxidative and inflammatory molecular, cellular, tissue events in the heart, brain, and arteries.
Collapse
Affiliation(s)
- Li Zhang
- Department of CardiologyThe First Affiliated Hospital of Guangdong Pharmaceutical UniversityGuangzhouChina
| | - Lisheng Liu
- National Centre for Cardiovascular DiseaseThe Beijing Hypertension League InstituteBeijingChina
| | - Mingyi Wang
- Laboratory of Cardiovascular ScienceIntramural Research ProgramNational Institute on AgingNational Institutes of HealthBRCBaltimoreMarylandUSA
| |
Collapse
|
39
|
Yu J, Zhao H, Qi X, Wei L, Li Z, Li C, Zhang X, Wu H. Dapagliflozin Mediates Plin5/PPARα Signaling Axis to Attenuate Cardiac Hypertrophy. Front Pharmacol 2021; 12:730623. [PMID: 34630108 PMCID: PMC8495133 DOI: 10.3389/fphar.2021.730623] [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: 06/25/2021] [Accepted: 09/08/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: The purpose of this study was to investigate the effect of dapagliflozin (DAPA), a sodium-glucose cotransporter 2 inhibitor, on relieving cardiac hypertrophy and its potential molecular mechanism. Methods: Cardiac hypertrophy induced by abdominal aortic constriction (AAC) in mice, dapagliflozin were administered in the drinking water at a dose of 25 mg/kg/d for 12 weeks was observed. Echocardiography was used to detect the changes of cardiac function, including LVEF, LVFS, LVEDd, LVEDs, HR and LV mass. Histological morphological changes were evaluated by Masson trichrome staining and wheat germ agglutinin (WGA) staining. The enrichment of differential genes and signal pathways after treatment was analyzed by gene microarray cardiomyocyte hypertrophy was induced by AngII (2 μM) and the protective effect of dapagliflozin (1 μM) was observed in vitro. The morphological changes of myocardial cells were detected by cTnI immunofluorescence staining. ELISA and qRT-PCR assays were performed to detect the expressions levels of cardiac hypertrophy related molecules. Results: After 12 weeks of treatment, DAPA significantly ameliorated cardiac function and inhibited cardiac hypertrophy in AAC-induced mice. In vitro, DAPA significantly inhibited abnormal hypertrophy in AngII-induced cardiacmyocytes. Both in vivo and in vitro experiments have confirmed that DAPA could mediate the Plin5/PPARα signaling axis to play a protective role in inhibiting cardiac hypertrophy. Conclusion: Dapagliflozin activated the Plin5/PPARα signaling axis and exerts a protective effect against cardiac hypertrophy.
Collapse
Affiliation(s)
- Jing Yu
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Huanhuan Zhao
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Xin Qi
- Department of Cardiology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China.,Nankai University School of Medicine, Tianjin, China
| | - Liping Wei
- Department of Cardiology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China.,Nankai University School of Medicine, Tianjin, China
| | - Zihao Li
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Chunpeng Li
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Xiaoying Zhang
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Hao Wu
- Department of Cardiology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China.,Nankai University School of Medicine, Tianjin, China
| |
Collapse
|
40
|
Zhao W, Zhao J, Zhang X, Fan N, Rong J. Upregulation of Small Ubiquitin-Like Modifier 2 and Protein SUMOylation as a Cardioprotective Mechanism Against Myocardial Ischemia-Reperfusion Injury. Front Pharmacol 2021; 12:731980. [PMID: 34588985 PMCID: PMC8473707 DOI: 10.3389/fphar.2021.731980] [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: 06/28/2021] [Accepted: 08/30/2021] [Indexed: 01/14/2023] Open
Abstract
Background: Small ubiquitin-like modifier (SUMO) proteins modify proteins through SUMOylation as an essential protein post-translational modification (PTM) for regulating redox status, inflammation, and cardiac fibrosis in myocardial infarction. This study aimed to investigate whether natural product puerarin could alleviate myocardial ischemia/reperfusion injury (MI-RI) by targeting protein SUMOylation. Methods: Mouse MI-RI model was induced by ligating the left anterior descending (LAD) coronary artery and subsequently treated with puerarin at the dose of 100 mg/kg. Rat cardiomyocyte H9c2 cells were challenged by hypoxia/reoxygenation and treated with puerarin at concentrations of 10, 20, and 40 μM. The infarction area of mouse hearts was assessed by 2% TTC staining. Cell damage was analyzed for the release of lactate dehydrogenase (LDH) in serum and cell culture medium. Western blot technique was employed to detect the expression of SUMO2, phospho-ERK, pro-inflammatory biomarker COX2, fibrosis index galectin-3, apoptosis-related protein cleaved PARP-1. The activation of the estrogen receptor (ER) pathway was assayed by the dual-luciferase reporter system. Results: The present study validated that puerarin effectively reduced myocardial infarct size and LDH release in the mouse MI-RI model. In the cell culture system, puerarin effectively decreased the release of LDH and the protein level of COX2, galectin-3, and cleaved PARP-1. Mechanistic studies revealed that puerarin increased the expression of SUMO2, SUMOylation of proteins and the activation of ER/ERK pathway in cardiomyocytes. ER, ERK and SUMO2 inhibitors attenuated the cardioprotective effects of puerarin. Conclusion: Puerarin may alleviate myocardial injury by promoting protein SUMOylation through ER/ERK/SUMO2-dependent mechanism.
Collapse
Affiliation(s)
- Wei Zhao
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, SAR China.,Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jia Zhao
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, SAR China
| | - Xiuying Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, SAR China
| | - Ni Fan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, SAR China
| | - Jianhui Rong
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, SAR China.,Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, China
| |
Collapse
|
41
|
Fang X, Dong S, Wu Y, He Y, Lu M, Shi D, Feng N, Yin S, Jiang Y, Zhang A, Ding Y, Zhang Q, Tang J, Zhang W, He X. Ameliorated biomechanical properties of carotid arteries by puerarin in spontaneously hypertensive rats. BMC Complement Med Ther 2021; 21:173. [PMID: 34154575 PMCID: PMC8216761 DOI: 10.1186/s12906-021-03345-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/02/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND An emerging body of evidence indicates that puerarin (PUE) plays an important role in the treatment of angina pectoris, myocardial ischemia-reperfusion injury, hypertension and other cardiovascular diseases, but how PUE affects the vascular remodeling of hypertensive rats has not been reported yet. This study aimed to investigate the effect and mechanism of PUE on carotid arteries of spontaneously hypertensive rats (SHR) to provide the basis for the clinical application of PUE. METHODS Thirty male SHR and six male Wistar Kyoto rats (WKY) aged 3 months were used in this study, SHR rats were randomly divided into 5 groups, PUE(40 or 80 mg/kg/d, ip) and telmisartan (TELMI) (30 mg/kg/d, ig) were administrated for 3 months. We use DMT myography pressure-diameter system to investigate biomechanical properties of carotid arteries, 10 μM pan-classical transient receptor potential channels (TRPCs) inhibitor SKF96365, 200 nM specific TRPC6 inhibitor SAR7334 and 100 μM Orai1 inhibitor ANCOA4 were used in the mechanical test. RESULTS PUE can significantly decrease systolic and diastolic blood pressure, long-term administration of PUE resulted in a mild reduction of thickness and inner diameter of carotid artery. PUE ameliorate NE-response and vascular remodeling mainly through inhibiting TRPCs channel activities of VSMC. CONCLUSION PUE can ameliorate biomechanical remodeling of carotid arteries through inhibiting TRPCs channel activities of VSMC in spontaneously hypertensive rats.
Collapse
Affiliation(s)
- Xiaoxia Fang
- Department of Neurology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000 China
| | - Sheng Dong
- Department of Anatomy, Hubei University of Medicine, Shiyan, 442000 China
| | - Yun Wu
- Department of Anatomy, Hubei University of Medicine, Shiyan, 442000 China
| | - Yun He
- Department of Ultrasound, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000 China
| | - Min Lu
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000 China
| | - Dandan Shi
- Department of Anatomy, Hubei University of Medicine, Shiyan, 442000 China
| | - Na Feng
- Department of Anatomy, Hubei University of Medicine, Shiyan, 442000 China
| | - Songhe Yin
- Department of Neurology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000 China
| | - Yan Jiang
- Department of Ultrasound, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000 China
| | - Anhua Zhang
- Department of Anatomy, Hubei University of Medicine, Shiyan, 442000 China
| | - Yan Ding
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000 China
| | - Qiufang Zhang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000 China
| | - Junming Tang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000 China
| | - Wenjun Zhang
- Department of Ultrasound, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000 China
| | - Xiju He
- Department of Anatomy, Hubei University of Medicine, Shiyan, 442000 China
- Department of Ultrasound, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000 China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000 China
| |
Collapse
|