1
|
Mohammadi K, Shafie D, Ghomashi N, Abdolizadeh A, Sadeghpour M. Kinin-kallikrein system: New perspectives in heart failure. Heart Fail Rev 2024; 29:729-737. [PMID: 38381277 DOI: 10.1007/s10741-024-10393-y] [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] [Accepted: 02/13/2024] [Indexed: 02/22/2024]
Abstract
Heart failure (HF) is a pervasive clinical challenge characterized by compromised cardiac function and reduced quality of life. The kinin-kallikrein system (KSS), a multifaceted peptide cascade, has garnered substantial attention due to its potential role in HF. Through activation of B1 and/or B2 receptors and downstream signaling, kinins modulate various physiological processes, including inflammation, coagulation, pain, blood pressure control, and vascular permeability. Notably, aberrations in KKS components have been linked to HF risk. The elevation of vasodilatory bradykinin (BK) due to kallikrein activity reduces preload and afterload, while concurrently fostering sodium reabsorption inhibition. However, kallikrein's conversion of prorenin to renin leads to angiotensinsII upregulation, resulting in vasoconstriction and fluid retention, alongside increased immune cell activity that fuels inflammation and cardiac remodeling. Importantly, prolonged KKS activation resulting from volume overload and tissue stretch contributes to cardiac collagen loss. The conventional renin-angiotensin-aldosterone system (RAAS) inhibitors used in HF management may inadvertently intensify KKS activity, exacerbating collagen depletion and cardiac remodeling. It is crucial to balance the KKS's role in acute cardiac damage, which may temporarily enhance function and metabolic parameters against its detrimental long-term effects. Thus, KKS blockade emerges as a promising strategy to impede HF progression. By attenuating the link between immune system function and tissue damage, KKS inhibition can potentially reduce cardiac remodeling and alleviate HF symptoms. However, the nuanced roles of BK in various acute conditions necessitate further investigation into the sustained benefits of kallikrein inhibitors in patients with chronic HF.
Collapse
Affiliation(s)
- Keivan Mohammadi
- Shahid Chamran Heart Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Davood Shafie
- Heart Failure Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Newsha Ghomashi
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Abdolizadeh
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Majid Sadeghpour
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
2
|
Bulut F, Tetiker AT, Çelikkol A, Yılmaz A, Ballica B. Low Antioxidant Enzyme Levels and Oxidative Stress in Laryngopharyngeal Reflux (LPR) Patients. J Voice 2023; 37:924-931. [PMID: 34253427 DOI: 10.1016/j.jvoice.2021.05.018] [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: 02/12/2021] [Revised: 04/11/2021] [Accepted: 05/18/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Laryngopharyngeal reflux disease (LPR) is a characterized by symptoms different from gastroesophageal reflux disease (GERD). LPR can causes chronic mucosal inflammation which may lead to an increase in cytokine production, and a systemic decrease in antioxidant enzyme levels. Our aim in this study is to evaluate antioxidant enzyme levels in patients with LPR. METHODS Reflux Symptom Index (RSI) questionnaire, extraesophageal symptom questionnaire which is included in RSI and Reflux Finding Score (RFS) evaluation with 70° rigid laryngoscope were performed to patients who applied to the otolaryngology clinic with a typical LPR complaint, and 60 patients who had an RSI score above 13 and an RFS score above 7 were included in the study. Thirty people consisting of healthy volunteers were included in the control group. Antioxidant enzyme SOD, GSH-Px and CAT levels were measured in the blood serum of the patients and compared with the control group. Results obtained from biochemical tests were expressed as mean ± SE. Descriptive statistical methods (mean ± standard error) were used for the independent t test for the control and study group. P < 0.05 was considered statistically significant. RESULTS In the LPR group, 28 (46%) were women, 32 (53%) were men, and age range was 21-60, average age was 36.45 ± 1.147.There was no significant difference between LPR and control group in terms of age, gender and Body Mass Index (BMI). In the LPR group, the lowest score for RSI was 14 and the highest score was 39. The average RSI was 23.67. RFS ranges from 8-22. The mean RFS was 13.50. A highly significant statistical correlation was observed between RSI and total RFS (P < 0.001). There was a significant difference between the antioxidant enzyme levels of the control group and the LPR group. Antioxidant enzyme levels of the control group were SOD 274.10 ± 26.836 U / L, GSH-Px 174.20 ± 20.699 µU / mL and CAT 42.2898 ± 20.699 KU / L. Antioxidant enzyme level results of the LPR group were SOD 147 ± 14.022 U / L (P < 0.01), GSH-Px 88.28 ± 9.113 µU / mL (P < 0.01) and CAT 12.67 ± 0.799 KU / L (P < 0.001). The RSI results ranges from 4 to 39 and the RFS from 8 to 22. Antioxidant enzyme levels demonstrated fairly consistent reliability with individual variables from both RFS and RFS. There was also a highly significant statistical correlation between RSI and RFS. CONCLUSION We found that the antioxidant enzymes SOD, GPX and catalase enzyme levels were significantly lower in LPR patients. Treatment modalities to reduce oxidative stress (OS) in LPR should be investigated.
Collapse
Affiliation(s)
- Fuat Bulut
- Otorhinolaryngology, Private Çorlu REYAP Hospital, Istanbul Rumeli University, Istanbul, Turkey.
| | | | - Aliye Çelikkol
- Department of Medical Biochemistry, Faculty of Medicine, Tekirdag Namık Kemal University, Tekirdag, Turkey
| | - Ahsen Yılmaz
- Department of Medical Biochemistry, Faculty of Medicine, Tekirdag Namık Kemal University, Tekirdag, Turkey
| | - Basak Ballica
- Bahcesehir University Medical Faculty, Istanbul, Turkey
| |
Collapse
|
3
|
Zhou K, Chen H, Wang XY, Xu YM, Liao YF, Qin YY, Ge XW, Zhang TT, Fang ZL, Fu BB, Xiao QZ, Zhu FQ, Chen SR, Liu XS, Luo QC, Gao S. Targeted pharmacokinetics and bioinformatics screening strategy reveals JAK2 as the main target for Xin-Ji-Er-Kang in treatment of MIR injury. Biomed Pharmacother 2022; 155:113792. [PMID: 36271569 DOI: 10.1016/j.biopha.2022.113792] [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: 08/07/2022] [Revised: 09/23/2022] [Accepted: 10/02/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Xin-Ji-Er-Kang (XJEK) is traditional Chinese formula presented excellent protective effects on several heart diseases, but the potential components and targets are still unclear. The aim of this study is to elucidate the effective components of XJEK and reveal its potential mechanism of cardioprotective effect in myocardial ischemia-reperfusion (MIR) injury. EXPERIMENTAL APPROACH Firstly, the key compounds in XJEK, plasma and heart tissue were analyzed by high resolution mass spectrometry. Bioinformatics studies were also involved to disclose the potential targets and the binding sites for the key compounds. Secondly, to study the protective effect of XJEK on MIR injury and related mechanism, mice subjected to MIR surgery and gavage administered with XJEK for 6 weeks. Cardiac function parameters and apoptosis level of cardiac tissue were assessed. The potential mechanism was further verified by knock down of target protein in vitro. RESULTS Pharmacokinetics studies showed that Sophora flavescens alkaloids, primarily composed with matrine, are the key component of XJEK. And, through bioinformatic analysis, we speculated JAK2 could be the potential target for XJEK, and could form stable hydrogen bonds with matrine. Administration of XJEK and matrine significantly improved heart function and reduced apoptosis of cardiomyocytes by increasing the phosphorylation of JAK2 and STAT3. The anti-apoptosis effect of XJEK and matrine was also observed on AC16 cells, and could be reversed by co-treatment with JAK2 inhibitor AG490 or knock-down of JAK2. CONCLUSION XJEK exerts cardioprotective effect on MIR injury, which may be associated with the activation of JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Kai Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Hua Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Xiao-Yu Wang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Yan-Mei Xu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Yu-Feng Liao
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Yuan-Yuan Qin
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Xue-Wan Ge
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Ting-Ting Zhang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Zhong-Lin Fang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Bei-Bei Fu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Qing-Zhong Xiao
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Feng-Qin Zhu
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230032, China
| | - Si-Rui Chen
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon Tong, Hong Kong SAR China
| | - Xue-Sheng Liu
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| | - Qi-Chao Luo
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| | - Shan Gao
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
4
|
The Relationship between Serum CXCL8 and ET-1 Expression Levels and Sepsis Complicated with Heart Failure. Cardiol Res Pract 2022; 2022:8570486. [PMID: 36065195 PMCID: PMC9440819 DOI: 10.1155/2022/8570486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/04/2022] [Accepted: 07/29/2022] [Indexed: 11/17/2022] Open
Abstract
Objective. The objective is to investigate the relationship between sepsis complicated with heart failure and the expression levels of CXC chemokine ligand 8 (CXCL8) and endothelin-1 (ET-1). Methods. A total of 128 sepsis patients accepted by the Ganzhou People’s Hospital from March 2019 to December 2021 were collected as observation objects, and they were separated into a simple sepsis group (86 cases) and a complicated heart failure group (42 cases) according to whether they were accompanied by heart failure or not. General data such as Sequential Organ Failure Assessment (SOFA) score and Acute Physiology and Chronic Health Evaluation II (APACHE II) were collected; the expression levels of serum CXCL8 and ET-1 were detected by enzyme-linked immunosorbent assay (ELISA); the cardiac function parameters such as left ventricular ejection fraction (LVEF), stroke volume (SV), cardiac output (CO), and cardiac index (CI) were measured by color Doppler ultrasound; the correlation between serum CXCL8 and ET-1 expression levels with clinical data and cardiac function parameters in patients with sepsis complicated with heart failure was analyzed by the Pearson correlation; and the influencing factors of sepsis complicated with heart failure were analyzed by the logistic regression analysis. Results. The serum CXCL8 and ET-1 expression levels, SOFA score, and APACHE II score in the complicated heart failure group were higher than those in the simple sepsis group (
), and LVEF, SV, CO, and CI in the complicated heart failure group were lower than those in the simple sepsis group (
). Serum CXCL8 was positively correlated with ET-1 in patients with sepsis complicated with heart failure (r = 0.531,
), and the two were positively correlated with SOFA score and APACHE II score (
) and were negatively correlated with LVEF, SV, CO, and CI (
). CXCL8 and ET-1 were independent risk factors for sepsis complicated with heart failure (
). Conclusion. The expression levels of serum CXCL8 and ET-1 in sepsis patients with heart failure are significantly increased, and both are risk factors for heart failure in sepsis patients.
Collapse
|
5
|
Sun LJ, Wang XY, Xia J, Xu YM, Liao YF, Qin YY, Ge XW, Zhao PW, Xu T, Zhu XL, Gao S, Xiao R, Liu XS, Zhou K. Xin-Ji-Er-Kang protects heart from ischemia-reperfusion injury by rebalancing lipid metabolism. Front Pharmacol 2022; 13:981766. [PMID: 36081937 PMCID: PMC9445194 DOI: 10.3389/fphar.2022.981766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/29/2022] [Indexed: 11/29/2022] Open
Abstract
Background and Purpose: We have previously reported a cardioprotective effect with Xin-Ji-Er-Kang (XJEK) treatment in mice with myocardial infarction (MI)-induced heart failure, but no report about its potential functions in myocardial ischemia-reperfusion (MIR) injury. Here we studied the therapeutic effects of XJEK on MIR injury and investigated the mechanisms involved. Experimental Approach: MIR model of Balb/c mice induced by left anterior descending coronary artery ligation for half an hour, followed by reperfusion, was utilized to study the potential therapeutic effects of XJEK on MIR-induced cardiac injury. Ultra-performance liquid chromatography tandem Orbitrap mass spectrometry platform was used for studying serum lipid metabolic signatures. Key Results: MIR caused cardiac dysfunctions, cardiac injury, myocardial fibrosis, and increased inflammation, and all the observed abnormalities caused by MIR were largely corrected by XJEK treatment. Mechanistically, XJEK exerts its cardioprotective effect in the context of MIR injury by suppressing MIR-induced inflammation and dysregulation of serum lipid metabolism. Conclusion and Implications: We have demonstrated for the first time that XJEK protects heart from MIR injury by restoring dysregulated lipidomics. Our data provide new evidence to support a therapeutic effect for XIEK on MIR-induced cardiac injury, and pave the way for exploring the therapeutic potential of XJEK in large animal study and early clinical trial.
Collapse
Affiliation(s)
- Li-Jun Sun
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xiao-Yu Wang
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jie Xia
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yan-Mei Xu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yu-Feng Liao
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yuan-Yuan Qin
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xue-Wan Ge
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Pei-Wen Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Tong Xu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xiao-Ling Zhu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shan Gao
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- *Correspondence: Shan Gao, ; Rui Xiao, ; Xue-Sheng Liu, ; Kai Zhou,
| | - Rui Xiao
- Ipswich Hospital, East Suffolk and North Essex NHS Foundation Trust, Ipswich, United Kingdom
- *Correspondence: Shan Gao, ; Rui Xiao, ; Xue-Sheng Liu, ; Kai Zhou,
| | - Xue-Sheng Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Shan Gao, ; Rui Xiao, ; Xue-Sheng Liu, ; Kai Zhou,
| | - Kai Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- *Correspondence: Shan Gao, ; Rui Xiao, ; Xue-Sheng Liu, ; Kai Zhou,
| |
Collapse
|
6
|
Yang S, Wang C, Ruan C, Chen M, Cao R, Sheng L, Chang N, Xu T, Zhao P, Liu X, Zhu F, Xiao Q, Gao S. Novel Insights into the Cardioprotective Effects of Calcitriol in Myocardial Infarction. Cells 2022; 11:1676. [PMID: 35626713 PMCID: PMC9139780 DOI: 10.3390/cells11101676] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 05/10/2022] [Accepted: 05/14/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Increasing evidence indicates that vitamin D deficiency negatively affects the cardiovascular system. Here we studied the therapeutic effects of calcitriol in myocardial infarction (MI) and investigated its underlying mechanisms. METHODS A MI model of Kun-ming mice induced by left anterior descending coronary artery ligation was utilized to study the potential therapeutic effects of calcitriol on MI. AC16 human cardiomyocyte-like cells treated with TNF-α were used for exploring the mechanisms that underlie the cardioprotective effects of calcitriol. RESULTS We observed that calcitriol reversed adverse cardiovascular function and cardiac remodeling in post-MI mice. Mechanistically, calcitriol suppressed MI-induced cardiac inflammation, ameliorated cardiomyocyte death, and promoted cardiomyocyte proliferation. Specifically, calcitriol exerted these cellular effects by upregulating Vitamin D receptor (VDR). Increased VDR directly interacted with p65 and retained p65 in cytoplasm, thereby dampening NF-κB signaling and suppressing inflammation. Moreover, up-regulated VDR was translocated into nuclei where it directly bound to IL-10 gene promoters to activate IL-10 gene transcription, further inhibiting inflammation. CONCLUSION We provide new insights into the cellular and molecular mechanisms underlying the cardioprotective effects of calcitriol, and we present comprehensive evidence to support the preventive and therapeutic effects of calcitriol on MI.
Collapse
Affiliation(s)
- Simin Yang
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China; (S.Y.); (C.R.); (M.C.); (R.C.); (L.S.); (N.C.); (T.X.); (P.Z.)
| | - Chunmiao Wang
- Department of Cardiology, The First Affiliated Hospital, Anhui Medical University, Hefei 230022, China;
| | - Chengshao Ruan
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China; (S.Y.); (C.R.); (M.C.); (R.C.); (L.S.); (N.C.); (T.X.); (P.Z.)
| | - Meiling Chen
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China; (S.Y.); (C.R.); (M.C.); (R.C.); (L.S.); (N.C.); (T.X.); (P.Z.)
- Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China;
| | - Ran Cao
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China; (S.Y.); (C.R.); (M.C.); (R.C.); (L.S.); (N.C.); (T.X.); (P.Z.)
- Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China;
| | - Liang Sheng
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China; (S.Y.); (C.R.); (M.C.); (R.C.); (L.S.); (N.C.); (T.X.); (P.Z.)
- Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China;
| | - Naiying Chang
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China; (S.Y.); (C.R.); (M.C.); (R.C.); (L.S.); (N.C.); (T.X.); (P.Z.)
- Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China;
| | - Tong Xu
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China; (S.Y.); (C.R.); (M.C.); (R.C.); (L.S.); (N.C.); (T.X.); (P.Z.)
| | - Peiwen Zhao
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China; (S.Y.); (C.R.); (M.C.); (R.C.); (L.S.); (N.C.); (T.X.); (P.Z.)
| | - Xuesheng Liu
- Department of Anesthesiology, The First Affiliated Hospital, Anhui Medical University, Hefei 230022, China;
| | - Fengqin Zhu
- Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, China;
| | - Qingzhong Xiao
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Shan Gao
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China; (S.Y.); (C.R.); (M.C.); (R.C.); (L.S.); (N.C.); (T.X.); (P.Z.)
| |
Collapse
|
7
|
Huanan G, Qiaoyan W, Shuping L. A smartphone-integrated dual-mode nanosensor based on Fe 3O 4@Au for rapid and highly selective detection of glutathione. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 271:120866. [PMID: 35033754 DOI: 10.1016/j.saa.2022.120866] [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: 12/04/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 06/14/2023]
Abstract
A simple, rapid and straightforward method for detecting reduced glutathione (GSH) was developed supported on smartphone analysis software package and a peroxide simulated catalyst nanoparticles (Fe3O4@Au) system. The nanocomposite was prepared by self-assembling technique, and the characterization was carried out using transmission electron microscopy, Fourier transforms infrared, and X-ray diffractometer. Fe3O4@Au materials have catalyzed the oxidation of a typical colorimetric substrate in the presence of H2O2, with the color changes from colorless to green oxidized. A smartphone with a free self-developed app referred to as "Color Capture" was accustomed live the RGB (red-greenblue) values of color intensity within the Fe3O4@Au system and computationally convert them GSH concentrations. The smartphone detection system showed high property and sensitivity of GSH detection. It gave a constant correlation (R2 = 0.9973) between the colour intensity of I and the GSH concentration, with a linear vary of 0-0.25 mmol/L, and a detection limit of 0.013 μmol/L. The results obtained were most consistent with the results obtained in ultraviolet spectrophotometry. The colorimetric system is based on smartphone analysis software developed to detect GSH in actual samples with potential application values.
Collapse
Affiliation(s)
- Guan Huanan
- College of Food Engineering, Harbin University of Commerce, Harbin 150076, People's Republic of China.
| | - Wu Qiaoyan
- College of Food Engineering, Harbin University of Commerce, Harbin 150076, People's Republic of China
| | - Liu Shuping
- College of Food Engineering, Harbin University of Commerce, Harbin 150076, People's Republic of China
| |
Collapse
|
8
|
Ling XX, Chen H, Fu BB, Ruan CS, Pana M, Zhou K, Fang ZR, Shao JT, Zhu FQ, Gao S. Xin-Ji-Er-Kang protects myocardial and renal injury in hypertensive heart failure in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 91:153675. [PMID: 34332285 DOI: 10.1016/j.phymed.2021.153675] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 05/27/2021] [Accepted: 07/14/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Xin-Ji-Er-Kang (XJEK) as a herbal formula of traditional Chinese medicine (TCM) has shown the protective effects on myocardial function as well as renal function in mouse models of myocardial infarction. HYPOTHESIS/PURPOSE We investigated the effects of XJEK on cardiovascular- and renal-function in a heart failure mouse model induced by high salt (HS) and the associated mechanisms. STUDY DESIGN For the purpose of assessing the effects of XJEK on a hypertensive heart failure model, mice were fed with 8% high salt diet. XJEK was administered by oral gavage for 8 weeks. Cardiovascular function parameters, renal function associated biomarkers and XJEK's impact on renin-angiotensin-aldosterone system (RAAS) activation were assessed. To determine the underlying mechanism, the calpain1/junctophilin-2 (JP2)/sarcoplasmic reticulum Ca2+ ATPase (SERCA2a) pathway was further studied in AC16 cells after angiotensin II-challenge or after calpastatin small interfering RNA (siRNA) transfection. RESULTS Mice on HS-diet exhibited hypertensive heart failure along with progressive kidney injury. Similar to fosinopril, XJEK ameliorated hypertension, cardiovascular-and renal- dysfunction in mice of HS-diet group. XJEK inhibited HS-induced activation of RAAS and reversed the abnormal expression pattern of calpain1and JP2 protein in heart tissues. XJEK significantly improved cell viability of angiotensin II-challenged AC16 cells. Moreover, XJEK's impact on calpain1/JP2 pathway was partly diminished in AC16 cells transfected with calpastatin siRNA. CONCLUSION XJEK was found to exert cardiovascular- and renal protection in HS-diet induced heart failure mouse model. XJEK inhibited HS-diet induced RAAS activation by inhibiting the activity and expression of calpain1 and protected the junctional membrane complex (JMC) in cardiomyocytes.
Collapse
Affiliation(s)
- Xin-Xin Ling
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China
| | - Hua Chen
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China
| | - Bei-Bei Fu
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China
| | - Cheng-Shao Ruan
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China
| | - Ming Pana
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China
| | - Kai Zhou
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China
| | - Zhi-Rui Fang
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China
| | - Jun-Tang Shao
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China.
| | - Feng-Qin Zhu
- Hefei Cancer Hospital, Chinese Academy of Science, Hefei 230032, China.
| | - Shan Gao
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
9
|
Wang Y, Zhen D, Fu D, Fu Y, Zhang X, Gong G, Wei C. 1, 8-cineole attenuates cardiac hypertrophy in heart failure by inhibiting the miR-206-3p/SERP1 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 91:153672. [PMID: 34385094 DOI: 10.1016/j.phymed.2021.153672] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 06/06/2021] [Accepted: 07/14/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND 1,8-Cineole (1,8-CIN) is a monoterpene found in diverse dietary and medicinal herbs that has been reported to be effective against cardiovascular diseases. PURPOSE The present research was designed to elucidate the treatment effects and the underlying mechanism of 1,8-CIN on heart failure (HF). METHOD An in vitro cardiac hypertrophy model and an in vivo heart failure (HF) model induced by isoprenaline (ISO) were established and treated with or without 1,8-CIN. In vitro miR-206-3p mimic or inhibitors were created. MiR-206-3p, SERP1 and related mRNAs or proteins were detected using qPCR or western blotting. Cell viability was tested by MTT assay, and apoptosis was measured using TUNEL assay, AO/EB assay and flow cytometry. Actin was stained with FITC-phalloidin. MiR-206-3p and related mRNAs or proteins in cardiac muscle tissues were measured using qPCR or western blotting, HE staining, Masson staining. RESULTS ISO subcutaneous injection increased cardiac hypertrophy, cytoplasmic vacuole formation, myofiber loss and fibrosis and decreased cardiomyocyte viability. 1,8-CIN treatment improved cardiomyocyte viability and reduced cardiac hypertrophy, cytoplasmic vacuole formation, myofibre loss and fibrosis. We found that 1,8-CIN attenuated apoptosis. We observed that expression of miR-206-3p was dramatically increased in ISO-exposed cardiomyocytes or ISO-treated rat hearts. MiR-206-3p was identified to target the 3'UTR of SERP1, resulting in the accumulation of un- or misfolded proteins, leading to endoplasmic reticulum (ER) stress. CONCLUSION These results suggest that 1,8-CIN reduces the apoptosis induced by ER stress through inhibiting miR-206-3p, which inhibits the expression of SERP1.
Collapse
Affiliation(s)
- Yu Wang
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, PR China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, P.R. China., Tongliao, Inner Mongolia, PR China
| | - Dong Zhen
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, PR China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, P.R. China., Tongliao, Inner Mongolia, PR China
| | - Danni Fu
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, PR China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, P.R. China., Tongliao, Inner Mongolia, PR China
| | - Yao Fu
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, PR China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, P.R. China., Tongliao, Inner Mongolia, PR China
| | - Xuan Zhang
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, PR China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, P.R. China., Tongliao, Inner Mongolia, PR China
| | - Guohua Gong
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, PR China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, P.R. China., Tongliao, Inner Mongolia, PR China; Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia, PR China.
| | - Chengxi Wei
- Medicinal Chemistry and Pharmacology Institute, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, PR China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, Inner Mongolia, P.R. China., Tongliao, Inner Mongolia, PR China.
| |
Collapse
|
10
|
Xie M, Tao W, Wu F, Wu K, Huang X, Ling G, Zhao C, Lv Q, Wang Q, Zhou X, Chen Y, Yuan Q, Chen Y. Anti-hypertensive and cardioprotective activities of traditional Chinese medicine-derived polysaccharides: A review. Int J Biol Macromol 2021; 185:917-934. [PMID: 34229020 DOI: 10.1016/j.ijbiomac.2021.07.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 02/05/2023]
Abstract
Cardiovascular diseases (CVDs), a leading cause of death in modern society, have become a major public health issue globally. Although numerous approaches have been proposed to reduce morbidity and mortality, the pursuit of pharmaceuticals with more preventive and/or therapeutic value remains a focus of attention. Being a vast treasure trove of natural drug molecules, Traditional Chinese Medicine (TCM) has a long history of clinical use in the prophylaxis and remedy of CVDs. Increasing lines of preclinical evidence have demonstrated the effectiveness of TCM-derived polysaccharides on hindering the progression of CVDs, e.g. hypertension, myocardial infarction. However, to the best of our knowledge, there are few reviews on the application of TCM-derived polysaccharides in combating CVDs. Hence, we provide an overview of primary literature on the anti-hypertensive and cardioprotective activities of herbal polysaccharides. Additionally, we also discuss the current limitations and propose a new hypothesis about how polysaccharides exert cardiovascular effects based on the metabolism of polysaccharides.
Collapse
Affiliation(s)
- Miaotian Xie
- Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Weili Tao
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Fengjia Wu
- Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Kunlin Wu
- Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Xiujie Huang
- Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Gensong Ling
- Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Chuanyi Zhao
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Qian Lv
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Qiongjin Wang
- Department of Pharmacology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Xianhuan Zhou
- Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Ying Chen
- Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Qin Yuan
- Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Yicun Chen
- Shantou University Medical College, Shantou 515041, Guangdong, China; Department of Pharmacology, Shantou University Medical College, Shantou 515041, Guangdong, China.
| |
Collapse
|
11
|
Zhang S, Zhang Y, Wang X, Wu L, Shen J, Gu M, Fang Z. Effects of Shenfu Qiangxin Drink on H 2O 2-induced oxidative stress, inflammation and apoptosis in neonatal rat cardiomyocytes and possible underlying mechanisms. Exp Ther Med 2021; 21:553. [PMID: 33850525 PMCID: PMC8027745 DOI: 10.3892/etm.2021.9985] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/26/2020] [Indexed: 12/13/2022] Open
Abstract
The aim of the present study was to investigate the effects of Shenfu Qiangxin Drink (SFQXD) on acute myocardial infarction (AMI) and identify the possible underlying mechanisms. Levels of reactive oxygen species (ROS) and inflammatory factors, including interleukin (IL)-6, IL-1β and tumor necrosis factor-α (TNF-α) in the blood samples of patients with AMI were measured using commercially available kits by visible spectrophotometry after SFQXD administration. The contents of phosphorylated (p-) forkhead box O3a (FOXO3a) was examined using an ELISA kit. In addition, a hydrogen peroxide (H2O2)-induced myocardial injury model was established in vitro using neonatal rat cardiomyocytes. Following treatment with SFQXD, the levels of intracellular ROS, cell apoptosis, oxidative stress- and inflammation-related markers were measured using commercially available kits by visible spectrophotometry. Additionally, western blot analysis was used to measure the expression of sirtuin-4 (SIRT4), p-FOXO3a, acetylated FOXO3a (ace-FOXO3a) and apoptosis-related genes (Bcl-2, Bax, BIM and cleaved caspase-3). Subsequently, to investigate the possible underlying regulatory mechanisms, SIRT4 expression was silenced by transfection with small hairpin RNA against SIRT4, following which changes in the extent of oxidative stress, inflammation and apoptosis were assessed. The levels of ROS and interleukin (IL)-1β were found to be significantly reduced, whilst FOXO3a phosphorylation was markedly increased following administration with SFQXD. In vitro, SFQXD dose-dependently inhibited H2O2-induced oxidative stress, inflammation and apoptosis in neonatal rat cardiomyocytes. In addition, FOXO3a phosphorylation was markedly upregulated whilst FOXO3a acetylation was downregulated following treatment of H2O2-induced primary neonatal cardiomyocytes with SFQXD. SIRT4 knockdown also markedly reversed the effects of SFQXD on oxidative stress, inflammation and apoptosis in neonatal rat cardiomyocytes. In conclusion, these findings demonstrated that SFQXD may alleviate oxidative stress-induced myocardial injury by potentially regulating SIRT4/FOXO3a signaling, suggesting that SFQXD may be of clinical value for the treatment of AMI.
Collapse
Affiliation(s)
- Sujie Zhang
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
- Department of Cardiology, Jiangsu Province Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Yiyan Zhang
- Department of Cardiology, Jiangsu Province Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Xindong Wang
- Department of Cardiology, Jiangsu Province Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Lixing Wu
- Department of Cardiology, Jiangsu Province Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Jianping Shen
- Department of Cardiology, Jiangsu Province Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Minglin Gu
- Department of Cardiology, Jiangsu Province Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Zhuyuan Fang
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| |
Collapse
|
12
|
Effect of high-intensity interval training on cardiac structure and function in rats with acute myocardial infarct. Biomed Pharmacother 2020; 131:110690. [PMID: 32890969 DOI: 10.1016/j.biopha.2020.110690] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Exercise training is beneficial for cardiac rehabilitation. Nevertheless, few study focused on the role of high-intensity interval training (HIIT) in cardiac repair. The current study aimed to elucidate the effect of HIIT on cardiac rehabilitation and the involved mechanisms after acute myocardial infarction (MI). METHODS A total of 65 male rats underwent coronary ligation or sham operation and were randomly assigned to 4 groups: sham (n = 10), sedentary (MI-Sed, n = 12), moderate-intensity continuous training (MI-MCT, n = 12) and HIIT (MI-HIIT, n = 12). One week after MI induction, adaptive training starts follow by formal training. After the experiment, cardiac functions were determined by echocardiography and hemodynamic measurements. Changes in infarct size, collagen accumulation, myofibroblasts, angiogenesis, inflammation level, endothelin-1 (ET-1), and renin-angiotensin-aldosterone system (RAAS) activities were measured. Data were analyzed by one-way ANOVA. RESULTS After MI, cardiac structure and function were significantly deteriorated. However, post-MI HIIT for 8 weeks had significantly ameliorated left ventricular end-diastolic pressure (LVEDP), LV systolic pressure (LVSP), and maximum peak velocities of relaxation (-dP/dtmax). Moreover, it preserved cardiac functions, reduced infarct size, protected the myocardium structure, increased angiogenesis and decreased the myofibroblasts and collagen accumulation. HIIT for 4 weeks had no effect on LVEDP, -dP/dtmax, infarct size and angiogenesis. Additionally, it induced inflammation response and repressed ET-1 and RAAS activities were found in myocardium and peripheral circulation after HIIT. CONCLUSION Our results suggested that post-MI HIIT had a positive role in cardiac repair, which might be linked with the induction of inflammation and inhibition of ET-1 and RAAS activities.
Collapse
|
13
|
Unveiling the Role of Inflammation and Oxidative Stress on Age-Related Cardiovascular Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1954398. [PMID: 32454933 PMCID: PMC7232723 DOI: 10.1155/2020/1954398] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/12/2020] [Accepted: 04/03/2020] [Indexed: 12/14/2022]
Abstract
The global population above 60 years has been growing exponentially in the last decades, which is accompanied by an increase in the prevalence of age-related chronic diseases, highlighting cardiovascular diseases (CVDs), such as hypertension, atherosclerosis, and heart failure. Aging is the main risk factor for these diseases. Such susceptibility to disease is explained, at least in part, by the increase of oxidative stress, in which it damages cellular components such as proteins, DNA, and lipids. In addition, the chronic inflammatory process in aging “inflammaging” also contributes to cell damage, creating a stressful environment which drives to the development of CVDs. Taken together, it is possible to identify the molecular connection between oxidative stress and the inflammatory process, especially by the crosstalk between the transcription factors Nrf-2 and NF-κB which are mediated by redox signalling and are involved in aging. Therapies that control this process are key targets in the prevention/combat of age-related CVDs. In this review, we show the basics of inflammation and oxidative stress, including the crosstalk between them, and the implications on age-related CVDs.
Collapse
|
14
|
Chen J, Xue R, Li L, Xiao LL, Shangguan J, Zhang W, Bai X, Liu G, Li L. Panax Notoginseng Saponins Protect Cardiac Myocytes Against Endoplasmic Reticulum Stress and Associated Apoptosis Through Mediation of Intracellular Calcium Homeostasis. Front Pharmacol 2019; 10:1013. [PMID: 31616293 PMCID: PMC6764115 DOI: 10.3389/fphar.2019.01013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 08/08/2019] [Indexed: 12/30/2022] Open
Abstract
Endoplasmic reticulum (ER) stress has been demonstrated to play important roles in the pathogenesis of various cardiovascular diseases. The ER stress pathway is therefore a promising therapeutic target in cardiovascular disease. Although Panax notoginseng saponins (PNS) are one of the patent medicines that are traditionally used to treat cardiovascular disorders, their effects on ER stress in cardiac myocytes remain unexploited so far. This study investigates the effects of PNS on ER stress and its associated cell apoptosis along with the related mechanism in cardiac myocytes. PNS compounds were identified via high-performance liquid chromatograph (HPLC) assay. PNS-pretreated H9c2 cells, HL-1 cells, and primary cultured neonatal rat cardiomyocytes were stimulated with thapsigargin (TG) to induce ER stress response and apoptosis. ER stress response was tested by immunofluorescence or immunoblot of the ER protein chaperones—calnexin, binding immunoglobulin protein (BiP) and the C/EBP homologous protein (CHOP). Cell viability was tested by methyl thiazolyl tetrazolium (MTT) assay. Cell apoptosis was detected by immunoblot of Cleaved caspase-3 and flow cytometry analysis of Annexin V/propidium iodide (PI) staining. Cytosolic, mitochondrial, and ER calcium dynamics were investigated by calcium imaging. Moreover, a ryanodine receptor type-2 (RyR2) overexpression stable cell line was generated to verify the mechanism of RyR2 involved in PNS in the inhibition of ER stress and cell apoptosis. We demonstrate here that PNS protected cardiac myocytes from ER stress response and associated cell death in a concentration-dependent manner. Importantly, PNS reduced the elevation of cytosolic calcium, mitochondria calcium, as well as ER calcium in response to either TG or histamine treatment. PNS protection in ER stress was regulated by RyR2 expression. In summary, PNS protection against TG-induced ER stress response and its associated cell apoptosis in cardiac myocytes is calcium dependent. Through the regulation of ER calcium release mediated by RyR2, a novel mechanism for PNS in the prevention of cardiovascular diseases is thereby identified.
Collapse
Affiliation(s)
- Jun Chen
- Vasculocardiology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rui Xue
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Li
- Vasculocardiology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Li Xiao
- Vasculocardiology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiahong Shangguan
- Vasculocardiology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenjing Zhang
- Vasculocardiology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xueyang Bai
- Vasculocardiology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Gangqiong Liu
- Vasculocardiology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ling Li
- Vasculocardiology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
15
|
Yan J, Yan J, Wang Y, Ling Y, Song X, Wang S, Liu H, Liu Q, Zhang Y, Yang P, Wang X, Chen A. Spermidine-enhanced autophagic flux improves cardiac dysfunction following myocardial infarction by targeting the AMPK/mTOR signalling pathway. Br J Pharmacol 2019; 176:3126-3142. [PMID: 31077347 PMCID: PMC6692641 DOI: 10.1111/bph.14706] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 04/17/2019] [Accepted: 04/29/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Spermidine, a natural polyamine, is abundant in mammalian cells and is involved in cell growth, proliferation, and regeneration. Recently, oral spermidine supplements were cardioprotective in age-related cardiac dysfunction, through enhancing autophagic flux. However, the effect of spermidine on myocardial injury and cardiac dysfunction following myocardial infarction (MI) remains unknown. EXPERIMENTAL APPROACH We determined the effects of spermidine in a model of MI, Sprague-Dawley rats with permanent ligation of the left anterior descending artery, and in cultured neonatal rat cardiomyocytes (NRCs) exposed to angiotensin II (Ang II). Cardiac function in vivo was assessed with echocardiography. In vivo and in vitro studies used histological and immunohistochemical techniques, along with western blots. KEY RESULTS Spermidine improved cardiomyocyte viability and decreased cell necrosis in NRCs treated with angiotensin II. In rats post-MI, spermidine reduced infarct size, improved cardiac function, and attenuated myocardial hypertrophy. Spermidine also suppressed the oxidative damage and inflammatory cytokines induced by MI. Moreover, spermidine enhanced autophagic flux and decreased apoptosis both in vitro and in vivo. The protective effects of spermidine on cardiomyocyte apoptosis and cardiac dysfunction were abolished by the autophagy inhibitor chloroquine, indicating that spermidine exerted cardioprotective effects at least partly through promoting autophagic flux, by activating the AMPK/mTOR signalling pathway. CONCLUSIONS AND IMPLICATIONS Our findings suggest that spermidine improved MI-induced cardiac dysfunction by promoting AMPK/mTOR-mediated autophagic flux.
Collapse
Affiliation(s)
- Jing Yan
- Laboratory of Heart Center and Department of Cardiology, Heart Center, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
- Laboratory of Heart Center, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular DiseaseGuangzhouChina
- Laboratory of Heart Center, Sino‐Japanese Cooperation Platform for Translational Research in Heart FailureGuangzhouChina
| | - Jian‐Yun Yan
- Laboratory of Heart Center and Department of Cardiology, Heart Center, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
- Laboratory of Heart Center, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular DiseaseGuangzhouChina
- Laboratory of Heart Center, Sino‐Japanese Cooperation Platform for Translational Research in Heart FailureGuangzhouChina
| | - Yu‐Xi Wang
- Laboratory of Heart Center and Department of Cardiology, Heart Center, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
- Laboratory of Heart Center, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular DiseaseGuangzhouChina
- Laboratory of Heart Center, Sino‐Japanese Cooperation Platform for Translational Research in Heart FailureGuangzhouChina
| | - Yuan‐Na Ling
- Laboratory of Heart Center and Department of Cardiology, Heart Center, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
- Laboratory of Heart Center, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular DiseaseGuangzhouChina
- Laboratory of Heart Center, Sino‐Japanese Cooperation Platform for Translational Research in Heart FailureGuangzhouChina
| | - Xu‐Dong Song
- Laboratory of Heart Center and Department of Cardiology, Heart Center, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
- Laboratory of Heart Center, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular DiseaseGuangzhouChina
- Laboratory of Heart Center, Sino‐Japanese Cooperation Platform for Translational Research in Heart FailureGuangzhouChina
| | - Si‐Yi Wang
- Laboratory of Heart Center and Department of Cardiology, Heart Center, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
- Laboratory of Heart Center, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular DiseaseGuangzhouChina
- Laboratory of Heart Center, Sino‐Japanese Cooperation Platform for Translational Research in Heart FailureGuangzhouChina
| | - Hai‐Qiong Liu
- Laboratory of Heart Center and Department of Cardiology, Heart Center, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
- Laboratory of Heart Center, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular DiseaseGuangzhouChina
- Laboratory of Heart Center, Sino‐Japanese Cooperation Platform for Translational Research in Heart FailureGuangzhouChina
| | - Qi‐Cai Liu
- Laboratory of Heart Center and Department of Cardiology, Heart Center, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
- Laboratory of Heart Center, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular DiseaseGuangzhouChina
- Laboratory of Heart Center, Sino‐Japanese Cooperation Platform for Translational Research in Heart FailureGuangzhouChina
| | - Ya Zhang
- Department of CardiologyXiangdong Affiliated Hospital of Hunan Normal UniversityZhuzhouHunanChina
| | - Ping‐Zhen Yang
- Laboratory of Heart Center and Department of Cardiology, Heart Center, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
- Laboratory of Heart Center, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular DiseaseGuangzhouChina
- Laboratory of Heart Center, Sino‐Japanese Cooperation Platform for Translational Research in Heart FailureGuangzhouChina
| | - Xian‐Bao Wang
- Laboratory of Heart Center and Department of Cardiology, Heart Center, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
- Laboratory of Heart Center, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular DiseaseGuangzhouChina
- Laboratory of Heart Center, Sino‐Japanese Cooperation Platform for Translational Research in Heart FailureGuangzhouChina
| | - Ai‐Hua Chen
- Laboratory of Heart Center and Department of Cardiology, Heart Center, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
- Laboratory of Heart Center, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular DiseaseGuangzhouChina
- Laboratory of Heart Center, Sino‐Japanese Cooperation Platform for Translational Research in Heart FailureGuangzhouChina
| |
Collapse
|
16
|
Lian FZ, Cheng P, Ruan CS, Ling XX, Wang XY, Pan M, Chen ML, Shen AZ, Gao S. Xin-Ji-Er-Kang ameliorates kidney injury following myocardial infarction by inhibiting oxidative stress via Nrf2/HO-1 pathway in rats. Biomed Pharmacother 2019; 117:109124. [DOI: 10.1016/j.biopha.2019.109124] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/04/2019] [Accepted: 06/12/2019] [Indexed: 12/11/2022] Open
|
17
|
Xin-Ji-Er-Kang Alleviates Myocardial Infarction-Induced Cardiovascular Remodeling in Rats by Inhibiting Endothelial Dysfunction. BIOMED RESEARCH INTERNATIONAL 2019; 2019:4794082. [PMID: 31341899 PMCID: PMC6614977 DOI: 10.1155/2019/4794082] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/29/2019] [Accepted: 05/26/2019] [Indexed: 12/17/2022]
Abstract
The present study was designed to elucidate the beneficial effects of XJEK on myocardial infarction (MI) in rats, especially through the amelioration of endothelial dysfunction (ED). 136 Sprague-Dawley rats were randomized into 13 groups: control group for 0wk (n = 8); sham groups for 2, 4, and 6 weeks (wk); MI groups for 2, 4, and 6 wk; MI+XJEK groups for 2, 4, and 6w k; MI+Fosinopril groups for 2, 4, and 6 wk (n = 8~10). In addition, 8 rats were treated for Evans blue staining and Tetrazolium chloride (TTC) staining to determine the infarct size. Cardiac function, ECG, and cardiac morphological changes were examined. Colorimetric analysis was employed to detect nitric oxide (NO), and enzyme-linked immunosorbent assay (ELISA) was applied to determine N-terminal probrain natriuretic peptide (NT-ProBNP), endothelin-1 (ET-1), angiotensin II (Ang II), asymmetric dimethylarginine (ADMA), tetrahydrobiopterin (BH4), and endothelial NO synthase (eNOS) content. The total eNOS and eNOS dimer/(dimer+monomer) ratios in cardiac tissues were detected by Western blot. We found that administration of XJEK markedly ameliorated cardiovascular remodeling (CR), which was manifested by decreased HW/BW ratio, CSA, and less collagen deposition after MI. XJEK administration also improved cardiac function by significant inhibition of the increased hemodynamic parameters in the early stage and by suppression of the decreased hemodynamic parameters later on. XJEK also continuously suppressed the increased NT-ProBNP content in the serum of MI rats. XJEK improved ED with stimulated eNOS activities, as well as upregulated NO levels, BH4 content, and eNOS dimer/(dimer+monomer) ratio in the cardiac tissues. XJEK downregulated ET-1, Ang II, and ADMA content obviously compared to sham group. In conclusion, XJEK may exert the protective effects on MI rats and could continuously ameliorate ED and reverse CR with the progression of MI over time.
Collapse
|
18
|
Du Y, Ge Y, Xu Z, Aa N, Gu X, Meng H, Lin Z, Zhu D, Shi J, Zhuang R, Wu X, Wang X, Yang Z. Hypoxia-Inducible Factor 1 alpha (HIF-1α)/Vascular Endothelial Growth Factor (VEGF) Pathway Participates in Angiogenesis of Myocardial Infarction in Muscone-Treated Mice: Preliminary Study. Med Sci Monit 2018; 24:8870-8877. [PMID: 30531686 PMCID: PMC6295139 DOI: 10.12659/msm.912051] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Angiogenesis plays a crucial role in myocardial infarction (MI) treatment by ameliorating myocardial remodeling, thus improving cardiac function and preventing heart failure. Muscone has been reported to have beneficial effects on cardiac remodeling in MI mice. However, the effects of muscone on angiogenesis in MI mice and its underlying mechanisms remain unknown. MATERIAL AND METHODS Mice were randomly divided into sham, MI, and MI+muscone groups. The MI mouse model was established by ligating the left anterior descending coronary artery. Mice in the sham group received the same procedure except for ligation. Mice were administered muscone or an equivalent volume of saline for 4 consecutive weeks. Cardiac function was evaluated by echocardiograph after MI for 2 and 4 weeks. Four weeks later, all mice were sacrificed and Masson's trichrome staining was used to assess myocardial fibrosis. Isolectin B4 staining was applied to evaluate the angiogenesis in mouse hearts. Immunohistochemistry, Western blot analysis, and quantitative real-time polymerase chain reaction (qPCR) were performed to analyze expression levels of HIF-1a and its downstream genes. RESULTS Compared with the MI group, muscone treatment significantly improved cardiac function and reduced myocardial fibrosis. Moreover, muscone enhanced angiogenesis in the peri-infarct region and p-VEGFR2 expression in the vascular endothelial cells. Western blot analysis and qPCR showed that muscone upregulated expression levels of HIF-1a and VEGFA. CONCLUSIONS Muscone improved cardiac function in MI mice through augmented angiogenesis. The potential mechanism of muscone treatment in regulating angiogenesis of MI mice was upregulating expression levels of HIF-1α and VEGFA.
Collapse
Affiliation(s)
- Yingqiang Du
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Yingbin Ge
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Zhihui Xu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Nan Aa
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Xin Gu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Haoyu Meng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| | - Zhou Lin
- Department of Cardiology, Wuxi No. 3 People's Hospital Affiliated to Nantong University, Wuxi, Jiangsu, China (mainland)
| | - Dongxiao Zhu
- Department of Cardiac Ultrasound, Wuxi No. 3 People's Hospital Affiliated to Nantong University, Wuxi, Jiangsu, China (mainland)
| | - Jingjing Shi
- Department of Cardiology, Wuxi No. 3 People's Hospital Affiliated to Nantong University, Wuxi, Jiangsu, China (mainland)
| | - Ruijuan Zhuang
- Department of Cardiology, Wuxi No. 3 People's Hospital Affiliated to Nantong University, Wuxi, Jiangsu, China (mainland)
| | - Xueming Wu
- Department of Cardiology, Wuxi No. 3 People's Hospital Affiliated to Nantong University, Wuxi, Jiangsu, China (mainland)
| | - Xiaoyan Wang
- Department of Cardiology, Wuxi No. 3 People's Hospital Affiliated to Nantong University, Wuxi, Jiangsu, China (mainland)
| | - Zhijian Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China (mainland)
| |
Collapse
|
19
|
Polunina EA, Voronina LP, Popov EA, Polunina OS. Analysis of levels of oxidative stress markers depending on the left ventricular ejection fraction in patients with chronic heart failure. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2018. [DOI: 10.15829/1728-8800-2018-5-34-39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Aim.To study and analyze the levels of oxidative stress (OS) markers (malondialdehyde (MDA), superoxide dismutase (SOD), advanced oxidation protein products (AOPPs)) depending on the left ventricular ejection fraction (LVEF) and functional class (FC) in patients with chronic heart failure (CHF).Material and methods.We examined 60 somatically healthy individuals and 345 patients with CHF, which were divided into three main groups depending on the LVEF and subgroups depending on FC. The levels of OS markers were determined in blood serum — MDA, SOD and AOPPs.Results.In the group of patients with preserved LVEF and FC II-IV CHF, levels of MDA and AOPPs were statistically significantly higher, and the SOD level was lower compared to the control group. In the group of patients with moderately reduced and reduced LVEF, the levels of MDA and AOPPs were statistically significantly higher, and SOD activity was lower compared with the control group and the group of patients with CHF and preserved LVEF. In patients with CHF with higher FC, there was a statistically significant increase of MDA and AOPPs levels and decrease of SOD activity. The most pronounced changes in the levels of above-mentioned markers were recorded in patients with reduced LVEF. According to the correlation analysis a direct relationship between the levels of markers of the OS and clinical manifestations of the disease was found.Conclusion.Changes in levels of MDA, SOD and AOPPs in patients with CHF were detected already in the early stages of the disease compared with the control group. In patients with higher FC CHF and preserved, moderately reduced and reduced LVEF, a statistically significant increase in the levels of MDA and AOPPs and a decrease of SOD activity were observed. The most pronounced changes in the levels of the markers were indicated in patients with reduced LVEF.
Collapse
Affiliation(s)
- E. A. Polunina
- Astrakhan State Medical University of the Ministry of Health
| | - L. P. Voronina
- Astrakhan State Medical University of the Ministry of Health
| | - E. A. Popov
- Astrakhan State Medical University of the Ministry of Health
| | - O. S. Polunina
- Astrakhan State Medical University of the Ministry of Health
| |
Collapse
|
20
|
Xu H, Li F. miR‑127 aggravates myocardial failure by promoting the TGF‑β1/Smad3 signaling. Mol Med Rep 2018; 18:4839-4846. [PMID: 30272299 PMCID: PMC6236281 DOI: 10.3892/mmr.2018.9514] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 07/23/2018] [Indexed: 12/24/2022] Open
Abstract
Myocardial failure has a negative impact on the quality of human life. MicroRNA (miR) expression abnormalities lead to the development of many pathological conditions, including myocardial failure, and therefore the present study primarily focused on the investigation of the functions of miR‑127 in the development of myocardial failure. The miR‑127 expression levels in serum samples from patients with myocardial failure were examined. Oil red O staining was used to analyze the characteristics of the myocardium of the patients. Immunohistochemistry was used to detect fatty acid synthase (FASN), stearoyl‑CoA desaturase‑1 (SCD1) and mitochondrial brown fat uncoupling protein 1 (UCP1) protein expression in the myocardium of the patients. Furthermore, C57BL/6J (B6) mice were induced with 15 mg/kg of doxorubicin. Echocardiography was used to detect the histopathological alterations of the myocardial cells by comparison of the myocardial tissues from the myocardial failure animal model and normal C57BL/6 mice. Reverse transcription‑quantitative polymerase chain reaction was used to detect the expression levels of miR‑127 following different induction periods and immunohistochemistry was used to detect the expression of transforming growth factor‑β1 (TGF‑β1) and mothers against decapentaplegic homolog 3 (Smad3). Immunofluorescence was used to detect the expression alterations TGF‑β1/Smad3 when miR‑127 overexpression or inhibition was established. The results of the present study indicated that myocardial failure resulted in an upregulated expression of miR‑127 and severe fat accumulation. FASN, SCD1 and UCP1 were highly expressed in the myocardial failure group compared with the control. Abdominal artery contraction and the ejection fraction were significantly reduced in the DOX‑induced B6 mice. The cardiomyocytes became hypertrophic, and left ventricular systolic pressure and left ventricular maximum ejection pressure were altered following DOX induction in B6 mice. The results confirmed that miR‑127 regulates the expression of TGF‑β1/Smad3. The potential pathological mechanism of the effect of miR‑127 may be based on the upregulation of the TGF‑β1/Smad3 signaling pathway.
Collapse
Affiliation(s)
- Hainian Xu
- Department of Cardiovascular Internal Medicine, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| | - Fengmei Li
- Department of Internal Medicine, Weifang Zuoshan Central Hospital, Weifang, Shandong 261041, P.R. China
| |
Collapse
|