1
|
Wang XC, Zhou Y, Chen HX, Hou HT, He GW, Yang Q. ER stress modulates Kv1.5 channels via PERK branch in HL-1 atrial myocytes: Relevance to atrial arrhythmogenesis and the effect of tetramethylpyrazine. Heliyon 2024; 10:e37767. [PMID: 39318794 PMCID: PMC11420496 DOI: 10.1016/j.heliyon.2024.e37767] [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: 04/19/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/26/2024] Open
Abstract
Endoplasmic reticulum (ER) stress is implicated in cardiac arrhythmia whereas the associated mechanisms remain inadequately understood. Kv1.5 channels are essential for atrial repolarization. Whether ER stress affects Kv1.5 channels is unknown. This study aimed to elucidate the response of Kv1.5 channels to ER stress by clarifying the unfolded protein response (UPR) branch responsible for the channel modulation. In addition, the effect of tetramethylpyrazine (TMP) on Kv1.5 channels was studied. Patch clamp and western-blot results revealed that exposure of HL-1 atrial myocytes to ER stress inducer tunicamycin upregulates Kv1.5 expression, increases Kv1.5 channel current (I Kur ) (14.91 ± 1.11 vs. 6.11 ± 1.31 pA/pF, P < 0.001), and shortened action potential duration (APD) (APD90: 82.79 ± 5.25 vs.121.11 ± 6.72 ms, P < 0.01), which could be reverted by ER stress inhibitors. Blockade of the PERK branch while not IRE1 and ATF6 branches of UPR downregulated Kv1.5 expression, accompanied by a decreased I Kur (9.03 ± 0.99 pA/pF) and a prolonged APD90 (113.69 ± 4.41 ms) (P < 0.01). PERK-mediated increases of Kv1.5 expression and I Kur were also observed in HL-1 cells incubated with thapsigargin. TMP suppressed the enhancement of I Kur (10.52 ± 0.97 vs. 17.52 ± 2.25 pA/pF, P < 0.05), prevented the shortening of APD (APD90: 110.16 ± 5.36 vs. 84.84 ± 4.58 ms, P < 0.05), and inhibited the upregulation of Kv1.5 triggered by ER stress. Our study suggests that ER stress induces upregulation and activation of Kv1.5 channels in atrial myocytes through the PERK branch of UPR. TMP prevents Kv1.5 upregulation/activation and the resultant APD shortening by inhibiting ER stress. These results may shed light on the mechanisms of atrial arrhythmogenesis and the antiarrhythmic effect of the traditional Chinese herb TMP.
Collapse
Affiliation(s)
- Xiang-Chong Wang
- Institute of Cardiovascular Diseases & Department of Cardiac Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, 300457, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, China
- Department of Pharmacology, Hebei Higher Education Institute Applied Technology Research Center on TCM Formula Preparation, Hebei International Cooperation Center for Ion channel Function and Innovative Traditional Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050091, China
- School of Medicine, Nankai University, Tianjin, 300457, China
| | - Yang Zhou
- Institute of Cardiovascular Diseases & Department of Cardiac Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, 300457, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, China
| | - Huan-Xin Chen
- Institute of Cardiovascular Diseases & Department of Cardiac Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, 300457, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, China
| | - Hai-Tao Hou
- Institute of Cardiovascular Diseases & Department of Cardiac Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, 300457, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, China
| | - Guo-Wei He
- Institute of Cardiovascular Diseases & Department of Cardiac Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, 300457, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, China
| | - Qin Yang
- Institute of Cardiovascular Diseases & Department of Cardiac Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College & Tianjin University, Tianjin, 300457, China
- Tianjin Key Laboratory of Molecular Regulation of Cardiovascular Diseases and Translational Medicine, Tianjin, China
| |
Collapse
|
2
|
Hong WM, Xie YW, Zhao MY, Yu TH, Wang LN, Xu WY, Gao S, Cai HB, Guo Y, Zhang F. Vasoprotective Effects of Hyperoside against Cerebral Ischemia/Reperfusion Injury in Rats: Activation of Large-Conductance Ca 2+-Activated K + Channels. Neural Plast 2023; 2023:5545205. [PMID: 37609123 PMCID: PMC10442186 DOI: 10.1155/2023/5545205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 06/29/2023] [Accepted: 07/19/2023] [Indexed: 08/24/2023] Open
Abstract
Hyperoside (Hyp), a kind of Chinese herbal medicine, exerts multiple therapeutic effects on many diseases. However, the role and mechanisms of Hyp in vascular pathophysiology in ischemic stroke need to be further established. The study aimed to investigate the role of (large-conductance Ca2+-activated K+) BK channels on the vasoprotection of Hyp against cerebral ischemia and reperfusion (I/R) injury in rats. The concentration gradient of Hyp was pretreated in both the middle cerebral artery occlusion and reperfusion model and oxygen-glucose deprivation/reoxygenation (OGD/R) model of primary vascular smooth muscle cells (VSMCs) in rats. A series of indicators were detected, including neurological deficit score, infarct volume, malondialdehyde (MDA), superoxide dismutase (SOD), cerebral blood flow (CBF), cell viability, membrane potential, and BK channels α- and β1-subunits expression. The results showed that Hyp significantly reduced infarct volume and ameliorated neurological dysfunction in I/R-injured rats. Besides, the effects of I/R-induced reduction of BK channels α- and β1-subunits expression were significantly reversed by Hyp in endothelial-denudated cerebral basilar arteries. Furthermore, the protective effect against I/R-induced increases of MDA and reduction of SOD as well as CBF induced by Hyp was significantly reversed by iberiotoxin (IbTX). In OGD/R-injured VSMCs, downregulated cellular viability and BK channels β1-subunits expression were remarkably reversed by Hyp. However, neither OGD/R nor Hyp affected BK channels α-subunits expression, and Hyp failed to induced hyperpolarization of VSMCs. Moreover, the protective effect against OGD/R-induced reduction of cell viability and SOD level and increases of MDA production induced by Hyp was significantly reversed by IbTX in VSMCs. The study indicates that Hyp has the therapeutic potential to improve vascular outcomes, and the mechanism is associated with suppressing oxidative stress and improving CBF through upregulating BK channels.
Collapse
Affiliation(s)
- Wen-Ming Hong
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
- School of Nursing, Anhui Medical University, Hefei 230032, China
- Open Project of Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei 230032, China
| | - Yue-Wu Xie
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China
| | - Meng-Yu Zhao
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Tian-Hang Yu
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Li-Na Wang
- School of Nursing, Anhui Medical University, Hefei 230032, China
| | - Wan-Yan Xu
- School of Nursing, Anhui Medical University, Hefei 230032, China
| | - Shen Gao
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Hua-Bao Cai
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Yan Guo
- Department of Pharmacology, Anhui Medical University, Hefei 230032, China
| | - Fang Zhang
- School of Nursing, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
3
|
Wu C, Duan X, Wang X, Wang L. Advances in the role of epigenetics in homocysteine-related diseases. Epigenomics 2023; 15:769-795. [PMID: 37718931 DOI: 10.2217/epi-2023-0207] [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] [Indexed: 09/19/2023] Open
Abstract
Homocysteine has a wide range of biological effects. However, the specific molecular mechanism of its pathogenicity is still unclear. The diseases induced by hyperhomocysteinemia (HHcy) are called homocysteine-related diseases. Clinical treatment of HHcy is mainly through folic acid and B-complex vitamins, which are not effective in reducing the associated end point events. Epigenetics is the alteration of heritable genes caused by DNA methylation, histone modification, noncoding RNAs and chromatin remodeling without altering the DNA sequence. In recent years the role of epigenetics in homocysteine-associated diseases has been gradually discovered. This article summarizes the latest evidence on the role of epigenetics in HHcy, providing new directions for its prevention and treatment.
Collapse
Affiliation(s)
- Chengyan Wu
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Xulei Duan
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Xuehui Wang
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Libo Wang
- The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| |
Collapse
|
4
|
Zhang Q, Chen Y, Wang Q, Wang Y, Feng W, Chai L, Liu J, Li D, Chen H, Qiu Y, Shen N, Shi X, Xie X, Li M. HMGB1-induced activation of ER stress contributes to pulmonary artery hypertension in vitro and in vivo. Respir Res 2023; 24:149. [PMID: 37268944 DOI: 10.1186/s12931-023-02454-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 05/18/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND HMGB1 and ER stress have been considered to participate in the progression of pulmonary artery hypertension (PAH). However, the molecular mechanism underlying HMGB1 and ER stress in PAH remains unclear. This study aims to explore whether HMGB1 induces pulmonary artery smooth muscle cells (PASMCs) functions and pulmonary artery remodeling through ER stress activation. METHODS Primary cultured PASMCs and monocrotaline (MCT)-induced PAH rats were applied in this study. Cell proliferation and migration were determined by CCK-8, EdU and transwell assay. Western blotting was conducted to detect the protein levels of protein kinase RNA-like endoplasmic reticulum kinase (PERK), activating transcription factor-4 (ATF4), seven in absentia homolog 2 (SIAH2) and homeodomain interacting protein kinase 2 (HIPK2). Hemodynamic measurements, immunohistochemistry staining, hematoxylin and eosin staining were used to evaluate the development of PAH. The ultrastructure of ER was observed by transmission electron microscopy. RESULTS In primary cultured PASMCs, HMGB1 reduced HIPK2 expression through upregulation of ER stress-related proteins (PERK and ATF4) and subsequently increased SIAH2 expression, which ultimately led to PASMC proliferation and migration. In MCT-induced PAH rats, interfering with HMGB1 by glycyrrhizin, suppression of ER stress by 4-phenylbutyric acid or targeting SIAH2 by vitamin K3 attenuated the development of PAH. Additionally, tetramethylpyrazine (TMP), as a component of traditional Chinese herbal medicine, reversed hemodynamic deterioration and vascular remodeling by targeting PERK/ATF4/SIAH2/HIPK2 axis. CONCLUSIONS The present study provides a novel insight to understand the pathogenesis of PAH and suggests that targeting HMGB1/PERK/ATF4/SIAH2/HIPK2 cascade might have potential therapeutic value for the prevention and treatment of PAH.
Collapse
Affiliation(s)
- Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Yuqian Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Qingting Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Yan Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Wei Feng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Limin Chai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Jin Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Danyang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Huan Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Yuanjie Qiu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Nirui Shen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Xiangyu Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Xinming Xie
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
5
|
Soluble epoxide hydrolase and TRPC3 channels jointly contribute to homocysteine-induced cardiac hypertrophy: Interrelation and regulation by C/EBPβ. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166643. [PMID: 36669577 DOI: 10.1016/j.bbadis.2023.166643] [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: 09/09/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023]
Abstract
OBJECTIVES Studies in certain cardiac hypertrophy models suggested the individual role of soluble epoxide hydrolase (sEH) and canonical transient receptor potential 3 (TRPC3) channels, however, whether they jointly mediate hypertrophic process remains unexplored. Hyperhomocysteinemia promotes cardiac hypertrophy while the involvement of sEH and TRPC3 channels remains unknown. This study aimed to explore the role of, and interrelation between sEH and TRPC3 channels in homocysteine-induced cardiac hypertrophy. METHODS Rats were fed methionine-enriched diet to induce hyperhomocysteinemia. H9c2 cells and neonatal rat cardiomyocytes were incubated with homocysteine. Cardiac hypertrophy was evaluated by echocardiography, histological examination, immunofluorescence imaging, and expressions of hypertrophic markers. Epoxyeicosatrienoic acids (EETs) were determined by ELISA. TRPC3 current was recorded by patch-clamp. Gene promotor activity was measured using dual-luciferase reporter assay. RESULTS Inhibition of sEH by 1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) urea (TPPU) reduced ventricular mass, lowered the expression of hypertrophic markers, decreased interstitial collagen deposition, and improved cardiac function in hyperhomocysteinemic rats, associated with restoration of EETs levels in myocardium. TPPU or knockdown of sEH suppressed TRPC3 transcription and translation as well as TRPC3 current that were enhanced by homocysteine. Exogenous 11,12-EET inhibited homocysteine-induced TRPC3 expression and cellular hypertrophy. Silencing C/EBPβ attenuated, while overexpressing C/EBPβ promoted homocysteine-induced hypertrophy and expressions of sEH and TRPC3, resulting respectively from inhibition or activation of sEH and TRPC3 gene promoters. CONCLUSIONS sEH and TRPC3 channels jointly contribute to homocysteine-induced cardiac hypertrophy. Homocysteine transcriptionally activates sEH and TRPC3 genes through a common regulatory element C/EBPβ. sEH activation leads to an upregulation of TRPC3 channels via a 11,12-EET-dependent manner.
Collapse
|
6
|
Jiang L, Li J, Reilly S, Xin H, Guo N, Zhang X. Role of organellar Ca2+-activated K+ channels in disease development. Life Sci 2023; 316:121433. [PMID: 36708987 DOI: 10.1016/j.lfs.2023.121433] [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: 11/09/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 01/27/2023]
Abstract
The organellar Ca2+-activated K+ channels share a similar ability to transfer the alteration of Ca2+ concentration to membrane conductance of potassium. Multiple effects of Ca2+-activated K+ channels on cell metabolism and complex signaling pathways during organ development have been explored. The organellar Ca2+-activated K+ channels are able to control the ionic equilibrium and are always associated with oxidative stress in different organelles and the whole cells. Some drugs targeting Ca2+-activated K+ channels have been tested for various diseases in clinical trials. In this review, the known roles of organellar Ca2+-activated K+ channels were described, and their effects on different diseases, particularly on diabetes, cardiovascular diseases, and neurological diseases were discussed. It was attempted to summarize the currently known operational modes with the involvement of organellar Ca2+-activated K+ channels. This review may assist scholars to more comprehensively understand organellar Ca2+-activated K+ channels and related diseases.
Collapse
Affiliation(s)
- Lan Jiang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Jiawei Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Svetlana Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Hong Xin
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Nan Guo
- Department of Pharmacy, Minhang hospital, Fudan University, Shanghai, China.
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Ginsenoside Rb1 from Panax notoginseng Suppressed TNF-α-Induced Matrix Metalloproteinase-9 via the Suppression of Double-Strand RNA-Dependent Protein Kinase (PKR)/NF-κB Pathway. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27228050. [PMID: 36432152 PMCID: PMC9692425 DOI: 10.3390/molecules27228050] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/12/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022]
Abstract
Chronic inflammation is commonly accompanied by the stimulation of matrix metalloproteinases (MMPs) production and the degradation of the extracellular matrix. The overexpression of MMP-9 (Gelatinase B) highly participates in the progression of pathetic cardiac remodeling and liver cancer metastasis. Panax notoginseng (Burkill) F. H. Chen (Sanqi), a widely used traditional Chinese medicinal herb, shows myocardial protective and anti-tumor effects. In this study, we examined the inhibitory effect of different PNG extracts on tumor necrosis factor (TNF)-α-induced MMP-9 expression in cardiac myoblast H9c2 cells. Using a bioassay-guided fractionation scheme, the most active extract was fractionated by silica gel column chromatography and high-performance liquid chromatography until an active compound was obtained. The compound was identified as Ginsenoside Rb1 by nuclear magnetic resonance. Ginsenoside Rb1 inhibited TNF-α-induced MMP-9 production in both H9c2 and liver carcinoma HepG-2 cells. Interestingly, it did not affect the MMP-2 (Gelatinase A) level and the cell proliferation of the two cell lines. The inhibitory effects of Ginsenoside Rb1 may be due to its modulation of double-strand RNA-dependent protein kinase and nuclear factor kappa B signaling pathways. The results reveal the potential use of Ginsenoside Rb1 for the treatment of inflammatory and MMP-9-related cardiac remodeling and metastasis of hepatocellular carcinomas.
Collapse
|
8
|
Tetramethylpyrazine: A review on its mechanisms and functions. Biomed Pharmacother 2022; 150:113005. [PMID: 35483189 DOI: 10.1016/j.biopha.2022.113005] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 11/21/2022] Open
Abstract
Ligusticum chuanxiong Hort (known as Chuanxiong in China, CX) is one of the most widely used and long-standing medicinal herbs in China. Tetramethylpyrazine (TMP) is an alkaloid and one of the active components of CX. Over the past few decades, TMP has been proven to possess several pharmacological properties. It has been used to treat a variety of diseases with excellent therapeutic effects. Here, the pharmacological characteristics and molecular mechanism of TMP in recent years are reviewed, with an emphasis on the signal-regulation mechanism of TMP. This review shows that TMP has many physiological functions, including anti-oxidant, anti-inflammatory, and anti-apoptosis properties; autophagy regulation; vasodilation; angiogenesis regulation; mitochondrial damage suppression; endothelial protection; reduction of proliferation and migration of vascular smooth muscle cells; and neuroprotection. At present, TMP is used in treating cardiovascular, nervous, and digestive system conditions, cancer, and other conditions and has achieved good curative effects. The therapeutic mechanism of TMP involves multiple targets, multiple pathways, and bidirectional regulation. TMP is, thus, a promising drug with great research potential.
Collapse
|
9
|
Sun W, Zhou Y, Xue H, Hou H, He G, Yang Q. Endoplasmic reticulum stress mediates homocysteine-induced hypertrophy of cardiac cells through activation of cyclic nucleotide phosphodiesterase 1C. Acta Biochim Biophys Sin (Shanghai) 2022; 54:388-399. [PMID: 35538034 PMCID: PMC9828163 DOI: 10.3724/abbs.2022009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Although the association of elevated homocysteine level with cardiac hypertrophy has been reported, the molecular mechanisms by which homocysteine induces cardiac hypertrophy remain inadequately understood. In this study we aim to uncover the roles of cyclic nucleotide phosphodiesterase 1 (PDE1) and endoplasmic reticulum (ER) stress and their relationship to advance the mechanistic understanding of homocysteine-induced cardiac cell hypertrophy. H9c2 cells and primary neonatal rat cardiomyocytes are exposed to homocysteine with or without ER stress inhibitor TUDCA or PDE1-specific inhibitor Lu AF58027, or transfected with siRNAs targeting PDE1 isoforms prior to homocysteine-exposure. Cell surface area is measured and ultrastructure is examined by transmission electron microscopy. Hypertrophic markers, PDE1 isoforms, and ER stress molecules are detected by q-PCR and western blot analysis. Intracellular cGMP and cAMP are measured by ELISA. The results show that homocysteine causes the enlargement of H9c2 cells, increases the expressions of hypertrophic markers β-MHC and ANP, upregulates PDE1A and PDE1C, promotes the expressions of ER stress molecules, and causes ER dilatation and degranulation. TUDCA and Lu AF58027 downregulate β-MHC and ANP, and alleviate cell enlargement. TUDCA decreases PDE1A and PDE1C levels. Silencing of PDE1C inhibits homocysteine-induced hypertrophy, whereas PDE1A knockdown has minor effect. Both cAMP and cGMP are decreased after homocysteine-exposure, while only cAMP is restored by Lu AF58027 and TUDCA. TUDCA and Lu AF58027 also inhibit cell enlargement, downregulate ANP, β-MHC and PDE1C, and enhance cAMP level in homocysteine-exposed primary cardiomyocytes. ER stress mediates homocysteine-induced hypertrophy of cardiac cells via upregulating PDE1C expression Cyclic nucleotide, especially cAMP, is the downstream mediator of the ER stress-PDE1C signaling axis in homocysteine-induced cell hypertrophy.
Collapse
Affiliation(s)
- Wentao Sun
- Center for Basic Medical Research & Department of Cardiovascular SurgeryTEDA International Cardiovascular HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300457China,The Institute of Cardiovascular DiseasesTianjin UniversityTianjin300457China
| | - Yang Zhou
- Center for Basic Medical Research & Department of Cardiovascular SurgeryTEDA International Cardiovascular HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300457China,The Institute of Cardiovascular DiseasesTianjin UniversityTianjin300457China
| | - Hongmei Xue
- Center for Basic Medical Research & Department of Cardiovascular SurgeryTEDA International Cardiovascular HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300457China,The Institute of Cardiovascular DiseasesTianjin UniversityTianjin300457China,Department of PhysiologyHebei Medical UniversityShijiazhuang050017China
| | - Haitao Hou
- Center for Basic Medical Research & Department of Cardiovascular SurgeryTEDA International Cardiovascular HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300457China,The Institute of Cardiovascular DiseasesTianjin UniversityTianjin300457China
| | - Guowei He
- Center for Basic Medical Research & Department of Cardiovascular SurgeryTEDA International Cardiovascular HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300457China,The Institute of Cardiovascular DiseasesTianjin UniversityTianjin300457China,Drug Research and Development CenterWannan Medical CollegeWuhu241002China,Department of SurgeryOregon Health and Science UniversityPortlandOR97239-3098USA
| | - Qin Yang
- Center for Basic Medical Research & Department of Cardiovascular SurgeryTEDA International Cardiovascular HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300457China,The Institute of Cardiovascular DiseasesTianjin UniversityTianjin300457China
| |
Collapse
|
10
|
Hu S, Wang S, He J, Bian Y. Tetramethylpyrazine alleviates endoplasmic reticulum stress‑activated apoptosis and related inflammation in chondrocytes. Mol Med Rep 2021; 25:12. [PMID: 34779501 PMCID: PMC8600404 DOI: 10.3892/mmr.2021.12528] [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: 05/22/2021] [Accepted: 09/07/2021] [Indexed: 11/25/2022] Open
Abstract
Excessive apoptosis of chondrocytes and degradation of the extracellular matrix (ECM) contribute to the typical pathological characteristics of osteoarthritis (OA). Various studies have reported that tetramethylpyrazine (TMP) protects against multiple disorders by inhibiting inflammation and oxidative stress. The present study investigated the effects of TMP on chondrocytes and evaluated the associated mechanisms. To determine the effect of TMP on OA and the underlying mechanisms, chondrocytes were incubated with TMP and IL-1β or thapsigargin (TG) Western blotting assays were performed to examine the expression levels of endoplasmic reticulum (ER) stress proteins, and TUNEL staining, fluorescence immunostaining and reverse transcription-quantitative PCR were used to determine the apoptosis levels, and catabolic and inflammatory factors. It was found that TMP protected chondrocytes by suppressing IL-1β-induced expression of glucose-regulated protein 78 (GRP78) and CHOP (an apoptotic protein). TMP regulated the TG-mediated upregulated expression of GRP78 and CHOP in the chondrocytes of rats, as well as markedly suppressed levels of ER stress-triggered inflammatory cytokines (TNF-α and IL-6). Furthermore, TMP modulated TG-induced changes in ECM catabolic metabolism in rat chondrocytes. Collectively, TMP alleviated ER-stress-activated apoptosis and related inflammation in chondrocytes, indicating that it has therapeutic potential for the treatment of OA.
Collapse
Affiliation(s)
- Shuai Hu
- Joint and Traumatology Department, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Sheng Wang
- Joint and Traumatology Department, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Jie He
- Joint and Traumatology Department, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Yangyang Bian
- Department of Trauma Medical Center, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570100, P.R. China
| |
Collapse
|
11
|
Yang Q, Shen F, Zhang F, Bai X, Zhang Y, Zhang H. The combination of two natural medicines, Chuanxiong and Asarum: A review of the chemical constituents and pharmacological activities. JOURNAL OF CHEMICAL RESEARCH 2021. [DOI: 10.1177/17475198211039130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Traditional Chinese medicine has been clinically used in China for many years, with experimental studies and clinical trials having demonstrated that it is safe and valid. Among many traditional natural medicines, Chuanxiong and Asarum have been proven to be effective in the treatment of relieving pain. Actually, as well as analgesic, they have common attributes, such as anti-inflammatory, cardiovascular benefits, and anticancer activities, with volatile oils being their major components. Furthermore, Chuanxiong and Asarum have been combined as drug pairs in the same prescription for thousands of years, with examples being Chuanxiong Chatiao San and Chuanxiongxixintang. More interestingly, their combination has better therapeutic effects on diseases than a single drug. After the combination of Chuanxiong and Asarum forms a blend, a series of changes take place in their chemical components, such as the contents of the main active ingredients, ferulic acid and ligustilide, increased significantly after this progress. At the same time, the pharmacological effects of the combination appearing to be more powerful, such as synergistic analgesic. This review focuses on the chemical constituents and pharmacological activities of Chuanxiong, Asarum, and Chuanxiong Asarum compositions.
Collapse
Affiliation(s)
- Qingcheng Yang
- College of Pharmacy, Dali University, Dali, P.R. China
- Department of Pharmacy, The First People’s Hospital of Kunming, Kunming, P.R. China
| | - Fangli Shen
- College of Pharmacy, Dali University, Dali, P.R. China
- Department of Pharmacy, The First People’s Hospital of Kunming, Kunming, P.R. China
| | - Fengqin Zhang
- College of Pharmacy, Dali University, Dali, P.R. China
| | - Xue Bai
- College of Pharmacy, Dali University, Dali, P.R. China
| | - Yanru Zhang
- College of Pharmacy, Dali University, Dali, P.R. China
| | - Haizhu Zhang
- College of Pharmacy, Dali University, Dali, P.R. China
- Western Yunnan Traditional Chinese Medicine and Ethnic Drug Engineering Center, Dali, P.R. China
| |
Collapse
|
12
|
Sun WT, Xue HM, Hou HT, Chen HX, Wang J, He GW, Yang Q. Homocysteine alters vasoreactivity of human internal mammary artery by affecting the K Ca channel family. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:625. [PMID: 33987323 PMCID: PMC8106027 DOI: 10.21037/atm-20-6821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Background Hyperhomocysteinemia is an independent risk factor for atherosclerotic heart disease. We previously demonstrated that disruption of calcium-activated potassium (KCa) channel activity is involved in homocysteine-induced dilatory dysfunction of porcine coronary arteries. Recently we reported that the KCa channel family, including large-, intermediate-, and small-conductance KCa (BKCa, IKCa, and SKCa) subtypes, are abundantly expressed in human internal mammary artery (IMA). In this study, we further investigated whether homocysteine affects the expression and functionality of the KCa channel family in this commonly used graft for coronary artery bypass surgery (CABG). Methods Residual IMA segments obtained from patients undergoing CABG were studied in a myograph for the role of KCa subtypes in both vasorelaxation and vasoconstriction. The expression and distribution of KCa subtypes were detected by Western blot and immunohistochemistry. Results Both the BKCa channel activator NS1619 and the IKCa/SKCa channel activator NS309 evoked significant IMA relaxation. Homocysteine exposure suppressed NS1619-induced relaxation whereas showed no influence on NS309-induced response. Blockade of BKCa but not IKCa and SKCa subtypes significantly suppressed acetylcholine-induced relaxation and enhanced U46619-induced contraction. Homocysteine compromised the vasodilating activity of the BKCa subtype in IMA, associated with a lowered protein level of the BKCa β1-subunit. Homocysteine potentiated the role of IKCa and SKCa subtypes in mediating endothelium-dependent relaxation without affecting the expression of these channels. Conclusions Homocysteine reduces the expression of BKCa β1-subunit and compromises the vasodilating activity of BKCa channels in IMA. Unlike BKCa, IKCa and SKCa subtypes are unessential for IMA vasoregulation, whereas the loss of BKCa functionality in hyperhomocysteinemia enhances the role of IKCa and SKCa subtypes in mediating endothelial dilator function. Targeting BKCa channels may form a strategy to improve the postoperative graft performance in CABG patients with hyperhomocysteinemia who receive IMA grafting.
Collapse
Affiliation(s)
- Wen-Tao Sun
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.,School of Medicine, Nankai University, Tianjin, China
| | - Hong-Mei Xue
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.,School of Medicine, Nankai University, Tianjin, China.,Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Hai-Tao Hou
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Huan-Xin Chen
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Jun Wang
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Guo-Wei He
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.,School of Pharmacy, Wannan Medical College, Wuhu, China.,Department of Surgery, Oregon Health and Science University, Portland, Oregon, USA
| | - Qin Yang
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| |
Collapse
|
13
|
Ma Q, Ma H, Xu F, Wang X, Sun W. Microfluidics in cardiovascular disease research: state of the art and future outlook. MICROSYSTEMS & NANOENGINEERING 2021; 7:19. [PMID: 34567733 PMCID: PMC8433381 DOI: 10.1038/s41378-021-00245-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/08/2021] [Accepted: 01/16/2021] [Indexed: 05/21/2023]
Abstract
Due to extremely severe morbidity and mortality worldwide, it is worth achieving a more in-depth and comprehensive understanding of cardiovascular diseases. Tremendous effort has been made to replicate the cardiovascular system and investigate the pathogenesis, diagnosis and treatment of cardiovascular diseases. Microfluidics can be used as a versatile primary strategy to achieve a holistic picture of cardiovascular disease. Here, a brief review of the application of microfluidics in comprehensive cardiovascular disease research is presented, with specific discussions of the characteristics of microfluidics for investigating cardiovascular diseases integrally, including the study of pathogenetic mechanisms, the development of accurate diagnostic methods and the establishment of therapeutic treatments. Investigations of critical pathogenetic mechanisms for typical cardiovascular diseases by microfluidic-based organ-on-a-chip are categorized and reviewed, followed by a detailed summary of microfluidic-based accurate diagnostic methods. Microfluidic-assisted cardiovascular drug evaluation and screening as well as the fabrication of novel delivery vehicles are also reviewed. Finally, the challenges with and outlook on further advancing the use of microfluidics technology in cardiovascular disease research are highlighted and discussed.
Collapse
Affiliation(s)
- Qingming Ma
- School of Pharmacy, Qingdao University, Qingdao, 266071 China
| | - Haixia Ma
- Center for Prenatal Diagnosis, Zibo Maternal and Child Health Care Hospital, Zibo, 255000 China
| | - Fenglan Xu
- Department of Clinical Pharmacy, The Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001 China
| | - Xinyu Wang
- Institute of Thermal Science and Technology, Shandong University, Jinan, 250061 China
| | - Wentao Sun
- Center for Basic Medical Research, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & School of Medicine, Nankai University, Tianjin, 300457 China
| |
Collapse
|
14
|
2,3,5,6-Tetramethylpyrazine protects retinal photoreceptors against endoplasmic reticulum stress by modulating ATF4-mediated inhibition of PRP aggregation. J Mol Med (Berl) 2021; 99:383-402. [PMID: 33409554 DOI: 10.1007/s00109-020-02017-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 11/23/2020] [Accepted: 11/25/2020] [Indexed: 01/17/2023]
Abstract
Endoplasmic reticulum (ER) stress is a common threat to photoreceptors during the pathogenesis of chronic retinopathies and often results in irreversible visual impairment. 2,3,5,6-Tetramethylpyrazine (TMP), which possesses many beneficial pharmacological activities, is a potential drug that could be used to protect photoreceptors. In the present study, we found that the cellular growth rate of 661 W cells cultured under low glucose conditions was lower than that of control cells, while the G2/M phase of the cell cycle was longer. We further found that the mitochondrial membrane potential (ΔΨm) was lower and that ER stress factor expression was increased in 661 W cells cultured under low glucose conditions. TMP reversed these trends. Visual function and cell counts in the outer nuclear layer (ONL) were low and the TUNEL-positive rate in the ONL was high in a C3H mouse model of spontaneous retinal degeneration. Similarly, visual function was decreased, and the TUNEL-positive rate in the ONL was increased in fasted C57/BL6j mice compared with control mice. On the other hand, ER stress factor expression was found to be increased in the retinas of both mouse models, as shown by reverse transcription real-time PCR (RT-qPCR) and western blotting. TMP reversed the physiological and molecular biological variations observed in both mouse models, and ATF4 expression was enhanced again. Further investigation by using western blotting illustrated that the proportion of insoluble prion protein (PRP) versus soluble PRP was reduced both in vitro and in vivo. Taken together, these results suggest that TMP increased the functions of photoreceptors by alleviating ER stress in vitro and in vivo, and the intrinsic mechanism was the ATF4-mediated inhibition of PRP aggregation. TMP may potentially be used clinically as a therapeutic agent to attenuate the functional loss of photoreceptors during the pathogenesis of chronic retinopathies. KEY MESSAGES: • Already known: TMP is a beneficial drug mainly used in clinic to enhance organ functions, and the intrinsic mechanism is still worthy of exploring. • New in the study: We discovered that TMP ameliorated retinal photoreceptors function via ER stress alleviation, which was promoted by ATF4-mediated inhibition of PRP aggregation. • Application prospect: In prospective clinical practices, TMP may potentially be used in the clinic as a therapeutic agent to attenuate the photoreceptors functional reduction in chronic retinopathies.
Collapse
|
15
|
Sun WT, Hou HT, Chen HX, Xue HM, Wang J, He GW, Yang Q. Calcium-activated potassium channel family in coronary artery bypass grafts. J Thorac Cardiovasc Surg 2019; 161:e399-e409. [PMID: 31928817 DOI: 10.1016/j.jtcvs.2019.11.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/06/2019] [Accepted: 11/08/2019] [Indexed: 01/18/2023]
Abstract
OBJECTIVES We examined the expression, distribution, and contribution to vasodilatation of the calcium-activated potassium (KCa) channel family in the commonly used coronary artery bypass graft internal thoracic artery (ITA) and saphenous vein (SV) to understand the role of large conductance KCa (BKCa), intermediate-conductance KCa (IKCa), and small-conductance KCa (SKCa) channel subtypes in graft dilating properties determined by endothelium-smooth muscle interaction that is essential to the postoperative performance of the graft. METHODS Real-time polymerase chain reaction and western blot were employed to detect the messenger RNA and protein level of KCa channel subtypes. Distribution of KCa channel subtypes was examined by immunohistochemistry. KCa subtype-mediated vasorelaxation was studied using wire myography. RESULTS Both ITA and SV express all KCa channel subtypes with each subtype distributed in both endothelium and smooth muscle. ITA and SV do not differ in the overall expression level of each KCa channel subtype, corresponding to comparable relaxant responses to respective subtype activators. In ITA, BKCa is more abundantly expressed in smooth muscle than in endothelium, whereas SKCa exhibits more abundance in the endothelium. In comparison, SV shows even distribution of KCa channel subtypes in the 2 layers. The BKCa subtype in the KCa family plays a significant role in vasodilatation of ITA, whereas its contribution in SV is quite limited. CONCLUSIONS KCa family is abundantly expressed in ITA and SV. There are differences between these 2 grafts in the abundance of KCa channel subtypes in the endothelium and the smooth muscle. The significance of the BKCa subtype in vasodilatation of ITA may suggest the potential of development of BKCa modulators for the prevention and treatment of ITA spasm during/after coronary artery bypass graft surgery.
Collapse
Affiliation(s)
- Wen-Tao Sun
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Hai-Tao Hou
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Huan-Xin Chen
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Hong-Mei Xue
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jun Wang
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Guo-Wei He
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China; School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China; Department of Surgery, Oregon Health and Science University, Portland, Ore
| | - Qin Yang
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China.
| |
Collapse
|
16
|
Imbalance of Homocysteine and H 2S: Significance, Mechanisms, and Therapeutic Promise in Vascular Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7629673. [PMID: 31885816 PMCID: PMC6893243 DOI: 10.1155/2019/7629673] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 10/16/2019] [Indexed: 12/28/2022]
Abstract
While the role of hyperhomocysteinemia in cardiovascular pathogenesis continuously draws attention, deficiency of hydrogen sulfide (H2S) has been growingly implicated in cardiovascular diseases. Generation of H2S is closely associated with the metabolism of homocysteine via key enzymes such as cystathionine β-synthase (CBS) and cystathionine γ-lyase (CSE). The level of homocysteine and H2S is regulated by each other. Metabolic switch in the activity of CBS and CSE may occur with a resultant operating preference change of these enzymes in homocysteine and H2S metabolism. This paper presented an overview regarding (1) linkage between the metabolism of homocysteine and H2S, (2) mutual regulation of homocysteine and H2S, (3) imbalance of homocysteine and H2S in cardiovascular disorders, (4) mechanisms underlying the protective effect of H2S against homocysteine-induced vascular injury, and (5) the current status of homocysteine-lowering and H2S-based therapies for cardiovascular disease. The metabolic imbalance of homocysteine and H2S renders H2S/homocysteine ratio a potentially reliable biomarker for cardiovascular disease and development of drugs or interventions targeting the interplay between homocysteine and H2S to maintain the endogenous balance of these two molecules may hold an even bigger promise for management of vascular disorders than targeting homocysteine or H2S alone.
Collapse
|