1
|
Wang F, Wang B, Gu X, Li X, Liu X, Li B. Chelerythrine triggers the prolongation of QT interval and induces cardiotoxicity by promoting the degradation of hERG channels. J Biol Chem 2025; 301:108023. [PMID: 39608718 PMCID: PMC11721429 DOI: 10.1016/j.jbc.2024.108023] [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: 02/07/2024] [Revised: 11/06/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024] Open
Abstract
Cardiotoxicity is a serious adverse reaction during drug treatment. The cardiac human ether-a-go-go-related gene (hERG) channels play a crucial role in driving cardiac action potential repolarization and are a key target for drug-induced cardiac toxicity. Chelerythrine (CHE) has anticancer effects on various human cancer cells. But little is known about its drug safety currently. The purpose of this study is to explore the key mechanism of cardiac toxicity induced by CHE under pathological conditions. CHE and hypoxia prolonged QT interval and action potential duration compared with control group in guinea pigs, as measured by BL-420S biological acquisition and processing system in conjunction with optical mapping technology. hERG current was measured by patch-clamp technique, and the interaction between ubiquitin molecules and hERG channels was assessed using immunoprecipitation method at the molecular level. The colocalization of proteins and the function of lysosomes were determined via confocal laser scanning microscopy. Further research indicates that CHE enhances the ubiquitination process of hERG proteins by catalyzing the formation of K63 ubiquitin chains, the ubiquitination modification disrupts hERG channel homeostasis, and promotes the degradation of the channel. Mechanistically, CHE accelerates the degradation of hERG channels through lysosomes via HDAC6, which may easily induce cardiotoxicity caused by prolonged QT interval under hypoxic conditions.
Collapse
Affiliation(s)
- Fang Wang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Baoqiang Wang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xiwei Gu
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xiaoxu Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xinyu Liu
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Baoxin Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province, China.
| |
Collapse
|
2
|
Liao J, Yang Z, Yang J, Lin H, Chen B, Fu H, Lin X, Lu B, Gao F. Investigating the cardiotoxicity of N-n-butyl haloperidol iodide: Inhibition mechanisms on hERG channels. Toxicology 2024; 508:153916. [PMID: 39128488 DOI: 10.1016/j.tox.2024.153916] [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: 07/07/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
The human Ether-à-go-go-Related Gene (hERG) encodes a protein responsible for forming the alpha subunit of the IKr channel, which plays a crucial role in cardiac repolarization. The proper functioning of hERG channels is paramount in maintaining a normal cardiac rhythm. Inhibition of these channels can result in the prolongation of the QT interval and potentially life-threatening arrhythmias. Cardiotoxicity is a primary concern in the field of drug development. N-n-Butyl haloperidol iodide (F2), a derivative of haloperidol, has been investigated for its therapeutic potential. However, the impact of this compound on cardiac toxicity, specifically on hERG channels, remains uncertain. This study employs computational and experimental methodologies to examine the inhibitory mechanisms of F2 on hERG channels. Molecular docking and molecular dynamics simulations commonly used techniques in computational biology to predict protein-ligand complexes' binding interactions and stability. In the context of the F2-hERG complex, these methods can provide valuable insights into the potential binding modes and strength of interaction between F2 and the hERG protein. On the other hand, electrophysiological assays are experimental techniques used to characterize the extent and nature of hERG channel inhibition caused by various compounds. By measuring the electrical activity of the hERG channel in response to different stimuli, these assays can provide important information about the functional effects of ligand binding to the channel. The study's key findings indicate that F2 interacts with the hERG channel by forming hydrogen bonding, π-cation interactions, and hydrophobic forces. This interaction leads to the inhibition of hERG currents in a concentration-dependent manner, with an IC50 of 3.75 μM. The results presented in this study demonstrate the potential cardiotoxicity of F2 and underscore the significance of considering hERG channel interactions during its clinical development. This study aims to provide comprehensive insights into the interaction between F2 and hERG, which will may guid us in the safe use of F2 and in the development of new derivatives with high efficiency while low toxicity.
Collapse
Affiliation(s)
- Jilin Liao
- Department of Pharmacy, the Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, China; Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Zhenyu Yang
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Jinhua Yang
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Hailing Lin
- Department of Pharmacy, the Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Bingxuan Chen
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Hongbo Fu
- Department of Pharmacy, the Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Xiaojie Lin
- Department of Pharmacy, the Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Binger Lu
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong 515041, China; Department of Pharmacy, the First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, China.
| | - Fenfei Gao
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong 515041, China.
| |
Collapse
|
3
|
Sun X, Zhou Q, Xiao C, Mao C, Liu Y, Chen G, Song Y. Role of post-translational modifications of Sp1 in cardiovascular diseases. Front Cell Dev Biol 2024; 12:1453901. [PMID: 39252788 PMCID: PMC11381397 DOI: 10.3389/fcell.2024.1453901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024] Open
Abstract
Specific protein 1 (Sp1) is pivotal in sustaining baseline transcription as well as modulating cell signaling pathways and transcription factors activity. Through interactions with various proteins, especially transcription factors, Sp1 controls the expression of target genes, influencing numerous biological processes. Numerous studies have confirmed Sp1's significant regulatory role in the pathogenesis of cardiovascular disorders. Post-translational modifications (PTMs) of Sp1, such as phosphorylation, ubiquitination, acetylation, glycosylation, SUMOylation, and S-sulfhydration, can enhance or modify its transcriptional activity and DNA-binding stability. These modifications also regulate Sp1 expression across different cell types. Sp1 is crucial in regulating non-coding gene expression and the activity of proteins in response to pathophysiological stimuli. Understanding Sp1 PTMs advances our knowledge of cell signaling pathways in controlling Sp1 stability during cardiovascular disease onset and progression. It also aids in identifying novel pharmaceutical targets and biomarkers essential for preventing and managing cardiovascular diseases.
Collapse
Affiliation(s)
- Xutao Sun
- Department of Synopsis of the Golden Chamber, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qi Zhou
- Department of Pharmacology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chengpu Xiao
- Department of Typhoid, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Caiyun Mao
- Department of Pharmacology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ying Liu
- The Second Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Guozhen Chen
- Department of Pediatrics, Yantai Yuhuangding Hospital, Shandong, China
| | - Yunjia Song
- Department of Pharmacology, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
4
|
Ding J, Fayyaz AI, Ding Y, Liang D, Luo M. Role of Specificity Protein 1 (SP1) in Cardiovascular Diseases: Pathological Mechanisms and Therapeutic Potentials. Biomolecules 2024; 14:807. [PMID: 39062521 PMCID: PMC11274404 DOI: 10.3390/biom14070807] [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: 06/12/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
In mammals, specificity protein 1 (SP1) was the first Cys2-His2 zinc finger transcription factor to be isolated within the specificity protein and Krüppel-like factor (Sp/KLF) gene family. SP1 regulates gene expression by binding to Guanine-Cytosine (GC)-rich sequences on promoter regions of target genes, affecting various cellular processes. Additionally, the activity of SP1 is markedly influenced by posttranslational modifications, such as phosphorylation, acetylation, glycosylation, and proteolysis. SP1 is implicated in the regulation of apoptosis, cell hypertrophy, inflammation, oxidative stress, lipid metabolism, plaque stabilization, endothelial dysfunction, fibrosis, calcification, and other pathological processes. These processes impact the onset and progression of numerous cardiovascular disorders, including coronary heart disease, ischemia-reperfusion injury, cardiomyopathy, arrhythmia, and vascular disease. SP1 emerges as a potential target for the prevention and therapeutic intervention of cardiac ailments. In this review, we delve into the biological functions, pathophysiological mechanisms, and potential clinical implications of SP1 in cardiac pathology to offer valuable insights into the regulatory functions of SP1 in heart diseases and unveil novel avenues for the prevention and treatment of cardiovascular conditions.
Collapse
Affiliation(s)
- Jie Ding
- School of Medicine, Tongji University, Shanghai 200092, China; (J.D.); (D.L.)
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Aminah I. Fayyaz
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; (A.I.F.); (Y.D.)
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; (A.I.F.); (Y.D.)
| | - Dandan Liang
- School of Medicine, Tongji University, Shanghai 200092, China; (J.D.); (D.L.)
- State Key Laboratory of Cardiovascular Diseases, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
- Shanghai Arrhythmia Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Ming Luo
- School of Medicine, Tongji University, Shanghai 200092, China; (J.D.); (D.L.)
| |
Collapse
|
5
|
Qi X, Yu X, Wei L, Jiang H, Dong J, Li H, Wei Y, Zhao L, Deng W, Guo W, Hu X, Li T. Novel α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptor (AMPAR) potentiator LT-102: A promising therapeutic agent for treating cognitive impairment associated with schizophrenia. CNS Neurosci Ther 2024; 30:e14713. [PMID: 38615362 PMCID: PMC11016348 DOI: 10.1111/cns.14713] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 03/07/2024] [Accepted: 03/23/2024] [Indexed: 04/16/2024] Open
Abstract
AIMS We aimed to evaluate the potential of a novel selective α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptor (AMPAR) potentiator, LT-102, in treating cognitive impairments associated with schizophrenia (CIAS) and elucidating its mechanism of action. METHODS The activity of LT-102 was examined by Ca2+ influx assays and patch-clamp in rat primary hippocampal neurons. The structure of the complex was determined by X-ray crystallography. The selectivity of LT-102 was evaluated by hERG tail current recording and kinase-inhibition assays. The electrophysiological characterization of LT-102 was characterized by patch-clamp recording in mouse hippocampal slices. The expression and phosphorylation levels of proteins were examined by Western blotting. Cognitive function was assessed using the Morris water maze and novel object recognition tests. RESULTS LT-102 is a novel and selective AMPAR potentiator with little agonistic effect, which binds to the allosteric site formed by the intradimer interface of AMPAR's GluA2 subunit. Treatment with LT-102 facilitated long-term potentiation in mouse hippocampal slices and reversed cognitive deficits in a phencyclidine-induced mouse model. Additionally, LT-102 treatment increased the protein level of brain-derived neurotrophic factor and the phosphorylation of GluA1 in primary neurons and hippocampal tissues. CONCLUSION We conclude that LT-102 ameliorates cognitive impairments in a phencyclidine-induced model of schizophrenia by enhancing synaptic function, which could make it a potential therapeutic candidate for CIAS.
Collapse
Affiliation(s)
- Xueyu Qi
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain‐Machine Integration, State Key Laboratory of Brain‐Machine IntelligenceZhejiang UniversityHangzhouChina
- NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang UniversityHangzhouChina
| | - Xueli Yu
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain‐Machine Integration, State Key Laboratory of Brain‐Machine IntelligenceZhejiang UniversityHangzhouChina
- NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang UniversityHangzhouChina
| | - Long Wei
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
| | - Han Jiang
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
| | - Jiangwen Dong
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
| | - Hongxing Li
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
| | - Yingying Wei
- The Psychiatric Laboratory, the State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Liansheng Zhao
- The Psychiatric Laboratory, the State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Wei Deng
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain‐Machine Integration, State Key Laboratory of Brain‐Machine IntelligenceZhejiang UniversityHangzhouChina
- NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang UniversityHangzhouChina
| | - Wanjun Guo
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain‐Machine Integration, State Key Laboratory of Brain‐Machine IntelligenceZhejiang UniversityHangzhouChina
- NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang UniversityHangzhouChina
| | - Xun Hu
- The Clinical Research Center and Department of Pathology, The Second Affiliated HospitalZhejiang University School of MedicineZhejiangHangzhouChina
| | - Tao Li
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital and School of Brain Science and Brain MedicineZhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain‐Machine Integration, State Key Laboratory of Brain‐Machine IntelligenceZhejiang UniversityHangzhouChina
- NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang UniversityHangzhouChina
| |
Collapse
|
6
|
Lu Z, Li S, Wei R, Li W, Huang Y, Yang T, Yan M. Quercetin is a foe in the heart by targeting the hERG potassium channel. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:1397-1404. [PMID: 39386239 PMCID: PMC11459348 DOI: 10.22038/ijbms.2024.77846.16848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/21/2024] [Indexed: 10/12/2024]
Abstract
Objectives Quercetin is a plant flavonoid known for its pharmacological activities, such as antioxidant, anti-inflammatory, and anti-cancer properties. However, there is limited information available regarding its potential toxicities. A previous study showed that quercetin can inhibit human ether-a-go-related gene (hERG, also named KCNH2) currents, which may lead to long QT syndrome, torsade de pointes (TdP), and even sudden cardiac death. This study aimed to investigate the effects of quercetin on hERG and its potential mechanism. Materials and Methods hERG currents and action potential duration (APD) were assessed using the patch clamp technique. Molecular docking was employed to elucidate the binding sites between quercetin and hERG. Transfection of wild-type or mutant plasmids was used to verify the results of molecular docking. Western blot was performed to determine the expression levels of hERG, transcription factor SP1, molecular chaperones HSP70 and HSP90, phosphorylated E3 ubiquitin ligase p-Nedd4-2, serum- and glucocorticoid-inducible kinase (SGK1), and phosphatidylinositol 3-kinase (PI3K). Immunoprecipitation was conducted to evaluate hERG ubiquitination. Results Quercetin acutely blocked hERG current by binding to F656 amino acid residue, subsequently accelerating channel inactivation. Long-term incubation of quercetin accelerates Nedd4-2-mediated ubiquitination degradation of hERG channels by inhibiting the PI3K/SGK1 signaling pathway. Moreover, the APD of human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) is significantly prolonged by 30 μM quercetin. Conclusion Quercetin has a potential risk of proarrhythmia, which provided useful information for the usage and development of quercetin as a medication.
Collapse
Affiliation(s)
- Zihao Lu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
- These authors contributed equally to this work
| | - Shuwen Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
- These authors contributed equally to this work
| | - Rui Wei
- Department of Pharmacy, Jiangsu Province Official Hospital, China
| | - Wenwen Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Yuqian Huang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Tingting Yang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Meng Yan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
7
|
Zhang R, Liu J, Xue G, Yang J, Li D, Tian T, Zhang X, Gao K, Pan Z. Forced activation of dystrophin transcription by CRISPR/dCas9 reduced arrhythmia susceptibility via restoring membrane Nav1.5 distribution. Gene Ther 2023; 30:142-149. [PMID: 35644811 DOI: 10.1038/s41434-022-00348-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 05/08/2022] [Accepted: 05/12/2022] [Indexed: 11/09/2022]
Abstract
Dystrophin deficiency due to genetic mutations causes cardiac abnormalities in Duchenne's muscular dystrophy. Dystrophin is also shown to be downregulated in conventional failing hearts. Whether restoration of dystrophin expression possesses any therapeutic potential for conventional heart failure (HF) remains to be examined. HF mouse model was generated by transverse aortic constriction (TAC). In vivo activation of dystrophin transcription was achieved by tail-vein injection of adeno-associated virus 9 carrying CRISPR/dCas system for dystrophin. We found that activation of dystrophin expression in TAC mice significantly reduced the susceptibility to arrhythmia of TAC mice and the mortality rate. We further demonstrated that over-expression of dystrophin increased cardiac conduction of hearts in TAC mice by optical mapping evaluation. Activation of dystrophin expression also increased peak sodium current in isolated ventricular myocytes from hearts of TAC mice as recorded by the patch-clamp technique. Immunoblotting and immunofluorescence showed that increased dystrophin transcription restored the membrane distribution of Nav1.5 in the hearts of TAC mice. In summary, correction of dystrophin downregulation by the CRISPR-dCas9 system reduced the susceptibility to arrhythmia of conventional HF mice through restoring Nav1.5 membrane distribution. This study paved the way to develop a new therapeutic strategy for HF-related ventricular arrhythmia.
Collapse
Affiliation(s)
- Ruixin Zhang
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Junwu Liu
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Genlong Xue
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, 150086, China.,Institute of Heart and Vascular Diseases, Department of Cardiology, and Central Laboratory, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiming Yang
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Desheng Li
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Tao Tian
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Xiaofang Zhang
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Kangyi Gao
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, 150086, China
| | - Zhenwei Pan
- Department of Pharmacology (The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin, 150086, China. .,Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, Beijing, 2019RU070, China. .,NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China.
| |
Collapse
|
8
|
Baltov B, Beyl S, Baburin I, Reinhardt J, Szkokan P, Garifulina A, Timin E, Kraushaar U, Potterat O, Hamburger M, Kügler P, Hering S. Assay for evaluation of proarrhythmic effects of herbal products: Case study with 12 Evodia preparations. Toxicol Rep 2023; 10:589-599. [PMID: 37213814 PMCID: PMC10196857 DOI: 10.1016/j.toxrep.2023.04.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/11/2023] [Accepted: 04/24/2023] [Indexed: 05/23/2023] Open
Abstract
Guidelines for preclinical drug development reduce the occurrence of arrhythmia-related side effects. Besides ample evidence for the presence of arrhythmogenic substances in plants, there is no consensus on a research strategy for the evaluation of proarrhythmic effects of herbal products. Here, we propose a cardiac safety assay for the detection of proarrhythmic effects of plant extracts based on the experimental approaches described in the Comprehensive In vitro Proarrhythmia Assay (CiPA). Microelectrode array studies (MEAs) and voltage sensing optical technique on human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) were combined with ionic current measurements in mammalian cell lines, In-silico simulations of cardiac action potentials (APs) and statistic regression analysis. Proarrhythmic effects of 12 Evodia preparations, containing different amounts of the hERG inhibitors dehydroevodiamine (DHE) and hortiamine were analysed. Extracts produced different prolongation of the AP, occurrence of early after depolarisations and triangulation of the AP in hiPSC-CMs depending on the contents of the hERG inhibitors. DHE and hortiamine dose-dependently prolonged the field potential duration in hiPSC-CMs studied with MEAs. In-silico simulations of ventricular AP support a scenario where proarrhythmic effects of Evodia extracts are predominantly caused by the content of the selective hERG inhibitors. Statistic regression analysis revealed a high torsadogenic risk for both compounds that was comparable to drugs assigned to the high-risk category in a CiPA study.
Collapse
Affiliation(s)
- Bozhidar Baltov
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
- ChanPharm GmbH, Am Kanal 27, 1110 Vienna, Austria
| | | | - Igor Baburin
- ChanPharm GmbH, Am Kanal 27, 1110 Vienna, Austria
| | - Jakob Reinhardt
- Pharmaceutical Biology, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | | | - Aleksandra Garifulina
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Eugen Timin
- ChanPharm GmbH, Am Kanal 27, 1110 Vienna, Austria
| | - Udo Kraushaar
- NMI Natural and Medical Sciences Institute at the University of Tuebingen, Reutlingen, Germany
| | - Olivier Potterat
- Pharmaceutical Biology, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Matthias Hamburger
- Pharmaceutical Biology, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Philipp Kügler
- University of Hohenheim, Institute of Applied Mathematics and Statistics and Computational Science Hub, 70599 Stuttgart, Germany
| | - Steffen Hering
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
- ChanPharm GmbH, Am Kanal 27, 1110 Vienna, Austria
- Correspondence to: Am Kanal 27,2/3/5–7, 1110 Vienna, Austria.
| |
Collapse
|
9
|
Zhang D, Lü J, Ren Z, Zhang X, Wu H, Sa R, Wang X, Wang Y, Lin Z, Zhang B. Potential cardiotoxicity induced by Euodiae Fructus: In vivo and in vitro experiments and untargeted metabolomics research. Front Pharmacol 2022; 13:1028046. [PMID: 36353487 PMCID: PMC9637925 DOI: 10.3389/fphar.2022.1028046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/05/2022] [Indexed: 09/16/2023] Open
Abstract
Background: Euodiae Fructus, a well-known herbal medicine, is widely used in Asia and has also gained in popularity in Western countries over the last decades. It has known side effects, which have been observed in clinical settings, but few studies have reported on its cardiotoxicity. Methods: In the present study, experiments using techniques of untargeted metabolomics clarify the hazardous effects of Euodiae Fructus on cardiac function and metabolism in rats in situations of overdosage and unsuitable syndrome differentiation. In vitro assays are conducted to observe the toxic effects of evodiamine and rutaecarpine, two main chemical constituents of Euodiae Fructus, in H9c2 and neonatal rat cardiomyocytes (NRCMs), with their signaling mechanisms analyzed accordingly. Results: The cardiac cytotoxicity of evodiamine and rutaecarpine in in vivo experiments is associated with remarkable alterations in lactate dehydrogenase, creatine kinase, and mitochondrial membrane potential; also with increased intensity of calcium fluorescence, decreased protein expression of the cGMP-PKG pathway in H9c2 cells, and frequency of spontaneous beat in NRCMs. Additionally, the results in rats with Yin deficiency receiving a high-dosage of Euodiae Fructus suggest obvious cardiac physiological dysfunction, abnormal electrocardiogram, pathological injuries, and decreased expression of PKG protein. At the level of endogenous metabolites, the cardiac side effects of overdose and irrational usage of Euodiae Fructus relate to 34 differential metabolites and 10 metabolic pathways involving among others, the purine metabolism, the glycerophospholipid metabolism, the glycerolipid metabolism, and the sphingolipid metabolism. Conclusion: These findings shed new light on the cardiotoxicity induced by Euodiae Fructus, which might be associated with overdose and unsuitable syndrome differentiation, that comes from modulating the cGMP-PKG pathway and disturbing the metabolic pathways of purine, lipid, and amino acid. Continuing research is needed to ensure pharmacovigilance for the safe administration of Chinese herbs in the future.
Collapse
Affiliation(s)
- Dan Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jintao Lü
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhixin Ren
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaomeng Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Centre for Pharmacovigilance and Rational Use of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Huanzhang Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Rina Sa
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Gansu Provincial Hospital, Lanzhou, China
| | - Xiaofang Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Centre for Pharmacovigilance and Rational Use of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhijian Lin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Centre for Pharmacovigilance and Rational Use of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Bing Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Centre for Pharmacovigilance and Rational Use of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
10
|
Xue H, Li Y, Zhao Z, Ren J, Yu W, Wang F, Li X, Li J, Xia Q, Zhang Y, Li B. Deacetylation mechanism and potential reversal strategy of long QT syndrome on hERG K + channel under hypoxia. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166487. [PMID: 35840042 DOI: 10.1016/j.bbadis.2022.166487] [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: 02/13/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 11/30/2022]
Abstract
Clinically, hypoxia is a major risk factor for long QT syndrome (LQTS), which is associated with many diseases, such as myocardial ischemia. LQTS can be caused by the deficiency of hERG, a potassium ion channel that plays a key role in cardiac repolarization. Modifications such as acetylation of histones or non-histone proteins can affect the protein expression. In the present study, we explored the mechanism underlying hypoxia-induced LQTS and a potential reversal strategy. Experiments were performed under hypoxia to determine transcriptional and post-transcriptional expression changes. We used real-time PCR, chromatin immunoprecipitation assay, and western blotting to determine the histones acetylation in the hERG gene and the mechanism. Molecular docking, western blotting, IP, and patch -clamp assay were performed to determine the acetylation and ubiquitination levels of hERG protein and the mechanism. hERG mRNA and protein expression were found to decrease under hypoxia. The histone deacetylation level increased under hypoxia at both H3K27 and H4 of the hERG gene. HDAC1 and HDAC2 are the key enzymes for the mechanism. HDAC6 directly interacts with hERG. The acetylation level of hERG decreased and the ubiquitination level of hERG increased under hypoxia. The inhibitors of HDAC1, HDAC2, and HDAC6 could reverse the reduction of hERG mRNA and hERG protein expression under hypoxia. In conclusion, deacetylation of hERG gene-associated histones and hERG protein might be the mechanisms for LQTS in patients with hypoxia, and the inhibition of HDAC1, HDAC2, and HDAC6 might be a promising reversal strategy for reducing hERG expression under different pathological conditions.
Collapse
Affiliation(s)
- Hui Xue
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yuexin Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Zhengrong Zhao
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Jiacheng Ren
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Wenting Yu
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Fang Wang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xianghua Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Jiaxin Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Qianqian Xia
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yuxin Zhang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Baoxin Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China.
| |
Collapse
|
11
|
Role and mechanism of chaperones calreticulin and ERP57 in restoring trafficking to mutant HERG‑A561V protein. Int J Mol Med 2021; 48:159. [PMID: 34212985 PMCID: PMC8262656 DOI: 10.3892/ijmm.2021.4992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 05/20/2021] [Indexed: 02/06/2023] Open
Abstract
Long QT syndrome type 2 is caused by a mutation in the human‑ether‑a‑go‑go‑related gene (HERG) gene encoding the rapidly activating delayed rectifier K‑current. HERG is a key cell membrane glycoprotein; however, whether the maturation process of HERG protein involves key molecules derived from the calnexin (CNX)/calreticulin (CRT) cycle and how these molecules work remains unknown. Using western blotting, the present study screened the key molecules CNX/CRT/endoplasmic reticulum protein 57 (ERP57) involved in this cycle, and it was revealed that the protein expression levels of CNX/CRT/ERP57 in wild‑type (WT)/A561V cells were increased compared with those in WT cells (n=3; P<0.05). Additionally, a co‑immunoprecipitation experiment was used to reveal that the ability of CNX/ERP57/CRT to interact with HERG was significantly increased in A561V and WT/A561V cells (n=3; P<0.05). A plasmid lacking the bb' domain of ERP57 was constructed and it was demonstrated that the key site of ERP57 binding to CRT and immature HERG protein is the bb' domain. The whole‑cell patch‑clamp technique detected that the tail current density increased by 46% following overexpression of CRT and by 53% following overexpression of ERP57 in WT/A561V cells. Overexpression of CRT and ERP57 could increased HERG protein levels on the membrane detected by confocal imaging. Furthermore, overexpression of ERP57 and CRT proteins could restore the HERG‑A561V mutant protein trafficking process and rescue the dominant‑negative suppression of WT. Overall, ERP57/CRT served a crucial role in the HERG‑A561V mutant protein trafficking deficiency and degradation process.
Collapse
|
12
|
Zhan G, Wang F, Ding YQ, Li XH, Li YX, Zhao ZR, Li JX, Liu Y, Zhao X, Yan CC, Li BX. Rutaecarpine targets hERG channels and participates in regulating electrophysiological properties leading to ventricular arrhythmia. J Cell Mol Med 2021; 25:4938-4949. [PMID: 33939251 PMCID: PMC8178274 DOI: 10.1111/jcmm.16292] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/11/2020] [Accepted: 12/29/2020] [Indexed: 01/12/2023] Open
Abstract
Drug-mediated or medical condition-mediated disruption of hERG function accounts for the main cause of acquired long-QT syndrome (acLQTs), which predisposes affected individuals to ventricular arrhythmias (VA) and sudden death. Many Chinese herbal medicines, especially alkaloids, have risks of arrhythmia in clinical application. The characterized mechanisms behind this adverse effect are frequently associated with inhibition of cardiac hERG channels. The present study aimed to assess the potent effect of Rutaecarpine (Rut) on hERG channels. hERG-HEK293 cell was applied for evaluating the effect of Rut on hERG channels and the underlying mechanism. hERG current (IhERG ) was measured by patch-clamp technique. Protein levels were analysed by Western blot, and the phosphorylation of Sp1 was determined by immunoprecipitation. Optical mapping and programmed electrical stimulation were used to evaluate cardiac electrophysiological activities, such as APD, QT/QTc, occurrence of arrhythmia, phase singularities (PSs), and dominant frequency (DF). Our results demonstrated that Rut reduced the IhERG by binding to F656 and Y652 amino acid residues of hERG channel instantaneously, subsequently accelerating the channel inactivation, and being trapped in the channel. The level of hERG channels was reduced by incubating with Rut for 24 hours, and Sp1 in nucleus was inhibited simultaneously. Mechanismly, Rut reduced threonine (Thr)/ tyrosine (Tyr) phosphorylation of Sp1 through PI3K/Akt pathway to regulate hERG channels expression. Cell-based model unables to fully reveal the pathological process of arrhythmia. In vivo study, we found that Rut prolonged QT/QTc intervals and increased induction rate of ventricular fibrillation (VF) in guinea pig heart after being dosed Rut for 2 weeks. The critical reasons led to increased incidence of arrhythmias eventually were prolonged APD90 and APD50 and the increase of DF, numbers of PSs, incidence of early after-depolarizations (EADs). Collectively, the results of this study suggest that Rut could reduce the IhERG by binding to hERG channels through F656 and Y652 instantaneously. While, the PI3K/Akt/Sp1 axis may play an essential role in the regulation of hERG channels, from the perspective of the long-term effects of Rut (incubating for 24 hours). Importantly, the changes of electrophysiological properties by Rut were the main cause of VA.
Collapse
Affiliation(s)
- Ge Zhan
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Fang Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yun-Qi Ding
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xiang-Hua Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yue-Xin Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Zheng-Rong Zhao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Jia-Xin Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yan Liu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xin Zhao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Cai-Chuan Yan
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Bao-Xin Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| |
Collapse
|