1
|
Zhang HN, Zhang M, Tian W, Quan W, Song F, Liu SY, Liu XX, Mo D, Sun Y, Gao YY, Ye W, Feng YD, Xing CY, Ye C, Zhou L, Meng JR, Cao W, Li XQ. Canonical transient receptor potential channel 1 aggravates myocardial ischemia-and-reperfusion injury by upregulating reactive oxygen species. J Pharm Anal 2023; 13:1309-1325. [PMID: 38174113 PMCID: PMC10759261 DOI: 10.1016/j.jpha.2023.08.018] [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: 04/03/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 01/05/2024] Open
Abstract
The canonical transient receptor potential channel (TRPC) proteins form Ca2+-permeable cation channels that are involved in various heart diseases. However, the roles of specific TRPC proteins in myocardial ischemia/reperfusion (I/R) injury remain poorly understood. We observed that TRPC1 and TRPC6 were highly expressed in the area at risk (AAR) in a coronary artery ligation induced I/R model. Trpc1-/- mice exhibited improved cardiac function, lower serum Troponin T and serum creatine kinase level, smaller infarct volume, less fibrotic scars, and fewer apoptotic cells after myocardial-I/R than wild-type or Trpc6-/- mice. Cardiomyocyte-specific knockdown of Trpc1 using adeno-associated virus 9 mitigated myocardial I/R injury. Furthermore, Trpc1 deficiency protected adult mouse ventricular myocytes (AMVMs) and HL-1 cells from death during hypoxia/reoxygenation (H/R) injury. RNA-sequencing-based transcriptome analysis revealed differential expression of genes related to reactive oxygen species (ROS) generation in Trpc1-/- cardiomyocytes. Among these genes, oxoglutarate dehydrogenase-like (Ogdhl) was markedly downregulated. Moreover, Trpc1 deficiency impaired the calcineurin (CaN)/nuclear factor-kappa B (NF-κB) signaling pathway in AMVMs. Suppression of this pathway inhibited Ogdhl upregulation and ROS generation in HL-1 cells under H/R conditions. Chromatin immunoprecipitation assays confirmed NF-κB binding to the Ogdhl promoter. The cardioprotective effect of Trpc1 deficiency was canceled out by overexpression of NF-κB and Ogdhl in cardiomyocytes. In conclusion, our findings reveal that TRPC1 is upregulated in the AAR following myocardial I/R, leading to increased Ca2+ influx into associated cardiomyocytes. Subsequently, this upregulates Ogdhl expression through the CaN/NF-κB signaling pathway, ultimately exacerbating ROS production and aggravating myocardial I/R injury.
Collapse
Affiliation(s)
- Hui-Nan Zhang
- Department of Health Management, Second Affiliated Hospital, Fourth Military Medical University, Xi'an, 710038, China
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Meng Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Shaanxi Key Laboratory of “Qin Medicine” Research and Development, Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710032, China
| | - Wen Tian
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Shaanxi Key Laboratory of “Qin Medicine” Research and Development, Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710032, China
| | - Wei Quan
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Shaanxi Key Laboratory of “Qin Medicine” Research and Development, Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710032, China
| | - Fan Song
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Institute of Materia Medica, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Shao-Yuan Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Shaanxi Key Laboratory of “Qin Medicine” Research and Development, Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710032, China
| | - Xiao-Xiao Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Dan Mo
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Yang Sun
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Yuan-Yuan Gao
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Wen Ye
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Shaanxi Key Laboratory of “Qin Medicine” Research and Development, Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710032, China
| | - Ying-Da Feng
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Shaanxi Key Laboratory of “Qin Medicine” Research and Development, Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710032, China
| | - Chang-Yang Xing
- Department of Ultrasound Diagnostics, Second Affiliated Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Chen Ye
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Lei Zhou
- Department of Pharmacy, School of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jing-Ru Meng
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Shaanxi Key Laboratory of “Qin Medicine” Research and Development, Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710032, China
| | - Wei Cao
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Department of Pharmacy, School of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xiao-Qiang Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
- Shaanxi Key Laboratory of “Qin Medicine” Research and Development, Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710032, China
| |
Collapse
|
2
|
Cai D, Wang X, Sun Y, Fan H, Zhou J, Yang Z, Qiu H, Wang J, Su J, Gong T, Jiang C, Liang P. Patient-specific iPSC-derived cardiomyocytes reveal aberrant activation of Wnt/β-catenin signaling in SCN5A-related Brugada syndrome. Stem Cell Res Ther 2023; 14:241. [PMID: 37679791 PMCID: PMC10486057 DOI: 10.1186/s13287-023-03477-3] [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: 01/12/2023] [Accepted: 08/29/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Mutations in the cardiac sodium channel gene SCN5A cause Brugada syndrome (BrS), an arrhythmic disorder that is a leading cause of sudden death and lacks effective treatment. An association between SCN5A and Wnt/β-catenin signaling has been recently established. However, the role of Wnt/β-catenin signaling in BrS and underlying mechanisms remains unknown. METHODS Three healthy control subjects and one BrS patient carrying a novel frameshift mutation (T1788fs) in the SCN5A gene were recruited in this study. Control and BrS patient-specific induced pluripotent stem cells (iPSCs) were generated from skin fibroblasts using nonintegrated Sendai virus. All iPSCs were differentiated into cardiomyocytes using monolayer-based differentiation protocol. Action potentials and sodium currents were recorded from control and BrS iPSC-derived cardiomyocytes (iPSC-CMs) by single-cell patch clamp. RESULTS BrS iPSC-CMs exhibited increased burden of arrhythmias and abnormal action potential profile featured by slower depolarization, decreased action potential amplitude, and increased beating interval variation. Moreover, BrS iPSC-CMs showed cardiac sodium channel (Nav1.5) loss-of-function as compared to control iPSC-CMs. Interestingly, the electrophysiological abnormalities and Nav1.5 loss-of-function observed in BrS iPSC-CMs were accompanied by aberrant activation of Wnt/β-catenin signaling. Notably, inhibition of Wnt/β-catenin significantly rescued Nav1.5 defects and arrhythmic phenotype in BrS iPSC-CMs. Mechanistically, SCN5A-encoded Nav1.5 interacts with β-catenin, and reduced expression of Nav1.5 leads to re-localization of β-catenin in BrS iPSC-CMs, which aberrantly activates Wnt/β-catenin signaling to suppress SCN5A transcription. CONCLUSIONS Our findings suggest that aberrant activation of Wnt/β-catenin signaling contributes to the pathogenesis of SCN5A-related BrS and point to Wnt/β-catenin as a potential therapeutic target.
Collapse
Affiliation(s)
- Dongsheng Cai
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qingchun East Road, Hangzhou, 310016, China
| | - Xiaochen Wang
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Yaxun Sun
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qingchun East Road, Hangzhou, 310016, China
| | - Hangping Fan
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Jingjun Zhou
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Zongkuai Yang
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Hangyuan Qiu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qingchun East Road, Hangzhou, 310016, China
| | - Jue Wang
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Jun Su
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Tingyu Gong
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Chenyang Jiang
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qingchun East Road, Hangzhou, 310016, China.
| | - Ping Liang
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China.
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China.
| |
Collapse
|
3
|
Streiff ME, Corbin AC, Ahmad AA, Hunter C, Sachse FB. TRPC1 channels underlie stretch-modulated sarcoplasmic reticulum calcium leak in cardiomyocytes. Front Physiol 2022; 13:1056657. [PMID: 36620209 PMCID: PMC9817106 DOI: 10.3389/fphys.2022.1056657] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/12/2022] [Indexed: 12/25/2022] Open
Abstract
Transient receptor potential canonical 1 (TRPC1) channels are Ca2+-permeable ion channels expressed in cardiomyocytes. An involvement of TRPC1 channels in cardiac diseases is widely established. However, the physiological role of TRPC1 channels and the mechanisms through which they contribute to disease development are still under investigation. Our prior work suggested that TRPC1 forms Ca2+ leak channels located in the sarcoplasmic reticulum (SR) membrane. Prior studies suggested that TRPC1 channels in the cell membrane are mechanosensitive, but this was not yet investigated in cardiomyocytes or for SR localized TRPC1 channels. We applied adenoviral transfection to overexpress or suppress TRPC1 expression in neonatal rat ventricular myocytes (NRVMs). Transfections were evaluated with RT-qPCR, western blot, and fluorescent imaging. Single-molecule localization microscopy revealed high colocalization of exogenously expressed TRPC1 and the sarco/endoplasmic reticulum Ca2+ ATPase (SERCA2). To test our hypothesis that TRPC1 channels contribute to mechanosensitive Ca2+ SR leak, we directly measured SR Ca2+ concentration ([Ca2+]SR) using adenoviral transfection with a novel ratiometric genetically encoded SR-targeting Ca2+ sensor. We performed fluorescence imaging to quantitatively assess [Ca2+]SR and leak through TRPC1 channels of NRVMs cultured on stretchable silicone membranes. [Ca2+]SR was increased in cells with suppressed TRPC1 expression vs. control and Transient receptor potential canonical 1-overexpressing cells. We also detected a significant reduction in [Ca2+]SR in cells with Transient receptor potential canonical 1 overexpression when 10% uniaxial stretch was applied. These findings indicate that TRPC1 channels underlie the mechanosensitive modulation of [Ca2+]SR. Our findings are critical for understanding the physiological role of TRPC1 channels and support the development of pharmacological therapies for cardiac diseases.
Collapse
Affiliation(s)
- Molly E. Streiff
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Andrea C. Corbin
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Azmi A. Ahmad
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Chris Hunter
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
| | - Frank B. Sachse
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT, United States
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
4
|
Sabourin J, Beauvais A, Luo R, Montani D, Benitah JP, Masson B, Antigny F. The SOCE Machinery: An Unbalanced Knowledge between Left and Right Ventricular Pathophysiology. Cells 2022; 11:cells11203282. [PMID: 36291148 PMCID: PMC9600889 DOI: 10.3390/cells11203282] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/09/2022] [Accepted: 10/15/2022] [Indexed: 11/16/2022] Open
Abstract
Right ventricular failure (RVF) is the most important prognostic factor for morbidity and mortality in pulmonary arterial hypertension (PAH) or pulmonary hypertension (PH) caused by left heart diseases. However, right ventricle (RV) remodeling is understudied and not targeted by specific therapies. This can be partly explained by the lack of basic knowledge of RV remodeling. Since the physiology and hemodynamic function of the RV differ from those of the left ventricle (LV), the mechanisms of LV dysfunction cannot be generalized to that of the RV, albeit a knowledge of these being helpful to understanding RV remodeling and dysfunction. Store-operated Ca2+ entry (SOCE) has recently emerged to participate in the LV cardiomyocyte Ca2+ homeostasis and as a critical player in Ca2+ mishandling in a pathological context. In this paper, we highlight the current knowledge on the SOCE contribution to the LV and RV dysfunctions, as SOCE molecules are present in both compartments. he relative lack of studies on RV dysfunction indicates the necessity of further investigations, a significant challenge over the coming years.
Collapse
Affiliation(s)
- Jessica Sabourin
- Signalisation et Physiopathologie Cardiovasculaire, Inserm, Université Paris-Saclay, UMR-S 1180, 91400 Orsay, France
- Correspondence: (J.S.); (F.A.); Tel.: +(33)-180-006-302 (J.S.); +(33)-140-942-299 (F.A.)
| | - Antoine Beauvais
- Faculté de Médecine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France
- Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Université Paris-Saclay, Inserm, UMR-S 999, 92350 Le Plessis-Robinson, France
- Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Rui Luo
- Signalisation et Physiopathologie Cardiovasculaire, Inserm, Université Paris-Saclay, UMR-S 1180, 91400 Orsay, France
| | - David Montani
- Faculté de Médecine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France
- Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Université Paris-Saclay, Inserm, UMR-S 999, 92350 Le Plessis-Robinson, France
- Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Jean-Pierre Benitah
- Signalisation et Physiopathologie Cardiovasculaire, Inserm, Université Paris-Saclay, UMR-S 1180, 91400 Orsay, France
| | - Bastien Masson
- Faculté de Médecine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France
- Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Université Paris-Saclay, Inserm, UMR-S 999, 92350 Le Plessis-Robinson, France
- Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Fabrice Antigny
- Faculté de Médecine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France
- Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Université Paris-Saclay, Inserm, UMR-S 999, 92350 Le Plessis-Robinson, France
- Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
- Correspondence: (J.S.); (F.A.); Tel.: +(33)-180-006-302 (J.S.); +(33)-140-942-299 (F.A.)
| |
Collapse
|
5
|
Fakhar M, Najumuddin, Zahid S, Rashid S. Structural basis of Klotho binding to VEGFR2 and TRPC1 and repurposing calcium channel blockers as TRPC1 antagonists for the treatment of age-related cardiac hypertrophy. Arch Biochem Biophys 2022; 719:109171. [DOI: 10.1016/j.abb.2022.109171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 12/13/2021] [Accepted: 02/28/2022] [Indexed: 11/15/2022]
|
6
|
Lu T, Zhang Y, Su Y, Zhou D, Xu Q. Role of store-operated Ca2+ entry in cardiovascular disease. Cell Commun Signal 2022; 20:33. [PMID: 35303866 PMCID: PMC8932232 DOI: 10.1186/s12964-022-00829-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/14/2022] [Indexed: 01/01/2023] Open
Abstract
Store-operated channels (SOCs) are highly selective Ca2+ channels that mediate Ca2+ influx in non-excitable and excitable (i.e., skeletal and cardiac muscle) cells. These channels are triggered by Ca2+ depletion of the endoplasmic reticulum and sarcoplasmic reticulum, independently of inositol 1,4,5-trisphosphate (InsP3), which is involved in cell growth, differentiation, and gene transcription. When the Ca2+ store is depleted, stromal interaction molecule1 (STIM1) as Ca2+ sensor redistributes into discrete puncta near the plasma membrane and activates the protein Ca2+ release activated Ca2+ channel protein 1 (Orai1). Accumulating evidence suggests that SOC is associated with several physiological roles in endothelial dysfunction and vascular smooth muscle proliferation that contribute to the progression of cardiovascular disease. This review mainly elaborates on the contribution of SOC in the vasculature (endothelial cells and vascular smooth muscle cells). We will further retrospect the literature implicating a critical role for these proteins in cardiovascular disease.
Collapse
Affiliation(s)
- Ting Lu
- Department of Cardiology, Chongqing Fifth People's Hospital, No. 24 Renji Road, Chongqing, 400000, China
| | - Yihua Zhang
- Department of Cardiology, Chongqing Fifth People's Hospital, No. 24 Renji Road, Chongqing, 400000, China
| | - Yong Su
- Department of Cardiology, Chongqing Fifth People's Hospital, No. 24 Renji Road, Chongqing, 400000, China
| | - Dayan Zhou
- Department of Cardiology, Chongqing Fifth People's Hospital, No. 24 Renji Road, Chongqing, 400000, China
| | - Qiang Xu
- Department of Cardiology, Chongqing Fifth People's Hospital, No. 24 Renji Road, Chongqing, 400000, China.
| |
Collapse
|
7
|
Tang Y, Xu Z, Chen X, Wang N, Deng X, Peng L, Chen Q, Cai H. Effects of Enalapril on TLR2/NF- κB Signaling Pathway and Inflammatory Factors in Rabbits with Chronic Heart Failure. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:9594607. [PMID: 34335842 PMCID: PMC8294953 DOI: 10.1155/2021/9594607] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/01/2021] [Indexed: 11/29/2022]
Abstract
Chronic heart failure (CHF) refers to the state of persistent heart failure, which is a complex clinical syndrome of various advanced heart diseases. The toll-like receptor 2 (TLR2)/nuclear transcription factor-κB (NF-κB) signal transduction pathway is one of the pathological mechanisms of CHF. Adriamycin can significantly induce the upregulation of TLR2 expression. Angiotensin-converting enzyme inhibitors (ACEI) are commonly used drugs for the treatment of CHF. In our study, the CHF model was established by injection of doxorubicin into the rabbit ear vein. The effect of enalapril on the TLR2/NF-κB signaling pathway in CHF rabbits has been analyzed and determined. Our research results showed that enalapril reduced the inflammatory response by inhibiting the activation of the TLR2/NF-κB signaling pathway, thereby improving cardiac structure, myocardial remodeling, and cardiac function.
Collapse
Affiliation(s)
- Yanping Tang
- Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Zelin Xu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, China
| | - Xinyu Chen
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, China
| | - Nan Wang
- Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Xu Deng
- Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Liqi Peng
- Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Qingyang Chen
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, China
| | - Huzhi Cai
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, China
| |
Collapse
|
8
|
Val‐Blasco A, Gil‐Fernández M, Rueda A, Pereira L, Delgado C, Smani T, Ruiz Hurtado G, Fernández‐Velasco M. Ca 2+ mishandling in heart failure: Potential targets. Acta Physiol (Oxf) 2021; 232:e13691. [PMID: 34022101 DOI: 10.1111/apha.13691] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/14/2022]
Abstract
Ca2+ mishandling is a common feature in several cardiovascular diseases such as heart failure (HF). In many cases, impairment of key players in intracellular Ca2+ homeostasis has been identified as the underlying mechanism of cardiac dysfunction and cardiac arrhythmias associated with HF. In this review, we summarize primary novel findings related to Ca2+ mishandling in HF progression. HF research has increasingly focused on the identification of new targets and the contribution of their role in Ca2+ handling to the progression of the disease. Recent research studies have identified potential targets in three major emerging areas implicated in regulation of Ca2+ handling: the innate immune system, bone metabolism factors and post-translational modification of key proteins involved in regulation of Ca2+ handling. Here, we describe their possible contributions to the progression of HF.
Collapse
Affiliation(s)
| | | | - Angélica Rueda
- Department of Biochemistry Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV‐IPN) México City Mexico
| | - Laetitia Pereira
- INSERM UMR‐S 1180 Laboratory of Ca Signaling and Cardiovascular Physiopathology University Paris‐Saclay Châtenay‐Malabry France
| | - Carmen Delgado
- Instituto de Investigaciones Biomédicas Alberto Sols Madrid Spain
- Department of Metabolism and Cell Signalling Biomedical Research Institute "Alberto Sols" CSIC‐UAM Madrid Spain
| | - Tarik Smani
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV) Madrid Spain
- Department of Medical Physiology and Biophysics University of Seville Seville Spain
- Group of Cardiovascular Pathophysiology Institute of Biomedicine of Seville University Hospital of Virgen del Rocío, University of Seville, CSIC Seville Spain
| | - Gema Ruiz Hurtado
- Cardiorenal Translational Laboratory Institute of Research i+12 University Hospital 12 de Octubre Madrid Spain
- CIBER‐CV University Hospita1 12 de Octubre Madrid Spain
| | - Maria Fernández‐Velasco
- La Paz University Hospital Health Research Institute IdiPAZ Madrid Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV) Madrid Spain
| |
Collapse
|
9
|
Jiang X, Chen Y, Liu X, Ye L, Yu M, Shen Z, Lei W, Hu S. Uncovering Inherited Cardiomyopathy With Human Induced Pluripotent Stem Cells. Front Cell Dev Biol 2021; 9:672039. [PMID: 34079803 PMCID: PMC8166268 DOI: 10.3389/fcell.2021.672039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/06/2021] [Indexed: 11/13/2022] Open
Abstract
In the past decades, researchers discovered the contribution of genetic defects to the pathogenesis of primary cardiomyopathy and tried to explain the pathogenesis of these diseases by establishing a variety of disease models. Although human heart tissues and primary cardiomyocytes have advantages in modeling human heart diseases, they are difficult to obtain and culture in vitro. Defects developed in genetically modified animal models are notably different from human diseases at the molecular level. The advent of human induced pluripotent stem cells (hiPSCs) provides an unprecedented opportunity to further investigate the pathogenic mechanisms of inherited cardiomyopathies in vitro using patient-specific hiPSC-derived cardiomyocytes. In this review, we will make a summary of recent advances in in vitro inherited cardiomyopathy modeling using hiPSCs.
Collapse
Affiliation(s)
- Xue Jiang
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, China
| | - Yihuan Chen
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, China
| | - Xiaofeng Liu
- The Affiliated Haian Hospital of Nantong University, Nantong, China
| | - Lingqun Ye
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, China
| | - Miao Yu
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, China
| | - Zhenya Shen
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, China
| | - Wei Lei
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, China
| | - Shijun Hu
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, China
| |
Collapse
|
10
|
Mani I. Genome editing in cardiovascular diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2021; 181:289-308. [PMID: 34127197 DOI: 10.1016/bs.pmbts.2021.01.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Genetic modification at the molecular level in somatic cells, germline, and animal models requires for different purposes, such as introducing desired mutation, deletion of alleles, and insertion of novel genes in the genome. Various genome-editing tools are available to accomplish these alterations, such as zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR associated (Cas) system. CRISPR-Cas system is an emerging technology, which is being used in biological and medical sciences, including in the cardiovascular field. It assists to identify the mechanism of various cardiovascular disease occurrence, such as hypertrophic cardiomyopathy (HCM), dilated cardiomyopathy (DCM), and arrhythmogenic cardiomyopathy (ACM). Furthermore, it has been advantages to edit various genes simultaneously and can also be used to treat and prevent several human diseases. This chapter explores the use of the scientific and therapeutic potential of a CRISPR-Cas system to edit the various cardiovascular disease-associated genes to understand the pathways involved in disease progression and treatment.
Collapse
Affiliation(s)
- Indra Mani
- Department of Microbiology, Gargi College, University of Delhi, New Delhi, India.
| |
Collapse
|
11
|
Sun Y, Nascimento Da Conceicao V, Ahamad N, Madesh M, Singh BB. Spatial localization of SOCE channels and its modulators regulate neuronal physiology and contributes to pathology. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
12
|
Chen X, Sooch G, Demaree IS, White FA, Obukhov AG. Transient Receptor Potential Canonical (TRPC) Channels: Then and Now. Cells 2020; 9:E1983. [PMID: 32872338 PMCID: PMC7565274 DOI: 10.3390/cells9091983] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 12/13/2022] Open
Abstract
Twenty-five years ago, the first mammalian Transient Receptor Potential Canonical (TRPC) channel was cloned, opening the vast horizon of the TRPC field. Today, we know that there are seven TRPC channels (TRPC1-7). TRPCs exhibit the highest protein sequence similarity to the Drosophila melanogaster TRP channels. Similar to Drosophila TRPs, TRPCs are localized to the plasma membrane and are activated in a G-protein-coupled receptor-phospholipase C-dependent manner. TRPCs may also be stimulated in a store-operated manner, via receptor tyrosine kinases, or by lysophospholipids, hypoosmotic solutions, and mechanical stimuli. Activated TRPCs allow the influx of Ca2+ and monovalent alkali cations into the cytosol of cells, leading to cell depolarization and rising intracellular Ca2+ concentration. TRPCs are involved in the continually growing number of cell functions. Furthermore, mutations in the TRPC6 gene are associated with hereditary diseases, such as focal segmental glomerulosclerosis. The most important recent breakthrough in TRPC research was the solving of cryo-EM structures of TRPC3, TRPC4, TRPC5, and TRPC6. These structural data shed light on the molecular mechanisms underlying TRPCs' functional properties and propelled the development of new modulators of the channels. This review provides a historical overview of the major advances in the TRPC field focusing on the role of gene knockouts and pharmacological tools.
Collapse
Affiliation(s)
- Xingjuan Chen
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China;
| | - Gagandeep Sooch
- The Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (G.S.); (I.S.D.)
| | - Isaac S. Demaree
- The Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (G.S.); (I.S.D.)
| | - Fletcher A. White
- The Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alexander G. Obukhov
- The Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (G.S.); (I.S.D.)
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
13
|
Guo Q, Wang J, Sun R, He Z, Chen Q, Liu W, Wu M, Bao J, Liu Z, Wang J, Zhang Y. Comprehensive Construction of a Circular RNA-Associated Competing Endogenous RNA Network Identified Novel Circular RNAs in Hypertrophic Cardiomyopathy by Integrated Analysis. Front Genet 2020; 11:764. [PMID: 32849787 PMCID: PMC7399352 DOI: 10.3389/fgene.2020.00764] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/29/2020] [Indexed: 11/20/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM), the most common heritable cardiomyopathy, is associated with a high risk of sudden cardiac death. The complexity and behavior of the circular RNA (circRNA)-associated competing endogenous RNA (ceRNA) network in HCM have not been thoroughly elucidated. Plasma circRNA and messenger RNA (mRNA) expression profiles were acquired by using a microarray. Weighted correlation network analysis (WGCNA) and linear models for microarray data (Limma) were used to analyze microarray data. Gene modules, consisting of genes with high correlations, were detected and represented by a designated color. The ceRNA network, including circRNA, microRNA (miRNA), and mRNA, was constructed based on the “ceRNA hypothesis” using an integrated systems biology method. By WGCNA, two modules, namely magenta and red modules, were identified as being positively correlated with HCM. In the combined analysis of WGCNA and Limma, 36 hub circRNAs in the magenta module and 83 hub circRNAs in the red module were significantly upregulated compared with the controls. By coexpression analysis, 270 circRNA–mRNA pairs were identified with a coefficient ≥0.9 and p < 0.05. With Starbase and miRWalk tools, circRNA–miRNA pairs and miRNA–mRNA pairs were predicted. Once these pairs were combined, the ceRNA network with 6 circRNAs, 29 miRNAs, and 6 mRNAs was constructed. Functional analysis demonstrated that these circRNAs in the ceRNA network were associated with calcium-release channel activity and muscle filament sliding. Our study provided a global perspective and systematic analysis of the circRNA-associated ceRNA network in HCM. The identified circRNAs hsa_circ_0043762, hsa_circ_0036248, and hsa_circ_0071269 may be key regulators involved in HCM pathogenesis.
Collapse
Affiliation(s)
- Qi Guo
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Junjie Wang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Runlu Sun
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Zhijian He
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Qian Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Wenhao Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Maoxiong Wu
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Jinlan Bao
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Zhaoyu Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Jingfeng Wang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Yuling Zhang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| |
Collapse
|
14
|
Camacho Londoño JE, Kuryshev V, Zorn M, Saar K, Tian Q, Hübner N, Nawroth P, Dietrich A, Birnbaumer L, Lipp P, Dieterich C, Freichel M. Transcriptional signatures regulated by TRPC1/C4-mediated Background Ca 2+ entry after pressure-overload induced cardiac remodelling. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2020; 159:86-104. [PMID: 32738354 DOI: 10.1016/j.pbiomolbio.2020.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/03/2020] [Accepted: 07/21/2020] [Indexed: 01/17/2023]
Abstract
AIMS After summarizing current concepts for the role of TRPC cation channels in cardiac cells and in processes triggered by mechanical stimuli arising e.g. during pressure overload, we analysed the role of TRPC1 and TRPC4 for background Ca2+ entry (BGCE) and for cardiac pressure overload induced transcriptional remodelling. METHODS AND RESULTS Mn2+-quench analysis in cardiomyocytes from several Trpc-deficient mice revealed that both TRPC1 and TRPC4 are required for BGCE. Electrically-evoked cell shortening of cardiomyocytes from TRPC1/C4-DKO mice was reduced, whereas parameters of cardiac contractility and relaxation assessed in vivo were unaltered. As pathological cardiac remodelling in mice depends on their genetic background, and the development of cardiac remodelling was found to be reduced in TRPC1/C4-DKO mice on a mixed genetic background, we studied TRPC1/C4-DKO mice on a C57BL6/N genetic background. Cardiac hypertrophy was reduced in those mice after chronic isoproterenol infusion (-51.4%) or after one week of transverse aortic constriction (TAC; -73.0%). This last manoeuvre was preceded by changes in the pressure overload induced transcriptional program as analysed by RNA sequencing. Genes encoding specific collagens, the Mef2 target myomaxin and the gene encoding the mechanosensitive channel Piezo2 were up-regulated after TAC in wild type but not in TRPC1/C4-DKO hearts. CONCLUSIONS Deletion of the TRPC1 and TRPC4 channel proteins protects against development of pathological cardiac hypertrophy independently of the genetic background. To determine if the TRPC1/C4-dependent changes in the pressure overload induced alterations in the transcriptional program causally contribute to cardio-protection needs to be elaborated in future studies.
Collapse
Affiliation(s)
- Juan E Camacho Londoño
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, 69120, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, 69120, Germany.
| | - Vladimir Kuryshev
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, 69120, Heidelberg, Germany; Innere Medizin III, Bioinformatik und Systemkardiologie, Klaus Tschira Institute for Computational Cardiology, Ruprecht-Karls-Universität Heidelberg, 69120, Heidelberg, Germany
| | - Markus Zorn
- Department of Medicine I and Clinical Chemistry, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Kathrin Saar
- Max-Delbrück-Centrum für Molekulare Medizin (MDC), 13125, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13347, Berlin, Germany
| | - Qinghai Tian
- Medical Faculty, Centre for Molecular Signalling (PZMS), Institute for Molecular Cell Biology and Research Center for Molecular Imaging and Screening, Saarland University, 66421 Homburg/Saar, Germany
| | - Norbert Hübner
- Max-Delbrück-Centrum für Molekulare Medizin (MDC), 13125, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13347, Berlin, Germany; Berlin Institute of Health (BIH), 10178, Berlin, Germany; Charité -Universitätsmedizin, 10117, Berlin, Germany
| | - Peter Nawroth
- Department of Medicine I and Clinical Chemistry, University Hospital Heidelberg, 69120, Heidelberg, Germany; German Center for Diabetes Research (DZD), Germany; Institute for Diabetes and Cancer IDC Helmholtz Center Munich, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Dept. of Medicine I, Heidelberg University Hospital, Heidelberg, Germany
| | - Alexander Dietrich
- Walther-Straub-Institut für Pharmakologie und Toxikologie, Member of the German Center for Lung Research (DZL), Ludwig-Maximilians-Universität, 80336, München, Germany
| | - Lutz Birnbaumer
- Laboratory of Neurobiology, NIEHS, North Carolina, USA and Institute of Biomedical Research (BIOMED), Catholic University of Argentina, C1107AFF Buenos Aires, Argentina
| | - Peter Lipp
- Medical Faculty, Centre for Molecular Signalling (PZMS), Institute for Molecular Cell Biology and Research Center for Molecular Imaging and Screening, Saarland University, 66421 Homburg/Saar, Germany
| | - Christoph Dieterich
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, 69120, Germany; Innere Medizin III, Bioinformatik und Systemkardiologie, Klaus Tschira Institute for Computational Cardiology, Ruprecht-Karls-Universität Heidelberg, 69120, Heidelberg, Germany
| | - Marc Freichel
- Pharmakologisches Institut, Ruprecht-Karls-Universität Heidelberg, 69120, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, 69120, Germany.
| |
Collapse
|
15
|
TRPC Channels in Cardiac Plasticity. Cells 2020; 9:cells9020454. [PMID: 32079284 PMCID: PMC7072762 DOI: 10.3390/cells9020454] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 01/21/2023] Open
Abstract
The heart flexibly changes its structure in response to changing environments and oxygen/nutrition demands of the body. Increased and decreased mechanical loading induces hypertrophy and atrophy of cardiomyocytes, respectively. In physiological conditions, these structural changes of the heart are reversible. However, chronic stresses such as hypertension or cancer cachexia cause irreversible remodeling of the heart, leading to heart failure. Accumulating evidence indicates that calcium dyshomeostasis and aberrant reactive oxygen species production cause pathological heart remodeling. Canonical transient receptor potential (TRPC) is a nonselective cation channel subfamily whose multimodal activation or modulation of channel activity play important roles in a plethora of cellular physiology. Roles of TRPC channels in cardiac physiology have been reported in pathological cardiac remodeling. In this review, we summarize recent findings regarding the importance of TRPC channels in flexible cardiac remodeling (i.e., cardiac plasticity) in response to environmental stresses and discuss questions that should be addressed in the near future.
Collapse
|
16
|
TRPC Channels: Dysregulation and Ca 2+ Mishandling in Ischemic Heart Disease. Cells 2020; 9:cells9010173. [PMID: 31936700 PMCID: PMC7017417 DOI: 10.3390/cells9010173] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/03/2020] [Accepted: 01/08/2020] [Indexed: 12/17/2022] Open
Abstract
Transient receptor potential canonical (TRPC) channels are ubiquitously expressed in excitable and non-excitable cardiac cells where they sense and respond to a wide variety of physical and chemical stimuli. As other TRP channels, TRPC channels may form homo or heterotetrameric ion channels, and they can associate with other membrane receptors and ion channels to regulate intracellular calcium concentration. Dysfunctions of TRPC channels are involved in many types of cardiovascular diseases. Significant increase in the expression of different TRPC isoforms was observed in different animal models of heart infarcts and in vitro experimental models of ischemia and reperfusion. TRPC channel-mediated increase of the intracellular Ca2+ concentration seems to be required for the activation of the signaling pathway that plays minor roles in the healthy heart, but they are more relevant for cardiac responses to ischemia, such as the activation of different factors of transcription and cardiac hypertrophy, fibrosis, and angiogenesis. In this review, we highlight the current knowledge regarding TRPC implication in different cellular processes related to ischemia and reperfusion and to heart infarction.
Collapse
|
17
|
Valenti MT, Serena M, Carbonare LD, Zipeto D. CRISPR/Cas system: An emerging technology in stem cell research. World J Stem Cells 2019; 11:937-956. [PMID: 31768221 PMCID: PMC6851009 DOI: 10.4252/wjsc.v11.i11.937] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 08/12/2019] [Accepted: 09/11/2019] [Indexed: 02/06/2023] Open
Abstract
The identification of new and even more precise technologies for modifying and manipulating the genome has been a challenge since the discovery of the DNA double helix. The ability to modify selectively specific genes provides a powerful tool for characterizing gene functions, performing gene therapy, correcting specific genetic mutations, eradicating diseases, engineering cells and organisms to achieve new and different functions and obtaining transgenic animals as models for studying specific diseases. Clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 technology has recently revolutionized genome engineering. The application of this new technology to stem cell research allows disease models to be developed to explore new therapeutic tools. The possibility of translating new systems of molecular knowledge to clinical research is particularly appealing for addressing degenerative diseases. In this review, we describe several applications of CRISPR/Cas9 to stem cells related to degenerative diseases. In addition, we address the challenges and future perspectives regarding the use of CRISPR/Cas9 as an important technology in the medical sciences.
Collapse
Affiliation(s)
- Maria Teresa Valenti
- Department of Medicine, Section of Internal Medicine D, University of Verona, Verona 37134, Italy.
| | - Michela Serena
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Luca Dalle Carbonare
- Department of Medicine, Section of Internal Medicine D, University of Verona, Verona 37134, Italy
| | - Donato Zipeto
- Department of Neurosciences, Biomedicine and Movement Sciences, Laboratory of Molecular Biology, Verona 37134, Italy
| |
Collapse
|