1
|
Namekata I, Tamura M, Kase J, Hamaguchi S, Tanaka H. Cardioprotective Effect against Ischemia-Reperfusion Injury of PAK-200, a Dihydropyridine Analog with an Inhibitory Effect on Cl - but Not Ca 2+ Current. Biomolecules 2023; 13:1719. [PMID: 38136589 PMCID: PMC10741401 DOI: 10.3390/biom13121719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
We examined the effects of a dihydropyridine analog, PAK-200, on guinea pig myocardium during experimental ischemia and reperfusion. In isolated ventricular cardiomyocytes, PAK-200 (1 μM) had no effect on the basal peak inward and steady-state currents but inhibited the isoprenaline-induced time-independent Cl- current. In the right atria, PAK-200 had no effect on the beating rate and the chronotropic response to isoprenaline. In an ischemia-reperfusion model with coronary-perfused right ventricular tissue, a decrease in contractile force and a rise in tension were observed during a period of 30-min no-flow ischemia. Upon reperfusion, contractile force returned to less than 50% of preischemic values. PAK-200 had no effect on the decline in contractile force during the no-flow ischemia but reduced the rise in resting tension. PAK-200 significantly improved the recovery of contractile force after reperfusion to about 70% of the preischemic value. PAK-200 was also shown to attenuate the decrease in tissue ATP during ischemia. Treatment of ventricular myocytes with an ischemia-mimetic solution resulted in depolarization of the mitochondrial membrane potential and an increase in cytoplasmic and mitochondrial Ca2+ concentrations. PAK-200 significantly delayed these changes. Thus, PAK-200 inhibits the cAMP-activated chloride current in cardiac muscle and may have protective effects against ischemia-reperfusion injury through novel mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Hikaru Tanaka
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University, 2-2-1 Miyama Funabashi, Chiba 274-8510, Japan; (I.N.); (M.T.); (J.K.); (S.H.)
| |
Collapse
|
2
|
Husti Z, Varró A, Baczkó I. Arrhythmogenic Remodeling in the Failing Heart. Cells 2021; 10:cells10113203. [PMID: 34831426 PMCID: PMC8623396 DOI: 10.3390/cells10113203] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/05/2021] [Accepted: 11/11/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic heart failure is a clinical syndrome with multiple etiologies, associated with significant morbidity and mortality. Cardiac arrhythmias, including ventricular tachyarrhythmias and atrial fibrillation, are common in heart failure. A number of cardiac diseases including heart failure alter the expression and regulation of ion channels and transporters leading to arrhythmogenic electrical remodeling. Myocardial hypertrophy, fibrosis and scar formation are key elements of arrhythmogenic structural remodeling in heart failure. In this article, the mechanisms responsible for increased arrhythmia susceptibility as well as the underlying changes in ion channel, transporter expression and function as well as alterations in calcium handling in heart failure are discussed. Understanding the mechanisms of arrhythmogenic remodeling is key to improving arrhythmia management and the prevention of sudden cardiac death in patients with heart failure.
Collapse
Affiliation(s)
- Zoltán Husti
- Department of Pharmacology and Pharmacotherapy, University of Szeged, 6720 Szeged, Hungary; (Z.H.); (A.V.)
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, 6720 Szeged, Hungary
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, University of Szeged, 6720 Szeged, Hungary; (Z.H.); (A.V.)
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, 6720 Szeged, Hungary
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research Network, 6720 Szeged, Hungary
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, University of Szeged, 6720 Szeged, Hungary; (Z.H.); (A.V.)
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, 6720 Szeged, Hungary
- Correspondence:
| |
Collapse
|
3
|
James AF. Enigmatic variations: The many facets of CFTR function in the heart. Acta Physiol (Oxf) 2020; 230:e13525. [PMID: 32562586 DOI: 10.1111/apha.13525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Andrew F. James
- School of Physiology Pharmacology & Neuroscience University of Bristol Bristol UK
| |
Collapse
|
4
|
CFTR deficiency causes cardiac dysplasia during zebrafish embryogenesis and is associated with dilated cardiomyopathy. Mech Dev 2020; 163:103627. [PMID: 32574800 DOI: 10.1016/j.mod.2020.103627] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/04/2020] [Accepted: 06/18/2020] [Indexed: 02/08/2023]
Abstract
Mutations in the CFTR gene cause cystic fibrosis (CF) with myocardial dysfunction. However, it remains unknown whether CF-related heart disease is a secondary effect of pulmonary disease, or an intrinsic primary defect in the heart. Here, we used zebrafish, which lack lung tissue, to investigate the role of CFTR in cardiogenesis. Our findings demonstrated that the loss of CFTR impairs cardiac development from the cardiac progenitor stage, resulting in cardiac looping defects, a dilated atrium, pericardial edema, and a decrease in heart rate. Furthermore, we found that cardiac development was perturbed in wild-type embryos treated with a gating-specific CFTR channel inhibitor, CFTRinh-172, at the blastula stage of development, but not at later stages. Gene expression analysis of blastulas indicated that transcript levels, including mRNAs associated with cardiovascular diseases, were significantly altered in embryos derived from cftr mutants relative to controls. To evaluate the role of CFTR in human heart failure, we performed a genetic association study on individuals with dilated cardiomyopathy and found that the I556V mutation in CFTR, which causes a channel defect, was associated with the disease. Similar to other well-studied channel-defective CFTR mutants, CFTR I556V mRNA failed to restore cardiac dysplasia in mutant embryos. The present study revealed an important role for the CFTR ion channel in regulating cardiac development during early embryogenesis, supporting the hypothesis that CF-related heart disease results from an intrinsic primary defect in the heart.
Collapse
|
5
|
Hansen TH, Yan Y, Ahlberg G, Vad OB, Refsgaard L, Dos Santos JL, Mutsaers N, Svendsen JH, Olesen MS, Bentzen BH, Schmitt N. A Novel Loss-of-Function Variant in the Chloride Ion Channel Gene Clcn2 Associates with Atrial Fibrillation. Sci Rep 2020; 10:1453. [PMID: 31996765 PMCID: PMC6989500 DOI: 10.1038/s41598-020-58475-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 01/15/2020] [Indexed: 11/09/2022] Open
Abstract
Atrial Fibrillation (AF) is the most common cardiac arrhythmia. Its pathogenesis is complex and poorly understood. Whole exome sequencing of Danish families with AF revealed a novel four nucleotide deletion c.1041_1044del in CLCN2 shared by affected individuals. We aimed to investigate the role of genetic variation of CLCN2 encoding the inwardly rectifying chloride channel ClC-2 as a risk factor for the development of familiar AF. The effect of the CLCN2 variant was evaluated by electrophysiological recordings on transiently transfected cells. We used quantitative PCR to assess CLCN2 mRNA expression levels in human atrial and ventricular tissue samples. The nucleotide deletion CLCN2 c.1041_1044del results in a frame-shift and premature stop codon. The truncated ClC-2 p.V347fs channel does not conduct current. Co-expression with wild-type ClC-2, imitating the heterozygote state of the patients, resulted in a 50% reduction in macroscopic current, suggesting an inability of truncated ClC-2 protein to form channel complexes with wild type channel subunits. Quantitative PCR experiments using human heart tissue from healthy donors demonstrated that CLCN2 is expressed across all four heart chambers. Our genetic and functional data points to a possible link between loss of ClC-2 function and an increased risk of developing AF.
Collapse
Affiliation(s)
- Thea Hyttel Hansen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,ALK-Abelló A/S, 2970, Hørsholm, Denmark
| | - Yannan Yan
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gustav Ahlberg
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Laboratory for Molecular Cardiology, Department of Cardiology, The Heart Centre, Righospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Oliver Bundgaard Vad
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Laboratory for Molecular Cardiology, Department of Cardiology, The Heart Centre, Righospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lena Refsgaard
- Laboratory for Molecular Cardiology, Department of Cardiology, The Heart Centre, Righospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Joana Larupa Dos Santos
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nancy Mutsaers
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Hastrup Svendsen
- Laboratory for Molecular Cardiology, Department of Cardiology, The Heart Centre, Righospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten Salling Olesen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Laboratory for Molecular Cardiology, Department of Cardiology, The Heart Centre, Righospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Bo Hjorth Bentzen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nicole Schmitt
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
6
|
Shu X, Fan Y, Li S, Jin Y, Xia C, Huang C. Anion binding and fluoride ion induced conformational changes in bisurea receptors. NEW J CHEM 2020. [DOI: 10.1039/c9nj05785d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Two types of bisurea receptors, containing either 2,6-substituted phenyl or 2,6-substituted pyridine, are prepared, and their anion binding properties are investigated.
Collapse
Affiliation(s)
- Xi Shu
- College of Chemistry
- Sichuan University
- Chengdu 610064
- P. R. China
| | - Yu Fan
- College of Chemistry
- Sichuan University
- Chengdu 610064
- P. R. China
| | - Shoujian Li
- College of Chemistry
- Sichuan University
- Chengdu 610064
- P. R. China
| | - Yongdong Jin
- College of Chemistry
- Sichuan University
- Chengdu 610064
- P. R. China
| | - Chuanqin Xia
- College of Chemistry
- Sichuan University
- Chengdu 610064
- P. R. China
| | - Chao Huang
- College of Chemistry
- Sichuan University
- Chengdu 610064
- P. R. China
| |
Collapse
|
7
|
Verkerk AO, Lodder EM, Wilders R. Aquaporin Channels in the Heart-Physiology and Pathophysiology. Int J Mol Sci 2019; 20:ijms20082039. [PMID: 31027200 PMCID: PMC6514906 DOI: 10.3390/ijms20082039] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/19/2019] [Accepted: 04/23/2019] [Indexed: 12/11/2022] Open
Abstract
Mammalian aquaporins (AQPs) are transmembrane channels expressed in a large variety of cells and tissues throughout the body. They are known as water channels, but they also facilitate the transport of small solutes, gasses, and monovalent cations. To date, 13 different AQPs, encoded by the genes AQP0–AQP12, have been identified in mammals, which regulate various important biological functions in kidney, brain, lung, digestive system, eye, and skin. Consequently, dysfunction of AQPs is involved in a wide variety of disorders. AQPs are also present in the heart, even with a specific distribution pattern in cardiomyocytes, but whether their presence is essential for proper (electro)physiological cardiac function has not intensively been studied. This review summarizes recent findings and highlights the involvement of AQPs in normal and pathological cardiac function. We conclude that AQPs are at least implicated in proper cardiac water homeostasis and energy balance as well as heart failure and arsenic cardiotoxicity. However, this review also demonstrates that many effects of cardiac AQPs, especially on excitation-contraction coupling processes, are virtually unexplored.
Collapse
Affiliation(s)
- Arie O Verkerk
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
- Department of Experimental Cardiology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Elisabeth M Lodder
- Department of Experimental Cardiology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| |
Collapse
|
8
|
Macrocyclic bis-urea receptor: Synthesis, crystal structure and phosphate binding properties. Tetrahedron Lett 2019. [DOI: 10.1016/j.tetlet.2019.02.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
9
|
Bader Ul Ain H, Saeed F, Ahmad N, Imran A, Niaz B, Afzaal M, Imran M, Tufail T, Javed A. Functional and health-endorsing properties of wheat and barley cell wall’s non-starch polysaccharides. INTERNATIONAL JOURNAL OF FOOD PROPERTIES 2018. [DOI: 10.1080/10942912.2018.1489837] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Huma Bader Ul Ain
- Institute of Home & Food Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Farhan Saeed
- Institute of Home & Food Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Nazir Ahmad
- Institute of Home & Food Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Ali Imran
- Institute of Home & Food Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Bushra Niaz
- Institute of Home & Food Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muhammad Afzaal
- Institute of Home & Food Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muhammad Imran
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore-Lahore, Lahore, Pakistan
| | - Tabussam Tufail
- Institute of Home & Food Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Ahsan Javed
- Institute of Home & Food Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| |
Collapse
|
10
|
Xiao GS, Zhang YH, Wang Y, Sun HY, Baumgarten CM, Li GR. Noradrenaline up-regulates volume-regulated chloride current by PKA-independent cAMP/exchange protein activated by cAMP pathway in human atrial myocytes. Br J Pharmacol 2018; 175:3422-3432. [PMID: 29900525 DOI: 10.1111/bph.14392] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 05/29/2018] [Accepted: 06/05/2018] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND AND PURPOSE Adrenergic regulation of cell volume-regulated chloride current (ICl.vol ) is species-dependent. The present study investigates the mechanism underlying adrenergic regulation of ICl.vol in human atrial myocytes. EXPERIMENTAL APPROACH Conventional whole-cell patch voltage-clamp techniques were used to record membrane current in human atrial myocytes. ICl.vol was evoked by hyposmotic bath solution (0.6 times isosmotic, 0.6 T). KEY RESULTS ICl.vol was augmented by noradrenaline (1 μM) during cell swelling in 0.6 T but not under isosmotic (1 T) conditions. Up-regulation of ICl.vol in 0.6 T was blocked by the β-adrenoceptor antagonist propranolol (2 μM), but not by the α1 -adrenoceptor antagonist prazosin (2 μM). This β-adrenergic response involved cAMP but was independent of PKA; the protein kinase inhibitor H-89 (2 μM) or PKI (10 μM in pipette solution) failed to prevent ICl.vol up-regulation by noradrenaline. Moreover, the PI3K/PKB inhibitor LY294002 (50 μM) and the PKG inhibitor KT5823 (10 μM) did not affect noradrenaline-induced increases in ICl.vol . Interestingly, the exchange protein directly activated by cAMP (Epac) agonist 8-pCPT-2'-O-Me-cAMP (50 μM) also up-regulated ICl.vol , and the noradrenaline-induced increase of ICl.vol in 0.6 T was reversed or prevented by the Epac inhibitor ESI-09 (10 μM). CONCLUSION AND IMPLICATIONS These data show that ICl.vol evoked by cell swelling of human atrial myocytes is up-regulated by noradrenaline via a PKA-independent cAMP/Epac pathway in human atrial myocytes. cAMP/Epac-induced ICl.vol is expected to shorten action potential duration during human atrial myocytes swelling and may be involved in abnormal cardiac electrical activity during cardiac pathologies that evoke β-adrenoceptor signalling.
Collapse
Affiliation(s)
- Guo-Sheng Xiao
- Xiamen Cardiovascular Hospital, Medical College of Xiamen University, Xiamen, Fujian, China.,Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, China
| | - Yan-Hui Zhang
- Xiamen Cardiovascular Hospital, Medical College of Xiamen University, Xiamen, Fujian, China.,Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, China
| | - Yan Wang
- Xiamen Cardiovascular Hospital, Medical College of Xiamen University, Xiamen, Fujian, China
| | - Hai-Ying Sun
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, China
| | - Clive M Baumgarten
- Department of Physiology and Biophysics, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA
| | - Gui-Rong Li
- Xiamen Cardiovascular Hospital, Medical College of Xiamen University, Xiamen, Fujian, China.,Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, China
| |
Collapse
|
11
|
Qiu LY, Duan GL, Yan YF, Li YY, Wang H, Xiao L, Liao ZP, Chen HP. Sasanquasaponin induces increase of Cl‑/HCO3‑ exchange of anion exchanger 3 and promotes intracellular Cl‑ efflux in hypoxia/reoxygenation cardiomyocytes. Mol Med Rep 2017; 16:2953-2961. [PMID: 28677776 DOI: 10.3892/mmr.2017.6882] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 04/21/2017] [Indexed: 11/06/2022] Open
Abstract
Anion exchanger 3 (AE3) is known to serve crucial roles in maintaining intracellular chloride homeostasis by facilitating the reversible electroneutral exchange of Cl‑ for HCO3‑ across the plasma membrane. Our previous studies reported that sasanquasaponin (SQS) can inhibit hypoxia/reoxygenation (H/R)‑induced elevation of intracellular Cl‑ concentration ([Cl‑]i) and elicit cardioprotection by favoring Cl‑/HCO3‑ exchange of AE3. However, the molecular basis for SQS‑induced increase of Cl‑/HCO3‑ exchange of AE3 remains unclear. The present study demonstrated that SQS activates protein kinase Cε (PKCε) and stimulates the phosphorylation of AE3 in H9c2 cells. Notably, SQS‑induced AE3 phosphorylation was blocked by the PKCε selective inhibitor εV1‑2, and a S67A mutation of AE3, indicating that SQS could promote phosphorylation of Ser67 of AE3 via a PKCε‑dependent regulatory signaling pathway. Additionally, both inhibition of PKCε by εV1‑2 and S67A mutation of AE3 eradicated the SQS‑induced increase of AE3 activity, reversed the inhibitory effect of SQS on H/R‑induced elevation of [Cl‑]i, Ca2+ overload and generation of reactive oxygen species, and eliminated SQS‑induced cardioprotection. In conclusion, PKCε‑dependent phosphorylation of serine 67 on AE3 may be responsible for the increase of Cl‑/HCO3‑ exchange of AE3 and intracellular chloride efflux by SQS, and contributes to the cardioprotection of SQS against H/R in H9c2 cells.
Collapse
Affiliation(s)
- Ling-Yu Qiu
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Guang-Ling Duan
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yu-Feng Yan
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, P.R. China
| | - Yuan-Yuan Li
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Huan Wang
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ling Xiao
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhang-Ping Liao
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - He-Ping Chen
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
12
|
Zhang XG, Zhao L, Zhang Y, Li YY, Wang H, Duan GL, Xiao L, Li XR, Chen HP. Extracellular Cl --free-induced cardioprotection against hypoxia/reoxygenation is associated with attenuation of mitochondrial permeability transition pore. Biomed Pharmacother 2016; 86:637-644. [PMID: 28033580 DOI: 10.1016/j.biopha.2016.12.048] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/04/2016] [Accepted: 12/12/2016] [Indexed: 12/21/2022] Open
Abstract
The isotonic substitution of extracellular chloride by gluconate (extracellular Cl--free) has been demonstrated to elicit cardioprotection by attenuating ischaemia/reperfusion-induced elevation of intracellular chloride ion concentration ([Cl-]i). However, the downstream mechanism underlying the cardioprotective effect of extracellular Cl--free is not fully established. Here, it was investigated whether extracellular Cl--free attenuates mitochondrial dysfunction after hypoxia/reoxygenation (H/R) and whether mitochondrial permeability transition pore (mPTP) plays a key role in the extracellular Cl--free cardioprotection. H9c2 cells were incubated with or without Cl--free solution, in which Cl- was replaced with equimolar gluconate, during H/R. The involvement of mPTP was determined with atractyloside (Atr), a specific mPTP opener. The results showed that extracellular Cl--free attenuated H/R-induced the elevation of [Cl-]i, accompanied by increase of cell viability and reduction of lactate dehydrogenase release. Moreover, extracellular Cl--free inhibited mPTP opening, and improved mitochondria function, as indicated by preserved mitochondrial membrane potential and respiratory chain complex activities, decreased mitochondrial reactive oxygen species generation, and increased ATP content. Intriguingly, pharmacologically opening of the mPTP with Atr attenuated all the protective effects caused by extracellular Cl--free, including suppression of mPTP opening, maintenance of mitochondrial membrane potential, and subsequent improvement of mitochondrial function. These results indicated that extracellular Cl--free protects mitochondria from H/R injury in H9c2 cells and inhibition of mPTP opening is a crucial step in mediating the cardioprotection of extracellular Cl--free.
Collapse
Affiliation(s)
- Xian-Gui Zhang
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, People's Republic of China
| | - Le Zhao
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, People's Republic of China
| | - Yi Zhang
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, People's Republic of China
| | - Yuan-Yuan Li
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, People's Republic of China
| | - Huan Wang
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, People's Republic of China
| | - Guang-Ling Duan
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, People's Republic of China
| | - Lin Xiao
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, People's Republic of China
| | - Xiao-Ran Li
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, People's Republic of China
| | - He-Ping Chen
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, People's Republic of China.
| |
Collapse
|
13
|
Sasanquasaponin-induced cardioprotection involves inhibition of mPTP opening via attenuating intracellular chloride accumulation. Fitoterapia 2016; 116:1-9. [PMID: 27838499 DOI: 10.1016/j.fitote.2016.11.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 10/26/2016] [Accepted: 11/06/2016] [Indexed: 11/22/2022]
Abstract
Sasanquasaponin (SQS) has been reported to elicit cardioprotection by suppressing hypoxia/reoxygenation (H/R)-induced elevation of intracellular chloride ion concentration ([Cl-]i). Given that the increased [Cl-]i is involved to modulate the mitochondrial permeability transition pore (mPTP), we herein sought to further investigate the role of mPTP in the cardioprotective effect of SQS on H/R injury. H9c2 cells were incubated for 24h with or without 10μM SQS followed by H/R. The involvement of mPTP was determined with a specific mPTP agonist atractyloside (ATR). The results showed that SQS attenuated H/R-induced the elevation of [Cl-]i, accompanied by reduction of lactate dehydrogenase release and increase of cell viability. Moreover, SQS suppressed mPTP opening, and protected mitochondria, as indicated by preserved mitochondrial membrane potential and respiratory chain complex activities, decreased mitochondrial reactive oxygen species generation, and increased ATP content. Interestingly, extracellular Cl--free condition created by replacing Cl- with equimolar gluconate resulted in a decrease in [Cl-]i and induced protective effects similar to SQS preconditioning, whereas pharmacologically opening of the mPTP with ATR abolished all the protective effects induced by SQS or Cl--free, including suppression of mPTP opening, maintenance of mitochondrial membrane potential, and subsequent improvement of mitochondrial function. The above results allow us to conclude that SQS-induced cardioprotection may be mediated by preserving the mitochondrial function through preventing mPTP opening via inhibition of H/R-induced elevation of [Cl-]i.
Collapse
|
14
|
Lu YY, Li NL, Jia LP, Ma RN, Jia WL, Tao XQ, Cui H, Wang HS. The synthesis of Ag@CQDs composite and its electrochemiluminescence application for the highly selective and sensitive detection of chloride. J Electroanal Chem (Lausanne) 2016. [DOI: 10.1016/j.jelechem.2016.05.045] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
15
|
Protein kinase C enhances the swelling-induced chloride current in human atrial myocytes. ACTA ACUST UNITED AC 2016; 36:383-388. [PMID: 27376808 DOI: 10.1007/s11596-016-1596-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 04/19/2016] [Indexed: 12/18/2022]
Abstract
Swelling-activated chloride currents (ICl.swell) are thought to play a role in several physiologic and pathophysiologic processes and thus represent a target for therapeutic approaches. However, the mechanism of ICl.swell regulation remains unclear. In this study, we used the whole-cell patch-clamp technique to examine the role of protein kinase C (PKC) in the regulation of ICl.swell in human atrial myocytes. Atrial myocytes were isolated from the right atrial appendages of patients undergoing coronary artery bypass and enzymatically dissociated. ICl.swell was evoked in hypotonic solution and recorded using the whole-cell patch-clamp technique. The PKC agonist phorbol dibutyrate (PDBu) enhanced ICl.swell in a concentration-dependent manner, which was reversed in isotonic solution and by a chloride current inhibitor, 9-anthracenecarboxylicacid. Furthermore, the PKC inhibitor bis-indolylmaleimide attenuated the effect and 4α-PDBu, an inactive PDBu analog, had no effect on ICl.swell. These results, obtained using the whole-cell patch-clamp technique, demonstrate the ability of PKC to activate ICl,swell in human atrial myocytes. This observation was consistent with a previous study using a single-channel patch-clamp technique, but differed from some findings in other species.
Collapse
|
16
|
Exchange of chemical signals between cardiac cells. Fundamental role on cell communication and metabolic cooperation. Exp Cell Res 2016; 346:130-6. [PMID: 27237090 DOI: 10.1016/j.yexcr.2016.05.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 05/09/2016] [Accepted: 05/10/2016] [Indexed: 01/14/2023]
Abstract
The exchange of chemical signals between cardiac cells and its relevance for cell communication and metabolic cooperation was reviewed. The role of gap junctions on the transfer of chemical information was discussed as well as the different factors involved in its regulation including changes in cell volume, high glucose, activation of the renin angiotensin aldosterone system including the intracrine effect of renin and angiotensin II on chemical coupling and cardiac energetics. Finally, the possible role of epigenetic changes of the renin angiotensin aldosterone system (RAAS) on the expression of components of the RAAS was discussed. The evidence available leads to the conception of the heart as a metabolic syncytium in which glucose as well nucleotides and hormones can flow from cell-to-cell though gap junctions, providing a new vision of how alterations in metabolic cooperation can induce cardiac diseases. These findings represent a stimulus for future research in this important area of cardiac physiology and pathology.
Collapse
|
17
|
Qiu LY, Chen HP, Yan YF, Li YY, Wang H, Liao ZP, Huang QR. Sasanquasaponin promotes cellular chloride efflux and elicits cardioprotection via the PKCε pathway. Mol Med Rep 2016; 13:3597-603. [PMID: 26956211 DOI: 10.3892/mmr.2016.4984] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 02/06/2016] [Indexed: 11/06/2022] Open
Abstract
Sasanquasaponin (SQS) is an active component of Camellia oleifera Abel. A recent study by our group demonstrated that SQS was able to inhibit ischemia/reperfusion‑induced elevation of the intracellular chloride ion concentration ([Cl‑]i) and exerted cardioprotective effects; however, the underlying intracellular signal transduction mechanisms have yet to be elucidated. As protein kinase C ε (PKCε) is able to mediate Cl‑ homeostasis, the present study investigated its possible involvement in the effects of SQS on cardiomyocytes subjected to ischemia/reperfusion injury. Cardiomyocytes were pre‑treated with or without SQS or SQS plus εV1‑2, a selective PKCε inhibitor, followed by simulated ischemia/reperfusion (sI/R). The effects on cell viability, PKCε phosphorylation levels, [Cl‑]i, mitochondrial membrane potential and reactive oxygen species (ROS) production were assessed using an MTS assay, western blot analysis, colorimetric assays and flow cytometry. The results revealed that treatment with SQS prior to sI/R increased the viability of cardiomyocytes, and efficiently attenuated lactate dehydrogenase and creatine phosphokinase release induced by sI/R. In addition, SQS promoted PKCε phosphorylation and inhibited sI/R‑induced elevation of [Cl‑]i, paralleled by the attenuation of mitochondrial membrane potential loss and ROS generation. However, when the cardiomyocytes were treated with εV1‑2 prior to SQS pre‑conditioning, the cardioprotection induced by SQS was reduced and the inhibitory effects of SQS on sI/R‑induced elevation of [Cl‑]i, production of ROS and loss of mitochondrial membrane potential were also attenuated. These findings indicated that SQS may inhibit sI/R‑induced elevation of [Cl‑]i through the PKCε signaling pathway to elicit cardioprotection in cultured cardiomyocytes.
Collapse
Affiliation(s)
- Ling-Yu Qiu
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - He-Ping Chen
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yu-Feng Yan
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yuan-Yuan Li
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Huan Wang
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhang-Ping Liao
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Qi-Ren Huang
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
18
|
Ye Z, Wu MM, Wang CY, Li YC, Yu CJ, Gong YF, Zhang J, Wang QS, Song BL, Yu K, Hartzell HC, Duan DD, Zhao D, Zhang ZR. Characterization of Cardiac Anoctamin1 Ca²⁺-Activated Chloride Channels and Functional Role in Ischemia-Induced Arrhythmias. J Cell Physiol 2015; 230:337-46. [PMID: 24962810 DOI: 10.1002/jcp.24709] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 06/20/2014] [Indexed: 12/17/2022]
Abstract
Anoctamin1 (ANO1) encodes a Ca(2+)-activated chloride (Cl(-)) channel (CaCC) in variety tissues of many species. Whether ANO1 expresses and functions as a CaCC in cardiomyocytes remain unknown. The objective of this study is to characterize the molecular and functional expression of ANO1 in cardiac myocytes and the role of ANO1-encoded CaCCs in ischemia-induced arrhythmias in the heart. Quantitative real-time RT-PCR, immunofluorescence staining assays, and immunohistochemistry identified the molecular expression, location, and distribution of ANO1 in mouse ventricular myocytes (mVMs). Patch-clamp recordings combined with pharmacological analyses found that ANO1 was responsible for a Ca(2+)-activated Cl(-) current (I(Cl.Ca)) in cardiomyocytes. Myocardial ischemia led to a significant increase in the current density of I(Cl.Ca), which was inhibited by a specific ANO1 inhibitor, T16A(inh)-A01, and an antibody targeting at the pore area of ANO1. Moreover, cardiomyocytes isolated from mice with ischemia-induced arrhythmias had an accelerated early phase 1 repolarization of action potentials (APs) and a deeper "spike and dome" compared to control cardiomyocytes from non-ischemia mice. Application of the antibody targeting at ANO1 pore prevented the ischemia-induced early phase 1 repolarization acceleration and caused a much shallower "spike and dome". We conclude that ANO1 encodes CaCC and plays a significant role in the phase 1 repolarization of APs in mVMs. The ischemia-induced increase in ANO1 expression may be responsible for the increased density of I(Cl.Ca) in the ischemic heart and may contribute, at least in part, to ischemia-induced arrhythmias.
Collapse
Affiliation(s)
- Zhen Ye
- Departments of Clinical Pharmacy and Cardiology, The 2nd Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P. R. China
| | - Ming-Ming Wu
- Departments of Clinical Pharmacy and Cardiology, The 2nd Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P. R. China
| | - Chun-Yu Wang
- Departments of Clinical Pharmacy and Cardiology, The 2nd Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P. R. China
| | - Yan-Chao Li
- Departments of Clinical Pharmacy and Cardiology, The 2nd Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P. R. China
| | - Chang-Jiang Yu
- Departments of Clinical Pharmacy and Cardiology, The 2nd Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P. R. China
| | - Yuan-Feng Gong
- Departments of Clinical Pharmacy and Cardiology, The 2nd Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P. R. China
| | - Jun Zhang
- Departments of Clinical Pharmacy and Cardiology, The 2nd Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P. R. China
| | - Qiu-Shi Wang
- Departments of Clinical Pharmacy and Cardiology, The 2nd Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P. R. China
| | - Bin-Lin Song
- Departments of Clinical Pharmacy and Cardiology, The 2nd Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P. R. China
| | - Kuai Yu
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia
| | - H Criss Hartzell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia
| | - Dayue Darrel Duan
- Laboratory of Cardiovascular Phenomics, Department of Pharmacology, Center for Molecular Medicine, School of Medicine University of Nevada, Reno, Nevada
| | - Dan Zhao
- Departments of Clinical Pharmacy and Cardiology, The 2nd Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P. R. China
| | - Zhi-Ren Zhang
- Departments of Clinical Pharmacy and Cardiology, The 2nd Affiliated Hospital, Harbin Medical University, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, P. R. China
| |
Collapse
|
19
|
Leblanc N, Forrest AS, Ayon RJ, Wiwchar M, Angermann JE, Pritchard HAT, Singer CA, Valencik ML, Britton F, Greenwood IA. Molecular and functional significance of Ca(2+)-activated Cl(-) channels in pulmonary arterial smooth muscle. Pulm Circ 2015; 5:244-68. [PMID: 26064450 DOI: 10.1086/680189] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 07/22/2014] [Indexed: 12/31/2022] Open
Abstract
Increased peripheral resistance of small distal pulmonary arteries is a hallmark signature of pulmonary hypertension (PH) and is believed to be the consequence of enhanced vasoconstriction to agonists, thickening of the arterial wall due to remodeling, and increased thrombosis. The elevation in arterial tone in PH is attributable, at least in part, to smooth muscle cells of PH patients being more depolarized and displaying higher intracellular Ca(2+) levels than cells from normal subjects. It is now clear that downregulation of voltage-dependent K(+) channels (e.g., Kv1.5) and increased expression and activity of voltage-dependent (Cav1.2) and voltage-independent (e.g., canonical and vanilloid transient receptor potential [TRPC and TRPV]) Ca(2+) channels play an important role in the functional remodeling of pulmonary arteries in PH. This review focuses on an anion-permeable channel that is now considered a novel excitatory mechanism in the systemic and pulmonary circulations. It is permeable to Cl(-) and is activated by a rise in intracellular Ca(2+) concentration (Ca(2+)-activated Cl(-) channel, or CaCC). The first section outlines the biophysical and pharmacological properties of the channel and ends with a description of the molecular candidate genes postulated to encode for CaCCs, with particular emphasis on the bestrophin and the newly discovered TMEM16 and anoctamin families of genes. The second section provides a review of the various sources of Ca(2+) activating CaCCs, which include stimulation by mobilization from intracellular Ca(2+) stores and Ca(2+) entry through voltage-dependent and voltage-independent Ca(2+) channels. The third and final section summarizes recent findings that suggest a potentially important role for CaCCs and the gene TMEM16A in PH.
Collapse
Affiliation(s)
- Normand Leblanc
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Abigail S Forrest
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Ramon J Ayon
- Department of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Michael Wiwchar
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Jeff E Angermann
- School of Community Health Sciences, University of Nevada, Reno, Nevada, USA
| | - Harry A T Pritchard
- Vascular Biology Research Centre, Institute of Cardiovascular and Cell Sciences, St. George's University of London, London, United Kingdom
| | - Cherie A Singer
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Maria L Valencik
- Department of Biochemistry and Molecular Biology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Fiona Britton
- Department of Physiology, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Iain A Greenwood
- Vascular Biology Research Centre, Institute of Cardiovascular and Cell Sciences, St. George's University of London, London, United Kingdom
| |
Collapse
|
20
|
Wezenberg SJ, Vlatković M, Kistemaker JCM, Feringa BL. Multi-State Regulation of the Dihydrogen Phosphate Binding Affinity to a Light- and Heat-Responsive Bis-Urea Receptor. J Am Chem Soc 2014; 136:16784-7. [DOI: 10.1021/ja510700j] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Sander J. Wezenberg
- Centre
for Systems Chemistry,
Stratingh Institute for Chemistry, University of Groningen, Nijenborgh
4, 9747 AG, Groningen, The Netherlands
| | - Matea Vlatković
- Centre
for Systems Chemistry,
Stratingh Institute for Chemistry, University of Groningen, Nijenborgh
4, 9747 AG, Groningen, The Netherlands
| | - Jos C. M. Kistemaker
- Centre
for Systems Chemistry,
Stratingh Institute for Chemistry, University of Groningen, Nijenborgh
4, 9747 AG, Groningen, The Netherlands
| | - Ben L. Feringa
- Centre
for Systems Chemistry,
Stratingh Institute for Chemistry, University of Groningen, Nijenborgh
4, 9747 AG, Groningen, The Netherlands
| |
Collapse
|
21
|
Li Y, Du X. Effects of α1-adrenoceptor agonist phenylephrine on swelling-activated chloride currents in human atrial myocytes. J Membr Biol 2014; 248:7-18. [PMID: 25155614 DOI: 10.1007/s00232-014-9723-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 08/14/2014] [Indexed: 10/24/2022]
Abstract
Swelling-activated chloride currents (ICl.swell) play an important role in cardiac electrophysiology and arrhythmogenesis. However, the regulation of these currents has not been clarified to date. In this research, we focused on the function of phenylephrine, an α1-adrenoceptor agonist, in the regulation of I(Cl.swell) in human atrial myocytes. We recorded I(Cl.swell) evoked by a hypotonic bath solution with the whole-cell patch-clamp technique. We found that I(Cl.swell) increased over time, and it was difficult to achieve absolute steady state. Phenylephrine potentiated I(Cl.swell) from -1.00 ± 0.51 pA/pF at -90 mV and 2.58 ± 1.17 pA/pF at +40 mV to -1.46 ± 0.70 and 3.84 ± 1.67 pA/pF, respectively (P < 0.05, n = 6), and the upward trend in ICl.swell was slowed after washout. This effect was concentration-dependent, and the α1-adrenoceptor antagonist prazosin shifted the dose-effect curve rightward. Addition of prazosin or the protein kinase C (PKC) inhibitor bisindolylmaleimide (BIM) attenuated the effect of phenylephrine. The PKC activator phorbol 12,13-dibutyrate (PDBu) activated I(Cl.swell) from -1.69 ± 1.67 pA/pF at -90 mV and 5.58 ± 6.36 pA/pF at +40 mV to -2.41 ± 1.95 pA/pF and 7.05 ± 6.99 pA/pF, respectively (P < 0.01 at -90 mV and P < 0.05 at +40 mV; n = 6). In conclusion, the α1-adrenoceptor agonist phenylephrine augmented I(Cl.swell), a result that differs from previous reports in other animal species. The effect was attenuated by BIM and mimicked by PDBu, which indicates that phenylephrine might modulate I(Cl,swell) in a PKC-dependent manner.
Collapse
Affiliation(s)
- Yetao Li
- Department of Cardiovascular Surgery, People's Hospital of Guizhou Province, Guiyang, 550002, China
| | | |
Collapse
|
22
|
Adkins GB, Curtis MJ. Potential role of cardiac chloride channels and transporters as novel therapeutic targets. Pharmacol Ther 2014; 145:67-75. [PMID: 25160469 DOI: 10.1016/j.pharmthera.2014.08.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 08/01/2014] [Indexed: 02/06/2023]
Abstract
The heart and blood vessels express a range of anion currents (e.g. ICl.PKA) and symporter/antiporters (e.g. Cl(-)/HCO3(-) exchanger) that translocate chloride (Cl(-)). They have been proposed to contribute to a variety of physiological processes including cellular excitability, cell volume homeostasis and apoptosis. Additionally there is evidence that Cl(-) currents or transporters may play a role in cardiac pathophysiology. Arrhythmogenesis, the process of cardiac ischaemic preconditioning, and the adaptive remodelling process in myocardial hypertrophy and heart failure have all been linked to such channels or transporters. We have explored the possibility that selective targeting of one or more of these may provide benefit in cardiovascular disease. Existing evidence points to an emerging role of cardiac cell anion channels as potential therapeutic targets, the 'disease-specificity' of which may represent a substantial improvement on current targets. However, the limitations of current techniques hitherto applied (such as developmental compensation in gene-modified animals) and pharmacological agents (which do not at present possess sufficient selectivity for the adequate probing of function) have thus far hindered translation to the introduction of new therapy.
Collapse
|
23
|
Diaz RJ, Harvey K, Boloorchi A, Hossain T, Hinek A, Backx PH, Wilson GJ. Enhanced cell volume regulation: a key mechanism in local and remote ischemic preconditioning. Am J Physiol Cell Physiol 2014; 306:C1191-9. [DOI: 10.1152/ajpcell.00259.2013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have previously shown that ischemic preconditioning (IPC) protection against necrosis in whole hearts and in both fresh and cultured cardiomyocytes, as well as the improved regulatory volume decrease to hypoosmotic swelling in cardiomyocytes, is abrogated through Cl− channel blockade, pointing to a role for enhanced cell volume regulation in IPC. To further define this cardioprotective mechanism, cultured rabbit ventricular cardiomyocytes were preconditioned either by 10-min simulated ischemia (SI) followed by 10-min simulated reperfusion (SR), by 10-min exposure/10-min washout of remote IPC (rIPC) plasma dialysate (from rabbits subjected to repetitive limb ischemia), or by adenoviral transfection with the constitutively active PKC-ε gene. These interventions were done before cardiomyocytes were subjected to either 60- or 75-min SI/60-min SR to assess cell necrosis (by trypan blue staining), 30-min SI to assess ischemic cell swelling, or 30-min hypoosmotic (200 mosM) stress to assess cell volume regulation. Necrosis after SI/SR and both SI- and hypoosmotic stress-induced swelling was reduced in preconditioned cardiomyocytes compared with control cardiomyocytes (neither preconditioned nor transfected). These effects on necrosis and cell swelling were blocked by either Cl− channel blockade or dominant negative knockdown of inwardly rectifying K+ channels with adenoviruses, suggesting that Cl− and K+ movements across the sarcolemma are critical for cell volume regulation and, thereby, cell survival under hypoxic/ischemic conditions. Our results define enhanced cell volume regulation as a key common mechanism of cardioprotection by preconditioning in cardiomyocytes.
Collapse
Affiliation(s)
- Roberto J. Diaz
- Division of Physiology and Experimental Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Pediatric Laboratory Medicine and Pathology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kordan Harvey
- Division of Physiology and Experimental Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; and
| | - Azadeh Boloorchi
- Division of Physiology and Experimental Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; and
| | - Taneya Hossain
- Division of Physiology and Experimental Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Alina Hinek
- Division of Physiology and Experimental Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Peter H. Backx
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Gregory J. Wilson
- Division of Physiology and Experimental Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Pediatric Laboratory Medicine and Pathology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; and
| |
Collapse
|
24
|
Bi MM, Hong S, Zhou HY, Wang HW, Wang LN, Zheng YJ. Chloride channelopathies of ClC-2. Int J Mol Sci 2013; 15:218-49. [PMID: 24378849 PMCID: PMC3907807 DOI: 10.3390/ijms15010218] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 11/14/2013] [Accepted: 12/16/2013] [Indexed: 12/15/2022] Open
Abstract
Chloride channels (ClCs) have gained worldwide interest because of their molecular diversity, widespread distribution in mammalian tissues and organs, and their link to various human diseases. Nine different ClCs have been molecularly identified and functionally characterized in mammals. ClC-2 is one of nine mammalian members of the ClC family. It possesses unique biophysical characteristics, pharmacological properties, and molecular features that distinguish it from other ClC family members. ClC-2 has wide organ/tissue distribution and is ubiquitously expressed. Published studies consistently point to a high degree of conservation of ClC-2 function and regulation across various species from nematodes to humans over vast evolutionary time spans. ClC-2 has been intensively and extensively studied over the past two decades, leading to the accumulation of a plethora of information to advance our understanding of its pathophysiological functions; however, many controversies still exist. It is necessary to analyze the research findings, and integrate different views to have a better understanding of ClC-2. This review focuses on ClC-2 only, providing an analytical overview of the available literature. Nearly every aspect of ClC-2 is discussed in the review: molecular features, biophysical characteristics, pharmacological properties, cellular function, regulation of expression and function, and channelopathies.
Collapse
Affiliation(s)
- Miao Miao Bi
- Department of Ophthalmology, the Second Hospital of Jilin University, Jilin University, Changchun 130041, Jilin, China.
| | - Sen Hong
- Department of Ophthalmology, the Second Hospital of Jilin University, Jilin University, Changchun 130041, Jilin, China.
| | - Hong Yan Zhou
- Department of Ophthalmology, the Second Hospital of Jilin University, Jilin University, Changchun 130041, Jilin, China.
| | - Hong Wei Wang
- Department of Ophthalmology, the Second Hospital of Jilin University, Jilin University, Changchun 130041, Jilin, China.
| | - Li Na Wang
- Department of Ophthalmology, the Second Hospital of Jilin University, Jilin University, Changchun 130041, Jilin, China.
| | - Ya Juan Zheng
- Department of Ophthalmology, the Second Hospital of Jilin University, Jilin University, Changchun 130041, Jilin, China.
| |
Collapse
|
25
|
Abstract
Forward genetic studies have identified several chloride (Cl-) channel genes, including CFTR, ClC-2, ClC-3, CLCA, Bestrophin, and Ano1, in the heart. Recent reverse genetic studies using gene targeting and transgenic techniques to delineate the functional role of cardiac Cl- channels have shown that Cl- channels may contribute to cardiac arrhythmogenesis, myocardial hypertrophy and heart failure, and cardioprotection against ischemia reperfusion. The study of physiological or pathophysiological phenotypes of cardiac Cl- channels, however, is complicated by the compensatory changes in the animals in response to the targeted genetic manipulation. Alternatively, tissue-specific conditional or inducible knockout or knockin animal models may be more valuable in the phenotypic studies of specific Cl- channels by limiting the effect of compensation on the phenotype. The integrated function of Cl- channels may involve multiprotein complexes of the Cl- channel subproteome. Similar phenotypes can be attained from alternative protein pathways within cellular networks, which are influenced by genetic and environmental factors. The phenomics approach, which characterizes phenotypes as a whole phenome and systematically studies the molecular changes that give rise to particular phenotypes achieved by modifying the genotype under the scope of genome/proteome/phenome, may provide more complete understanding of the integrated function of each cardiac Cl- channel in the context of health and disease.
Collapse
Affiliation(s)
- Dayue Darrel Duan
- The Laboratory of Cardiovascular Phenomics, Department of Pharmacology, University of Nevada, School of Medicine, Reno, Nevada, USA.
| |
Collapse
|
26
|
Ramsay WJ, Ronson TK, Clegg JK, Nitschke JR. Bidirectional Regulation of Halide Binding in a Heterometallic Supramolecular Cube. Angew Chem Int Ed Engl 2013; 52:13439-43. [DOI: 10.1002/anie.201307478] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Indexed: 01/07/2023]
|
27
|
Ramsay WJ, Ronson TK, Clegg JK, Nitschke JR. Bidirectional Regulation of Halide Binding in a Heterometallic Supramolecular Cube. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201307478] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
28
|
Zhang H, Zhu L, Zuo W, Luo H, Mao J, Ye D, Li Y, Liu S, Wei Y, Ye W, Chen L, Wang L. The ClC-3 chloride channel protein is a downstream target of cyclin D1 in nasopharyngeal carcinoma cells. Int J Biochem Cell Biol 2013; 45:672-83. [DOI: 10.1016/j.biocel.2012.12.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 12/16/2012] [Indexed: 12/26/2022]
|
29
|
Epac activator critically regulates action potential duration by decreasing potassium current in rat adult ventricle. J Mol Cell Cardiol 2013; 57:96-105. [PMID: 23376036 DOI: 10.1016/j.yjmcc.2013.01.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 12/21/2012] [Accepted: 01/17/2013] [Indexed: 02/08/2023]
Abstract
Sympathetic stimulation is an important modulator of cardiac function via the classic cAMP-dependent signaling pathway, PKA. Recently, this paradigm has been challenged by the discovery of a family of guanine nucleotide exchange proteins directly activated by cAMP (Epac), acting in parallel to the classic signaling pathway. In cardiac myocytes, Epac activation is known to modulate Ca(2+) cycling yet their actions on cardiac ionic currents remain poorly characterized. This study attempts to address this paucity of information using the patch clamp technique to record action potential (AP) and ionic currents on rat ventricular myocytes. Epac was selectively activated by 8-CPT-AM (acetoxymethyl ester form of 8-CPT). AP amplitude, maximum depolarization rate and resting membrane amplitude were unaltered by 8-CPT-AM, strongly suggesting that Na(+) current and inward rectifier K(+) current are not regulated by Epac. In contrast, AP duration was significantly increased by 8-CPT-AM (prolongation of duration at 50% and 90% of repolarization by 41±10% and 43±8% respectively, n=11). L-type Ca(2+) current density was unaltered by 8-CPT-AM (n=16) so this cannot explain the action potential lengthening. However, the steady state component of K(+) current was significantly inhibited by 8-CPT-AM (-38±6%, n=15), while the transient outward K(+) current was unaffected by 8-CPT-AM. These effects were PKA-independent since they were observed in the presence of PKA inhibitor KT5720. Isoprenaline (100nM) induced a significant prolongation of AP duration, even in the presence of KT5720. This study provides the first evidence that the cAMP-binding protein Epac critically modulates cardiac AP duration by decreasing steady state K(+) current. These observations may be relevant to diseases in which Epac is upregulated, like cardiac hypertrophy.
Collapse
|
30
|
Yamazaki J, Okamura K, Uehara K, Hatta M. CLCA splicing isoform associated with adhesion through β1-integrin and its scaffolding protein: specific expression in undifferentiated epithelial cells. J Biol Chem 2013; 288:4831-43. [PMID: 23297403 DOI: 10.1074/jbc.m112.396481] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We previously found that a rat CLCA homologue (rCLCA-f) modulates Ca(2+)-dependent Cl(-) transport in the ductal cells of the rat submandibular gland. CLCA proteins have been shown to be multifunctional, with roles in, for example, cell adhesion. Here, we describe the mRNA and protein expressions of a splicing isoform of rat rCLCA (rCLCA-t). This isoform is a 514-amino acid protein containing a C-terminal 59-amino acid that is distinct from the rCLCA-f sequence. Immunohistochemistry revealed rCLCA-t to be located in the basal cells of the rat submandibular gland excretory duct and the stratum basale of rat epidermis, whereas rCLCA-f was detected in cells during the process of differentiation. In a heterologous expression system, rCLCA-t was found to be a membrane protein present predominantly in the perinuclear region, and not to be either present on the cell surface or secreted. rCLCA-t failed to enhance ionomycin-induced Cl(-) conductance (unlike rCLCA-f). When compared with rCLCA-f, it weakened cell attachment to a greater extent and in a manner that was evidently modulated by intracellular Ca(2+), protein kinase C, and β(1)-integrin. rCLCA-t was found to associate with RACK1 (receptor for activated C kinase) and to reduce expression of mature β(1)-integrin. Treatment of rat skin with rCLCA-t siRNA increased the expression of β(1)-integrin in the stratum basale of the epidermis. These results are consistent with cell-specific splicing of rCLCA mRNA playing a role in the modulation of the adhesive potential of undifferentiated epithelial cells.
Collapse
Affiliation(s)
- Jun Yamazaki
- Department of Physiological Science and Molecular Biology, Fukuoka Dental College, Sawara-ku, Fukuoka 814-0193, Japan.
| | | | | | | |
Collapse
|
31
|
Kang XL, Zhang M, Liu J, Lv XF, Tang YB, Guan YY. Differences between femoral artery and vein smooth muscle cells in volume-regulated chloride channels. Can J Physiol Pharmacol 2012. [PMID: 23181279 DOI: 10.1139/y2012-117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The purpose of the present study was to compare the differences between the role of volume-regulated Cl⁻ channels (VRCCs) in veins and arteries. We used the whole cell patch clamp and fluorescence imaging techniques to evaluate swelling-induced Cl⁻ current (I(Cl,vol)) and changes in the intracellular concentrations of Cl⁻ ([Cl⁻](i)) induced by hypotonic solutions in rat femoral artery cells (FASMCs) and vein smooth muscle cells (FVSMCs). I(Cl,vol) and [Cl⁻](i) decline induced by hypotonic solution were more prominent in FASMCs than in FVSMCs. I(Cl,vol) and the alterations in [Cl⁻](i) were gradually increased as the number of cell passages increased. However, the regulatory function of tyrosine protein phosphorylation in volume-regulated chloride movement is prominent in veins. The expression of ClC-3 was higher in FASMCs than in FVSMCs. VRCC activity is more pronounced in rat femoral arteries than in veins. VRCC activity and tyrosine protein phosphorylation regulative function increase gradually as vascular cells switch from contractile to proliferative phenotypes.
Collapse
Affiliation(s)
- Xiao-Long Kang
- Department of Pharmacology, Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, 74 Zhongshan 2 Road, Guangzhou 510089, People's Republic of China
| | | | | | | | | | | |
Collapse
|
32
|
Li P, Xiao H, Tang B. A Near-infrared Fluorescent Probe for Selective Simultaneous Detection of Fe2+and Cl−in Living Cells. CHINESE J CHEM 2012. [DOI: 10.1002/cjoc.201200652] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
33
|
Kumagai K, Imai S, Toyoda F, Okumura N, Isoya E, Matsuura H, Matsusue Y. 17β-Oestradiol inhibits doxorubicin-induced apoptosis via block of the volume-sensitive Cl(-) current in rabbit articular chondrocytes. Br J Pharmacol 2012; 166:702-20. [PMID: 22142024 DOI: 10.1111/j.1476-5381.2011.01802.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Chondrocyte apoptosis contributes to disruption of cartilage integrity in osteoarthritis. Recent evidence suggested that the volume-sensitive organic osmolyte/anion channel [volume-sensitive (outwardly rectifying) Cl(-) current (I(Cl,vol) )] plays a functional role in the development of cell shrinkage associated with apoptosis (apoptotic volume decrease) in several cell types. In this study, we investigated the cellular effects of 17β-oestradiol on doxorubicin-induced apoptotic responses in rabbit articular chondrocytes. EXPERIMENTAL APPROACH Whole-cell membrane currents and cross-sectional area were measured from chondrocytes using a patch-clamp method and microscopic cell imaging, respectively. Caspase-3/7 activity was assayed as an index of apoptosis. KEY RESULTS Addition of doxorubicin (1 µM) to isosmotic bath solution rapidly activated the Cl(-) current with properties similar to those of I(Cl,vol) in chondrocytes. Doxorubicin also gradually decreased the cross-sectional area of chondrocytes, followed by enhanced caspase-3/7 activity; both of these responses were totally abolished by the I(Cl,vol) blocker DCPIB (20 µM). Pretreatment of chondrocytes with 17β-oestradiol (1 nM) for short (approximately 10 min) and long (24 h) periods almost completely prevented the doxorubicin-induced activation of I(Cl,vol) and subsequent elevation of caspase-3/7 activity. These effects of 17β-oestradiol were significantly attenuated by the oestrogen receptor blocker ICI 182780 (10 µM), as well as the phosphatidyl inositol-3-kinase (PI3K) inhibitors wortmannin (100 nM) and LY294002 (20 µM). Testosterone (10 nM) had no effect on the doxorubicin-induced Cl(-) current. CONCLUSIONS AND IMPLICATIONS 17β-Oestradiol prevents the doxorubicin-induced cell shrinkage mediated through activation of I(Cl,vol) and subsequent induction of apoptosis signals, through a membrane receptor-dependent PI3K pathway in rabbit articular chondrocytes.
Collapse
Affiliation(s)
- Kousuke Kumagai
- Department of Orthopaedic Surgery, Shiga University of Medical Science, Otsu, Shiga, Japan
| | | | | | | | | | | | | |
Collapse
|
34
|
Cystic fibrosis: insight into CFTR pathophysiology and pharmacotherapy. Clin Biochem 2012; 45:1132-44. [PMID: 22698459 DOI: 10.1016/j.clinbiochem.2012.05.034] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2012] [Revised: 05/15/2012] [Accepted: 05/28/2012] [Indexed: 12/14/2022]
Abstract
Cystic fibrosis is the most common life-threatening recessively inherited disease in Caucasians. Due to early provision of care in specialized reference centers and more comprehensive care, survival has improved over time. Despite great advances in supportive care and in our understanding of its pathophysiology, there is still no cure for the disease. Therapeutic strategies aimed at rescuing the abnormal protein are either being sought after or under investigation. This review highlights salient insights into pathophysiology and candidate molecules suitable for CFTR pharmacotherapy. Clinical trials using Ataluren, VX-809 and ivacaftor have provided encouraging data. Preclinical data with inhibitors of phosphodiesterase type 5, such as sildenafil and analogs, have highlighted their potential for CFTR pharmacotherapy. Because sildenafil and analogs are in clinical use for other clinical applications, research on this class of drugs might speed up the development of new therapies for CF.
Collapse
|
35
|
Li P, Xie T, Fan N, Li K, Tang B. Ratiometric fluorescence imaging for distinguishing chloride concentration between normal and ischemic ventricular myocytes. Chem Commun (Camb) 2012; 48:2077-9. [DOI: 10.1039/c1cc15258k] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
36
|
Bozeat ND, Xiang SY, Ye LL, Yao TY, Duan ML, Burkin DJ, Lamb FS, Duan DD. Activation of volume regulated chloride channels protects myocardium from ischemia/reperfusion damage in second-window ischemic preconditioning. Cell Physiol Biochem 2011; 28:1265-78. [PMID: 22179014 DOI: 10.1159/000335858] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2011] [Indexed: 02/03/2023] Open
Abstract
Activation of volume regulated chloride channels (VRCCs) has been shown to be cardioprotective in ischemic preconditioning (IPC) of isolated hearts but the underlying molecular mechanisms remain unclear. Recent independent studies support that ClC-3, a ClC voltage-gated chloride channel, may function as a key component of the VRCCs. Thus, ClC-3 knockout (Clcn3(-/-)) mice and their age-matched heterozygous (Clcn3(+/-)) and wild-type (Clcn3(+/+)) littermates were used to test whether activation of VRCCs contributes to cardioprotection in early and/or second-window IPC. Targeted disruption of ClC-3 gene caused a decrease in the body weight but no changes in heart/body weight ratio. Telemetry ECG and echocardiography revealed no differences in ECG and cardiac function under resting conditions among all groups. Under treadmill stress (10 m/min for 10 min), the Clcn3(-/-) mice had significant slower heart rate (648±12 bpm) than Clcn3(+/+) littermates (737±19 bpm, n=6, P<0.05). Ex vivo IPC in the isolated working-heart preparations protected cardiac function during reperfusion and significantly decreased apoptosis and infarct size in all groups. In vivo early IPC significantly reduced infarct size in all groups including Clcn3(-/-) mice (22.7±3.7% vs control 40.1±4.3%, n=22, P=0.004). Second-window IPC significantly reduced apoptosis and infarction in Clcn3(+/+) (22.9±3.2% vs 45.7±5.4%, n=22, P<0.001) and Clcn3(+/-) mice (27.5±4.1% vs 42.2±5.7%, n=15, P<0.05) but not in Clcn3(-/-) littermates (39.8±4.9% vs 41.5±8.2%, n=13, P>0.05). Impaired cell volume regulation of the Clcn3(-/-) myocytes may contribute to the failure of cardioprotection by second-window IPC. These results strongly support that activation of VRCCs may play an important cardioprotective role in second-window IPC.
Collapse
Affiliation(s)
- Nathan D Bozeat
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada 89557-0318, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Pandit SV, Kaur K, Zlochiver S, Noujaim SF, Furspan P, Mironov S, Shibayama J, Anumonwo J, Jalife J. Left-to-right ventricular differences in I(KATP) underlie epicardial repolarization gradient during global ischemia. Heart Rhythm 2011; 8:1732-9. [PMID: 21723845 PMCID: PMC3244837 DOI: 10.1016/j.hrthm.2011.06.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Accepted: 06/21/2011] [Indexed: 01/30/2023]
Abstract
BACKGROUND The ionic mechanisms of electrical heterogeneity in the ischemic ventricular epicardium remain poorly understood. OBJECTIVE This study sought to test the hypothesis that the adenosine triphosphate (ATP)-activated K+ current (I(KATP)) plays an important role in mediating repolarization differences between the right ventricle (RV) and left ventricle (LV) during global ischemia. METHODS Electrical activity in Langendorff-perfused guinea pig hearts was recorded optically during control, ischemia, and reperfusion. Patch-clamp experiments were used to quantify I(KATP) density in isolated myocytes. Molecular correlates of I(KATP) (Kir6/SUR) were probed via reverse transcriptase-polymerase chain reaction. The role of I(KATP) in modulating repolarization was studied using computer simulations. RESULTS Action potential duration (APD) was similar between LV and RV in control hearts, but significantly different in global ischemia. Pretreatment of hearts with 10 μM glibenclamide (I(KATP) blocker) abolished the APD gradient during ischemia. In the absence of ischemia, pinacidil (I(KATP) opener) tended to shorten the APD more in the LV, and caused a small but significant increase in APD dispersion. In voltage clamp experiments, the density of the whole-cell current activated by pinacidil at depolarized potentials was significantly larger in LV, compared with RV epicardial myocytes. The mRNA levels of Kir6.1/Kir6.2 were significantly higher in LV compared with RV. Simulations showed that I(KATP) is the main determinant of LV-RV APD gradient, whereas cell-to-cell coupling modified the spatial distribution of this APD gradient. CONCLUSION I(KATP) is an important determinant of the epicardial LV-RV APD gradient during global ischemia, in part due to a higher current density and molecular expression in the LV.
Collapse
Affiliation(s)
- Sandeep V Pandit
- Center for Arrhythmia Research, University of Michigan, Ann Arbor, Michigan 48108, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Chen HP, He M, Mei ZJ, Huang QR, Peng W, Huang M. Anion exchanger 3 is required for sasanquasaponin to inhibit ischemia/reperfusion-induced elevation of intracellular Cl− concentration and to elicit cardioprotection. J Cell Biochem 2011; 112:2803-12. [DOI: 10.1002/jcb.23195] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
39
|
O'Driscoll KE, Pipe RA, Britton FC. Increased complexity of Tmem16a/Anoctamin 1 transcript alternative splicing. BMC Mol Biol 2011; 12:35. [PMID: 21824394 PMCID: PMC3170211 DOI: 10.1186/1471-2199-12-35] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Accepted: 08/08/2011] [Indexed: 01/09/2023] Open
Abstract
Background TMEM16A (Anoctamin 1; ANO1) is an eight transmembrane protein that functions as a calcium-activated chloride channel. TMEM16A in human exhibits alternatively spliced exons (6b, 13 and 15), which confer important roles in the regulation of channel function. Mouse Tmem16a is reported to consist of 25 exons that code for a 956 amino acid protein. In this study our aim was to provide details of mouse Tmem16a genomic structure and to investigate if Tmem16a transcript undergoes alternative splicing to generate channel diversity. Results We identified Tmem16a transcript variants consisting of alternative exons 6b, 10, 13, 14, 15 and 18. Our findings indicate that many of these exons are expressed in various combinations and that these splicing events are mostly conserved between mouse and human. In addition, we confirmed the expression of these exon variants in other mouse tissues. Additional splicing events were identified including a novel conserved exon 13b, tandem splice sites of exon 1 and 21 and two intron retention events. Conclusion Our results suggest that Tmem16a gene is significantly more complex than previously described. The complexity is especially evident in the region spanning exons 6 through 16 where a number of the alternative splicing events are thought to affect calcium sensitivity, voltage dependence and the kinetics of activation and deactivation of this calcium-activated chloride channel. The identification of multiple Tmem16a splice variants suggests that alternative splicing is an exquisite mechanism that operates to diversify TMEM16A channel function in both physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Kate E O'Driscoll
- Department of Physiology and Cell Biology, 1664 North Virginia Street, University of Nevada School of Medicine, Reno, Nevada 89557-0046, USA
| | | | | |
Collapse
|
40
|
Effects of ion channels on proliferation in cultured human cardiac fibroblasts. J Mol Cell Cardiol 2011; 51:198-206. [DOI: 10.1016/j.yjmcc.2011.05.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Revised: 04/24/2011] [Accepted: 05/11/2011] [Indexed: 01/01/2023]
|
41
|
Yamamoto S, Kita S, Iyoda T, Yamada T, Iwamoto T. New molecular mechanisms for cardiovascular disease: cardiac hypertrophy and cell-volume regulation. J Pharmacol Sci 2011; 116:343-9. [PMID: 21757844 DOI: 10.1254/jphs.10r31fm] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Cardiac hypertrophy is an increase in the muscle volume of the ventricle due to the enlargement of cardiac cells. Physiological cardiac hypertrophy is the normal response to healthy exercise, and pathological hypertrophy is the response to increased stress such as hypertension. Intracellular and extracellular aniosmotic conditions also change cell volume. Since persistent cell swelling or cell shrinkage during aniosmotic conditions results in cell death, the ability to regulate cell volume is important for the maintenance of cellular homeostasis. Cell swelling activates a regulatory volume decrease (RVD) response in which solute leakage pathways are stimulated and solute with water exits cells, reducing the cell volume towards the original value. In cardiac cells, one of the essential factors for cell-volume regulation is the volume-regulated anion channel (VRAC). However, the relationship between cardiac hypertrophy and cell-volume regulation is not clear. In this review, we introduce our recent findings showing that the impairment of VRAC current is exhibited in ventricular cells from mice with cardiac hypertrophy induced by transverse aortic constriction. Similar results were shown in caveolin-3-deficient mice, which develop cardiac hypertrophy without pressure overload. These results suggest that VRAC will be a new target for protection from the development of cardiac hypertrophy.
Collapse
Affiliation(s)
- Shintaro Yamamoto
- Department of Pharmacology, School of Medicine, Fukuoka University, Japan.
| | | | | | | | | |
Collapse
|
42
|
Winslow RL, Cortassa S, O'Rourke B, Hashambhoy YL, Rice JJ, Greenstein JL. Integrative modeling of the cardiac ventricular myocyte. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2011; 3:392-413. [PMID: 20865780 PMCID: PMC3110595 DOI: 10.1002/wsbm.122] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cardiac electrophysiology is a discipline with a rich 50-year history of experimental research coupled with integrative modeling which has enabled us to achieve a quantitative understanding of the relationships between molecular function and the integrated behavior of the cardiac myocyte in health and disease. In this paper, we review the development of integrative computational models of the cardiac myocyte. We begin with a historical overview of key cardiac cell models that helped shape the field. We then narrow our focus to models of the cardiac ventricular myocyte and describe these models in the context of their subcellular functional systems including dynamic models of voltage-gated ion channels, mitochondrial energy production, ATP-dependent and electrogenic membrane transporters, intracellular Ca dynamics, mechanical contraction, and regulatory signal transduction pathways. We describe key advances and limitations of the models as well as point to new directions for future modeling research. WIREs Syst Biol Med 2011 3 392-413 DOI: 10.1002/wsbm.122
Collapse
Affiliation(s)
- Raimond L Winslow
- Institute of Computational Medicine and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
| | | | | | | | | | | |
Collapse
|
43
|
Xiang SY, Ye LL, Duan LLM, Liu LH, Ge ZD, Auchampach JA, Gross GJ, Duan DD. Characterization of a critical role for CFTR chloride channels in cardioprotection against ischemia/reperfusion injury. Acta Pharmacol Sin 2011; 32:824-33. [PMID: 21642951 PMCID: PMC3217336 DOI: 10.1038/aps.2011.61] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Accepted: 04/18/2011] [Indexed: 01/08/2023]
Abstract
AIM To further characterize the functional role of cystic fibrosis transmembrane conductance regulator (CFTR) in early and late (second window) ischemic preconditioning (IPC)- and postconditioning (POC)-mediated cardioprotection against ischemia/reperfusion (I/R) injury. METHODS CFTR knockout (CFTR(-/-)) mice and age- and gender-matched wild-type (CFTR(+/+)) and heterozygous (CFTR(+/-)) mice were used. In in vivo studies, the animals were subjected to a 30-min coronary occlusion followed by a 40-min reperfusion. In ex vivo (isolate heart) studies, a 45-min global ischemia was applied. To evaluate apoptosis, the level of activated caspase 3 and TdT-mediated dUTP-X nick end labeling (TUNEL) were examined. RESULTS In the in vivo I/R models, early IPC significantly reduced the myocardial infarct size in wild-type (CFTR(+/+)) (from 40.4% ± 5.3% to 10.4% ± 2.0%, n=8, P<0.001) and heterozygous (CFTR(+/-)) littermates (from 39.4% ± 2.4% to 15.4% ± 5.1%, n=6, P<0.001) but failed to protect CFTR knockout (CFTR(-/-)) mice from I/R induced myocardial infarction (46.9% ± 6.2% vs 55.5% ± 7.8%, n=6, P>0.5). Similar results were observed in the in vivo late IPC experiments. Furthermore, in both in vivo and ex vivo I/R models, POC significantly reduced myocardial infarction in wild-type mice, but not in CFTR knockout mice. In ex vivo I/R models, targeted inactivation of CFTR gene abolished the protective effects of IPC against I/R-induced apoptosis. CONCLUSION These results provide compelling evidence for a critical role for CFTR Cl(-) channels in IPC- and POC-mediated cardioprotection against I/R-induced myocardial injury.
Collapse
Affiliation(s)
- Sunny Yang Xiang
- Department of Pharmacology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Linda L Ye
- Department of Pharmacology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Li-lu Marie Duan
- Department of Pharmacology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Li-hui Liu
- Department of Pharmacology, University of Nevada School of Medicine, Reno, NV 89557, USA
- Institute of Clinical Pharmacology, Central South University Xiangya School of Medicine, Changsha 410078, China
| | - Zhi-dong Ge
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - John A Auchampach
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Garrett J Gross
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Dayue Darrel Duan
- Department of Pharmacology, University of Nevada School of Medicine, Reno, NV 89557, USA
| |
Collapse
|
44
|
Abstract
ClC-3 is a member of the ClC voltage-gated chloride (Cl(-)) channel superfamily. Recent studies have demonstrated the abundant expression and pleiotropy of ClC-3 in cardiac atrial and ventricular myocytes, vascular smooth muscle cells, and endothelial cells. ClC-3 Cl(-) channels can be activated by increase in cell volume, direct stretch of β1-integrin through focal adhesion kinase and many active molecules or growth factors including angiotensin II and endothelin-1-mediated signaling pathways, Ca(2+)/calmodulin-dependent protein kinase II and reactive oxygen species. ClC-3 may function as a key component of the volume-regulated Cl(-) channels, a superoxide anion transport and/or NADPH oxidase interaction partner, and a regulator of many other transporters. ClC-3 has been implicated in the regulation of electrical activity, cell volume, proliferation, differentiation, migration, apoptosis and intracellular pH. This review will highlight the major findings and recent advances in the study of ClC-3 Cl(-) channels in the cardiovascular system and discuss their important roles in cardiac and vascular remodeling during hypertension, myocardial hypertrophy, ischemia/reperfusion, and heart failure.
Collapse
Affiliation(s)
- Dayue Darrel Duan
- Laboratory of Cardiovascular Phenomics, Center of Biomedical Research Excellence, Department of Pharmacology, University of Nevada School of Medicine, Reno, NV 89557, USA.
| |
Collapse
|
45
|
Mechanisms of chloride in cardiomyocyte anoxia-reoxygenation injury: the involvement of oxidative stress and NF-kappaB activation. Mol Cell Biochem 2011; 355:201-9. [DOI: 10.1007/s11010-011-0855-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Accepted: 04/20/2011] [Indexed: 10/18/2022]
|
46
|
Barman PP, Choisy SCM, Gadeberg HC, Hancox JC, James AF. Cardiac ion channel current modulation by the CFTR inhibitor GlyH-101. Biochem Biophys Res Commun 2011; 408:12-7. [PMID: 21439936 DOI: 10.1016/j.bbrc.2011.03.089] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Accepted: 03/21/2011] [Indexed: 11/30/2022]
Abstract
The role in the heart of the cardiac isoform of the cystic fibrosis transmembrane conductance regulator (CFTR), which underlies a protein kinase A-dependent Cl(-) current (I(Cl.PKA)) in cardiomyocytes, remains unclear. The identification of a CFTR-selective inhibitor would provide an important tool for the investigation of the contribution of CFTR to cardiac electrophysiology. GlyH-101 is a glycine hydrazide that has recently been shown to block CFTR channels but its effects on cardiomyocytes are unknown. Here the action of GlyH-101 on cardiac I(Cl.PKA) and on other ion currents has been established. Whole-cell patch-clamp recordings were made from rabbit isolated ventricular myocytes. GlyH-101 blocked I(Cl.PKA) in a concentration- and voltage-dependent fashion (IC(50) at +100 mV=0.3 ± 1.5 μM and at -100 mV=5.1 ± 1.3 μM). Woodhull analysis suggested that GlyH-101 blocks the open pore of cardiac CFTR channels at an electrical distance of 0.15 ± 0.03 from the external membrane surface. A concentration of GlyH-101 maximally effective against I(Cl.PKA) (30 μM) was tested on other cardiac ion currents. Inward current at -120 mV, comprised predominantly of the inward-rectifier background K(+) current, I(K1), was reduced by ∼43% (n=5). Under selective recording conditions, the Na(+) current (I(Na)) was markedly inhibited by GlyH-101 over the entire voltage range (with a fractional block at -40 mV of ∼82%; n=8). GlyH-101 also produced a voltage-dependent inhibition of L-type Ca(2+) channel current (I(Ca,L)); fractional block at +10 mV of ∼49% and of ∼28% at -10 mV; n=11, with a ∼-3 mV shift in the voltage-dependence of I(Ca,L) activation. Thus, this study demonstrates for the first time that GlyH-101 blocks cardiac I(Cl.PKA) channels in a similar fashion to that reported for recombinant CFTR. However, inhibition of other cardiac conductances may limit its use as a CFTR-selective blocker in the heart.
Collapse
Affiliation(s)
- Palash P Barman
- Cardiovascular Research Laboratories, School of Physiology and Pharmacology and Bristol Heart Institute, University of Bristol, Bristol BS8 1TD, UK
| | | | | | | | | |
Collapse
|
47
|
Ichishima K, Yamamoto S, Iwamoto T, Ehara T. alpha-Adrenoceptor-mediated depletion of phosphatidylinositol 4, 5-bisphosphate inhibits activation of volume-regulated anion channels in mouse ventricular myocytes. Br J Pharmacol 2010; 161:193-206. [PMID: 20718750 DOI: 10.1111/j.1476-5381.2010.00896.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND AND PURPOSE Volume-regulated anion channels (VRACs) play an important role in cell-volume regulation. alpha(1)-Adrenoceptor stimulation by phenylephrine (PE) suppressed the hypotonic activation of VRAC current in mouse ventricular cells and regulatory volume decrease (RVD) was also absent in PE-treated cells. We examined whether the effects of alpha(1)-adrenoceptor stimuli on VRAC current were modulated by phosphatidylinositol signalling. EXPERIMENTAL APPROACH Whole-cell patch-clamp method was used to record the hypotonicity-induced VRAC current in mouse ventricular cells. RVD was analyzed by videomicroscopic measurement of cell images. KEY RESULTS The attenuation of VRAC current by PE was suppressed by alpha(1A)-adrenoceptor antagonists (prazosin and WB-4101), anti-G(q) protein antibody and a specific phosphoinositide-specific phospholipase C (PLC) inhibitor (U-73122), but not by antagonists for alpha(1B)-, alpha(1D)- or beta-adrenoceptor, or protein kinase C inhibitors. The inhibition of VRAC by PE was antagonized by intracellular excess phosphatidylinositol 4,5-bisphosphate (PIP(2)), while intracellular anti-PIP(2) antibody (PIP(2) Ab) inhibited the activation of VRAC currents. When cells were loaded with phosphatidylinositol 3,4,5-trisphosphate (PIP(3)) with or without PIP(2) Ab, PE little affected the VRAC current. Extracellular m-3M3FBS (an activator of PLC) suppressed VRAC in the absence of PE, and this effect was reversed by intracellular excess PIP(2). CONCLUSIONS AND IMPLICATIONS Our results indicate that the stimulation of alpha(1A)-adrenoceptors by PE inhibited the activation of cardiac VRAC current via PIP(3) depletion brought about by PLC-dependent reduction of membrane PIP(2) level.
Collapse
Affiliation(s)
- K Ichishima
- Department of Physiology, Faculty of Medicine, Saga University, Saga, Japan
| | | | | | | |
Collapse
|
48
|
De Mello WC. Angiotensin (1-7) reduces the cell volume of swollen cardiac cells and decreases the swelling-dependent chloride current. Implications for cardiac arrhythmias and myocardial ischemia. Peptides 2010; 31:2322-4. [PMID: 20816713 DOI: 10.1016/j.peptides.2010.08.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2010] [Revised: 08/25/2010] [Accepted: 08/25/2010] [Indexed: 11/29/2022]
Abstract
The influence of angiotensin II and angiotensin (1-17) on cell volume and on the activation of ionic channels including the swelling-dependent chloride channel was reviewed. Particular emphasis was given to the influence of the balance between the ACE-angiotensin II and of the ACE2-angiotensin (1-7)-Mas receptor axis on heart cell volume regulation and on the swelling-dependent chloride current. The implications for myocardial ischemia and cardiac arrhythmias are discussed.
Collapse
Affiliation(s)
- Walmor C De Mello
- Medical Sciences Campus, UPR, School of Medicine, PO BOX 365067, San Juan, PR 00936-5067, USA.
| |
Collapse
|
49
|
De Mello WC. Cell swelling impairs dye coupling in adult rat ventricular myocytes. Cell volume as a regulator of cell communication. Mol Cell Biochem 2010; 343:107-13. [PMID: 20512611 DOI: 10.1007/s11010-010-0504-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Accepted: 05/15/2010] [Indexed: 11/25/2022]
Abstract
The influence of cell swelling on cell communication was investigated in cardiomyocytes isolated from the ventricle of adult rats. Measurements of dye coupling were performed in cell pairs using intracellular dialysis of Lucifer Yellow CH. The pipette was attached to one cell of the pair and after a gig ohm seal was achieved, the membrane was ruptured by a brief suction allowing the dye to diffuse from the pipette into the cell. Fluorescence of the dye in the injected as well as in non-dialyzed cell of the pair was continuously monitored. The results indicate that in cell pairs exposed to hypotonic solution the cell volume was increased by about 60% within 35 min and the dye coupling was significantly reduced by cell swelling. Calculation of gap junction permeability (P(j)) assuming an the intracellular volume accessible to intracellular diffusion of the dye as 12% of total cell volume, showed an average P(j) value of 0.16 ± 0.04 × 10(-4) cm/s (n = 35) in the control and 0.89 ± 1.1 × 10(-5) cm (n = 40) for cells exposed to hypotonic solution (P < 0.05). Similar results were found assuming intracellular volumes accessible to the dye of 20 and 30% of total cell volume, respectively. Cell swelling did not change the rate of intracellular diffusion of the dye. The results which indicate that cell volume is an important regulator of gap junction permeability, have important implications to myocardial ischemia and heart failure as well as to heart pharmacology because changes in cell volume caused by drugs and transmitters can impair cell communication with consequent generation of slow conduction and cardiac arrhythmias.
Collapse
Affiliation(s)
- Walmor C De Mello
- Department of Pharmacology, School of Medicine, UPR, San Juan, PR 00936-5067.
| |
Collapse
|
50
|
CLC-3 chloride channels in the pulmonary vasculature. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 661:237-47. [PMID: 20204734 DOI: 10.1007/978-1-60761-500-2_15] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Volume-sensitive outwardly rectifying anion channels (VSOACs) are expressed in pulmonary artery smooth muscle cells (PASMCs) and have been implicated in cell proliferation, growth, apoptosis and protection against oxidative stress. In this chapter, we review the properties of native VSOACs in PASMCs, and consider the evidence that ClC-3, a member of the ClC superfamily of voltage dependent Cl- channels, may be responsible for native VSOACs in PASMCs. Finally, we examine whether or not native VSOACs and heterologously expressed ClC-3 channels function as bona fide chloride channels or as chloride/proton antiporters.
Collapse
|