1
|
Garcia E, Claudi L, La Chica Lhoëst MT, Polishchuk A, Samouillan V, Benitez Amaro A, Pinero J, Escolà-Gil JC, Sabidó E, Leta R, Vilades D, Llorente Cortes V. Reduced blood EPAC1 protein levels as a marker of severe coronary artery disease: the role of hypoxic foam cell-transformed smooth muscle cells. J Transl Med 2025; 23:523. [PMID: 40346550 PMCID: PMC12063457 DOI: 10.1186/s12967-025-06513-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 04/18/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Vascular smooth muscle cells loaded with cholesterol (foam-VSMCs) play a crucial role in the progression of human atherosclerosis. Exchange Protein Directly Activated by cAMP 1 (EPAC1) is a critical protein in the regulation of vascular tone, endothelial function, and inflammation. Our objectives were to identify proteins specifically secreted by foam human coronary VSMCs (foam-hcVSMC) to evaluate their potential as circulating biomarkers for diagnosing coronary artery disease (CAD), and to ascertain the mechanisms underlying their levels in the blood of patients with CAD. METHODS AND RESULTS Differential proteomics identified EPAC1 as a differential foam-hcVSMC-secreted protein. Circulating EPAC1 levels were measured by ELISA in blood from 202 patients with suspected CAD who underwent coronary computed tomography angiography (CCTA). Blood EPAC1 levels were significantly lower in CAD patients compared to controls (p < 0.001). EPAC1 levels were reduced in both men and women with severe CAD (SIS > 4) compared to those with moderate CAD (SIS 1-4). ROC analysis identified 9.16 ng/ml as the optimal EPAC1 cut-off for severe CAD. At this threshold, EPAC1 predicted severe CAD (SIS > 4) with 69.6% sensitivity and 79.4% specificity, outperforming hs-CRP and hs-TnT in predicting CAD severity. Real-time PCR and Western blot analysis revealed that human foam-SMCs under hypoxic conditions exhibited a significant reduction in EPAC1 mRNA (p = 0.013) and protein (p < 0.001) levels. CONCLUSIONS These findings suggest that circulating EPAC1 protein levels lower than 9.16 ng/mL are predictive of severe CAD in humans. Hypoxic foam-SMCs, characteristic of advanced atherosclerotic lesions, exhibit diminished production of EPAC1, potentially contributing to the decreased circulating EPAC1 levels in patients with severe CAD.
Collapse
Affiliation(s)
- Eduardo Garcia
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, 08041, Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, 08041, Barcelona, Spain
| | - Lene Claudi
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, 08041, Barcelona, Spain
| | - Maria Teresa La Chica Lhoëst
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, 08041, Barcelona, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08041, Barcelona, Spain
| | - Anna Polishchuk
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, 08041, Barcelona, Spain
| | - Valerie Samouillan
- CIRIMAT, Université de Toulouse, Université Paul Sabatier, Equipe PHYPOL, 31062, Toulouse, France
| | - Aleyda Benitez Amaro
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, 08041, Barcelona, Spain
| | - Janet Pinero
- Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences (DCEXS), Hospital del Mar Medical Research Institute (IMIM), Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Joan Carles Escolà-Gil
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, 08041, Barcelona, Spain
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08041, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029, Madrid, Spain
| | - Eduard Sabidó
- Proteomics Unit, Centre de Regulació Genòmica, Barcelona Institute of Science and Technology; Universitat Pompeu i Fabra (UPF), Barcelona, Spain
| | - Ruben Leta
- Cardiac Imaging Unit, Department of Cardiology, Hospital de la Santa Creu i Sant Pau, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - David Vilades
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, 08041, Barcelona, Spain
- Cardiac Imaging Unit, Department of Cardiology, Hospital de la Santa Creu i Sant Pau, Universitat Autonoma de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Cardiovasculares CIBERCV, Institute of Health Carlos III, 28029, Madrid, Spain
| | - Vicenta Llorente Cortes
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain.
- Institut de Recerca Sant Pau (IR SANT PAU), Sant Quintí 77-79, 08041, Barcelona, Spain.
- CIBER de Enfermedades Cardiovasculares CIBERCV, Institute of Health Carlos III, 28029, Madrid, Spain.
| |
Collapse
|
2
|
Hou P, Zhao L, Zhong L, Shi J, Wang HZ, Gao J, Liu H, Zuckerman J, Cohen IS, Cui J. The fully activated open state of KCNQ1 controls the cardiac "fight-or-flight" response. PNAS NEXUS 2024; 3:pgae452. [PMID: 39434867 PMCID: PMC11492796 DOI: 10.1093/pnasnexus/pgae452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 10/02/2024] [Indexed: 10/23/2024]
Abstract
The cardiac KCNQ1 + KCNE1 (IKs) channel regulates heart rhythm under both normal and stress conditions. Under stress, the β-adrenergic stimulation elevates the intracellular cyclic adenosine monophosphate (cAMP) level, leading to KCNQ1 phosphorylation by protein kinase A and increased IKs, which shortens action potentials to adapt to accelerated heart rate. An impaired response to the β-adrenergic stimulation due to KCNQ1 mutations is associated with the occurrence of a lethal congenital long QT syndrome (type 1, also known as LQT1). However, the underlying mechanism of β-adrenergic stimulation of IKs remains unclear, impeding the development of new therapeutics. Here, we find that the unique properties of KCNQ1 channel gating with two distinct open states are key to this mechanism. KCNQ1's fully activated open (AO) state is more sensitive to cAMP than its intermediate open state. By enhancing the AO state occupancy, the small molecules ML277 and C28 are found to effectively enhance the cAMP sensitivity of the KCNQ1 channel, independent of KCNE1 association. This finding of enhancing AO state occupancy leads to a potential novel strategy to rescue the response of IKs to β-adrenergic stimulation in LQT1 mutants. The success of this approach is demonstrated in cardiac myocytes and also in a high-risk LQT1 mutation. In conclusion, the present study not only uncovers the key role of the AO state in IKs channel phosphorylation, but also provides a target for antiarrhythmic strategy.
Collapse
Affiliation(s)
- Panpan Hou
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, Washington University, St. Louis, MO 63130, USA
| | - Lu Zhao
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, Washington University, St. Louis, MO 63130, USA
| | - Ling Zhong
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, Washington University, St. Louis, MO 63130, USA
| | - Jingyi Shi
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, Washington University, St. Louis, MO 63130, USA
| | - Hong Zhan Wang
- Department of Physiology and Biophysics, Institute for Molecular Cardiology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Junyuan Gao
- Department of Physiology and Biophysics, Institute for Molecular Cardiology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Huilin Liu
- Department of Physiology and Biophysics, Institute for Molecular Cardiology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Joan Zuckerman
- Department of Physiology and Biophysics, Institute for Molecular Cardiology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Ira S Cohen
- Department of Physiology and Biophysics, Institute for Molecular Cardiology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jianmin Cui
- Department of Biomedical Engineering, Center for the Investigation of Membrane Excitability Disorders, Washington University, St. Louis, MO 63130, USA
| |
Collapse
|
3
|
Boileve A, Romito O, Hof T, Levallois A, Brard L, d'Hers S, Fouchet A, Simard C, Guinamard R, Brette F, Sallé L. EPAC1 and 2 inhibit K + currents via PLC/PKC and NOS/PKG pathways in rat ventricular cardiomyocytes. Am J Physiol Cell Physiol 2024; 327:C557-C570. [PMID: 38985989 DOI: 10.1152/ajpcell.00582.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 05/29/2024] [Accepted: 06/19/2024] [Indexed: 07/12/2024]
Abstract
The exchange protein directly activated by cAMP (EPAC) has been implicated in cardiac proarrhythmic signaling pathways including spontaneous diastolic Ca2+ leak from sarcoplasmic reticulum and increased action potential duration (APD) in isolated ventricular cardiomyocytes. The action potential (AP) lengthening following acute EPAC activation is mainly due to a decrease of repolarizing steady-state K+ current (IKSS) but the mechanisms involved remain unknown. This study aimed to assess the role of EPAC1 and EPAC2 in the decrease of IKSS and to investigate the underlying signaling pathways. AP and K+ currents were recorded with the whole cell configuration of the patch-clamp technique in freshly isolated rat ventricular myocytes. EPAC1 and EPAC2 were pharmacologically activated with 8-(4-chlorophenylthio)-2'-O-methyl-cAMP acetoxymethyl ester (8-CPTAM, 10 µmol/L) and inhibited with R-Ce3F4 and ESI-05, respectively. Inhibition of EPAC1 and EPAC2 significantly decreased the effect of 8-CPTAM on APD and IKSS showing that both EPAC isoforms are involved in these effects. Unexpectedly, calmodulin-dependent protein kinase II (CaMKII) inhibition by AIP or KN-93, and Ca2+ chelation by intracellular BAPTA, did not impact the response to 8-CPTAM. However, inhibition of PLC/PKC and nitric oxide synthase (NOS)/PKG pathways partially prevents the 8-CPTAM-dependent decrease of IKSS. Finally, the cumulative inhibition of PKC and PKG blocked the 8-CPTAM effect, suggesting that these two actors work along parallel pathways to regulate IKSS upon EPAC activation. On the basis of such findings, we propose that EPAC1 and EPAC2 are involved in APD lengthening by inhibiting a K+ current via both PLC/PKC and NOS/PKG pathways. This may have pathological implications since EPAC is upregulated in diseases such as cardiac hypertrophy.NEW & NOTEWORHTY Exchange protein directly activated by cAMP (EPAC) proteins modulate ventricular electrophysiology at the cellular level. Both EPAC1 and EPAC2 isoforms participate in this effect. Mechanistically, PLC/PKC and nitric oxide synthase (NO)/PKG pathways are involved in regulating K+ repolarizing current whereas the well-known downstream effector of EPAC, calmodulin-dependent protein kinase II (CaMKII), does not participate. This may have pathological implications since EPAC is upregulated in diseases such as cardiac hypertrophy. Thus, EPAC inhibition may be a new approach to prevent arrhythmias under pathological conditions.
Collapse
Affiliation(s)
- Arthur Boileve
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Olivier Romito
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Thomas Hof
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Aurélia Levallois
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Laura Brard
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Sarah d'Hers
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Alexandre Fouchet
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Christophe Simard
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Romain Guinamard
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| | - Fabien Brette
- PhyMedExp, INSERM U1046, CNRS 9412, Université de Montpellier, Montpellier, France
| | - Laurent Sallé
- UR 4650 PSIR, GIP Cyceron, Caen, France
- Normandie University, Caen, France
- UNICAEN, Caen, France
| |
Collapse
|
4
|
Hou P, Zhao L, Zhong L, Shi J, Wang HZ, Gao J, Liu H, Zuckerman J, Cohen IS, Cui J. The fully activated open state of KCNQ1 controls the cardiac "fight-or-flight" response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.02.601749. [PMID: 39005479 PMCID: PMC11244952 DOI: 10.1101/2024.07.02.601749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The cardiac KCNQ1+KCNE1 (I Ks ) channel regulates heart rhythm in both normal and stress conditions. Under stress, the β-adrenergic stimulation elevates the intracellular cAMP level, leading to KCNQ1 phosphorylation by protein kinase A and increased I Ks , which shortens action potentials to adapt to accelerated heart rate. An impaired response to the β-adrenergic stimulation due to KCNQ1 mutations is associated with the occurrence of a lethal congenital long QT syndrome (type 1, also known as LQT1). However, the underlying mechanism of β-adrenergic stimulation of I Ks remains unclear, impeding the development of new therapeutics. Here we find that the unique properties of KCNQ1 channel gating with two distinct open states are key to this mechanism. KCNQ1's fully activated open (AO) state is more sensitive to cAMP than its' intermediate open (IO) state. By enhancing the AO state occupancy, the small molecules ML277 and C28 are found to effectively enhance the cAMP sensitivity of the KCNQ1 channel, independent of KCNE1 association. This finding of enhancing AO state occupancy leads to a potential novel strategy to rescue the response of I Ks to β-adrenergic stimulation in LQT1 mutants. The success of this approach is demonstrated in cardiac myocytes and also in a high-risk LQT1 mutation. In conclusion the present study not only uncovers the key role of the AO state in I Ks channel phosphorylation, but also provides a new target for anti-arrhythmic strategy. Significance statement The increase of I Ks potassium currents with adrenalin stimulation is important for "fight-or-flight" responses. Mutations of the IKs channel reducing adrenalin responses are associated with more lethal form of the type-1 long-QT syndrome (LQT). The alpha subunit of the IKs channel, KCNQ1 opens in two distinct open states, the intermediate-open (IO) and activated-open (AO) states, following a two-step voltage sensing domain (VSD) activation process. We found that the AO state, but not the IO state, is responsible for the adrenalin response. Modulators that specifically enhance the AO state occupancy can enhance adrenalin responses of the WT and LQT-associated mutant channels. These results reveal a mechanism of state dependent modulation of ion channels and provide an anti-arrhythmic strategy.
Collapse
|
5
|
van Weperen VYH, Ripplinger CM, Vaseghi M. Autonomic control of ventricular function in health and disease: current state of the art. Clin Auton Res 2023; 33:491-517. [PMID: 37166736 PMCID: PMC10173946 DOI: 10.1007/s10286-023-00948-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/20/2023] [Indexed: 05/12/2023]
Abstract
PURPOSE Cardiac autonomic dysfunction is one of the main pillars of cardiovascular pathophysiology. The purpose of this review is to provide an overview of the current state of the art on the pathological remodeling that occurs within the autonomic nervous system with cardiac injury and available neuromodulatory therapies for autonomic dysfunction in heart failure. METHODS Data from peer-reviewed publications on autonomic function in health and after cardiac injury are reviewed. The role of and evidence behind various neuromodulatory therapies both in preclinical investigation and in-use in clinical practice are summarized. RESULTS A harmonic interplay between the heart and the autonomic nervous system exists at multiple levels of the neuraxis. This interplay becomes disrupted in the setting of cardiovascular disease, resulting in pathological changes at multiple levels, from subcellular cardiac signaling of neurotransmitters to extra-cardiac, extra-thoracic remodeling. The subsequent detrimental cycle of sympathovagal imbalance, characterized by sympathoexcitation and parasympathetic withdrawal, predisposes to ventricular arrhythmias, progression of heart failure, and cardiac mortality. Knowledge on the etiology and pathophysiology of this condition has increased exponentially over the past few decades, resulting in a number of different neuromodulatory approaches. However, significant knowledge gaps in both sympathetic and parasympathetic interactions and causal factors that mediate progressive sympathoexcitation and parasympathetic dysfunction remain. CONCLUSIONS Although our understanding of autonomic imbalance in cardiovascular diseases has significantly increased, specific, pivotal mediators of this imbalance and the recognition and implementation of available autonomic parameters and neuromodulatory therapies are still lagging.
Collapse
Affiliation(s)
- Valerie Y H van Weperen
- Division of Cardiology, Department of Medicine, UCLA Cardiac Arrythmia Center, University of California, 100 Medical Plaza, Suite 660, Los Angeles, CA, 90095, USA
| | | | - Marmar Vaseghi
- Division of Cardiology, Department of Medicine, UCLA Cardiac Arrythmia Center, University of California, 100 Medical Plaza, Suite 660, Los Angeles, CA, 90095, USA.
| |
Collapse
|
6
|
Guillot B, Boileve A, Walton R, Harfoush A, Conte C, Sainte-Marie Y, Charron S, Bernus O, Recalde A, Sallé L, Brette F, Lezoualc'h F. Inhibition of EPAC1 signaling pathway alters atrial electrophysiology and prevents atrial fibrillation. Front Physiol 2023; 14:1120336. [PMID: 36909224 PMCID: PMC9992743 DOI: 10.3389/fphys.2023.1120336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/09/2023] [Indexed: 02/24/2023] Open
Abstract
Introduction: Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia and is associated with increased mortality and morbidity. The Exchange Protein directly Activated by cAMP (EPAC), has been implicated in pro-arrhythmic signaling pathways in the atria, but the underlying mechanisms remain unknown. Methods: In this study, we investigated the involvement of EPAC1 and EPAC2 isoforms in the genesis of AF in wild type (WT) mice and knockout (KO) mice for EPAC1 or EPAC2. We also employed EPAC pharmacological modulators to selectively activate EPAC proteins (8-CPT-AM; 10 μM), or inhibit either EPAC1 (AM-001; 20 μM) or EPAC2 (ESI-05; 25 μM). Transesophageal stimulation was used to characterize the induction of AF in vivo in mice. Optical mapping experiments were performed on isolated mouse atria and cellular electrophysiology was examined by whole-cell patch-clamp technique. Results: In wild type mice, we found 8-CPT-AM slightly increased AF susceptibility and that this was blocked by the EPAC1 inhibitor AM-001 but not the EPAC2 inhibitor ESI-05. Consistent with this, in EPAC1 KO mice, occurrence of AF was observed in 3/12 (vs. 4/10 WT littermates) and 4/10 in EPAC2 KO (vs. 5/10 WT littermates). In wild type animals, optical mapping experiments revealed that 8-CPT-AM perfusion increased action potential duration even in the presence of AM-001 or ESI-05. Interestingly, 8-CPT-AM perfusion decreased conduction velocity, an effect blunted by AM-001 but not ESI-05. Patch-clamp experiments demonstrated action potential prolongation after 8-CPT-AM perfusion in both wild type and EPAC1 KO mice and this effect was partially prevented by AM-001 in WT. Conclusion: Together, these results indicate that EPAC1 and EPAC2 signaling pathways differentially alter atrial electrophysiology but only the EPAC1 isoform is involved in the genesis of AF. Selective blockade of EPAC1 with AM-001 prevents AF in mice.
Collapse
Affiliation(s)
- Bastien Guillot
- IHU LIRYC -CRCTB U1045, Pessac, France.,INSERM U1045 -Université de Bordeaux, Bordeaux, France
| | - Arthur Boileve
- UR 4650 PSIR, GIP Cyceron, Caen, France.,Université de Caen-Normandie, Caen, France
| | - Richard Walton
- IHU LIRYC -CRCTB U1045, Pessac, France.,INSERM U1045 -Université de Bordeaux, Bordeaux, France
| | - Alexandre Harfoush
- UR 4650 PSIR, GIP Cyceron, Caen, France.,Université de Caen-Normandie, Caen, France
| | - Caroline Conte
- Université de Toulouse-Paul Sabatier, Toulouse, France.,Institut des maladies métaboliques et cardiovasculaires, INSERM UMR-1297, Toulouse, France
| | - Yannis Sainte-Marie
- Université de Toulouse-Paul Sabatier, Toulouse, France.,Institut des maladies métaboliques et cardiovasculaires, INSERM UMR-1297, Toulouse, France
| | - Sabine Charron
- IHU LIRYC -CRCTB U1045, Pessac, France.,INSERM U1045 -Université de Bordeaux, Bordeaux, France
| | - Olivier Bernus
- IHU LIRYC -CRCTB U1045, Pessac, France.,INSERM U1045 -Université de Bordeaux, Bordeaux, France
| | - Alice Recalde
- IHU LIRYC -CRCTB U1045, Pessac, France.,INSERM U1045 -Université de Bordeaux, Bordeaux, France
| | - Laurent Sallé
- UR 4650 PSIR, GIP Cyceron, Caen, France.,Université de Caen-Normandie, Caen, France
| | - Fabien Brette
- IHU LIRYC -CRCTB U1045, Pessac, France.,INSERM U1045 -Université de Bordeaux, Bordeaux, France.,PhyMedExp, INSERM U1046, CNRS 9412, Université de Montpellier, Montpellier, France
| | - Frank Lezoualc'h
- Université de Toulouse-Paul Sabatier, Toulouse, France.,Institut des maladies métaboliques et cardiovasculaires, INSERM UMR-1297, Toulouse, France
| |
Collapse
|
7
|
Huang Z, Liu L, Zhang J, Conde K, Phansalkar J, Li Z, Yao L, Xu Z, Wang W, Zhou J, Bi G, Wu F, Seeley RJ, Scott MM, Zhan C, Pang ZP, Liu J. Glucose-sensing glucagon-like peptide-1 receptor neurons in the dorsomedial hypothalamus regulate glucose metabolism. SCIENCE ADVANCES 2022; 8:eabn5345. [PMID: 35675406 PMCID: PMC9177072 DOI: 10.1126/sciadv.abn5345] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 04/21/2022] [Indexed: 05/23/2023]
Abstract
Glucagon-like peptide-1 (GLP-1) regulates energy homeostasis via activation of the GLP-1 receptors (GLP-1Rs) in the central nervous system. However, the mechanism by which the central GLP-1 signal controls blood glucose levels, especially in different nutrient states, remains unclear. Here, we defined a population of glucose-sensing GLP-1R neurons in the dorsomedial hypothalamic nucleus (DMH), by which endogenous GLP-1 decreases glucose levels via the cross-talk between the hypothalamus and pancreas. Specifically, we illustrated the sufficiency and necessity of DMHGLP-1R in glucose regulation. The activation of the DMHGLP-1R neurons is mediated by a cAMP-PKA-dependent inhibition of a delayed rectifier potassium current. We also dissected a descending control of DMHGLP-1R -dorsal motor nucleus of the vagus nerve (DMV)-pancreas activity that can regulate glucose levels by increasing insulin release. Thus, our results illustrate how central GLP-1 action in the DMH can induce a nutrient state-dependent reduction in blood glucose level.
Collapse
Affiliation(s)
- Zhaohuan Huang
- National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Anhui, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Ling Liu
- National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Anhui, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Jian Zhang
- CAS Key Laboratory of Brain Function and Diseases, Life Science School, University of Science and Technology of China, Anhui, China
| | - Kristie Conde
- Child Health Institute of New Jersey, Rutgers University Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Jay Phansalkar
- Child Health Institute of New Jersey, Rutgers University Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Zhongzhong Li
- National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Anhui, China
| | - Lei Yao
- CAS Key Laboratory of Brain Function and Diseases, Life Science School, University of Science and Technology of China, Anhui, China
| | - Zihui Xu
- Child Health Institute of New Jersey, Rutgers University Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Wei Wang
- Department of Endocrinology and Laboratory for Diabetes, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui, China
| | - Jiangning Zhou
- CAS Key Laboratory of Brain Function and Diseases, Life Science School, University of Science and Technology of China, Anhui, China
| | - Guoqiang Bi
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
- CAS Key Laboratory of Brain Function and Diseases, Life Science School, University of Science and Technology of China, Anhui, China
| | - Feng Wu
- National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Anhui, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Randy J. Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michael M. Scott
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Cheng Zhan
- Department of Hematology, The First Affiliated Hospital, Life Science School, University of Science and Technology of China, Anhui, China
- National Institute of Biological Sciences, Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 100084, China
| | - Zhiping P. Pang
- Child Health Institute of New Jersey, Rutgers University Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
- Department of Neuroscience and Cell Biology, Rutgers University Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Ji Liu
- National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Anhui, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
- CAS Key Laboratory of Brain Function and Diseases, Life Science School, University of Science and Technology of China, Anhui, China
| |
Collapse
|
8
|
An X, Lee J, Kim GH, Kim HJ, Pyo HJ, Kwon I, Cho H. Modulation of I Ks channel-PIP 2 interaction by PRMT1 plays a critical role in the control of cardiac repolarization. J Cell Physiol 2022; 237:3069-3079. [PMID: 35580065 PMCID: PMC9543859 DOI: 10.1002/jcp.30775] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 04/18/2022] [Accepted: 04/25/2022] [Indexed: 11/12/2022]
Abstract
Recent studies have shown that protein arginine methyltransferase 1 (PRMT1) is highly expressed in the human heart, and loss of PRMT1 contributes to cardiac remodeling in the heart failure. However, the functional importance of PRMT1 in cardiac ion channels remains uncertain. The slow activating delayed rectifier K+ (IKs) channel is a cardiac K+ channel composed of KCNQ1 and KCNE1 subunits and is a new therapeutic target for treating lethal arrhythmias in many cardiac pathologies, especially heart failure. Here, we demonstrate that PRMT1 is a critical regulator of the IKs channel and cardiac rhythm. In the guinea pig ventricular myocytes, treatment with furamidine, a PRMT1‐specific inhibitor, prolonged the action potential duration (APD). We further show that this APD prolongation was attributable to IKs reduction. In HEK293T cells expressing human KCNQ1 and KCNE1, inhibiting PRMT1 via furamidine reduced IKs and concurrently decreased the arginine methylation of KCNQ1, a pore‐forming α‐subunit. Evidence presented here indicates that furamidine decreased IKs mainly by lowering the affinity of IKs channels for the membrane phospholipid, phosphatidylinositol 4,5‐bisphosphate (PIP2), which is crucial for pore opening. Finally, applying exogenous PIP2 to cardiomyocytes prevented the furamidine‐induced IKs reduction and APD prolongation. Taken together, these results indicate that PRMT1 positively regulated IKs activity through channel–PIP2 interaction, thereby restricting excessive cardiac action potential.
Collapse
Affiliation(s)
- Xue An
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Jiwon Lee
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Ga Hye Kim
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Hyun-Ji Kim
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Hyun-Jeong Pyo
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Ilmin Kwon
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Hana Cho
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Korea
| |
Collapse
|
9
|
Tan YQ, Li J, Chen HW. Epac, a positive or negative signaling molecule in cardiovascular diseases. Pharmacotherapy 2022; 148:112726. [DOI: 10.1016/j.biopha.2022.112726] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 02/08/2023]
|
10
|
van Weperen VYH, Vos MA, Ajijola OA. Autonomic modulation of ventricular electrical activity: recent developments and clinical implications. Clin Auton Res 2021; 31:659-676. [PMID: 34591191 PMCID: PMC8629778 DOI: 10.1007/s10286-021-00823-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/12/2021] [Indexed: 12/19/2022]
Abstract
PURPOSE This review aimed to provide a complete overview of the current stance and recent developments in antiarrhythmic neuromodulatory interventions, focusing on lifethreatening vetricular arrhythmias. METHODS Both preclinical studies and clinical studies were assessed to highlight the gaps in knowledge that remain to be answered and the necessary steps required to properly translate these strategies to the clinical setting. RESULTS Cardiac autonomic imbalance, characterized by chronic sympathoexcitation and parasympathetic withdrawal, destabilizes cardiac electrophysiology and promotes ventricular arrhythmogenesis. Therefore, neuromodulatory interventions that target the sympatho-vagal imbalance have emerged as promising antiarrhythmic strategies. These strategies are aimed at different parts of the cardiac neuraxis and directly or indirectly restore cardiac autonomic tone. These interventions include pharmacological blockade of sympathetic neurotransmitters and neuropeptides, cardiac sympathetic denervation, thoracic epidural anesthesia, and spinal cord and vagal nerve stimulation. CONCLUSION Neuromodulatory strategies have repeatedly been demonstrated to be highly effective and very promising anti-arrhythmic therapies. Nevertheless, there is still much room to gain in our understanding of neurocardiac physiology, refining the current neuromodulatory strategic options and elucidating the chronic effects of many of these strategic options.
Collapse
Affiliation(s)
- Valerie Y H van Weperen
- Department of Medical Physiology, Universitair Medisch Centrum Utrecht, Utrecht, The Netherlands
- UCLA Cardiac Arrhythmia Center, UCLA Neurocardiology Research Center, UCLA Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, University of California, 100 Medical Plaza, Suite 660, Westwood Blvd, Los Angeles, CA, 90095-1679, USA
| | - Marc A Vos
- Department of Medical Physiology, Universitair Medisch Centrum Utrecht, Utrecht, The Netherlands
| | - Olujimi A Ajijola
- UCLA Cardiac Arrhythmia Center, UCLA Neurocardiology Research Center, UCLA Neurocardiology Research Program of Excellence, David Geffen School of Medicine at UCLA, University of California, 100 Medical Plaza, Suite 660, Westwood Blvd, Los Angeles, CA, 90095-1679, USA.
| |
Collapse
|
11
|
Yang W, Ma X, Zhu Y, Meng X, Tian R, Yang Z. Paraquat but not diquat induces TGF-β expression and thus activates calcium-NFAT axis for epithelial-mesenchymal transition. Toxicol Res (Camb) 2021; 10:733-741. [PMID: 34484664 PMCID: PMC8403590 DOI: 10.1093/toxres/tfab055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/04/2021] [Accepted: 05/24/2021] [Indexed: 12/27/2022] Open
Abstract
Paraquat (PQ) and diquat (DQ), two highly efficient herbicides sharing similar chemical backbone, both induce reactive oxygen species and are highly toxic to humans and livestock, however, PQ but not DQ poisoning result in pulmonary fibrosis, the leading cause of high mortality rate in patients suffering PQ toxicity. Understanding the unique mechanism of PQ different from DQ therefore would provide potential strategies to reduce PQ-induced pulmonary fibrosis. Here, we identified that PQ but not DQ continuously upregulates TGF-β expression in alveolar type II (AT II) cells. Importantly, such high expression of TGF-β increases cytosolic calcium levels and further promotes the activation of calcineurin-NFAT axis. TGF-β mainly activates NFATc1 and NFATc2, but not NFATc3 or NFATc4. Administration of the inhibitors targeting cytosolic calcium or calcineurin largely reverses PQ-induced epithelial-mesenchymal transition (EMT), whereas DQ has little effects on activation of NFAT and EMT. Ultimately, PQ poisoned patients exhibit significantly reduced blood calcium levels compared to DQ poisoning, possibly via the large usage of calcium by AT II cells. All in all, we found a vicious cycle that the upregulated TGF-β in PQ-induced EMT further aggravates EMT via promotion of the calcium-calcineurin axis, which could be potential drug targets for treating PQ-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Wenyu Yang
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, 650 Xingsongjiang Road, Shanghai 201620, China
| | - Xinrun Ma
- Institute of clinical Immunology, Center for Translational Medicine, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, 650 Xingsongjiang Road, Shanghai 201620, China
| | - Yong Zhu
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, 650 Xingsongjiang Road, Shanghai 201620, China
| | - Xiaoxiao Meng
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, 650 Xingsongjiang Road, Shanghai 201620, China
| | - Rui Tian
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, 650 Xingsongjiang Road, Shanghai 201620, China
| | - Zhengfeng Yang
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, 650 Xingsongjiang Road, Shanghai 201620, China
- Institute of clinical Immunology, Center for Translational Medicine, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, 650 Xingsongjiang Road, Shanghai 201620, China
| |
Collapse
|
12
|
Zhu D, Zhu R, Zhou X, Shi E, Zhang X, Zhou Z, Li D, Zou J, Wang Y. Exchange proteins directly activated by cAMP mediate cardiac repolarization and arrhythmogenesis during chronic heart failure. Can J Physiol Pharmacol 2021; 99:729-736. [PMID: 33175603 DOI: 10.1139/cjpp-2020-0122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Most sudden cardiac death in chronic heart failure (CHF) is caused by malignant ventricular arrhythmia (VA); however, the molecular mechanism remains unclear. This study aims to explore the effect of exchange proteins directly activated by cAMP (Epac) on VA in CHF and the potential molecular mechanism. Transaortic constriction was performed to prepare CHF guinea pigs. Epac activation model was obtained with 8-pCPT administration. Programmed electrical stimulation (PES) was performed to detect effective refractory period (ERP) or induce VA. Isolated adult cardiomyocytes were treated with 8-pCPT and (or) the Epac inhibitor. Cellular electrophysiology was examined by whole-cell patch clamp. With Epac activation, corrected QT duration was lengthened by 12.6%. The 8-pCPT increased action potential duration (APD) (APD50: 236.9 ± 18.07 ms vs. 328.8 ± 11.27 ms, p < 0.05; APD90: 264.6 ± 18.22 ms vs. 388.6 ± 6.47 ms, p < 0.05) and decreased rapid delayed rectifier potassium (IKr) current (tail current density: 1.1 ± 0.08 pA/pF vs. 0.7 ± 0.03 pA/pF, p < 0.05). PES induced more malignant arrhythmias in the 8-pCPT group than in the control group (3/4 vs. 0/8, p < 0.05). The selective Epac1 inhibitor CE3F4 rescued the drop in IKr after 8-pCPT stimulation (tail current density: 0.5 ± 0.02 pA/pF vs. 0.6 ± 0.03 pA/pF, p < 0.05). In conclusion, Epac1 regulates IKr, APD, and ERP in guinea pigs, which could contribute to the proarrhythmic effect of Epac1 in CHF.
Collapse
Affiliation(s)
- Didi Zhu
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, China
| | - Rui Zhu
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaozhu Zhou
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Erdan Shi
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinwei Zhang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zihao Zhou
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dongcheng Li
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiangang Zou
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yao Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
13
|
Koracevic G, Stojanovic M, Lovic D, Zdravkovic M, Sakac D. Certain beta blockers (e.g., bisoprolol) may be reevaluated in hypertension guidelines for patients with left ventricular hypertrophy to diminish the ventricular arrhythmic risk. J Hum Hypertens 2021; 35:564-576. [PMID: 33654234 DOI: 10.1038/s41371-021-00505-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 01/29/2021] [Accepted: 02/04/2021] [Indexed: 12/18/2022]
Abstract
Hypertensive left ventricular hypertrophy (HTN LVH) is associated with almost threefold increased risk of ventricular tachycardia (VT)/ventricular fibrillation (VF). Furthermore, HTN LVH increases the risk of sudden cardiac death (SCD). The reverse LV remodeling due to efficient antihypertensive therapy lowers the incidence rates of cardiovascular events and SCD and the vast majority of available arterial hypertension (HTN) guidelines recommend renin angiotensin system (RAS) blockers and calcium channel blockers (CCBs) for HTN LVH aiming for LVH regression. On the other hand, beta blockers (BBs) as a class are not recommended in HTN LVH due to their insufficient capacity to reverse LVH remodeling even though they are recommended as the first-line drugs for prevention/treatment of VT/VF (in general, unrelated to HTN LVH). Moreover, BBs are the best antiarrhythmic (against VT/VF) among antihypertensive drugs. Despite that, BBs are currently not recommended for LVH treatment in HTN Guidelines. It is important to prevent VT/VF in patients at high risk, such as those with HTN LVH. Therefore, certain BBs (such as Bisoprolol) may be reevaluated in guidelines for HTN (in the section of HTN LVH).
Collapse
Affiliation(s)
- Goran Koracevic
- Department for Cardiovascular Diseases, Clinical Center Nis, Nis, Serbia.,Faculty of Medicine, University of Nis, Nis, Serbia
| | | | - Dragan Lovic
- Clinic for Internal Diseases Inter Medica, Nis, Serbia.,Singidunum University, School of Medicine, Belgrade, Serbia
| | - Marija Zdravkovic
- University Hospital Medical Center Bezanijska Kosa, Belgrade, Serbia
| | - Dejan Sakac
- Institute for Cardiovascular Diseases of Vojvodina, Sremska Kamenica & Medical Faculty Novi Sad, Novi Sad, Serbia
| |
Collapse
|
14
|
GRKs and Epac1 Interaction in Cardiac Remodeling and Heart Failure. Cells 2021; 10:cells10010154. [PMID: 33466800 PMCID: PMC7830799 DOI: 10.3390/cells10010154] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/25/2022] Open
Abstract
β-adrenergic receptors (β-ARs) play a major role in the physiological regulation of cardiac function through signaling routes tightly controlled by G protein-coupled receptor kinases (GRKs). Although the acute stimulation of β-ARs and the subsequent production of cyclic AMP (cAMP) have beneficial effects on cardiac function, chronic stimulation of β-ARs as observed under sympathetic overdrive promotes the development of pathological cardiac remodeling and heart failure (HF), a leading cause of mortality worldwide. This is accompanied by an alteration in cAMP compartmentalization and the activation of the exchange protein directly activated by cAMP 1 (Epac1) signaling. Among downstream signals of β-ARs, compelling evidence indicates that GRK2, GRK5, and Epac1 represent attractive therapeutic targets for cardiac disease. Here, we summarize the pathophysiological roles of GRK2, GRK5, and Epac1 in the heart. We focus on their signalosome and describe how under pathological settings, these proteins can cross-talk and are part of scaffolded nodal signaling systems that contribute to a decreased cardiac function and HF development.
Collapse
|
15
|
Wu X, Larsson HP. Insights into Cardiac IKs (KCNQ1/KCNE1) Channels Regulation. Int J Mol Sci 2020; 21:ijms21249440. [PMID: 33322401 PMCID: PMC7763278 DOI: 10.3390/ijms21249440] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/05/2020] [Accepted: 12/09/2020] [Indexed: 12/19/2022] Open
Abstract
The delayed rectifier potassium IKs channel is an important regulator of the duration of the ventricular action potential. Hundreds of mutations in the genes (KCNQ1 and KCNE1) encoding the IKs channel cause long QT syndrome (LQTS). LQTS is a heart disorder that can lead to severe cardiac arrhythmias and sudden cardiac death. A better understanding of the IKs channel (here called the KCNQ1/KCNE1 channel) properties and activities is of great importance to find the causes of LQTS and thus potentially treat LQTS. The KCNQ1/KCNE1 channel belongs to the superfamily of voltage-gated potassium channels. The KCNQ1/KCNE1 channel consists of both the pore-forming subunit KCNQ1 and the modulatory subunit KCNE1. KCNE1 regulates the function of the KCNQ1 channel in several ways. This review aims to describe the current structural and functional knowledge about the cardiac KCNQ1/KCNE1 channel. In addition, we focus on the modulation of the KCNQ1/KCNE1 channel and its potential as a target therapeutic of LQTS.
Collapse
|
16
|
Hyperglycemia regulates cardiac K + channels via O-GlcNAc-CaMKII and NOX2-ROS-PKC pathways. Basic Res Cardiol 2020; 115:71. [PMID: 33237428 DOI: 10.1007/s00395-020-00834-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 11/11/2020] [Indexed: 12/14/2022]
Abstract
Chronic hyperglycemia and diabetes lead to impaired cardiac repolarization, K+ channel remodeling and increased arrhythmia risk. However, the exact signaling mechanism by which diabetic hyperglycemia regulates cardiac K+ channels remains elusive. Here, we show that acute hyperglycemia increases inward rectifier K+ current (IK1), but reduces the amplitude and inactivation recovery time of the transient outward K+ current (Ito) in mouse, rat, and rabbit myocytes. These changes were all critically dependent on intracellular O-GlcNAcylation. Additionally, IK1 amplitude and Ito recovery effects (but not Ito amplitude) were prevented by the Ca2+/calmodulin-dependent kinase II (CaMKII) inhibitor autocamtide-2-related inhibitory peptide, CaMKIIδ-knockout, and O-GlcNAc-resistant CaMKIIδ-S280A knock-in. Ito reduction was prevented by inhibition of protein kinase C (PKC) and NADPH oxidase 2 (NOX2)-derived reactive oxygen species (ROS). In mouse models of chronic diabetes (streptozotocin, db/db, and high-fat diet), heart failure, and CaMKIIδ overexpression, both Ito and IK1 were reduced in line with the downregulated K+ channel expression. However, IK1 downregulation in diabetes was markedly attenuated in CaMKIIδ-S280A. We conclude that acute hyperglycemia enhances IK1 and Ito recovery via CaMKIIδ-S280 O-GlcNAcylation, but reduces Ito amplitude via a NOX2-ROS-PKC pathway. Moreover, chronic hyperglycemia during diabetes and CaMKII activation downregulate K+ channel expression and function, which may further increase arrhythmia susceptibility.
Collapse
|
17
|
Shugg T, Hudmon A, Overholser BR. Neurohormonal Regulation of I Ks in Heart Failure: Implications for Ventricular Arrhythmogenesis and Sudden Cardiac Death. J Am Heart Assoc 2020; 9:e016900. [PMID: 32865116 PMCID: PMC7726975 DOI: 10.1161/jaha.120.016900] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Heart failure (HF) results in sustained alterations in neurohormonal signaling, including enhanced signaling through the sympathetic nervous system and renin-angiotensin-aldosterone system pathways. While enhanced sympathetic nervous system and renin-angiotensin-aldosterone system activity initially help compensate for the failing myocardium, sustained signaling through these pathways ultimately contributes to HF pathophysiology. HF remains a leading cause of mortality, with arrhythmogenic sudden cardiac death comprising a common mechanism of HF-related death. The propensity for arrhythmia development in HF occurs secondary to cardiac electrical remodeling that involves pathological regulation of ventricular ion channels, including the slow component of the delayed rectifier potassium current, that contribute to action potential duration prolongation. To elucidate a mechanistic explanation for how HF-mediated electrical remodeling predisposes to arrhythmia development, a multitude of investigations have investigated the specific regulatory effects of HF-associated stimuli, including enhanced sympathetic nervous system and renin-angiotensin-aldosterone system signaling, on the slow component of the delayed rectifier potassium current. The objective of this review is to summarize the current knowledge related to the regulation of the slow component of the delayed rectifier potassium current in response to HF-associated stimuli, including the intracellular pathways involved and the specific regulatory mechanisms.
Collapse
Affiliation(s)
- Tyler Shugg
- Division of Clinical PharmacologyIndiana University School of MedicineIndianapolisIN
| | - Andy Hudmon
- Department of Medicinal Chemistry and Molecular PharmacologyPurdue University College of PharmacyWest LafayetteIN
| | - Brian R. Overholser
- Division of Clinical PharmacologyIndiana University School of MedicineIndianapolisIN
- Department of Pharmacy PracticePurdue University College of PharmacyIndianapolisIN
| |
Collapse
|
18
|
Formoso K, Lezoualc'h F, Mialet-Perez J. Role of EPAC1 Signalosomes in Cell Fate: Friends or Foes? Cells 2020; 9:E1954. [PMID: 32854274 PMCID: PMC7563956 DOI: 10.3390/cells9091954] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/21/2020] [Accepted: 08/22/2020] [Indexed: 02/06/2023] Open
Abstract
The compartmentation of signaling processes is accomplished by the assembly of protein complexes called signalosomes. These signaling platforms colocalize enzymes, substrates, and anchoring proteins into specific subcellular compartments. Exchange protein directly activated by cAMP 1 (EPAC1) is an effector of the second messenger, 3',5'-cyclic adenosine monophosphate (cAMP) that is associated with multiple roles in several pathologies including cardiac diseases. Both EPAC1 intracellular localization and molecular partners are key players in the regulation of cell fate, which may have important therapeutic potential. In this review, we summarize the recent findings on EPAC1 structure, regulation, and pharmacology. We describe the importance of EPAC1 subcellular distribution in its biological action, paying special attention to its nuclear localization and mechanism of action leading to cardiomyocyte hypertrophy. In addition, we discuss the role of mitochondrial EPAC1 in the regulation of cell death. Depending on the cell type and stress condition, we present evidence that supports either a protective or detrimental role of EPAC1 activation.
Collapse
Affiliation(s)
- Karina Formoso
- INSERM UMR-1048, Institute of Metabolic and Cardiovascular Diseases, and Université de Toulouse III-Paul Sabatier, 31432 Toulouse, France
| | - Frank Lezoualc'h
- INSERM UMR-1048, Institute of Metabolic and Cardiovascular Diseases, and Université de Toulouse III-Paul Sabatier, 31432 Toulouse, France
| | - Jeanne Mialet-Perez
- INSERM UMR-1048, Institute of Metabolic and Cardiovascular Diseases, and Université de Toulouse III-Paul Sabatier, 31432 Toulouse, France
| |
Collapse
|
19
|
van der Horst J, Greenwood IA, Jepps TA. Cyclic AMP-Dependent Regulation of Kv7 Voltage-Gated Potassium Channels. Front Physiol 2020; 11:727. [PMID: 32695022 PMCID: PMC7338754 DOI: 10.3389/fphys.2020.00727] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/04/2020] [Indexed: 01/08/2023] Open
Abstract
Voltage-gated Kv7 potassium channels, encoded by KCNQ genes, have major physiological impacts cardiac myocytes, neurons, epithelial cells, and smooth muscle cells. Cyclic adenosine monophosphate (cAMP), a well-known intracellular secondary messenger, can activate numerous downstream effector proteins, generating downstream signaling pathways that regulate many functions in cells. A role for cAMP in ion channel regulation has been established, and recent findings show that cAMP signaling plays a role in Kv7 channel regulation. Although cAMP signaling is recognized to regulate Kv7 channels, the precise molecular mechanism behind the cAMP-dependent regulation of Kv7 channels is complex. This review will summarize recent research findings that support the mechanisms of cAMP-dependent regulation of Kv7 channels.
Collapse
Affiliation(s)
- Jennifer van der Horst
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Iain A Greenwood
- Molecular and Clinical Sciences Institute, St. George's University of London, London, United Kingdom
| | - Thomas A Jepps
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
20
|
Joca HC, Santos-Miranda A, Joviano-Santos JV, Maia-Joca RPM, Brum PC, Williams GSB, Cruz JS. Chronic Sympathetic Hyperactivity Triggers Electrophysiological Remodeling and Disrupts Excitation-Contraction Coupling in Heart. Sci Rep 2020; 10:8001. [PMID: 32409748 PMCID: PMC7224293 DOI: 10.1038/s41598-020-64949-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/20/2020] [Indexed: 12/31/2022] Open
Abstract
The sympathetic nervous system is essential for maintenance of cardiac function via activation of post-junctional adrenergic receptors. Prolonged adrenergic receptor activation, however, has deleterious long-term effects leading to hypertrophy and the development of heart failure. Here we investigate the effect of chronic adrenergic receptors activation on excitation-contraction coupling (ECC) in ventricular cardiomyocytes from a previously characterized mouse model of chronic sympathetic hyperactivity, which are genetically deficient in the adrenoceptor α2A and α2C genes (ARDKO). When compared to wild-type (WT) cardiomyocytes, ARDKO displayed reduced fractional shortening (~33%) and slower relaxation (~20%). Furthermore, ARDKO cells exhibited several electrophysiological changes such as action potential (AP) prolongation (~50%), reduced L-type calcium channel (LCC) current (~33%), reduced outward potassium (K+) currents (~30%), and increased sodium/calcium exchanger (NCX) activity (~52%). Consistent with reduced contractility and calcium (Ca2+) currents, the cytosolic Ca2+ ([Ca2+]i) transient from ARDKO animals was smaller and decayed slower. Importantly, no changes were observed in membrane resting potential, AP amplitude, or the inward K+ current. Finally, we modified our existing cardiac ECC computational model to account for changes in the ARDKO heart. Simulations suggest that cellular changes in the ARDKO heart resulted in variable and dyssynchronous Ca2+-induced Ca2+ release therefore altering [Ca2+]i transient dynamics and reducing force generation. In conclusion, chronic sympathetic hyperactivity impairs ECC by changing the density of several ionic currents (and thus AP repolarization) causing altered Ca2+ dynamics and contractile activity. This demonstrates the important role of ECC remodeling in the cardiac dysfunction secondary to chronic sympathetic activity.
Collapse
Affiliation(s)
- Humberto C Joca
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Artur Santos-Miranda
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Department of Biophysics, Universidade Federal de Sao Paulo, Sao Paulo, SP, Brazil
| | | | - Rebeca P M Maia-Joca
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Patricia C Brum
- School of Physical Education and Sport, University of São Paulo, São Paulo, SP, Brazil
| | - George S B Williams
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jader S Cruz
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
21
|
Cardiomyocyte calcium handling in health and disease: Insights from in vitro and in silico studies. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2020; 157:54-75. [PMID: 32188566 DOI: 10.1016/j.pbiomolbio.2020.02.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/31/2019] [Accepted: 02/29/2020] [Indexed: 02/07/2023]
Abstract
Calcium (Ca2+) plays a central role in cardiomyocyte excitation-contraction coupling. To ensure an optimal electrical impulse propagation and cardiac contraction, Ca2+ levels are regulated by a variety of Ca2+-handling proteins. In turn, Ca2+ modulates numerous electrophysiological processes. Accordingly, Ca2+-handling abnormalities can promote cardiac arrhythmias via various mechanisms, including the promotion of afterdepolarizations, ion-channel modulation and structural remodeling. In the last 30 years, significant improvements have been made in the computational modeling of cardiomyocyte Ca2+ handling under physiological and pathological conditions. However, numerous questions involving the Ca2+-dependent regulation of different macromolecular complexes, cross-talk between Ca2+-dependent regulatory pathways operating over a wide range of time scales, and bidirectional interactions between electrophysiology and mechanics remain to be addressed by in vitro and in silico studies. A better understanding of disease-specific Ca2+-dependent proarrhythmic mechanisms may facilitate the development of improved therapeutic strategies. In this review, we describe the fundamental mechanisms of cardiomyocyte Ca2+ handling in health and disease, and provide an overview of currently available computational models for cardiomyocyte Ca2+ handling. Finally, we discuss important uncertainties and open questions about cardiomyocyte Ca2+ handling and highlight how synergy between in vitro and in silico studies may help to answer several of these issues.
Collapse
|
22
|
Surinkaew S, Aflaki M, Takawale A, Chen Y, Qi XY, Gillis MA, Shi YF, Tardif JC, Chattipakorn N, Nattel S. Exchange protein activated by cyclic-adenosine monophosphate (Epac) regulates atrial fibroblast function and controls cardiac remodelling. Cardiovasc Res 2020; 115:94-106. [PMID: 30016400 DOI: 10.1093/cvr/cvy173] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 07/13/2018] [Indexed: 12/21/2022] Open
Abstract
Aims Heart failure (HF) produces left atrial (LA)-selective fibrosis and promotes atrial fibrillation. HF also causes adrenergic activation, which contributes to remodelling via a variety of signalling molecules, including the exchange protein activated by cAMP (Epac). Here, we evaluate the effects of Epac1-signalling on LA fibroblast (FB) function and its potential role in HF-related atrial remodelling. Methods and results HF was induced in adult male mongrel dogs by ventricular tachypacing (VTP). Epac1-expression decreased in LA-FBs within 12 h (-3.9-fold) of VTP onset. The selective Epac activator, 8-pCPT (50 µM) reduced, whereas the Epac blocker ESI-09 (1 µM) enhanced, collagen expression in LA-FBs. Norepinephrine (1 µM) decreased Epac1-expression, an effect blocked by prazosin, and increased FB collagen production. The β-adrenoceptor (AR) agonist isoproterenol increased Epac1 expression, an effect antagonized by ICI (β2-AR-blocker), but not by CGP (β1-AR-blocker). β-AR-activation with isoproterenol decreased collagen expression, an effect mimicked by the β2-AR-agonist salbutamol and blocked by the Epac1-antagonist ESI-09. Transforming growth factor-β1, known to be activated in HF, suppressed Epac1 expression, an effect blocked by the Smad3-inhibitor SIS3. To evaluate effects on atrial fibrosis in vivo, mice subjected to myocardial infarction (MI) received the Epac-activator Sp-8-pCPT or vehicle for 2 weeks post-MI; Sp-8-pCPT diminished LA fibrosis and attenuated cardiac dysfunction. Conclusions HF reduces LA-FB Epac1 expression. Adrenergic activation has complex effects on FBs, with α-AR-activation suppressing Epac1-expression and increasing collagen expression, and β2-AR-activation having opposite effects. Epac1-activation reduces cardiac dysfunction and LA fibrosis post-MI. Thus, Epac1 signalling may be a novel target for the prevention of profibrillatory cardiac remodelling.
Collapse
Affiliation(s)
- Sirirat Surinkaew
- Department of Medicine, and Research Center Montreal Heart Institute and Université de Montréal, 5000 Belanger St. E., Montreal, Quebec, Canada.,Faculty of Associated Medical Sciences, Biomedical Technology Research Center, Chiang Mai University, 110 Inthawarorot Rd, Chiang Mai, Thailand
| | - Mona Aflaki
- Department of Medicine, and Research Center Montreal Heart Institute and Université de Montréal, 5000 Belanger St. E., Montreal, Quebec, Canada.,Faculty of Medicine, Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada
| | - Abhijit Takawale
- Department of Medicine, and Research Center Montreal Heart Institute and Université de Montréal, 5000 Belanger St. E., Montreal, Quebec, Canada.,Faculty of Medicine, Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada
| | - Yu Chen
- Department of Medicine, and Research Center Montreal Heart Institute and Université de Montréal, 5000 Belanger St. E., Montreal, Quebec, Canada
| | - Xiao-Yan Qi
- Department of Medicine, and Research Center Montreal Heart Institute and Université de Montréal, 5000 Belanger St. E., Montreal, Quebec, Canada
| | - Marc-Antoine Gillis
- Department of Medicine, and Research Center Montreal Heart Institute and Université de Montréal, 5000 Belanger St. E., Montreal, Quebec, Canada
| | - Yan-Fen Shi
- Department of Medicine, and Research Center Montreal Heart Institute and Université de Montréal, 5000 Belanger St. E., Montreal, Quebec, Canada
| | - Jean-Claude Tardif
- Department of Medicine, and Research Center Montreal Heart Institute and Université de Montréal, 5000 Belanger St. E., Montreal, Quebec, Canada
| | - Nipon Chattipakorn
- Department of Physiology, Faculty of Medicine, Cardiac Electrophysiology Research and Training Center, Chiang Mai University, 110 Inthawarorot Rd, Chiang Mai, Thailand
| | - Stanley Nattel
- Department of Medicine, and Research Center Montreal Heart Institute and Université de Montréal, 5000 Belanger St. E., Montreal, Quebec, Canada.,Faculty of Medicine, Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada.,Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Hufelandstrasse 55, Essen Germany
| |
Collapse
|
23
|
The Epac1 Protein: Pharmacological Modulators, Cardiac Signalosome and Pathophysiology. Cells 2019; 8:cells8121543. [PMID: 31795450 PMCID: PMC6953115 DOI: 10.3390/cells8121543] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/22/2019] [Accepted: 11/24/2019] [Indexed: 12/11/2022] Open
Abstract
The second messenger 3′,5′-cyclic adenosine monophosphate (cAMP) is one of the most important signalling molecules in the heart as it regulates many physiological and pathophysiological processes. In addition to the classical protein kinase A (PKA) signalling route, the exchange proteins directly activated by cAMP (Epac) mediate the intracellular functions of cAMP and are now emerging as a new key cAMP effector in cardiac pathophysiology. In this review, we provide a perspective on recent advances in the discovery of new chemical entities targeting the Epac1 isoform and illustrate their use to study the Epac1 signalosome and functional characterisation in cardiac cells. We summarize the role of Epac1 in different subcompartments of the cardiomyocyte and discuss how cAMP–Epac1 specific signalling networks may contribute to the development of cardiac diseases. We also highlight ongoing work on the therapeutic potential of Epac1-selective small molecules for the treatment of cardiac disorders.
Collapse
|
24
|
Wang J, Dai M, Cao Q, Yu Q, Luo Q, Shu L, Zhang Y, Bao M. Carotid baroreceptor stimulation suppresses ventricular fibrillation in canines with chronic heart failure. Basic Res Cardiol 2019; 114:41. [PMID: 31502080 DOI: 10.1007/s00395-019-0750-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 09/06/2019] [Indexed: 12/16/2022]
Abstract
Carotid baroreceptor stimulation (CBS) has been shown to improve cardiac dysfunction and pathological structure remodelling. This study aimed to investigate the effects of CBS on the ventricular electrophysiological properties in canines with chronic heart failure (CHF). Thirty-eight beagles were randomized into control (CON), CHF, low-level CBS (LL-CBS), and moderate-level CBS (ML-CBS) groups. The CHF model was established with 6 weeks of rapid right ventricular pacing (RVP), and concomitant LL-CBS and ML-CBS were applied in the LL-CBS and ML-CBS groups, respectively. After 6 weeks of RVP, ventricular electrophysiological parameters and left stellate ganglion (LSG) neural activity and function were measured. Autonomic neural remodelling in the LSG and left ventricle (LV) and ionic remodelling in the LV were detected. Compared with the CHF group, both LL-CBS and ML-CBS decreased spatial dispersion of action potential duration (APD), suppressed APD alternans, reduced ventricular fibrillation (VF) inducibility, and inhibited enhanced LSG neural discharge and function. Only ML-CBS significantly inhibited ventricular repolarization prolongation and increased the VF threshold. Moreover, ML-CBS inhibited the increase in growth-associated protein-43 and tyrosine hydroxylase-positive nerve fibre densities in LV, increased acetylcholinesterase protein expression in LSG, and decreased nerve growth factor protein expression in LSG and LV. Chronic RVP resulted in a remarkable reduction in protein expression encoding both potassium and L-type calcium currents; these changes were partly amended by ML-CBS and LL-CBS. In conclusion, CBS suppresses VF in CHF canines, potentially by modulating autonomic nerve and ion channels. In addition, the effects of ML-CBS on ventricular electrophysiological properties, autonomic remodelling, and ionic remodelling were superior to those of LL-CBS.
Collapse
Affiliation(s)
- Jing Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Mingyan Dai
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Quan Cao
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Qiao Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Qiang Luo
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Ling Shu
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Yijie Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China
| | - Mingwei Bao
- Department of Cardiology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei, People's Republic of China.
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China.
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, People's Republic of China.
| |
Collapse
|
25
|
Grandi E, Ripplinger CM. Antiarrhythmic mechanisms of beta blocker therapy. Pharmacol Res 2019; 146:104274. [PMID: 31100336 DOI: 10.1016/j.phrs.2019.104274] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 05/04/2019] [Accepted: 05/13/2019] [Indexed: 02/07/2023]
Abstract
Sympathetic activity plays an important role in modulation of cardiac rhythm. Indeed, while exerting positive tropic effects in response to physiologic and pathologic stressors, β-adrenergic stimulation influences cardiac electrophysiology and can lead to disturbances of the heart rhythm and potentially lethal arrhythmias, particularly in pathological settings. For this reason, β-blockers are widely utilized clinically as antiarrhythmics. In this review, the molecular mechanisms of β-adrenergic action in the heart, the cellular and tissue level cardiac responses to β-adrenergic stimulation, and the clinical use of β-blockers as antiarrhythmic agents are reviewed. We emphasize the complex interaction between cardiomyocyte signaling, contraction, and electrophysiology occurring over multiple time- and spatial-scales during pathophysiological responses to β-adrenergic stimulation. An integrated understanding of this complex system is essential for optimizing therapies aimed at preventing arrhythmias.
Collapse
Affiliation(s)
- Eleonora Grandi
- Department of Pharmacology, University of California Davis, United States.
| | | |
Collapse
|
26
|
Hegyi B, Bers DM, Bossuyt J. CaMKII signaling in heart diseases: Emerging role in diabetic cardiomyopathy. J Mol Cell Cardiol 2019; 127:246-259. [PMID: 30633874 DOI: 10.1016/j.yjmcc.2019.01.001] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 01/04/2019] [Indexed: 02/07/2023]
Abstract
Calcium/calmodulin-dependent protein kinase II (CaMKII) is upregulated in diabetes and significantly contributes to cardiac remodeling with increased risk of cardiac arrhythmias. Diabetes is frequently associated with atrial fibrillation, coronary artery disease, and heart failure, which may further enhance CaMKII. Activation of CaMKII occurs downstream of neurohormonal stimulation (e.g. via G-protein coupled receptors) and involve various posttranslational modifications including autophosphorylation, oxidation, S-nitrosylation and O-GlcNAcylation. CaMKII signaling regulates diverse cellular processes in a spatiotemporal manner including excitation-contraction and excitation-transcription coupling, mechanics and energetics in cardiac myocytes. Chronic activation of CaMKII results in cellular remodeling and ultimately arrhythmogenic alterations in Ca2+ handling, ion channels, cell-to-cell coupling and metabolism. This review addresses the detrimental effects of the upregulated CaMKII signaling to enhance the arrhythmogenic substrate and trigger mechanisms in the heart. We also briefly summarize preclinical studies using kinase inhibitors and genetically modified mice targeting CaMKII in diabetes. The mechanistic understanding of CaMKII signaling, cardiac remodeling and arrhythmia mechanisms may reveal new therapeutic targets and ultimately better treatment in diabetes and heart disease in general.
Collapse
Affiliation(s)
- Bence Hegyi
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - Donald M Bers
- Department of Pharmacology, University of California Davis, Davis, CA, USA.
| | - Julie Bossuyt
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| |
Collapse
|
27
|
Chen M, Yin D, Guo S, Xu DZ, Wang Z, Chen Z, Rubart-von der Lohe M, Lin SF, Everett Iv TH, Weiss JN, Chen PS. Sex-specific activation of SK current by isoproterenol facilitates action potential triangulation and arrhythmogenesis in rabbit ventricles. J Physiol 2018; 596:4299-4322. [PMID: 29917243 DOI: 10.1113/jp275681] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 06/11/2018] [Indexed: 01/09/2023] Open
Abstract
KEY POINTS It is unknown if a sex difference exists in cardiac apamin-sensitive small conductance Ca2+ -activated K+ (SK) current (IKAS ). There is no sex difference in IKAS in the basal condition. However, there is larger IKAS in female rabbit ventricles than in male during isoproterenol infusion. IKAS activation by isoproterenol leads to action potential triangulation in females, indicating its abundant activation at early phases of repolarization. IKAS activation in females induces negative Ca2+ -voltage coupling and promotes electromechanically discordant phase 2 repolarization alternans. IKAS is important in the mechanisms of ventricular fibrillation in females during sympathetic stimulation. ABSTRACT Sex has a large influence on cardiac electrophysiological properties. Whether sex differences exist in apamin-sensitive small conductance Ca2+ -activated K+ (SK) current (IKAS ) remains unknown. We performed optical mapping, transmembrane potential, patch clamp, western blot and immunostaining in 62 normal rabbit ventricles, including 32 females and 30 males. IKAS blockade by apamin only minimally prolonged action potential (AP) duration (APD) in the basal condition for both sexes, but significantly prolonged APD in the presence of isoproterenol in females. Apamin prolonged APD at the level of 25% repolarization (APD25 ) more prominently than APD at the level of 80% repolarization (APD80 ), consequently reversing isoproterenol-induced AP triangulation in females. In comparison, apamin prolonged APD to a significantly lesser extent in males and failed to restore the AP plateau during isoproterenol infusion. IKAS in males did not respond to the L-type calcium current agonist BayK8644, but was amplified by the casein kinase 2 (CK2) inhibitor 4,5,6,7-tetrabromobenzotriazole. In addition, whole-cell outward IKAS densities in ventricular cardiomyocytes were significantly larger in females than in males. SK channel subtype 2 (SK2) protein expression was higher and the CK2/SK2 ratio was lower in females than in males. IKAS activation in females induced negative intracellular Ca2+ -voltage coupling, promoted electromechanically discordant phase 2 repolarization alternans and facilitated ventricular fibrillation (VF). Apamin eliminated the negative Ca2+ -voltage coupling, attenuated alternans and reduced VF inducibility, phase singularities and dominant frequencies in females, but not in males. We conclude that β-adrenergic stimulation activates ventricular IKAS in females to a much greater extent than in males. IKAS activation plays an important role in ventricular arrhythmogenesis in females during sympathetic stimulation.
Collapse
Affiliation(s)
- Mu Chen
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dechun Yin
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuai Guo
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dong-Zhu Xu
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Cardiovascular Division, Institute of Clinical Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Zhuo Wang
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhenhui Chen
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Michael Rubart-von der Lohe
- Department of Pediatrics, Riley Heart Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shien-Fong Lin
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Institute of Biomedical Engineering, National Chiao-Tung University, Hsin-Chu, Taiwan
| | - Thomas H Everett Iv
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - James N Weiss
- Departments of Medicine (Cardiology) and Physiology, University of California, Los Angeles, CA, USA
| | - Peng-Sheng Chen
- Krannert Institute of Cardiology and Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
28
|
Yuan YH, Zheng XM, He XH, Liu LP, Xu W, Xia XH, Luo JH, Lyu M, Zhu QL, Wang S, Wu S. [Establishment of cardiac remodeling model in FVB/N mice by intraperitoneal injection of isoproterenol]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2018; 20:508-513. [PMID: 29972128 PMCID: PMC7389954 DOI: 10.7499/j.issn.1008-8830.2018.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/08/2018] [Indexed: 06/08/2023]
Abstract
OBJECTIVE To explore the feasibility of intraperitoneal injection of isoproterenol (ISO) to induce cardiac remodeling in FVB/N mice. METHODS Forty-eight FVB/N mice were divided into back subcutaneous saline group (subcutaneous saline group), intraperitoneal saline group, back subcutaneous ISO group (subcutaneous ISO group), and intraperitoneal ISO group according to the route of administration of saline or ISO. ISO (30 μg/g body weight/day) was given to the subcutaneous ISO group and the intraperitoneal ISO group, twice daily with an interval of 12 hours, for 14 consecutive days. The subcutaneous saline group and the intraperitoneal saline group were injected with an equal volume of saline. The left ventricular end-diastolic posterior wall thickness was measured by echocardiography, and the ratio of heart weight to tibia length was determined. Hematoxylin-eosin staining was used to determine the myocardial fiber diameter. Picric-sirius red staining was used to determine the myocardial collagen deposition area. Quantitative real-time PCR was used to measure the mRNA expression of collagen I. RESULTS Compared with the subcutaneous ISO, subcutaneous saline, and intraperitoneal saline groups, the intraperitoneal ISO group had increased sizes of the cardiac cavity and the heart. Compared with the subcutaneous saline and intraperitoneal saline groups, the subcutaneous ISO group showed no significant changes in the gross morphology of the cardiac cavity and the heart. The intraperitoneal ISO group showed significant increases in the ratio of heart weight to tibia length, myocardial fiber diameter, left ventricular end-diastolic posterior wall thickness, myocardial collagen area percentage, and the mRNA expression of collagen I compared with the subcutaneous ISO, subcutaneous saline, and intraperitoneal saline groups (P<0.01). There were no significant differences in the above five indices between the subcutaneous ISO group and the subcutaneous saline and intraperitoneal saline groups (P>0.05). No significant difference in the mortality rate was found between the subcutaneous ISO and intraperitoneal ISO groups (P>0.05). CONCLUSIONS Intraperitoneal injection of ISO can induce cardiac hypertrophy and fibrosis in FVB/N mice.
Collapse
Affiliation(s)
- Yong-Hua Yuan
- Department of Pediatric Cardiology, Hunan People's Hospital, Changsha 410005, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
A novel role of the antitumor agent tricyclodecan-9-yl-xanthogenate as an open channel blocker of KCNQ1/KCNE1. Eur J Pharmacol 2018; 824:99-107. [DOI: 10.1016/j.ejphar.2018.02.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/08/2018] [Accepted: 02/09/2018] [Indexed: 12/26/2022]
|
30
|
Affiliation(s)
- Jordi Heijman
- From the Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, The Netherlands (J.H.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Canada (J.-B.G., S.N.); University Hospital of Saint-Étienne, University Jean Monnet, Saint-Étienne, France (J.-B.G.); Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen (D.D., S.N.); and
| | - Jean-Baptiste Guichard
- From the Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, The Netherlands (J.H.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Canada (J.-B.G., S.N.); University Hospital of Saint-Étienne, University Jean Monnet, Saint-Étienne, France (J.-B.G.); Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen (D.D., S.N.); and
| | - Dobromir Dobrev
- From the Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, The Netherlands (J.H.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Canada (J.-B.G., S.N.); University Hospital of Saint-Étienne, University Jean Monnet, Saint-Étienne, France (J.-B.G.); Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen (D.D., S.N.); and
| | - Stanley Nattel
- From the Department of Cardiology, Cardiovascular Research Institute Maastricht, Faculty of Health, Medicine, and Life Sciences, Maastricht University, The Netherlands (J.H.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Canada (J.-B.G., S.N.); University Hospital of Saint-Étienne, University Jean Monnet, Saint-Étienne, France (J.-B.G.); Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen (D.D., S.N.); and
| |
Collapse
|
31
|
Laudette M, Zuo H, Lezoualc'h F, Schmidt M. Epac Function and cAMP Scaffolds in the Heart and Lung. J Cardiovasc Dev Dis 2018; 5:jcdd5010009. [PMID: 29401660 PMCID: PMC5872357 DOI: 10.3390/jcdd5010009] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 01/25/2018] [Accepted: 01/29/2018] [Indexed: 12/13/2022] Open
Abstract
Evidence collected over the last ten years indicates that Epac and cAMP scaffold proteins play a critical role in integrating and transducing multiple signaling pathways at the basis of cardiac and lung physiopathology. Some of the deleterious effects of Epac, such as cardiomyocyte hypertrophy and arrhythmia, initially described in vitro, have been confirmed in genetically modified mice for Epac1 and Epac2. Similar recent findings have been collected in the lung. The following sections will describe how Epac and cAMP signalosomes in different subcellular compartments may contribute to cardiac and lung diseases.
Collapse
Affiliation(s)
- Marion Laudette
- Inserm UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université Toulouse III, 31432 Toulouse, France.
| | - Haoxiao Zuo
- Department of Molecular Pharmacology, University of Groningen, 9713AV Groningen, The Netherlands.
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, 9713AV Groningen, The Netherlands.
| | - Frank Lezoualc'h
- Inserm UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université Toulouse III, 31432 Toulouse, France.
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, 9713AV Groningen, The Netherlands.
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, 9713AV Groningen, The Netherlands.
| |
Collapse
|
32
|
Shugg T, Johnson DE, Shao M, Lai X, Witzmann F, Cummins TR, Rubart-Von-der Lohe M, Hudmon A, Overholser BR. Calcium/calmodulin-dependent protein kinase II regulation of I Ks during sustained β-adrenergic receptor stimulation. Heart Rhythm 2018; 15:895-904. [PMID: 29410121 DOI: 10.1016/j.hrthm.2018.01.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Indexed: 01/21/2023]
Abstract
BACKGROUND Sustained β-adrenergic receptor (β-AR) stimulation causes pathophysiological changes during heart failure (HF), including inhibition of the slow component of the delayed rectifier potassium current (IKs). Aberrant calcium handling, including increased activation of calcium/calmodulin-dependent protein kinase II (CaMKII), contributes to arrhythmia development during HF. OBJECTIVE The purpose of this study was to investigate CaMKII regulation of KCNQ1 (pore-forming subunit of IKs) during sustained β-AR stimulation and associated functional implications on IKs. METHODS KCNQ1 phosphorylation was assessed using liquid chromatography-tandem mass spectrometry after sustained β-AR stimulation with isoproterenol (ISO). Peptide fragments corresponding to KCNQ1 residues were synthesized to identify CaMKII phosphorylation at the identified sites. Dephosphorylated (alanine) and phosphorylated (aspartic acid) mimics were introduced at identified residues. Whole-cell, voltage-clamp experiments were performed in human endothelial kidney 293 cells coexpressing wild-type or mutant KCNQ1 and KCNE1 (auxiliary subunit) during ISO treatment or lentiviral δCaMKII overexpression. RESULTS Novel KCNQ1 carboxy-terminal sites were identified with enhanced phosphorylation during sustained β-AR stimulation at T482 and S484. S484 peptides demonstrated the strongest δCaMKII phosphorylation. Sustained β-AR stimulation reduced IKs activation (P = .02 vs control) similar to the phosphorylated mimic (P = .62 vs sustained β-AR). Individual phosphorylated mimics at S484 (P = .04) but not at T482 (P = .17) reduced IKs function. Treatment with CN21 (CaMKII inhibitor) reversed the reductions in IKs vs CN21-Alanine control (P < .01). δCaMKII overexpression reduced IKs similar to ISO treatment in wild type (P < .01) but not in the dephosphorylated S484 mimic (P = .99). CONCLUSION CaMKII regulates KCNQ1 at S484 during sustained β-AR stimulation to inhibit IKs. The ability of CaMKII to inhibit IKs may contribute to arrhythmogenicity during HF.
Collapse
Affiliation(s)
- Tyler Shugg
- Department of Pharmacy Practice, College of Pharmacy, Purdue University, West Lafayette, Indiana
| | - Derrick E Johnson
- Department of Biochemistry and Molecular Biology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana
| | - Minghai Shao
- Department of Pharmacy Practice, College of Pharmacy, Purdue University, West Lafayette, Indiana
| | - Xianyin Lai
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Frank Witzmann
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Theodore R Cummins
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana
| | - Michael Rubart-Von-der Lohe
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Andy Hudmon
- Department of Biochemistry and Molecular Biology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana
| | - Brian R Overholser
- Department of Pharmacy Practice, College of Pharmacy, Purdue University, West Lafayette, Indiana; Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
33
|
Yao H, Ji CC, Cheng YJ, Chen XM, Liu LJ, Fan J, Wu SH. Mutation in KCNE1 associated to early repolarization syndrome by modulation of slowly activating delayed rectifier K + current. Exp Cell Res 2018; 363:315-320. [DOI: 10.1016/j.yexcr.2018.01.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/11/2018] [Accepted: 01/20/2018] [Indexed: 01/13/2023]
|
34
|
Leroy J, Vandecasteele G, Fischmeister R. Cyclic AMP signaling in cardiac myocytes. CURRENT OPINION IN PHYSIOLOGY 2018. [DOI: 10.1016/j.cophys.2017.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
35
|
Lezcano N, Mariángelo JIE, Vittone L, Wehrens XHT, Said M, Mundiña-Weilenmann C. Early effects of Epac depend on the fine-tuning of the sarcoplasmic reticulum Ca 2+ handling in cardiomyocytes. J Mol Cell Cardiol 2018; 114:1-9. [PMID: 29037982 PMCID: PMC5801154 DOI: 10.1016/j.yjmcc.2017.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 09/19/2017] [Accepted: 10/09/2017] [Indexed: 12/30/2022]
Abstract
In cardiac muscle, signaling through cAMP governs many fundamental cellular functions, including contractility, relaxation and automatism. cAMP cascade leads to the activation of the classic protein kinase A but also to the stimulation of the recently discovered exchange protein directly activated by cAMP (Epac). The role of Epac in the regulation of intracellular Ca2+ homeostasis and contractility in cardiac myocytes is still matter of debate. In this study we showed that the selective Epac activator, 8-(4-chloro-phenylthio)-2'-O-methyladenosine-3', 5'-cyclic monophosphate (8-CPT), produced a positive inotropic effect when adult rat cardiac myocytes were stabilized at low [Ca2+]o (0.5mM), no changes at 1mM [Ca2+]o and a negative inotropic effect when [Ca2+]o was increased to 1.8mM. These effects were associated to parallel variations in sarcoplasmic reticulum (SR) Ca2+ content. At all [Ca2+]o studied, 8-CPT induced an increase in Ca2+ spark frequency and enhanced CaMKII autophosphorylation and the CaMKII-dependent phosphorylation of SR proteins: phospholamban (PLN, at Thr17 site) and ryanodine receptor (RyR2, at Ser2814 site). We used transgenic mice lacking PLN CaMKII phosphorylation site (PLN-DM) and knock-in mice with an inactivated CaMKII site S2814 on RyR2 (RyR2-S2814A) to investigate the involvement of these processes in the effects of Epac stimulation. In PLN-DM mice, 8-CPT failed to induce the positive inotropic effect at low [Ca2+]o and RyR2-S2814A mice showed no propensity to arrhythmic events when compared to wild type mice myocytes. We conclude that stimulation of Epac proteins could have either beneficial or deleterious effects depending on the steady-state Ca2+ levels at which the myocyte is functioning, favoring the prevailing mechanism of SR Ca2+ handling (uptake vs. leak) in the different situations.
Collapse
Affiliation(s)
- N Lezcano
- Centro de Investigaciones Cardiovasculares, CCT-CONICET La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Argentina
| | - J I E Mariángelo
- Centro de Investigaciones Cardiovasculares, CCT-CONICET La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Argentina
| | - L Vittone
- Centro de Investigaciones Cardiovasculares, CCT-CONICET La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Argentina
| | - X H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, United States
| | - M Said
- Centro de Investigaciones Cardiovasculares, CCT-CONICET La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Argentina
| | - C Mundiña-Weilenmann
- Centro de Investigaciones Cardiovasculares, CCT-CONICET La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Argentina.
| |
Collapse
|
36
|
Fujita T, Umemura M, Yokoyama U, Okumura S, Ishikawa Y. The role of Epac in the heart. Cell Mol Life Sci 2017; 74:591-606. [PMID: 27549789 PMCID: PMC11107744 DOI: 10.1007/s00018-016-2336-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 07/21/2016] [Accepted: 08/09/2016] [Indexed: 02/08/2023]
Abstract
As one of the most important second messengers, 3',5'-cyclic adenosine monophosphate (cAMP) mediates various extracellular signals including hormones and neurotransmitters, and induces appropriate responses in diverse types of cells. Since cAMP was formerly believed to transmit signals through only two direct target molecules, protein kinase A and the cyclic nucleotide-gated channel, the sensational discovery in 1998 of another novel direct effecter of cAMP [exchange proteins directly activated by cAMP (Epac)] attracted a great deal of scientific interest in cAMP signaling. Numerous studies on Epac have since disclosed its important functions in various tissues in the body. Recently, observations of genetically manipulated mice in various pathogenic models have begun to reveal the in vivo significance of previous in vitro or cellular-level findings. Here, we focused on the function of Epac in the heart. Accumulating evidence has revealed that both Epac1 and Epac2 play important roles in the structure and function of the heart under physiological and pathological conditions. Accordingly, developing the ability to regulate cAMP-mediated signaling through Epac may lead to remarkable new therapies for the treatment of cardiac diseases.
Collapse
Affiliation(s)
- Takayuki Fujita
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Masanari Umemura
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Utako Yokoyama
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Satoshi Okumura
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshihiro Ishikawa
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| |
Collapse
|
37
|
Screening, verification, and analysis of biomarkers for drug-induced cardiac toxicity in vitro based on RTCA coupled with PCR Array technology. Toxicol Lett 2017; 268:17-25. [PMID: 28099878 DOI: 10.1016/j.toxlet.2017.01.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 01/11/2017] [Accepted: 01/13/2017] [Indexed: 12/23/2022]
Abstract
Cardiotoxicity is one of the most serious side effects of new drugs. Early detection of the drug induced cardiotoxicity based on the biomarkers provides an important preventative strategy for detecting potential cardiotoxicity of candidate drugs. In this study, we aim to identify the predictive genomics biomarkers for drug-induced cardiac toxicity based on the RTCA coupled with PCR Array technology in primary cells. Three prototypical cardiotoxic compounds (doxorubicin, isoproterenol, ouabain) with different mechanisms were firstly real-time monitored to diagnose the cytotoxicity by using the RTCA, while the functional alterations of cardiomyocytes were also monitored by analyzing the beating frequency of cardiomyocytes. Then cardiac specific toxicity gene expression changes were studied by using the technology of PCR Array, which can detect the changes of 84 cardiac functions related genes. Rps6kb1 was identified to be the common cardiac biomarkers by using multivariate statistical and integration analyses. The biomarker was further verified by selecting other drugs with or without cardiotoxicity, and the results showed that the gene exhibited specific changes in cardiac toxicity. Moreover, IPA was applied to combine relevant pathways of Rps6kb1, and identify the main types of cardiac toxicity. These results would further enrich the evaluating strategy of drug-induced cardiotoxicity in vitro, and Rps6kb1 could be used as the specific biomarker of cardiotoxcity during safety assessment of the novel drug candidates.
Collapse
|
38
|
Lezoualc'h F, Fazal L, Laudette M, Conte C. Cyclic AMP Sensor EPAC Proteins and Their Role in Cardiovascular Function and Disease. Circ Res 2016; 118:881-97. [PMID: 26941424 DOI: 10.1161/circresaha.115.306529] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
cAMP is a universal second messenger that plays central roles in cardiovascular regulation influencing gene expression, cell morphology, and function. A crucial step toward a better understanding of cAMP signaling came 18 years ago with the discovery of the exchange protein directly activated by cAMP (EPAC). The 2 EPAC isoforms, EPAC1 and EPAC2, are guanine-nucleotide exchange factors for the Ras-like GTPases, Rap1 and Rap2, which they activate independently of the classical effector of cAMP, protein kinase A. With the development of EPAC pharmacological modulators, many reports in the literature have demonstrated the critical role of EPAC in the regulation of various cAMP-dependent cardiovascular functions, such as calcium handling and vascular tone. EPAC proteins are coupled to a multitude of effectors into distinct subcellular compartments because of their multidomain architecture. These novel cAMP sensors are not only at the crossroads of different physiological processes but also may represent attractive therapeutic targets for the treatment of several cardiovascular disorders, including cardiac arrhythmia and heart failure.
Collapse
Affiliation(s)
- Frank Lezoualc'h
- From the Department of Cardiac and Renal Remodeling of the Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-1048, Toulouse, France (F.L., L.F., M.L., C.C.); and Université Toulouse III-Paul Sabatier, Toulouse, France (F.L., L.F., M.L., C.C.).
| | - Loubina Fazal
- From the Department of Cardiac and Renal Remodeling of the Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-1048, Toulouse, France (F.L., L.F., M.L., C.C.); and Université Toulouse III-Paul Sabatier, Toulouse, France (F.L., L.F., M.L., C.C.)
| | - Marion Laudette
- From the Department of Cardiac and Renal Remodeling of the Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-1048, Toulouse, France (F.L., L.F., M.L., C.C.); and Université Toulouse III-Paul Sabatier, Toulouse, France (F.L., L.F., M.L., C.C.)
| | - Caroline Conte
- From the Department of Cardiac and Renal Remodeling of the Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM), UMR-1048, Toulouse, France (F.L., L.F., M.L., C.C.); and Université Toulouse III-Paul Sabatier, Toulouse, France (F.L., L.F., M.L., C.C.)
| |
Collapse
|
39
|
Gardner RT, Ripplinger CM, Myles RC, Habecker BA. Molecular Mechanisms of Sympathetic Remodeling and Arrhythmias. Circ Arrhythm Electrophysiol 2016; 9:e001359. [PMID: 26810594 DOI: 10.1161/circep.115.001359] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Ryan T Gardner
- From the Department of Physiology and Pharmacology and Knight Cardiovascular Institute, Oregon Health and Science University, Portland (R.T.G., B.A.H.); Department of Pharmacology, School of Medicine, University of California, Davis (C.M.R.); and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (R.C.M.)
| | - Crystal M Ripplinger
- From the Department of Physiology and Pharmacology and Knight Cardiovascular Institute, Oregon Health and Science University, Portland (R.T.G., B.A.H.); Department of Pharmacology, School of Medicine, University of California, Davis (C.M.R.); and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (R.C.M.)
| | - Rachel C Myles
- From the Department of Physiology and Pharmacology and Knight Cardiovascular Institute, Oregon Health and Science University, Portland (R.T.G., B.A.H.); Department of Pharmacology, School of Medicine, University of California, Davis (C.M.R.); and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (R.C.M.)
| | - Beth A Habecker
- From the Department of Physiology and Pharmacology and Knight Cardiovascular Institute, Oregon Health and Science University, Portland (R.T.G., B.A.H.); Department of Pharmacology, School of Medicine, University of California, Davis (C.M.R.); and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (R.C.M.).
| |
Collapse
|
40
|
Moghtadaei M, Polina I, Rose RA. Electrophysiological effects of natriuretic peptides in the heart are mediated by multiple receptor subtypes. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 120:37-49. [DOI: 10.1016/j.pbiomolbio.2015.12.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 11/25/2015] [Accepted: 12/02/2015] [Indexed: 12/13/2022]
|
41
|
Domínguez-Rodríguez A, Ruiz-Hurtado G, Sabourin J, Gómez AM, Alvarez JL, Benitah JP. Proarrhythmic effect of sustained EPAC activation on TRPC3/4 in rat ventricular cardiomyocytes. J Mol Cell Cardiol 2015. [PMID: 26219954 DOI: 10.1016/j.yjmcc.2015.07.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The Exchange Protein directly Activated by cAMP (EPAC) participates to the pathological signaling of cardiac hypertrophy and heart failure, in which the role of Ca(2+) entry through the Transient Receptor Potential Canonical (TRPC) channels begin to be appreciated. Here we studied whether EPAC activation could influence the activity and/or expression of TRPC channels in cardiac myocytes. In adult rat ventricular myocytes treated for 4 to 6h with the selective EPAC activator, 8-pCPT (10μM), we observed by Fluo-3 confocal fluorescence a Store-Operated Ca(2+) Entry (SOCE) like-activity, which was blunted by co-incubation with EPAC inhibitors (ESI-05 and CE3F4 at 10 μM). This SOCE-like activity, which was very small in control incubated cells, was sensitive to 30-μM SKF-96365. Molecular screening showed a specific upregulation of TRPC3 and C4 protein isoforms after 8-pCPT treatment. Moreover, sustained EPAC activation favored proarrhythmic Ca(2+) waves, which were reduced either by co-incubation with EPAC inhibitors or bath perfusion with TRPC inhibitors. Our study provides the first evidence that sustained selective EPAC activation leads to an increase in TRPC3 and C4 protein expression and induces a proarrhythmic SOCE-like activity in adult rat ventricular cardiomyocytes, which might be of importance during the development of cardiac diseases.
Collapse
Affiliation(s)
- Alejandro Domínguez-Rodríguez
- Inserm, UMR-S 1180, Châtenay-Malabry, France; Faculté de Pharmacie, Université Paris Sud, Labex LERMIT, DHU TORINO, Châtenay-Malabry, France; Institute of Biomedicine of Seville, Seville, Spain
| | - Gema Ruiz-Hurtado
- Inserm, UMR-S 1180, Châtenay-Malabry, France; Faculté de Pharmacie, Université Paris Sud, Labex LERMIT, DHU TORINO, Châtenay-Malabry, France
| | - Jessica Sabourin
- Inserm, UMR-S 1180, Châtenay-Malabry, France; Faculté de Pharmacie, Université Paris Sud, Labex LERMIT, DHU TORINO, Châtenay-Malabry, France
| | - Ana Maria Gómez
- Inserm, UMR-S 1180, Châtenay-Malabry, France; Faculté de Pharmacie, Université Paris Sud, Labex LERMIT, DHU TORINO, Châtenay-Malabry, France
| | - Julio L Alvarez
- Inserm, UMR-S 1180, Châtenay-Malabry, France; Faculté de Pharmacie, Université Paris Sud, Labex LERMIT, DHU TORINO, Châtenay-Malabry, France; Laboratorio de Electrofisiología. Instituto de Cardiología y Cirugía Cardiovascular, La Habana, Cuba
| | - Jean-Pierre Benitah
- Inserm, UMR-S 1180, Châtenay-Malabry, France; Faculté de Pharmacie, Université Paris Sud, Labex LERMIT, DHU TORINO, Châtenay-Malabry, France.
| |
Collapse
|
42
|
Lv Y, Bai S, Zhang H, Zhang H, Meng J, Li L, Xu Y. Aldosterone down-regulates the slowly activated delayed rectifier potassium current in adult guinea pig cardiomyocytes. Br J Pharmacol 2015; 172:5596-608. [PMID: 25857626 DOI: 10.1111/bph.13163] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 03/25/2015] [Accepted: 03/31/2015] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE There is emerging evidence that the mineralocorticoid hormone aldosterone is associated with arrhythmias in cardiovascular disease. However, the effect of aldosterone on the slowly activated delayed rectifier potassium current (IK s ) remains poorly understood. The present study was designed to investigate the modulation of IK s by aldosterone. EXPERIMENTAL APPROACH Adult guinea pigs were treated with aldosterone for 28 days via osmotic pumps. Standard glass microelectrode recordings and whole-cell patch-clamp techniques were used to record action potentials in papillary muscles and IK s in ventricular cardiomyocytes. KEY RESULTS The aldosterone-treated animals exhibited a prolongation of the QT interval and action potential duration with a higher incidence of early afterdepolarizations. Patch-clamp recordings showed a significant down-regulation of IK s density in the ventricular myocytes of these treated animals. These aldosterone-induced electrophysiological changes were fully prevented by a combined treatment with spironolactone, a mineralocorticoid receptor (MR) antagonist. In addition, in in vitro cultured ventricular cardiomyocytes, treatment with aldosterone (sustained exposure for 24 h) decreased the IK s density in a concentration-dependent manner. Furthermore, a significant corresponding reduction in the mRNA/protein expression of IKs channel pore and auxiliary subunits, KCNQ1 and KCNE1 was detected in ventricular tissue from the aldosterone-treated animals. CONCLUSIONS AND IMPLICATIONS Aldosterone down-regulates IK s by inhibiting the expression of KCNQ1 and KCNE1, thus delaying the ventricular repolarization. These results provide new insights into the mechanism underlying K(+) channel remodelling in heart disease and may explain the highly beneficial effects of MR antagonists in HF.
Collapse
Affiliation(s)
- Yankun Lv
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, Hebei, China.,Heart Center, Hebei General Hospital, Shijiazhuang, China
| | - Song Bai
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, Hebei, China
| | - Hua Zhang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, Hebei, China
| | - Hongxue Zhang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, Hebei, China
| | - Jing Meng
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, Hebei, China
| | - Li Li
- Heart Center, Hebei General Hospital, Shijiazhuang, China
| | - Yanfang Xu
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang, Hebei, China
| |
Collapse
|
43
|
Novel Epac fluorescent ligand reveals distinct Epac1 vs. Epac2 distribution and function in cardiomyocytes. Proc Natl Acad Sci U S A 2015; 112:3991-6. [PMID: 25829540 DOI: 10.1073/pnas.1416163112] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Exchange proteins directly activated by cAMP (Epac1 and Epac2) have been recently recognized as key players in β-adrenergic-dependent cardiac arrhythmias. Whereas Epac1 overexpression can lead to cardiac hypertrophy and Epac2 activation can be arrhythmogenic, it is unknown whether distinct subcellular distribution of Epac1 vs. Epac2 contributes to differential functional effects. Here, we characterized and used a novel fluorescent cAMP derivate Epac ligand 8-[Pharos-575]-2'-O-methyladenosine-3',5'-cyclic monophosphate (Φ-O-Me-cAMP) in mice lacking either one or both isoforms (Epac1-KO, Epac2-KO, or double knockout, DKO) to assess isoform localization and function. Fluorescence of Φ-O-Me-cAMP was enhanced by binding to Epac. Unlike several Epac-specific antibodies tested, Φ-O-Me-cAMP exhibited dramatically reduced signals in DKO myocytes. In WT, the apparent binding affinity (Kd = 10.2 ± 0.8 µM) is comparable to that of cAMP and nonfluorescent Epac-selective agonist 8-(4-chlorophenylthio)-2-O-methyladenosine-3'-,5'-cyclicmonophosphate (OMe-CPT). Φ-O-Me-cAMP readily entered intact myocytes, but did not activate PKA and its binding was competitively inhibited by OMe-CPT, confirming its Epac specificity. Φ-O-Me-cAMP is a weak partial agonist for purified Epac, but functioned as an antagonist for four Epac signaling pathways in myocytes. Epac2 and Epac1 were differentially concentrated along T tubules and around the nucleus, respectively. Epac1-KO abolished OMe-CPT-induced nuclear CaMKII activation and export of transcriptional regulator histone deacetylase 5. In conclusion, Epac1 is localized and functionally involved in nuclear signaling, whereas Epac2 is located at the T tubules and regulates arrhythmogenic sarcoplasmic reticulum Ca leak.
Collapse
|
44
|
Xiong Q, Cao Q, Zhou Q, Xie J, Shen Y, Wan R, Yu J, Yan S, Marian AJ, Hong K. Arrhythmogenic cardiomyopathy in a patient with a rare loss-of-function KCNQ1 mutation. J Am Heart Assoc 2015; 4:e001526. [PMID: 25616976 PMCID: PMC4330077 DOI: 10.1161/jaha.114.001526] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Ventricular tachycardia (VT) is a common manifestation of advanced cardiomyopathies. In a subset of patients with dilated cardiomyopathy, VT is the initial and the cardinal manifestation of the disease. The molecular genetic basis of this subset of dilated cardiomyopathy is largely unknown. METHODS AND RESULTS We identified 10 patients with dilated cardiomyopathy who presented with VT and sequenced 14 common causal genes for cardiomyopathies and arrhythmias. Functional studies included cellular patch clamp, confocal microscopy, and immunoblotting. We identified nonsynonymous variants in 4 patients, including a rare missense p.R397Q mutation in the KCNQ1 gene in a 60-year-old man who presented with incessant VT and had mild cardiac dysfunction. The p.R397Q mutation was absent in an ethnically matched control group, affected a conserved amino acid, and was predicted by multiple algorithms to be pathogenic. Co-expression of the mutant KCNQ1 with its partner unit KCNE1 was associated with reduced tail current density of slowly activating delayed rectifier K(+) current (IKs). The mutation reduced membrane localization of the protein. CONCLUSIONS Dilated cardiomyopathy with an initial presentation of VT may be a forme fruste of arrhythmogenic cardiomyopathy caused by mutations in genes encoding the ion channels. The findings implicate KCNQ1 as a possible causal gene for arrhythmogenic cardiomyopathy.
Collapse
Affiliation(s)
- Qinmei Xiong
- Cardiovascular Department, the Second Affiliated Hospital of Nanchang University, Nanchang, China (Q.X., Q.Z., Y.S., J.Y., S.Y., K.H.)
| | - Qing Cao
- The Key Laboratory of Molecular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, China (Q.C., J.X., R.W., K.H.)
| | - Qiongqiong Zhou
- Cardiovascular Department, the Second Affiliated Hospital of Nanchang University, Nanchang, China (Q.X., Q.Z., Y.S., J.Y., S.Y., K.H.)
| | - Jinyan Xie
- The Key Laboratory of Molecular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, China (Q.C., J.X., R.W., K.H.)
| | - Yang Shen
- Cardiovascular Department, the Second Affiliated Hospital of Nanchang University, Nanchang, China (Q.X., Q.Z., Y.S., J.Y., S.Y., K.H.)
| | - Rong Wan
- The Key Laboratory of Molecular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, China (Q.C., J.X., R.W., K.H.)
| | - Jianhua Yu
- Cardiovascular Department, the Second Affiliated Hospital of Nanchang University, Nanchang, China (Q.X., Q.Z., Y.S., J.Y., S.Y., K.H.)
| | - Sujuan Yan
- Cardiovascular Department, the Second Affiliated Hospital of Nanchang University, Nanchang, China (Q.X., Q.Z., Y.S., J.Y., S.Y., K.H.)
| | - Ali J Marian
- Center for Cardiovascular Genetics at The University of Texas Health Science Center-Houston and Texas Heart Institute, Houston, TX (A.J.M.)
| | - Kui Hong
- Cardiovascular Department, the Second Affiliated Hospital of Nanchang University, Nanchang, China (Q.X., Q.Z., Y.S., J.Y., S.Y., K.H.) The Key Laboratory of Molecular Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, China (Q.C., J.X., R.W., K.H.)
| |
Collapse
|
45
|
Grimes MT, Powell M, Gutierrez SM, Darby-King A, Harley CW, McLean JH. Epac activation initiates associative odor preference memories in the rat pup. ACTA ACUST UNITED AC 2015; 22:74-82. [PMID: 25593293 PMCID: PMC4341366 DOI: 10.1101/lm.037101.114] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Here we examine the role of the exchange protein directly activated by cAMP (Epac) in β-adrenergic-dependent associative odor preference learning in rat pups. Bulbar Epac agonist (8-pCPT-2-O-Me-cAMP, or 8-pCPT) infusions, paired with odor, initiated preference learning, which was selective for the paired odor. Interestingly, pairing odor with Epac activation produced both short-term (STM) and long-term (LTM) odor preference memories. Training using β-adrenergic-activation paired with odor recruited rapid and transient ERK phosphorylation consistent with a role for Epac activation in normal learning. An ERK antagonist prevented intermediate-term memory (ITM) and LTM, but not STM. Epac agonist infusions induced ERK phosphorylation in the mitral cell layer, in the inner half of the dendritic external plexiform layer, in the glomeruli and, patchily, among granule cells. Increased CREB phosphorylation in the mitral and granule cell layers was also seen. Simultaneous blockade of both ERK and CREB pathways prevented any long-term β-adrenergic activated odor preference memory, while LTM deficits associated with blocking only one pathway were prevented by stronger β-adrenergic activation. These results suggest that Epac and PKA play parallel and independent, as well as likely synergistic, roles in creating cAMP-dependent associative memory in rat pups. They further implicate a novel ERK-independent pathway in the mediation of STM by Epac.
Collapse
Affiliation(s)
- Matthew T Grimes
- Division of BioMedical Sciences, Memorial University of Newfoundland, St. John's, Newfoundland, A1B 3V6 Canada
| | - Maria Powell
- Division of BioMedical Sciences, Memorial University of Newfoundland, St. John's, Newfoundland, A1B 3V6 Canada
| | - Sandra Mohammed Gutierrez
- Division of BioMedical Sciences, Memorial University of Newfoundland, St. John's, Newfoundland, A1B 3V6 Canada
| | - Andrea Darby-King
- Division of BioMedical Sciences, Memorial University of Newfoundland, St. John's, Newfoundland, A1B 3V6 Canada
| | - Carolyn W Harley
- Department of Psychology, Memorial University of Newfoundland, St. John's, Newfoundland, A1B 3V6 Canada
| | - John H McLean
- Division of BioMedical Sciences, Memorial University of Newfoundland, St. John's, Newfoundland, A1B 3V6 Canada
| |
Collapse
|