28 research outputs found

    Kir2.1 Interactome Mapping Uncovers PKP4 as a Modulator of the Kir2.1-Regulated Inward Rectifier Potassium Currents

    Get PDF
    Kir2.1, a strong inward rectifier potassium channel encoded by the KCNJ2 gene, is a key regulator of the resting membrane potential of the cardiomyocyte and plays an important role in controlling ventricular excitation and action potential duration in the human heart. Mutations in KCNJ2 result in inheritable cardiac diseases in humans, e.g. the type-1 Andersen-Tawil syndrome (ATS1). Understanding the molecular mechanisms that govern the regulation of inward rectifier potassium currents by Kir2.1 in both normal and disease contexts should help uncover novel targets for therapeutic intervention in ATS1 and other Kir2.1-associated channelopathies. The information available to date on protein-protein interactions involving Kir2.1 channels remains limited. Additional efforts are necessary to provide a comprehensive map of the Kir2.1 interactome. Here we describe the generation of a comprehensive map of the Kir2.1 interactome using the proximity-labeling approach BioID. Most of the 218 high-confidence Kir2.1 channel interactions we identified are novel and encompass various molecular mechanisms of Kir2.1 function, ranging from intracellular trafficking to cross-talk with the insulin-like growth factor receptor signaling pathway, as well as lysosomal degradation. Our map also explores the variations in the interactome profiles of Kir2.1WT versus Kir2.1Δ314-315, a trafficking deficient ATS1 mutant, thus uncovering molecular mechanisms whose malfunctions may underlie ATS1 disease. Finally, using patch-clamp analysis, we validate the functional relevance of PKP4, one of our top BioID interactors, to the modulation of Kir2.1-controlled inward rectifier potassium currents. Our results validate the power of our BioID approach in identifying functionally relevant Kir2.1 interactors and underline the value of our Kir2.1 interactome as a repository for numerous novel biological hypotheses on Kir2.1 and Kir2.1-associated diseases.This work was supported by the National Institutes of Health (NIH) through the National Heart, Lung, and Blood Institute (NHLBI) grant R01HL122352 awarded to J.J., as well as the National Institute of General Medical Sciences (NIGMS) grant R01GM094231 and the National Cancer Institute (NCI) grant U24CA210967 awarded to A.I.N. R.K. is supported by the NCI support grant P30CA046592 awarded to the University of Michigan Rogel Cancer Center. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.S

    Paclitaxel mitigates structural alterations and cardiac conduction system defects in a mouse model of Hutchinson-Gilford progeria syndrome.

    Get PDF
    Hutchinson-Gilford progeria syndrome (HGPS) is an ultrarare laminopathy caused by expression of progerin, a lamin A variant, also present at low levels in non-HGPS individuals. HGPS patients age and die prematurely, predominantly from cardiovascular complications. Progerin-induced cardiac repolarization defects have been described previously, although the underlying mechanisms are unknown. We conducted studies in heart tissue from progerin-expressing LmnaG609G/G609G (G609G) mice, including microscopy, intracellular calcium dynamics, patch-clamping, in vivo magnetic resonance imaging, and electrocardiography. G609G mouse cardiomyocytes showed tubulin-cytoskeleton disorganization, t-tubular system disruption, sarcomere shortening, altered excitation-contraction coupling, and reductions in ventricular thickening and cardiac index. G609G mice exhibited severe bradycardia, and significant alterations of atrio-ventricular conduction and repolarization. Most importantly, 50% of G609G mice had altered heart rate variability, and sinoatrial block, both significant signs of premature cardiac aging. G609G cardiomyocytes had electrophysiological alterations, which resulted in an elevated action potential plateau and early afterdepolarization bursting, reflecting slower sodium current inactivation and long Ca+2 transient duration, which may also help explain the mild QT prolongation in some HGPS patients. Chronic treatment with low-dose paclitaxel ameliorated structural and functional alterations in G609G hearts. Our results demonstrate that tubulin-cytoskeleton disorganization in progerin-expressing cardiomyocytes causes structural, cardiac conduction, and excitation-contraction coupling defects, all of which can be partially corrected by chronic treatment with low dose paclitaxel.Work in V.A.’s laboratory is supported by grants from the Spanish Ministerio de Ciencia e Innovacio´n (MCIN) (SAF2016-79490-R, PID2019-108489RBI00) and the Instituto de Salud Carlos III (ISCIII) (AC17/00067) with cofunding from the European Regional Development Fund/Fondo Europeo de Desarrollo Regional (ERDF/FEDER, ‘Una manera de hacer Europa’), and the Progeria Research Foundation (Award PRF 2019–77). Work in J.J.’s laboratory is supported by the National Heart, Lung, and Blood Institute (R01 Grant HL122352), a CNIC ‘Severo Ochoa’ intramural competitive grant, and Fondos FEDER, Madrid, Spain. Work in D.F.-R.’s laboratory is supported by the Spanish MCIN (SAF2016-80324-R) and the ISCIII (AC17/00053). The CNIC is supported by the MCIN, the ISCIII, and the Pro CNIC Foundation.S

    Human influenza A virus causes myocardial and cardiac-specific conduction system infections associated with early inflammation and premature death.

    Get PDF
    Human influenza A virus (hIAV) infection is associated with important cardiovascular complications, although cardiac infection pathophysiology is poorly understood. We aimed to study the ability of hIAV of different pathogenicity to infect the mouse heart, and establish the relationship between the infective capacity and the associated in vivo, cellular and molecular alterations. We evaluated lung and heart viral titres in mice infected with either one of several hIAV strains inoculated intranasally. 3D reconstructions of infected cardiac tissue were used to identify viral proteins inside mouse cardiomyocytes, Purkinje cells, and cardiac vessels. Viral replication was measured in mouse cultured cardiomyocytes. Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) were used to confirm infection and study underlying molecular alterations associated with the in vivo electrophysiological phenotype. Pathogenic and attenuated hIAV strains infected and replicated in cardiomyocytes, Purkinje cells, and hiPSC-CMs. The infection was also present in cardiac endothelial cells. Remarkably, lung viral titres did not statistically correlate with viral titres in the mouse heart. The highly pathogenic human recombinant virus PAmut showed faster replication, higher level of inflammatory cytokines in cardiac tissue and higher viral titres in cardiac HL-1 mouse cells and hiPSC-CMs compared with PB2mut-attenuated virus. Correspondingly, cardiac conduction alterations were especially pronounced in PAmut-infected mice, associated with high mortality rates, compared with PB2mut-infected animals. Consistently, connexin43 and NaV1.5 expression decreased acutely in hiPSC-CMs infected with PAmut virus. YEM1L protease also decreased more rapidly and to lower levels in PAmut-infected hiPSC-CMs compared with PB2mut-infected cells, consistent with mitochondrial dysfunction. Human IAV infection did not increase myocardial fibrosis at 4-day post-infection, although PAmut-infected mice showed an early increase in mRNAs expression of lysyl oxidase. Human IAV can infect the heart and cardiac-specific conduction system, which may contribute to cardiac complications and premature death.JV is a PhD fellow of the La Caixa Foundation International Fellowship Programme (La Caixa/CNB). This work was supported by the European Molecular Biology Organizat ion (STF-7649 to AF), the Spanish Ministry of Science, Innovation and Universities (MCIU), (BFU2011-26175 and BFU2014-57797-R to AN), and the network Ciber de Enfermedades Respiratorias (CIBERES) including the Improvement and Mobilit y Programme. The CNIC is a Severo Ochoa Center of Excellence (SEV-2015-0505). CNIC is supported by MCIU and the Pro CNIC Foundation. This study was supported by grants from Fondo Europeo de Desarrollo Regional (CB16/11/00458), grants SAF2015-65607-R and SAF2016-80324-R from MCIU (A.H. and D.F-R.) and fellowship SVP-2014-068595 to J.A.N-A. This study was supported by Frankel Cardiovascular Centre, Michigan Medicine (Grant 332475). JJ is supported in part by the National Heart, Lung, and Blood Institute (R01 Grant HL122352). S.F.N is supported in part by the National Heart, Lung, and Blood Institute grants R21HL138064 and R01HL129136.S

    p38γ/δ activation alters cardiac electrical activity and predisposes to ventricular arrhythmia

    Get PDF
    We gratefully acknowledge L. Sen-Martín, J. Alegre-Cebollada (CNIC, Madrid) and L. Carrier (University Medical Center HamburgEppendorf and DZHK, Hamburg) for the cMyBP3-C KO cardiac tissue; D. Roiz-Valle and C. López-Otín (IUOPA; Universidad de Oviedo, Oviedo) for the LmnaG609G/G609G cardiac tissue; and R. J. Davis for the MKK6 KO mice. We thank G. Giovinazzo and the CNIC Pluripotent Cell Technology Unit (CNIC, Madrid) for the hiPSCs. We thank S. Bartlett and F. Chanut (CNIC, Madrid) for English editing, and R. R. Mondragon (University of Michigan, Ann Arbor) for technical support. We are grateful to R. J. Davis (University of Massachusetts Chan Medical School, Worcester), A. Padmanabhan (University of California, San Francisco) and M. Costa and C. López-Otín (IUOPA; Universidad de Oviedo, Oviedo) for critical reading of the manuscript. We thank the staf at the CNIC Genomics and Bioinformatics Units for technical support and help with data analysis and A. C. Silva for help with figure editing and design. This work was funded by a CNIC Intramural Project Severo Ochoa (Expediente 12- 2016 IGP) to G.S. and J.J. G.S. is supported by the following projects: PMP21/00057 IMPACT-2021, funded by the Instituto de Salud Carlos III (ISCIII), and PDC2021-121147-I00 and PID2019-104399RB-I00, funded by MCIN/AEI/10.13039/501100011033—all funded by the European Union (FEDER/FSE); ‘Una manera de hacer Europa’/‘El FSE invierte en tu futuro’/Next Generation EU and co-funded by the European Union/Plan de Recuperación, Transformación y Resiliencia (PRTR). R.R.B. is a fellow of the FPU Program (FPU17/03847). B.G.T. was a fellow of the FPI Severo Ochoa CNIC Program (SVP‐2013‐067639) and an American Heart Association Postdoctoral Fellow (18POST34080175). The following grants provided additional funding: Instituto de Salud Carlos III, PDC2021-121147-I00 Convocatoria: Proyectos Prueba de Concepto 2021 Ministerio de Ciencia e Innovación and PID2022-138525OB-I00 Ministerio de Ciencia e Innovación; US National Heart, Lung, and Blood Institute (R01 grant HL122352); Fondos FEDER, Madrid, Spain, and Fundación Bancaria ‘La Caixa (project HR19/52160013) to J.J.; American Heart Association Postdoctoral Fellowship 14POST17820005 to D.P.B.; and MICINN PGC2018-097019-B-I00, ISCIII-SGEFI/ERDF (PRB3-IPT17/0019, ProteoRed), the Fundació Marató TV3 (grant 122/C/2015) and ‘la Caixa’ Banking Foundation (project code HR17- 00247) to J.V. The CNIC is supported by the Instituto de Salud Carlos III (ISCIII), the Ministerio de Ciencia e Innovación (MCIN) and the Pro CNIC Foundation and is a Severo Ochoa Center of Excellence (grant CEX2020-001041-S, funded by MICIN/AEI/10.13039/501100011033).S

    TGF-β1, released by myofibroblasts, differentially regulates transcription and function of sodium and potassium channels in adult rat ventricular myocytes.

    Get PDF
    Cardiac injury promotes fibroblasts activation and differentiation into myofibroblasts, which are hypersecretory of multiple cytokines. It is unknown whether any of such cytokines are involved in the electrophysiological remodeling of adult cardiomyocytes. We cultured adult cardiomyocytes for 3 days in cardiac fibroblast conditioned medium (FCM) from adult rats. In whole-cell voltage-clamp experiments, FCM-treated myocytes had 41% more peak inward sodium current (I(Na)) density at -40 mV than myocytes in control medium (p<0.01). In contrast, peak transient outward current (I(to)) was decreased by ∼55% at 60 mV (p<0.001). Protein analysis of FCM demonstrated that the concentration of TGF-β1 was >3 fold greater in FCM than control, which suggested that FCM effects could be mediated by TGF-β1. This was confirmed by pre-treatment with TGF-β1 neutralizing antibody, which abolished the FCM-induced changes in both I(Na) and I(to). In current-clamp experiments TGF-β1 (10 ng/ml) prolonged the action potential duration at 30, 50, and 90 repolarization (p<0.05); at 50 ng/ml it gave rise to early afterdepolarizations. In voltage-clamp experiments, TGF-β1 increased I(Na) density in a dose-dependent manner without affecting voltage dependence of activation or inactivation. I(Na) density was -36.25±2.8 pA/pF in control, -59.17±6.2 pA/pF at 0.1 ng/ml (p<0.01), and -58.22±6.6 pA/pF at 1 ng/ml (p<0.01). In sharp contrast, I(to) density decreased from 22.2±1.2 pA/pF to 12.7±0.98 pA/pF (p<0.001) at 10 ng/ml. At 1 ng/ml TGF-β1 significantly increased SCN5A (Na(V)1.5) (+73%; p<0.01), while reducing KCNIP2 (Kchip2; -77%; p<0.01) and KCND2 (K(V)4.2; -50% p<0.05) mRNA levels. Further, the TGF-β1-induced increase in I(Na) was mediated through activation of the PI3K-AKT pathway via phosphorylation of FOXO1 (a negative regulator of SCN5A). TGF-β1 released by myofibroblasts differentially regulates transcription and function of the main cardiac sodium channel and of the channel responsible for the transient outward current. The results provide new mechanistic insight into the electrical remodeling associated with myocardial injury

    Constitutive Intracellular Na+ Excess in Purkinje Cells Promotes Arrhythmogenesis at Lower Levels of Stress Than Ventricular Myocytes From Mice With Catecholaminergic Polymorphic Ventricular Tachycardia

    Get PDF
    Background-In catecholaminergic polymorphic ventricular tachycardia (CPVT), cardiac Purkinje cells (PCs) appear more susceptible to Ca2+ dysfunction than ventricular myocytes (VMs). The underlying mechanisms remain unknown. Using a CPVT mouse (Ry(R2R4496C+/Cx40eGFP)), we tested whether PC intracellular Ca2+ ([Ca2+](i)) dysregulation results from a constitutive [Na+](i) surplus relative to VMs. Methods and Results-Simultaneous optical mapping of voltage and [Ca2+](i) in CPVT hearts showed that spontaneous Ca2+ release preceded pacing-induced triggered activity at subendocardial PCs. On simultaneous current-clamp and Ca2+ imaging, early and delayed afterdepolarizations trailed spontaneous Ca2+ release and were more frequent in CPVT PCs than CPVT VMs. As a result of increased activity of mutant ryanodine receptor type 2 channels, sarcoplasmic reticulum Ca2+ load, measured by caffeine-induced Ca2+ transients, was lower in CPVT VMs and PCs than respective controls, and sarcoplasmic reticulum fractional release was greater in both CPVT PCs and VMs than respective controls. [Na2+](i) was higher in both control and CPVT PCs than VMs, whereas the density of the Na+/Ca2+ exchanger current was not different between PCs and VMs. Computer simulations using a PC model predicted that the elevated [Na2+](i) of PCs promoted delayed afterdepolarizations, which were always preceded by spontaneous Ca2+ release events from hyperactive ryanodine receptor type 2 channels. Increasing [Na2+](i) monotonically increased delayed afterdepolarization frequency. Confocal imaging experiments showed that postpacing Ca2+ spark frequency was highest in intact CPVT PCs, but such differences were reversed on saponin-induced membrane permeabilization, indicating that differences in [Na2+](i) played a central role. Conclusions-In CPVT mice, the constitutive [Na2+](i) excess of PCs promotes triggered activity and arrhythmogenesis at lower levels of stress than VMs.This work was supported by National Heart, Lung, and Blood Institute grants P01-HL039707, P01-HL087226, and R01-HL122352; the Leducq Foundation: Transatlantic Network of Excellence Program on ``Structural Alterations in the Myocardium and the Substrate for Cardiac Fibrillation´´ (Dr Jalife); grants HL055438 and HL120108 (to Dr Valdivia); and American Heart Association grant 12SDG11480010 (Dr Deo).S

    Effects of FOXO1 overexpression on peak inward sodium current (I<sub>Na</sub>) after 72 hr of treatment.

    No full text
    <p>A) Current-voltage relationships for control (<u>black</u>) and FOXO1-CA (<u>blue</u>) expressing cells. B) Time dependent recovery of the channel. Shown in panel C, Voltage dependence of activation (m∞ curve) and inactivation (h∞ curve). Values are mean ± SE. N = 9−23 cells from 4 different isolations *indicates p<0.05, **indicates p<0.01, significant difference between control and treated cells.</p
    corecore