Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
Nat Commun ; 15(1): 526, 2024 Jan 16.
Article En | MEDLINE | ID: mdl-38228630

The inner ear is the hub where hair cells (HCs) transduce sound, gravity, and head acceleration stimuli to the brain. Hearing and balance rely on mechanosensation, the fastest sensory signals transmitted to the brain. The mechanoelectrical transducer (MET) channel is the entryway for the sound-balance-brain interface, but the channel-complex composition is not entirely known. Here, we report that the mouse utilizes Piezo1 (Pz1) and Piezo2 (Pz2) isoforms as MET-complex components. The Pz channels, expressed in HC stereocilia, and cell lines are co-localized and co-assembled with MET complex partners. Mice expressing non-functional Pz1 and Pz2 at the ROSA26 locus have impaired auditory and vestibular traits that can only be explained if the Pzs are integral to the MET complex. We suggest that Pz subunits constitute part of the MET complex and that interactions with other MET complex components yield functional MET units to generate HC MET currents.


Ear, Inner , Hair Cells, Auditory, Inner , Animals , Mice , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory/metabolism , Stereocilia/metabolism , Ear, Inner/metabolism , Hearing , Mechanotransduction, Cellular , Mammals/metabolism , Ion Channels/genetics , Ion Channels/metabolism
2.
JCI Insight ; 7(22)2022 11 22.
Article En | MEDLINE | ID: mdl-36509290

Sinoatrial node (SAN) cells are the heart's primary pacemaker. Their activity is tightly regulated by ß-adrenergic receptor (ß-AR) signaling. Adenylyl cyclase (AC) is a key enzyme in the ß-AR pathway that catalyzes the production of cAMP. There are current gaps in our knowledge regarding the dominant AC isoforms and the specific roles of Ca2+-activated ACs in the SAN. The current study tests the hypothesis that distinct AC isoforms are preferentially expressed in the SAN and compartmentalize within microdomains to orchestrate heart rate regulation during ß-AR signaling. In contrast to atrial and ventricular myocytes, SAN cells express a diverse repertoire of ACs, with ACI as the predominant Ca2+-activated isoform. Although ACI-KO (ACI-/-) mice exhibit normal cardiac systolic or diastolic function, they experience SAN dysfunction. Similarly, SAN-specific CRISPR/Cas9-mediated gene silencing of ACI results in sinus node dysfunction. Mechanistically, hyperpolarization-activated cyclic nucleotide-gated 4 (HCN4) channels form functional microdomains almost exclusively with ACI, while ryanodine receptor and L-type Ca2+ channels likely compartmentalize with ACI and other AC isoforms. In contrast, there were no significant differences in T-type Ca2+ and Na+ currents at baseline or after ß-AR stimulation between WT and ACI-/- SAN cells. Due to its central characteristic feature as a Ca2+-activated isoform, ACI plays a unique role in sustaining the rise of local cAMP and heart rates during ß-AR stimulation. The findings provide insights into the critical roles of the Ca2+-activated isoform of AC in sustaining SAN automaticity that is distinct from contractile cardiomyocytes.


Adenylyl Cyclases , Sinoatrial Node , Animals , Mice , Sinoatrial Node/metabolism , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Calcium/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Protein Isoforms/metabolism
3.
J Gen Physiol ; 152(12)2020 12 07.
Article En | MEDLINE | ID: mdl-33211795

Calmodulin (CaM) plays a critical role in intracellular signaling and regulation of Ca2+-dependent proteins and ion channels. Mutations in CaM cause life-threatening cardiac arrhythmias. Among the known CaM targets, small-conductance Ca2+-activated K+ (SK) channels are unique, since they are gated solely by beat-to-beat changes in intracellular Ca2+. However, the molecular mechanisms of how CaM mutations may affect the function of SK channels remain incompletely understood. To address the structural and functional effects of these mutations, we introduced prototypical human CaM mutations in human induced pluripotent stem cell-derived cardiomyocyte-like cells (hiPSC-CMs). Using structural modeling and molecular dynamics simulation, we demonstrate that human calmodulinopathy-associated CaM mutations disrupt cardiac SK channel function via distinct mechanisms. CaMD96V and CaMD130G mutants reduce SK currents through a dominant-negative fashion. By contrast, specific mutations replacing phenylalanine with leucine result in conformational changes that affect helix packing in the C-lobe, which disengage the interactions between apo-CaM and the CaM-binding domain of SK channels. Distinct mutant CaMs may result in a significant reduction in the activation of the SK channels, leading to a decrease in the key Ca2+-dependent repolarization currents these channels mediate. The findings in this study may be generalizable to other interactions of mutant CaMs with Ca2+-dependent proteins within cardiac myocytes.


Calmodulin , Induced Pluripotent Stem Cells , Small-Conductance Calcium-Activated Potassium Channels/physiology , Arrhythmias, Cardiac , Calcium/metabolism , Calmodulin/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Mutation
4.
Front Pharmacol ; 11: 1182, 2020.
Article En | MEDLINE | ID: mdl-32848785

BACKGROUND: Long QT syndrome 3 (LQT3) is caused by SCN5A mutations. Late sodium current (late I Na) inhibitors are current-specific to treat patients with LQT3, but the mechanisms underlying mexiletine (MEX) -sensitive (N1325S and R1623Q) and -insensitive (M1652R) mutations remains to be elucidated. METHODS: LQT3 patients with causative mutations were treated with oral MEX following i.v. lidocaine. Whole-cell patch-clamp techniques and molecular remodeling were used to determine the mechanisms underlying the sensitivity to MEX. RESULTS: Intravenous administration of lidocaine followed by MEX orally in LQT patients with N1325S and R1623Q sodium channel mutation shortened QTc interval, abolished arrhythmias, and completely normalized the ECG. In HEK293 cells, the steady-state inactivation curves of the M1652R channels were rightward shifted by 5.6 mV relative to the WT channel. In contrast, the R1623Q mutation caused a leftward shift of the steady-state inactivation curve by 15.2 mV compared with WT channel, and N1325S mutation did not affect steady-state inactivation (n = 5-13, P < 0.05). The extent of the window current was expanded in all three mutant channels compared with WT. All three mutations increased late I Na with the greatest amplitude in the M1652R channel (n = 9-15, P < 0.05). MEX caused a hyperpolarizing shift of the steady-state inactivation and delayed the recovery of all three mutant channels. Furthermore, it suppressed late I Na in N1325S and R1623Q to a greater extent compared to that of M1652R mutant channel. Mutations altered the sensitivity of Nav1.5 to MEX through allosteric mechanisms by changing the conformation of Nav1.5 to become more or less favorable for MEX binding. Late I Na inhibitors suppressed late I Na in N1325S and R1623Q to a greater extent than that in the M1652R mutation (n = 4-7, P < 0.05). CONCLUSION: The N1325S, R1623Q, and M1652R mutations are associated with a variable augmentation of late I Na, which was reversed by MEX. M1652R mutation changes the conformation of Nav1.5 that disrupt the inactivation of channel affecting MEX binding, corresponding to the poor response to MEX. The lidocaine test, molecular modeling, and drugs screening in cells expressing mutant channels are useful for predicting the effectiveness of late I Na inhibitors.

5.
Nucleic Acids Res ; 48(1): 304-315, 2020 01 10.
Article En | MEDLINE | ID: mdl-31754723

Influenza A viruses cause widespread human respiratory disease. The viral multifunctional NS1 protein inhibits host antiviral responses. This inhibition results from the binding of specific cellular antiviral proteins at various positions on the NS1 protein. Remarkably, binding of several proteins also requires the two amino-acid residues in the NS1 N-terminal RNA-binding domain (RBD) that are required for binding double-stranded RNA (dsRNA). Here we focus on the host restriction factor DHX30 helicase that is countered by the NS1 protein, and establish why the dsRNA-binding activity of NS1 is required for its binding to DHX30. We show that the N-terminal 152 amino-acid residue segment of DHX30, denoted DHX30N, possesses all the antiviral activity of DHX30 and contains a dsRNA-binding domain, and that the NS1-DHX30 interaction in vivo requires the dsRNA-binding activity of both DHX30N and the NS1 RBD. We demonstrate why this is the case using bacteria-expressed proteins: the DHX30N-NS1 RBD interaction in vitro requires the presence of a dsRNA platform that binds both NS1 RBD and DHX30N. We propose that a similar dsRNA platform functions in interactions of the NS1 protein with other proteins that requires these same two amino-acid residues required for NS1 RBD dsRNA-binding activity.


Host-Pathogen Interactions/genetics , RNA Helicases/genetics , RNA, Double-Stranded/genetics , Viral Nonstructural Proteins/genetics , Animals , Binding Sites , Cloning, Molecular , Dogs , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , HeLa Cells , Humans , Madin Darby Canine Kidney Cells , Models, Molecular , Nucleic Acid Conformation , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , RNA Helicases/chemistry , RNA Helicases/metabolism , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism
6.
J Biol Chem ; 291(5): 2499-509, 2016 Jan 29.
Article En | MEDLINE | ID: mdl-26515070

Calmodulin (CaM), a Ca(2+)-sensing protein, is constitutively bound to IQ domains of the C termini of human Kv7 (hKv7, KCNQ) channels to mediate Ca(2+)-dependent reduction of Kv7 currents. However, the mechanism remains unclear. We report that CaM binds to two isoforms of the hKv7.4 channel in a Ca(2+)-independent manner but that only the long isoform (hKv7.4a) is regulated by Ca(2+)/CaM. Ca(2+)/CaM mediate reduction of the hKv7.4a channel by decreasing the channel open probability and altering activation kinetics. We took advantage of a known missense mutation (G321S) that has been linked to progressive hearing loss to further examine the inhibitory effects of Ca(2+)/CaM on the Kv7.4 channel. Using multidisciplinary techniques, we demonstrate that the G321S mutation may destabilize CaM binding, leading to a decrease in the inhibitory effects of Ca(2+) on the channels. Our study utilizes an expression system to dissect the biophysical properties of the WT and mutant Kv7.4 channels. This report provides mechanistic insights into the critical roles of Ca(2+)/CaM regulation of the Kv7.4 channel under physiological and pathological conditions.


Calmodulin/chemistry , Gene Expression Regulation , KCNQ Potassium Channels/chemistry , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , Calcium/chemistry , Cricetinae , Cricetulus , Electrophysiology , Epitopes/chemistry , Humans , Immunoprecipitation , Ions , Kinetics , Models, Molecular , Molecular Sequence Data , Mutation , Mutation, Missense , Patch-Clamp Techniques , Protein Binding , Protein Isoforms/chemistry , Sequence Homology, Amino Acid
7.
J Biol Chem ; 290(8): 4663-4676, 2015 Feb 20.
Article En | MEDLINE | ID: mdl-25538241

Cav1.3 L-type Ca(2+) channel is known to be highly expressed in neurons and neuroendocrine cells. However, we have previously demonstrated that the Cav1.3 channel is also expressed in atria and pacemaking cells in the heart. The significance of the tissue-specific expression of the channel is underpinned by our previous demonstration of atrial fibrillation in a Cav1.3 null mutant mouse model. Indeed, a recent study has confirmed the critical roles of Cav1.3 in the human heart (Baig, S. M., Koschak, A., Lieb, A., Gebhart, M., Dafinger, C., Nürnberg, G., Ali, A., Ahmad, I., Sinnegger-Brauns, M. J., Brandt, N., Engel, J., Mangoni, M. E., Farooq, M., Khan, H. U., Nürnberg, P., Striessnig, J., and Bolz, H. J. (2011) Nat. Neurosci. 14, 77-84). These studies suggest that detailed knowledge of Cav1.3 may have broad therapeutic ramifications in the treatment of cardiac arrhythmias. Here, we tested the hypothesis that there is a functional cross-talk between the Cav1.3 channel and a small conductance Ca(2+)-activated K(+) channel (SK2), which we have documented to be highly expressed in human and mouse atrial myocytes. Specifically, we tested the hypothesis that the C terminus of Cav1.3 may translocate to the nucleus where it functions as a transcriptional factor. Here, we reported for the first time that the C terminus of Cav1.3 translocates to the nucleus where it functions as a transcriptional regulator to modulate the function of Ca(2+)-activated K(+) channels in atrial myocytes. Nuclear translocation of the C-terminal domain of Cav1.3 is directly regulated by intracellular Ca(2+). Utilizing a Cav1.3 null mutant mouse model, we demonstrate that ablation of Cav1.3 results in a decrease in the protein expression of myosin light chain 2, which interacts and increases the membrane localization of SK2 channels.


Calcium Channels, L-Type/metabolism , Cell Nucleus/metabolism , Gene Expression Regulation/physiology , Myocytes, Cardiac/metabolism , Transcription, Genetic/physiology , Active Transport, Cell Nucleus/physiology , Animals , Calcium Channels, L-Type/genetics , Cardiac Myosins/biosynthesis , Cardiac Myosins/genetics , Cell Nucleus/genetics , Heart Atria/cytology , Heart Atria/metabolism , Humans , Mice , Mice, Knockout , Myocytes, Cardiac/cytology , Myosin Light Chains/biosynthesis , Myosin Light Chains/genetics , Protein Structure, Tertiary
8.
Proc Natl Acad Sci U S A ; 111(27): 9989-94, 2014 Jul 08.
Article En | MEDLINE | ID: mdl-24951510

For an excitable cell to function properly, a precise number of ion channel proteins need to be trafficked to distinct locations on the cell surface membrane, through a network and anchoring activity of cytoskeletal proteins. Not surprisingly, mutations in anchoring proteins have profound effects on membrane excitability. Ca(2+)-activated K(+) channels (KCa2 or SK) have been shown to play critical roles in shaping the cardiac atrial action potential profile. Here, we demonstrate that filamin A, a cytoskeletal protein, augments the trafficking of SK2 channels in cardiac myocytes. The trafficking of SK2 channel is Ca(2+)-dependent. Further, the Ca(2+) dependence relies on another channel-interacting protein, α-actinin2, revealing a tight, yet intriguing, assembly of cytoskeletal proteins that orchestrate membrane expression of SK2 channels in cardiac myocytes. We assert that changes in SK channel trafficking would significantly alter atrial action potential and consequently atrial excitability. Identification of therapeutic targets to manipulate the subcellular localization of SK channels is likely to be clinically efficacious. The findings here may transcend the area of SK2 channel studies and may have implications not only in cardiac myocytes but in other types of excitable cells.


Calcium/metabolism , Filamins/metabolism , Membrane Proteins/metabolism , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Action Potentials , Animals , Animals, Newborn , Filamins/genetics , HEK293 Cells , Heart Atria/metabolism , Humans , Mice , Myocytes, Cardiac/metabolism , Protein Binding , RNA, Small Interfering/genetics
9.
Circ Res ; 112(12): 1567-76, 2013 Jun 07.
Article En | MEDLINE | ID: mdl-23609114

RATIONALE: Adenylyl cyclase (AC) represents one of the principal molecules in the ß-adrenergic receptor signaling pathway, responsible for the conversion of ATP to the second messenger, cAMP. AC types 5 (ACV) and 6 (ACVI) are the 2 main isoforms in the heart. Although highly homologous in sequence, these 2 proteins play different roles during the development of heart failure. Caveolin-3 is a scaffolding protein, integrating many intracellular signaling molecules in specialized areas called caveolae. In cardiomyocytes, caveolin is located predominantly along invaginations of the cell membrane known as t-tubules. OBJECTIVE: We take advantage of ACV and ACVI knockout mouse models to test the hypothesis that there is distinct compartmentalization of these isoforms in ventricular myocytes. METHODS AND RESULTS: We demonstrate that ACV and ACVI isoforms exhibit distinct subcellular localization. The ACVI isoform is localized in the plasma membrane outside the t-tubular region and is responsible for ß1-adrenergic receptor signaling-mediated enhancement of the L-type Ca(2+) current (ICa,L) in ventricular myocytes. In contrast, the ACV isoform is localized mainly in the t-tubular region where its influence on ICa,L is restricted by phosphodiesterase. We further demonstrate that the interaction between caveolin-3 with ACV and phosphodiesterase is responsible for the compartmentalization of ACV signaling. CONCLUSIONS: Our results provide new insights into the compartmentalization of the 2 AC isoforms in the regulation of ICa,L in ventricular myocytes. Because caveolae are found in most mammalian cells, the mechanism of ß- adrenergic receptor and AC compartmentalization may also be important for ß-adrenergic receptor signaling in other cell types.


Adenylyl Cyclases/metabolism , Calcium Channels, L-Type/metabolism , Heart Ventricles/enzymology , Myocytes, Cardiac/enzymology , Adenylyl Cyclases/deficiency , Adenylyl Cyclases/genetics , Adrenergic beta-Agonists/pharmacology , Amino Acid Sequence , Animals , Calcium Channels, L-Type/drug effects , Caveolin 3/metabolism , Cell Membrane/enzymology , Computer Simulation , Fluorescent Antibody Technique , Heart Ventricles/drug effects , Isoenzymes , Isoproterenol/pharmacology , Membrane Potentials , Mice , Mice, Knockout , Microscopy, Confocal , Molecular Sequence Data , Myocytes, Cardiac/drug effects , Phosphoric Diester Hydrolases/metabolism , Receptors, Adrenergic, beta-1/metabolism , Signal Transduction
...