Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 115(47): 12051-12056, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30397133

ABSTRACT

Genetically encoded inhibitors for voltage-dependent Ca2+ (CaV) channels (GECCIs) are useful research tools and potential therapeutics. Rad/Rem/Rem2/Gem (RGK) proteins are Ras-like G proteins that potently inhibit high voltage-activated (HVA) Ca2+ (CaV1/CaV2 family) channels, but their nonselectivity limits their potential applications. We hypothesized that nonselectivity of RGK inhibition derives from their binding to auxiliary CaVß-subunits. To investigate latent CaVß-independent components of inhibition, we coexpressed each RGK individually with CaV1 (CaV1.2/CaV1.3) or CaV2 (CaV2.1/CaV2.2) channels reconstituted in HEK293 cells with either wild-type (WT) ß2a or a mutant version (ß2a,TM) that does not bind RGKs. All four RGKs strongly inhibited CaV1/CaV2 channels reconstituted with WT ß2a By contrast, when channels were reconstituted with ß2a,TM, Rem inhibited only CaV1.2, Rad selectively inhibited CaV1.2 and CaV2.2, while Gem and Rem2 were ineffective. We generated mutant RGKs (Rem[R200A/L227A] and Rad[R208A/L235A]) unable to bind WT CaVß, as confirmed by fluorescence resonance energy transfer. Rem[R200A/L227A] selectively blocked reconstituted CaV1.2 while Rad[R208A/L235A] inhibited CaV1.2/CaV2.2 but not CaV1.3/CaV2.1. Rem[R200A/L227A] and Rad[R208A/L235A] both suppressed endogenous CaV1.2 channels in ventricular cardiomyocytes and selectively blocked 25 and 62%, respectively, of HVA currents in somatosensory neurons of the dorsal root ganglion, corresponding to their distinctive selectivity for CaV1.2 and CaV1.2/CaV2.2 channels. Thus, we have exploited latent ß-binding-independent Rem and Rad inhibition of specific CaV1/CaV2 channels to develop selective GECCIs with properties unmatched by current small-molecule CaV channel blockers.


Subject(s)
Calcium Channel Blockers/metabolism , Calcium Channels/genetics , Monomeric GTP-Binding Proteins/metabolism , Biophysical Phenomena , Calcium/metabolism , Calcium Channels/metabolism , Calcium Channels, L-Type/metabolism , Calcium Channels, N-Type/metabolism , HEK293 Cells , Humans , Ion Channel Gating/physiology , Myocytes, Cardiac/metabolism , Neurons/metabolism , Protein Engineering/methods , Substrate Specificity/genetics , ras Proteins/metabolism
2.
Channels (Austin) ; 10(5): 379-394, 2016 Sep 02.
Article in English | MEDLINE | ID: mdl-27115600

ABSTRACT

Rad/Rem/Rem2/Gem (RGK) proteins are Ras-like GTPases that potently inhibit all high-voltage-gated calcium (CaV1/CaV2) channels and are, thus, well-positioned to tune diverse physiological processes. Understanding how RGK proteins inhibit CaV channels is important for perspectives on their (patho)physiological roles and could advance their development and use as genetically-encoded CaV channel blockers. We previously reported that Rem can block surface CaV1.2 channels in 2 independent ways that engage distinct components of the channel complex: (1) by binding auxiliary ß subunits (ß-binding-dependent inhibition, or BBD); and (2) by binding the pore-forming α1C subunit N-terminus (α1C-binding-dependent inhibition, or ABD). By contrast, Gem uses only the BBD mechanism to block CaV1.2. Rem molecular determinants required for BBD CaV1.2 inhibition are the distal C-terminus and the guanine nucleotide binding G-domain which interact with the plasma membrane and CaVß, respectively. However, Rem determinants for ABD CaV1.2 inhibition are unknown. Here, combining fluorescence resonance energy transfer, electrophysiology, systematic truncations, and Rem/Gem chimeras we found that the same Rem distal C-terminus and G-domain also mediate ABD CaV1.2 inhibition, but with different interaction partners. Rem distal C-terminus interacts with α1C N-terminus to anchor the G-domain which likely interacts with an as-yet-unidentified site. In contrast to some previous studies, neither the C-terminus of Rem nor Gem was sufficient to inhibit CaV1/CaV2 channels. The results reveal that similar molecular determinants on Rem are repurposed to initiate 2 independent mechanisms of CaV1.2 inhibition.


Subject(s)
Calcium Channels, L-Type/physiology , Monomeric GTP-Binding Proteins/physiology , Animals , Cells, Cultured , HEK293 Cells , Heart Ventricles/cytology , Humans , Male , Myocytes, Cardiac/physiology , Rats, Sprague-Dawley
3.
Heart Rhythm ; 13(5): 1121-1130, 2016 05.
Article in English | MEDLINE | ID: mdl-26775140

ABSTRACT

BACKGROUND: The human ether-à-go-go-related gene (hERG 1a) potassium channel is critical for cardiac repolarization. hERG 1b, another variant subunit, co-assembles with hERG 1a, modulates channel biophysical properties and plays an important role in repolarization. Mutations of hERG 1a lead to type 2 long QT syndrome (LQT2), and increased risk for fatal arrhythmias. The functional consequences of these mutations in the presence of hERG 1b are not known. OBJECTIVE: To investigate whether hERG 1a mutants exert dominant negative gating and trafficking defects when co-expressed with hERG 1b. METHODS: Electrophysiology, co-immunoprecipitation, and fluorescence resonance energy transfer (FRET) experiments in HEK293 cells and guinea pig cardiomyocytes were used to assess the mutants on gating and trafficking. Mutations of 1a-G965X and 1a-R1014X, relevant to gating and trafficking were introduced in the C-terminus region. RESULTS: The hERG 1a mutants when expressed alone did not result in decreased current amplitude. Compared to wild-type hERG 1a currents, 1a-G965X currents were significantly larger, whereas those produced by the 1a-R1014X mutant were similar in magnitude. Only when co-expressed with wild-type hERG 1a and 1b did a mutant phenotype emerge, with a marked reduction in surface expression, current amplitude, and a corresponding positive shift in the V1/2 of the activation curve. Co-immunoprecipitation and FRET assays confirmed association of mutant and wild-type subunits. CONCLUSION: Heterologously expressed hERG 1a C-terminus truncation mutants, exert a dominant negative gating and trafficking effect only when co-expressed with hERG 1b. These findings may have potentially profound implications for LQT2 therapy.


Subject(s)
Ether-A-Go-Go Potassium Channels , Long QT Syndrome , Animals , Electrophysiological Phenomena , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/metabolism , Guinea Pigs , HEK293 Cells , Humans , Long QT Syndrome/genetics , Long QT Syndrome/physiopathology , Mutation , Myocytes, Cardiac/metabolism , Protein Transport/physiology
4.
PLoS One ; 7(5): e37079, 2012.
Article in English | MEDLINE | ID: mdl-22590648

ABSTRACT

Ca(V)1/Ca(V)2 channels, comprised of pore-forming α(1) and auxiliary (ß,α(2)δ) subunits, control diverse biological responses in excitable cells. Molecules blocking Ca(V)1/Ca(V)2 channel currents (I(Ca)) profoundly regulate physiology and have many therapeutic applications. Rad/Rem/Rem2/Gem GTPases (RGKs) strongly inhibit Ca(V)1/Ca(V)2 channels. Understanding how RGKs block I(Ca) is critical for insights into their physiological function, and may provide design principles for developing novel Ca(V)1/Ca(V)2 channel inhibitors. The RGK binding sites within Ca(V)1/Ca(V)2 channel complexes responsible for I(Ca) inhibition are ambiguous, and it is unclear whether there are mechanistic differences among distinct RGKs. All RGKs bind ß subunits, but it is unknown if and how this interaction contributes to I(Ca) inhibition. We investigated the role of RGK/ß interaction in Rem inhibition of recombinant Ca(V)1.2 channels, using a mutated ß (ß(2aTM)) selectively lacking RGK binding. Rem blocked ß(2aTM)-reconstituted channels (74% inhibition) less potently than channels containing wild-type ß(2a) (96% inhibition), suggesting the prevalence of both ß-binding-dependent and independent modes of inhibition. Two mechanistic signatures of Rem inhibition of Ca(V)1.2 channels (decreased channel surface density and open probability), but not a third (reduced maximal gating charge), depended on Rem binding to ß. We identified a novel Rem binding site in Ca(V)1.2 α(1C) N-terminus that mediated ß-binding-independent inhibition. The Ca(V)2.2 α(1B) subunit lacks the Rem binding site in the N-terminus and displays a solely ß-binding-dependent form of channel inhibition. Finally, we discovered an unexpected functional dichotomy amongst distinct RGKs- while Rem and Rad use both ß-binding-dependent and independent mechanisms, Gem and Rem2 use only a ß-binding-dependent method to inhibit Ca(V)1.2 channels. The results provide new mechanistic perspectives, and reveal unexpected variations in determinants, underlying inhibition of Ca(V)1.2/Ca(V)2.2 channels by distinct RGK GTPases.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Channels, N-Type/metabolism , Ion Channel Gating/physiology , Monomeric GTP-Binding Proteins/metabolism , Binding Sites , Calcium Channels, L-Type/genetics , Calcium Channels, N-Type/genetics , HEK293 Cells , Humans , Monomeric GTP-Binding Proteins/genetics , Protein Binding
5.
Nat Commun ; 3: 706, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22426227

ABSTRACT

The cardiac voltage-gated sodium channel (Na(V)1.5) underlies impulse conduction in the heart, and its depolarization-induced inactivation is essential in control of the duration of the QT interval of the electrocardiogram. Perturbation of Na(V)1.5 inactivation by drugs or inherited mutation can underlie and trigger cardiac arrhythmias. The carboxy terminus has an important role in channel inactivation, but complete structural information on its predicted structural domain is unknown. Here we measure interactions between the functionally critical distal carboxy terminus α-helix (H6) and the proximal structured EF-hand motif using transition-metal ion fluorescence resonance energy transfer. We measure distances at three loci along H6 relative to an intrinsic tryptophan, demonstrating the proximal-distal interaction in a contiguous carboxy terminus polypeptide. Using these data together with the existing Na(V)1.5 carboxy terminus nuclear magnetic resonance structure, we construct a model of the predicted structured region of the carboxy terminus. An arrhythmia-associated H6 mutant that impairs inactivation decreases fluorescence resonance energy transfer, indicating destabilization of the distal-proximal intramolecular interaction. These data provide a structural correlation to the pathological phenotype of the mutant channel.


Subject(s)
Heart Conduction System/physiology , Long QT Syndrome/genetics , Sodium Channels/chemistry , Sodium Channels/genetics , Amino Acid Motifs , Cell Membrane/metabolism , Fluorescence Resonance Energy Transfer , Heart/physiology , Humans , Ion Channel Gating , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation , Myocardium/metabolism , NAV1.5 Voltage-Gated Sodium Channel , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Secondary , Protein Structure, Tertiary , Sodium Channels/metabolism
6.
J Biol Chem ; 283(25): 17221-6, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18458091

ABSTRACT

Voltage-dependent calcium channels (VDCCs) play a pivotal role in normal excitation-contraction coupling in cardiac myocytes. These channels can be modulated through activation of beta-adrenergic receptors (beta-ARs), which leads to an increase in calcium current (I(Ca-L)) density through cardiac Ca(v)1 channels as a result of phosphorylation by cAMP-dependent protein kinase A. Changes in I(Ca-L) density and kinetics in heart failure often occur in the absence of changes in Ca(v)1 channel expression, arguing for the importance of post-translational modification of these channels in heart disease. The precise molecular mechanisms that govern the regulation of VDCCs and their cell surface localization remain unknown. Our data show that sustained beta-AR activation induces internalization of a cardiac macromolecular complex involving VDCC and beta-arrestin 1 (beta-Arr1) into clathrin-coated vesicles. Pretreatment of myocytes with pertussis toxin prevents the internalization of VDCCs, suggesting that G(i/o) mediates this response. A peptide that selectively disrupts the interaction between Ca(V)1.2 and beta-Arr1 and tyrosine kinase inhibitors readily prevent agonist-induced VDCC internalization. These observations suggest that VDCC trafficking is mediated by G protein switching to G(i) of the beta-AR, which plays a prominent role in various cardiac pathologies associated with a hyperadrenergic state, such as hypertrophy and heart failure.


Subject(s)
Arrestins/metabolism , Calcium Channels, L-Type/metabolism , Myocardium/metabolism , Receptors, Adrenergic, beta/metabolism , Animals , Cell Membrane/metabolism , Clathrin/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Kinetics , Models, Biological , Muscle Cells/metabolism , Peptides/chemistry , Protein Binding , Rats , Substrate Specificity , beta-Arrestin 1 , beta-Arrestins
7.
J Biol Chem ; 281(41): 31131-41, 2006 Oct 13.
Article in English | MEDLINE | ID: mdl-16912040

ABSTRACT

Many metabotropic receptors in the nervous system act through signaling pathways that result in the inhibition of voltage-dependent calcium channels. Our previous findings showed that activation of seven-transmembrane receptors results in the internalization of calcium channels. This internalization takes place within a few seconds, raising the question of whether the endocytic machinery is in close proximity to the calcium channel to cause such rapid internalization. Here we show that voltage-dependent calcium channels are pre-associated with arrestin, a protein known to play a role in receptor trafficking. Upon GABAB receptor activation, receptors are recruited to the arrestin-channel complex and internalized. beta-Arrestin 1 selectively binds to the SNARE-binding region of the calcium channel. Peptides containing the arrestin-binding site of the channel disrupt agonist-induced channel internalization. Taken together these data suggest a novel neuronal role for arrestin.


Subject(s)
Arrestin/physiology , Calcium Channels/metabolism , Animals , Arrestin/metabolism , Binding Sites , Calcium Channels, N-Type/metabolism , Chick Embryo , Endocytosis , Neurons/metabolism , Peptides/chemistry , Protein Binding , Protein Transport , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...