Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Channels (Austin) ; 12(1): 176-186, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29983085

RESUMO

INTRODUCTION: Individual mutations in the SCN5A-encoding cardiac sodium channel α-subunit usually cause a single cardiac arrhythmia disorder, some cause mixed biophysical or clinical phenotypes. Here we report an infant, female patient harboring a N406K mutation in SCN5A with a marked and mixed biophysical phenotype and assess pathogenic mechanisms. METHODS AND RESULTS: A patient suffered from recurrent seizures during sleep and torsades de pointes with a QTc of 530 ms. Mutational analysis identified a N406K mutation in SCN5A. The mutation was engineered by site-directed mutagenesis and heterologously expressed in HEK293 cells. After 48 hours incubation with and without mexiletine, macroscopic voltage-gated sodium current (INa) was measured with standard whole-cell patch clamp techniques. SCN5A-N406K elicited both a significantly decreased peak INa and a significantly increased late INa compared to wide-type (WT) channels. Furthermore, mexiletine both restored the decreased peak INa of the mutant channel and inhibited the increased late INa of the mutant channel. CONCLUSION: SCN5A-N406K channel displays both "gain-of-function" in late INa and "loss-of-function" in peak INa density contributing to a mixed biophysical phenotype. Moreover, our finding may provide the first example that mexiletine exerts a dual rescue of both "gain-of-function" and "loss-of-function" of the mutant sodium channel.


Assuntos
Antiarrítmicos/farmacologia , Doença do Sistema de Condução Cardíaco/tratamento farmacológico , Doença do Sistema de Condução Cardíaco/genética , Síndrome do QT Longo/tratamento farmacológico , Síndrome do QT Longo/genética , Mexiletina/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Doença do Sistema de Condução Cardíaco/metabolismo , Feminino , Células HEK293 , Humanos , Lactente , Síndrome do QT Longo/metabolismo , Mutação , Fenótipo
2.
PLoS One ; 10(4): e0124921, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25923670

RESUMO

BACKGROUND: SCN5A is a susceptibility gene for type 3 long QT syndrome, Brugada syndrome, and sudden infant death syndrome. INa dysfunction from mutated SCN5A can depend upon the splice variant background in which it is expressed and also upon environmental factors such as acidosis. S1787N was reported previously as a LQT3-associated mutation and has also been observed in 1 of 295 healthy white controls. Here, we determined the in vitro biophysical phenotype of SCN5A-S1787N in an effort to further assess its possible pathogenicity. METHODS AND RESULTS: We engineered S1787N in the two most common alternatively spliced SCN5A isoforms, the major isoform lacking a glutamine at position 1077 (Q1077del) and the minor isoform containing Q1077, and expressed these two engineered constructs in HEK293 cells for electrophysiological study. Macroscopic voltage-gated INa was measured 24 hours after transfection with standard whole-cell patch clamp techniques. We applied intracellular solutions with pH7.4 or pH6.7. S1787N in the Q1077 background had WT-like INa including peak INa density, activation and inactivation parameters, and late INa amplitude in both pH 7.4 and pH 6.7. However, with S1787N in the Q1077del background, the percentages of INa late/peak were increased by 2.1 fold in pH 7.4 and by 2.9 fold in pH 6.7 when compared to WT. CONCLUSION: The LQT3-like biophysical phenotype for S1787N depends on both the SCN5A splice variant and on the intracellular pH. These findings provide further evidence that the splice variant and environmental factors affect the molecular phenotype of cardiac SCN5A-encoded sodium channel (Nav1.5), has implications for the clinical phenotype, and may provide insight into acidosis-induced arrhythmia mechanisms.


Assuntos
Acidose/genética , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Splicing de RNA , Acidose/complicações , Acidose/patologia , Arritmias Cardíacas/etiologia , Sequência de Bases , Doença do Sistema de Condução Cardíaco , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Síndrome do QT Longo/genética , Síndrome do QT Longo/patologia , Potenciais da Membrana , Mutagênese Sítio-Dirigida , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Técnicas de Patch-Clamp , Fenótipo , Polimorfismo de Nucleotídeo Único , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
3.
J Mol Cell Cardiol ; 84: 104-11, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25918050

RESUMO

We evaluated whether phospholemman (PLM) regulates L-type Ca(2+) current (ICa) in mouse ventricular myocytes. Expression of α1-subunit of L-type Ca(2+) channels between wild-type (WT) and PLM knockout (KO) hearts was similar. Compared to WT myocytes, peak ICa (at -10 mV) from KO myocytes was ~41% larger, the inactivation time constant (τ(inact)) of ICa was ~39% longer, but deactivation time constant (τ(deact)) was similar. In the presence of isoproterenol (1 µM), peak ICa was ~48% larger and τ(inact) was ~144% higher in KO myocytes. With Ba(2+) as the permeant ion, PLM enhanced voltage-dependent inactivation but had no effect on τ(deact). To dissect the molecular determinants by which PLM regulated ICa, we expressed PLM mutants by adenovirus-mediated gene transfer in cultured KO myocytes. After 24h in culture, KO myocytes expressing green fluorescent protein (GFP) had significantly larger peak ICa and longer τ(inact) than KO myocytes expressing WT PLM; thereby independently confirming the observations in freshly isolated myocytes. Compared to KO myocytes expressing GFP, KO myocytes expressing the cytoplasmic domain truncation mutant (TM43), the non-phosphorylatable S68A mutant, the phosphomimetic S68E mutant, and the signature PFXYD to alanine (ALL5) mutant all resulted in lower peak ICa. Expressing PLM mutants did not alter expression of α1-subunit of L-type Ca(2+) channels in cultured KO myocytes. Our results suggested that both the extracellular PFXYD motif and the transmembrane domain of PLM but not the cytoplasmic tail were necessary for regulation of peak ICa amplitude. We conclude that PLM limits Ca(2+) influx in cardiac myocytes by reducing maximal ICa and accelerating voltage-dependent inactivation.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Proteínas de Membrana/metabolismo , Miócitos Cardíacos/metabolismo , Fosfoproteínas/metabolismo , Adenoviridae/metabolismo , Motivos de Aminoácidos , Animais , Células Cultivadas , Citoplasma/química , Cães , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Isoproterenol/farmacologia , Proteínas de Membrana/química , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Mutantes/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Fosfoproteínas/química , Fosfosserina/metabolismo , Estrutura Terciária de Proteína , Relação Estrutura-Atividade
4.
Mol Med Rep ; 7(2): 571-6, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23229127

RESUMO

The anomalous mole fraction effect (AMFE) is an important indicator of ion-ion interactions in the pore of voltage-gated Ca2+ channels (VGCCs). The residues at position 1144 that differ in several classes of VGCCs are important to the permeation of the pore. Phe-1144 (F, CaV1) was substituted with glycine (G, CaV2) and lysine (K, CaV3) and the effects of mutation on the voltage and concentration dependency of AMFE were observed. Whole-cell currents were recorded in external solutions with Ca2+ and Ba2+ at the indicated ratios with a total divalent cation concentration of 2, 10 or 20 mM, at holding potentials from -80 to -20 mV. Results showed the ratio of Ba2+ to Ca2+ currents determined at 2 mM to be different from that determined under higher concentrations for wild-type channels but this ratio was not different when tail currents were evoked at different potentials. AMFE was greatest at relatively positive potentials (-20 mV) and when the total divalent cation concentrations were kept low (2 mM). AMFE was attenuated for F/G while it was accentuated for F/K compared with wild-type, respectively. The results demonstrated that glycine and lysine substitutions of Phe-1144 affect AMFE through different mechanisms. Additionally, residues at position 1144 were shown to be major determinates of channel permeation of several classes of VGCCs.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Motivos de Aminoácidos , Substituição de Aminoácidos , Bário/metabolismo , Cálcio/metabolismo , Canais de Cálcio Tipo L/genética , Células HEK293 , Humanos , Transporte de Íons/fisiologia , Técnicas de Patch-Clamp , Transfecção
5.
Am J Physiol Heart Circ Physiol ; 302(3): H770-81, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22081699

RESUMO

Expression and activity of cardiac Na(+)/Ca(2+) exchanger (NCX1) are altered in many disease states. We engineered mice in which the phosphomimetic phospholemman S68E mutant (inhibits NCX1 but not Na(+)-K(+)-ATPase) was constitutively overexpressed in a cardiac-specific manner (conS68E). At 4-6 wk, conS68E mice exhibited severe bradycardia, ventricular arrhythmias, increased left ventricular (LV) mass, decreased cardiac output (CO), and ∼50% mortality compared with wild-type (WT) littermates. Protein levels of NCX1, calsequestrin, ryanodine receptor, and α(1)- and α(2)-subunits of Na(+)-K(+)-ATPase were similar, but sarco(endo)plasmic reticulum Ca(2+)-ATPase was lower, whereas L-type Ca(2+) channels were higher in conS68E hearts. Resting membrane potential and action potential amplitude were similar, but action potential duration was dramatically prolonged in conS68E myocytes. Diastolic intracellular Ca(2+) ([Ca(2+)](i)) was higher, [Ca(2+)](i) transient and maximal contraction amplitudes were lower, and half-time of [Ca(2+)](i) transient decline was longer in conS68E myocytes. Intracellular Na(+) reached maximum within 3 min after isoproterenol addition, followed by decline in WT but not in conS68E myocytes. Na(+)/Ca(2+) exchange, L-type Ca(2+), Na(+)-K(+)-ATPase, and depolarization-activated K(+) currents were decreased in conS68E myocytes. At 22 wk, bradycardia and increased LV mass persisted in conS68E survivors. Despite comparable baseline CO, conS68E survivors at 22 wk exhibited decreased chronotropic, inotropic, and lusitropic responses to isoproterenol. We conclude that constitutive overexpression of S68E mutant was detrimental, both in terms of depressed cardiac function and increased arrhythmogenesis.


Assuntos
Arritmias Cardíacas/fisiopatologia , Insuficiência Cardíaca , Proteínas de Membrana/genética , Fosfoproteínas/genética , Trocador de Sódio e Cálcio/genética , Potenciais de Ação/genética , Potenciais de Ação/fisiologia , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Bradicardia/genética , Bradicardia/metabolismo , Bradicardia/mortalidade , Cálcio/metabolismo , Canais de Cálcio Tipo L/fisiologia , Feminino , Expressão Gênica/fisiologia , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/mortalidade , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Contração Miocárdica/fisiologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/fisiologia , Fosfoproteínas/metabolismo , Potássio/metabolismo , Receptores Adrenérgicos beta/metabolismo , Sódio/metabolismo , Trocador de Sódio e Cálcio/metabolismo
6.
J Neurophysiol ; 107(7): 1942-51, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22205645

RESUMO

Ca(V)2.2 (N-type) and Ca(V)1.2 (L-type) calcium channels gate differently in response to membrane depolarization, which is critical to the unique physiological functions mediated by these channels. We wondered if the source for these differences could be identified. As a first step, we examined the effect of domain exchange between N-type and L-type channels on activation-deactivation kinetics, which were significantly different between these channels. Kinetic analysis of chimeric channels revealed N-channel-like deactivation for all chimeric channels containing N-channel domain III, while activation appeared to be a more distributed function across domains. This led us to hypothesize that domain III was an important regulator of N-channel closing. This idea was further examined with R-roscovitine, which is a trisubstituted purine that slows N-channel deactivation by exclusively binding to activated N-channels. L-channels lack this response to roscovitine, which allowed us to use N-L chimeras to test the role of domain III in roscovitine modulation of N-channel deactivation. In support of our hypothesis, all chimeric channels containing the N-channel domain III responded to roscovitine with slowed deactivation, while those chimeric channels with L-channel domain III did not. Thus a combination of kinetic and pharmacological evidence supports the hypothesis that domain III is an important regulator of N-channel closing. Our results support specialization of gating functions among calcium channel domains.


Assuntos
Fenômenos Biofísicos/fisiologia , Canais de Cálcio Tipo N/química , Canais de Cálcio Tipo N/fisiologia , Ativação do Canal Iônico/fisiologia , Animais , Fenômenos Biofísicos/efeitos dos fármacos , Fenômenos Biofísicos/genética , Canais de Cálcio Tipo N/genética , Estimulação Elétrica , Células HEK293 , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/genética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Proteínas Mutantes Quiméricas/genética , Técnicas de Patch-Clamp , Inibidores de Proteínas Quinases/farmacologia , Estrutura Terciária de Proteína , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Purinas/farmacologia , Coelhos , Roscovitina , Transfecção
7.
Ann Hum Genet ; 75(5): 559-68, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21790524

RESUMO

Surfactant protein-D (SP-D) is expressed on mucosal surfaces and functions in the innate immune response to microorganisms. We studied the genetic association of the two nonsynonymous SP-D single nucleotide polymorphisms (SNPs) rs721917 and rs2243639 in 256 inflammatory bowel disease (IBD) cases (123 CD and 133 UC) and 376 unrelated healthy individuals from an IBD population from Central Pennsylvania. Case-control analysis revealed a significant association of rs2243639 with susceptibility to Crohn's disease (CD) (p= 0.0036), but not ulcerative colitis (UC) (p= 0.883), and no association of rs721917 with CD (p= 0.328) or UC (p= 0.218). Using intestinal tissues from 19 individuals heterozygous for each SNP, we compared allelic expression of these two SNPs between diseased and matched normal tissues. rs2243639 exhibited balanced biallelic (BB) expression; while rs721917 exhibited differential allelic expression (BB 37%, imbalanced biallelic [IB] 45%, and dominant monoallelic [DM] 18%). Comparison of allelic expression pattern between diseased and matched normal tissues, 13 of 19 individuals (14 UC, 5 CD) showed a similar pattern. The six patients exhibiting a different pattern were all UC patients. The results suggest that differential allelic expression may affect penetrance of the SNP rs721917 disease-susceptibility allele in IBD. The potential impact of SP-D monoallelic expression on incomplete penetrance is discussed.


Assuntos
Doenças Inflamatórias Intestinais/genética , Polimorfismo de Nucleotídeo Único , Proteína D Associada a Surfactante Pulmonar/genética , Alelos , Estudos de Casos e Controles , Predisposição Genética para Doença , Humanos , Mucosa Intestinal/metabolismo , Penetrância , Polimorfismo de Fragmento de Restrição
8.
Am J Physiol Cell Physiol ; 299(5): C1203-11, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20720179

RESUMO

We have found that phospholemman (PLM) associates with and modulates the gating of cardiac L-type calcium channels (Wang et al., Biophys J 98: 1149-1159, 2010). The short 17 amino acid extracellular NH(2)-terminal domain of PLM contains a highly conserved PFTYD sequence that defines it as a member of the FXYD family of ion transport regulators. Although we have learned a great deal about PLM-dependent changes in calcium channel gating, little is known regarding the molecular mechanisms underlying the observed changes. Therefore, we investigated the role of the PFTYD segment in the modulation of cardiac calcium channels by individually replacing Pro-8, Phe-9, Thr-10, Tyr-11, and Asp-12 with alanine (P8A, F9A, T10A, Y11A, D12A). In addition, Asp-12 was changed to lysine (D12K) and cysteine (D12C). As expected, wild-type PLM significantly slows channel activation and deactivation and enhances voltage-dependent inactivation (VDI). We were surprised to find that amino acid substitutions at Thr-10 and Asp-12 significantly enhanced the ability of PLM to modulate Ca(V)1.2 gating. T10A exhibited a twofold enhancement of PLM-induced slowing of activation, whereas D12K and D12C dramatically enhanced PLM-induced increase of VDI. The PLM-induced slowing of channel closing was abrogated by D12A and D12C, whereas D12K and T10A failed to impact this effect. These studies demonstrate that the PFXYD motif is not necessary for the association of PLM with Ca(V)1.2. Instead, since altering the chemical and/or physical properties of the PFXYD segment alters the relative magnitudes of opposing PLM-induced effects on Ca(V)1.2 channel gating, PLM appears to play an important role in fine tuning the gating kinetics of cardiac calcium channels and likely plays an important role in shaping the cardiac action potential and regulating Ca(2+) dynamics in the heart.


Assuntos
Motivos de Aminoácidos , Substituição de Aminoácidos , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Ativação do Canal Iônico/genética , Proteínas de Membrana/metabolismo , Fosfoproteínas/metabolismo , Alanina/genética , Sequência de Aminoácidos , Animais , Linhagem Celular , Humanos , Proteínas de Membrana/genética , Dados de Sequência Molecular , Mutação , Técnicas de Patch-Clamp , Fosfoproteínas/genética , Treonina/genética
9.
Biophys J ; 98(7): 1149-59, 2010 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-20371314

RESUMO

Ca(2+) entry through L-type calcium channels (Ca(V)1.2) is critical in shaping the cardiac action potential and initiating cardiac contraction. Modulation of Ca(V)1.2 channel gating directly affects myocyte excitability and cardiac function. We have found that phospholemman (PLM), a member of the FXYD family and regulator of cardiac ion transport, coimmunoprecipitates with Ca(V)1.2 channels from guinea pig myocytes, which suggests PLM is an endogenous modulator. Cotransfection of PLM in HEK293 cells slowed Ca(V)1.2 current activation at voltages near the threshold for activation, slowed deactivation after long and strong depolarizing steps, enhanced the rate and magnitude of voltage-dependent inactivation (VDI), and slowed recovery from inactivation. However, Ca(2+)-dependent inactivation was not affected. Consistent with slower channel closing, PLM significantly increased Ca(2+) influx via Ca(V)1.2 channels during the repolarization phase of a human cardiac action potential waveform. Our results support PLM as an endogenous regulator of Ca(V)1.2 channel gating. The enhanced VDI induced by PLM may help protect the heart under conditions such as ischemia or tachycardia where the channels are depolarized for prolonged periods of time and could induce Ca(2+) overload. The time and voltage-dependent slowed deactivation could represent a gating shift that helps maintain Ca(2+) influx during the cardiac action potential waveform plateau phase.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Coração/fisiologia , Proteínas de Membrana/metabolismo , Fosfoproteínas/metabolismo , Potenciais de Ação , Animais , Cálcio/metabolismo , Cobaias , Ventrículos do Coração/patologia , Humanos , Íons , Isquemia/patologia , Modelos Biológicos , Células Musculares/metabolismo , Estrutura Terciária de Proteína , Taquicardia/patologia
10.
J Membr Biol ; 233(1-3): 23-33, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20098982

RESUMO

Glutamate scanning mutagenesis was used to assess the role of the calcicludine binding segment in regulating channel permeation and gating using both Ca(2+) and Ba(2+) as charge carriers. As expected, wild-type Ca(V)1.2 channels had a Ba(2+) conductance ~2x that in Ca(2+) (G(Ba)/G(Ca) = 2) and activation was ~10 mV more positive in Ca(2+) vs. Ba(2+). Of the 11 mutants tested, F1126E was the only one that showed unique permeation and gating properties compared to the wild type. F1126E equalized the Ca(V)1.2 channel conductance (G(Ba)/G(Ca) = 1) and activation voltage dependence between Ca(2+) and Ba(2+). Ba(2+) permeation was reduced because the interactions among multiple Ba(2+) ions and the pore were specifically altered for F1126E, which resulted in Ca(2+)-like ionic conductance and unitary current. However, the high-affinity block of monovalent cation flux was not altered for either Ca(2+) or Ba(2+). The half-activation voltage of F1126E in Ba(2+) was depolarized to match that in Ca(2+), which was unchanged from that in the wild type. As a result, the voltages for half-activation and half-inactivation of F1126E in Ba(2+) and Ca(2+) were similar to those of wild-type in Ca(2+). This effect was specific to F1126E since F1126A did not affect the half-activation voltage in either Ca(2+) or Ba(2+). These results indicate that residues in the outer vestibule of the Ca(V)1.2 channel pore are major determinants of channel gating, selectivity, and permeation.


Assuntos
Bário/metabolismo , Transporte Biológico/fisiologia , Canais de Cálcio Tipo L/metabolismo , Cálcio/metabolismo , Ativação do Canal Iônico/fisiologia , Transporte Biológico/genética , Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo L/genética , Linhagem Celular , Eletrofisiologia , Glutamatos/genética , Glutamatos/fisiologia , Humanos , Ativação do Canal Iônico/genética , Mutagênese Sítio-Dirigida , Técnicas de Patch-Clamp
11.
J Biol Chem ; 285(1): 43-53, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19887376

RESUMO

L-type (Ca(V)1.2) calcium channel antagonists play an important role in the treatment of cardiovascular disease. (R)-Roscovitine, a trisubstituted purine, has been shown to inhibit L-currents by slowing activation and enhancing inactivation. This study utilized molecular and pharmacological approaches to determine whether these effects result from (R)-roscovitine binding to a single site. Using the S enantiomer, we find that (S)-roscovitine enhances inactivation without affecting activation, which suggests multiple sites. This was further supported in studies using chimeric channels comprised of N- and L-channel domains. Those chimeras containing L-channel domains I and IV showed (R)-roscovitine-induced slowed activation like that of wild type L-channels, whereas chimeric channels containing L-channel domain I responded to (R)-roscovitine with enhanced inactivation. We conclude that (R)-roscovitine binds to distinct sites on L-type channels to slow activation and enhance inactivation. These sites appear to be unique from other calcium channel antagonist sites that reside within domains III and IV and are thus novel sites that could be exploited for future drug development. Trisubstituted purines could become a new class of drugs for the treatment of diseases related to hyperfunction of L-type channels, such as Torsades de Pointes.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Ativação do Canal Iônico , Purinas/metabolismo , Animais , Sítios de Ligação , Canais de Cálcio Tipo L/química , Linhagem Celular , Relação Dose-Resposta a Droga , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Estrutura Terciária de Proteína , Purinas/farmacologia , Coelhos , Ratos , Proteínas Recombinantes/metabolismo , Roscovitina
12.
Am J Physiol Gastrointest Liver Physiol ; 297(1): G34-42, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19423751

RESUMO

Sensitization of esophageal sensory afferents by inflammatory mediators plays an important role in esophageal nociception. We have shown esophageal mast cell activation induces long-lasting mechanical hypersensitivity in vagal nodose C-fibers. However, the roles of mast cell mediators and downstream ion channels in this process are unclear. Mast cell tryptase via protease-activated receptor 2 (PAR2)-mediated pathways sensitizes sensory nerves and induces hyperalgesia. Transient receptor potential A1 (TRPA1) plays an important role in mechanosensory transduction and nociception. Here we tested the hypothesis that mast cell activation via a PAR2-dependent mechanism sensitizes TRPA1 to induce mechanical hypersensitivity in esophageal vagal C-fibers. The expression profiles of PAR2 and TRPA1 in vagal nodose ganglia were determined by immunostaining, Western blot, and RT-PCR. Extracellular recordings from esophageal nodose neurons were performed in ex vivo guinea pig esophageal-vagal preparations. Action potentials evoked by esophageal distention and chemical perfusion were compared. Both PAR2 and TRPA1 expressions were identified in vagal nodose neurons by immunostaining, Western blot, and RT-PCR. Ninety-one percent of TRPA1-positive neurons were of small and medium diameters, and 80% coexpressed PAR2. Esophageal mast cell activation significantly enhanced the response of nodose C-fibers to esophageal distension (mechanical hypersensitivity). This was mimicked by PAR2-activating peptide, which sustained for 90 min after wash, but not by PAR2 reverse peptide. TRPA1 inhibitor HC-030031 pretreatment significantly inhibited mechanical hypersensitivity induced by either mast cell activation or PAR2 agonist. Collectively, our data provide new evidence that sensitizing TRPA1 via a PAR2-dependent mechanism plays an important role in mast cell activation-induced mechanical hypersensitivity of vagal nodose C-fibers in guinea pig esophagus.


Assuntos
Esôfago/inervação , Hiperalgesia/metabolismo , Hipersensibilidade/metabolismo , Mastócitos/metabolismo , Mecanotransdução Celular , Fibras Nervosas Amielínicas/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Nervo Vago/metabolismo , Vias Aferentes/metabolismo , Vias Aferentes/fisiopatologia , Animais , Modelos Animais de Doenças , Potenciais Evocados , Cobaias , Hiperalgesia/imunologia , Hiperalgesia/fisiopatologia , Hipersensibilidade/imunologia , Hipersensibilidade/fisiopatologia , Masculino , Mastócitos/imunologia , Gânglio Nodoso/metabolismo , Gânglio Nodoso/fisiopatologia , Ovalbumina , Estimulação Física , Pressão , Receptor PAR-2/metabolismo , Estimulação Química , Estresse Mecânico , Canais de Potencial de Receptor Transitório/genética , Triptases/metabolismo , Nervo Vago/fisiopatologia
13.
J Physiol ; 587(3): 551-65, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19074970

RESUMO

Timothy syndrome (TS) is a multiorgan dysfunction caused by a Gly to Arg substitution at position 406 (G406R) of the human CaV1.2 (L-type) channel. The TS phenotype includes severe arrhythmias that are thought to be triggered by impaired open-state voltage-dependent inactivation (OSvdI). The effect of the TS mutation on other L-channel gating mechanisms has yet to be investigated. We compared kinetic properties of exogenously expressed (HEK293 cells) rabbit cardiac L-channels with (G436R; corresponding to position 406 in human clone) and without (wild-type) the TS mutation. Our results surprisingly show that the TS mutation did not affect close-state voltage-dependent inactivation, which suggests different gating mechanisms underlie these two types of voltage-dependent inactivation. The TS mutation also significantly slowed activation at voltages less than 10 mV, and significantly slowed deactivation across all test voltages. Deactivation was slowed in the double mutant G436R/S439A, which suggests that phosphorylation of S439 was not involved. The L-channel agonist Bay K8644 increased the magnitude of both step and tail currents, but surprisingly failed to slow deactivation of TS channels. Our mathematical model showed that slowed deactivation plus impaired OSvdI combine to synergistically increase cardiac action potential duration that is a likely cause of arrhythmias in TS patients. Roscovitine, a tri-substituted purine that enhances L-channel OSvdI, restored TS-impaired OSvdI. Thus, inactivation-enhancing drugs are likely to improve cardiac arrhythmias and other pathologies afflicting TS patients.


Assuntos
Canais de Cálcio Tipo L , Ativação do Canal Iônico/fisiologia , Síndrome do QT Longo/fisiopatologia , Mutação de Sentido Incorreto , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/farmacologia , Animais , Agonistas dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Linhagem Celular , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Síndrome do QT Longo/genética , Modelos Teóricos , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Purinas/farmacologia , Coelhos , Roscovitina
14.
Biochemistry ; 46(25): 7590-8, 2007 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-17536837

RESUMO

How dihydropyridines modulate L-type voltage-gated Ca2+ channels is not known. Dihydropyridines bind cooperatively with Ca2+ binding to the selectivity filter, suggesting that they alter channel activity by promoting structural rearrangements in the pore. We used radioligand binding and patch-clamp electrophysiology to demonstrate that calcicludine, a toxin from the venom of the green mamba snake, binds in the outer vestibule of the pore and, like Ca2+, is a positive modulator of dihydropyridine binding. Data were fit using an allosteric scheme where dissociation constants for dihydropyridine and calcicludine binding, KDHP and KCaC, are linked via the coupling factor, alpha. Nine acidic amino acids located within the S5-Pore-helix segment of repeat III were sequentially changed to alanine in groups of three, resulting in the mutant channels, Mut-A, Mut-B, and Mut-C. Mut-A, whose substitutions are proximal to IIIS5, exhibits a 4.5-fold reduction in dihydropyridine binding and is insensitive to calcicludine binding. Block of Mut-A currents by calcicludine is indistinguishable from wild-type, indicating that KCaC is unchanged and that the coupling between dihydropyridine and calcicludine binding (i.e., alpha) is disrupted. Mut-B and Mut-C possess KDHP values that resemble that of the wild type. Mut-C, the most C-terminal of the mutant channels, is insensitive to calcicludine binding and block. KCaC values for the Mut-C single mutants, E1122A, D1127A, and D1129A, increase from 0.3 (wild type) to 1.14, 2.00, and 20.5 microM, respectively. Together, these findings suggest that dihydropyridine antagonist and calcicludine binding to L-type Ca2+ channels promote similar structural changes in the pore that stabilize the channel in a nonconducting, blocked state.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Di-Hidropiridinas/metabolismo , Venenos Elapídicos/metabolismo , Alanina/metabolismo , Regulação Alostérica , Sequência de Aminoácidos , Substituição de Aminoácidos , Canais de Cálcio Tipo L/genética , Linhagem Celular , Di-Hidropiridinas/antagonistas & inibidores , Dissulfetos/química , Venenos Elapídicos/química , Humanos , Cinética , Técnicas de Patch-Clamp
15.
Mol Pharmacol ; 70(2): 667-75, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16675661

RESUMO

Dihydropyridines (DHPs) are an important class of drugs, used extensively in the treatment of angina pectoris, hypertension, and arrhythmia. The molecular mechanism by which DHPs modulate Ca(2+) channel function is not known in detail. We have found that DHP binding is allosterically coupled to Ca(2+) binding to the selectivity filter of the skeletal muscle Ca(2+) channel Ca(V)1.1, which initiates excitation-contraction coupling and conducts L-type Ca(2+) currents. Increasing Ca(2+) concentrations from approximately 10 nM to 1 mM causes the DHP receptor site to shift from a low-affinity state to a high-affinity state with an EC(50) for Ca(2+) of 300 nM. Substituting each of the four negatively charged glutamate residues that form the ion selectivity filter with neutral glutamine or positively charged lysine residues results in mutant channels whose DHP binding affinities are decreased up to 10-fold and are up to 150-fold less sensitive to Ca(2+) than wild-type channels. Analysis of mutations of amino acid residues adjacent to the selectivity filter led to identification of Phe-1013 and Tyr-1021, whose mutation causes substantial changes in DHP binding. Thermo-dynamic mutant cycle analysis of these mutants demonstrates that Phe-1013 and Tyr-1021 are energetically coupled when a single Ca(2+) ion is bound to the channel pore. We propose that DHP binding stabilizes a nonconducting state containing a single Ca(2+) ion in the pore through which Phe-1013 and Tyr-1021 are energetically coupled. The selectivity filter in this energetically coupled high-affinity state is blocked by bound Ca(2+), which is responsible for the high-affinity inhibition of Ca(2+) channels by DHP antagonists.


Assuntos
Bloqueadores dos Canais de Cálcio/metabolismo , Canais de Cálcio Tipo L/metabolismo , Cálcio/metabolismo , Isradipino/metabolismo , Regulação Alostérica , Sequência de Aminoácidos , Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo L/efeitos dos fármacos , Isradipino/farmacologia , Dados de Sequência Molecular , Conformação Proteica , Relação Estrutura-Atividade
16.
J Biol Chem ; 281(14): 9076-85, 2006 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-16461353

RESUMO

TRPM2, a member of the transient receptor potential (TRP) superfamily, is a Ca(2+)-permeable channel activated by oxidative stress or tumor necrosis factoralpha involved in susceptibility to cell death. TRPM2 activation is dependent on the level of intracellular Ca(2+). We explored whether calmodulin (CaM) is the Ca(2+) sensor for TRPM2. HEK 293T cells were transfected with TRPM2 and wild type CaM or mutant CaM (CaM(MUT)) with substitutions of all four EF hands. Treatment of cells expressing TRPM2 with H(2)O(2) or tumor necrosis factor alpha resulted in a significant increase in intracellular calcium ([Ca(2+)](i)). This was not affected by coexpression of CaM, suggesting that endogenous CaM levels are sufficient for maximal response. Cotransfection of CaM(MUT) with TRPM2 dramatically inhibited the increase in [Ca(2+)](i), demonstrating the requirement for CaM in TRPM2 activation. Immunoprecipitation confirmed direct interaction of CaM and CaM(MUT) with TRPM2, and the Ca(2+) dependence of this association. CaM bound strongly to the TRPM2 N terminus (amino acids 1-730), but weakly to the C terminus (amino acids 1060-1503). CaM binding to an IQ-like motif (amino acids 406-416) in the TRPM2 N terminus was demonstrated utilizing gel shift, immunoprecipitation, biotinylated CaM overlay, and pull-down assays. A substitution mutant of the IQ-like motif of TRPM2 (TRPM2-IQ(MUT1)) reduced but did not eliminate CaM binding to TRPM2, suggesting the presence of at least one other CaM binding site. The functional importance of the TRPM2 IQ-like motif was demonstrated by treatment of TRPM2-IQ(MUT1)-expressing cells with H(2)O(2). The increase in [Ca(2+)](i) observed with wild type TRPM2 was absent and cell viability was preserved. These data demonstrate the requirement for CaM in TRPM2 activation. They suggest that Ca(2+) entering through TRPM2 enhances interaction of CaM with TRPM2 at the IQ-like motif in the N terminus, providing crucial positive feedback for channel activation.


Assuntos
Cálcio/farmacocinética , Calmodulina/fisiologia , Estresse Oxidativo , Canais de Cátion TRPM/fisiologia , Substituição de Aminoácidos , Calmodulina/genética , Técnicas de Cultura de Células , Morte Celular , Sobrevivência Celular , Humanos , Imunoprecipitação , Rim/citologia , Rim/embriologia , Mutação , Transfecção
17.
Biophys J ; 89(3): 1731-43, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15980164

RESUMO

Ba(2+) currents through Ca(V)1.2 Ca(2+) channels are typically twice as large as Ca(2+) currents. Replacing Phe-1144 in the pore-loop of domain III with glycine and lysine, and Tyr-1152 with lysine, reduces whole-cell G(Ba)/G(Ca) from 2.2 (wild-type) to 0.95, 1.21, and 0.90, respectively. Whole-cell and single-channel measurements indicate that reductions in G(Ba)/G(Ca) result specifically from a decrease in Ba(2+) conductance and not changes in V(h) or P(O). Half-maximal block of I(Li) is increased by 3.2-, 3.8-, and 1.6-fold in Ca(2+), and 3.8-, 4.2-, and 1.8-fold in Ba(2+) for F1144G, Y1152K, and F1144K, respectively. High affinity interactions of individual divalent cations to the pore are not important for determining G(Ba)/G(Ca), because the fold increases in IC(50) values for Ba(2+) and Ca(2+) are similar. On the contrary, conductance-concentration curves indicate that G(Ba)/G(Ca) is reduced because the interactions of multiple Ba(2+) ions in the mutant pores are altered. The complexity of these interactions is exemplified by the anomalous mole fraction effect, which is flattened for F1144G and FY/GK but accentuated for F1144K. In summary, the physicochemical properties of the amino acid residues at positions 1144 and 1152 are crucial to the pore's ability to distinguish between multiple Ba(2+) ions and Ca(2+) ions.


Assuntos
Bário/química , Canais de Cálcio Tipo L/fisiologia , Canais de Cálcio/química , Cálcio/química , Sequência de Aminoácidos , Transporte Biológico , Cálcio/metabolismo , Canais de Cálcio Tipo L/química , Cátions , Cátions Bivalentes , Linhagem Celular , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Ácido Glutâmico/química , Glicina/química , Humanos , Concentração de Íons de Hidrogênio , Concentração Inibidora 50 , Ativação do Canal Iônico , Íons , Cinética , Lisina/química , Potenciais da Membrana , Dados de Sequência Molecular , Mutação , Técnicas de Patch-Clamp , Fenilalanina/química , Ligação Proteica , Estrutura Terciária de Proteína , Tirosina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...