Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Biol Chem ; 300(7): 107470, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38879012

RESUMO

Resistance to inhibitors of cholinesterases (ric-8 proteins) are involved in modulating G-protein function, but little is known of their potential physiological importance in the heart. In the present study, we assessed the role of resistance to inhibitors of cholinesterase 8b (Ric-8b) in determining cardiac contractile function. We developed a murine model in which it was possible to conditionally delete ric-8b in cardiac tissue in the adult animal after the addition of tamoxifen. Deletion of ric-8b led to severely reduced contractility as measured using echocardiography days after administration of tamoxifen. Histological analysis of the ventricular tissue showed highly variable myocyte size, prominent fibrosis, and an increase in cellular apoptosis. RNA sequencing revealed transcriptional remodeling in response to cardiac ric-8b deletion involving the extracellular matrix and inflammation. Phosphoproteomic analysis revealed substantial downregulation of phosphopeptides related to myosin light chain 2. At the cellular level, the deletion of ric-8b led to loss of activation of the L-type calcium channel through the ß-adrenergic pathways. Using fluorescence resonance energy transfer-based assays, we showed ric-8b protein selectively interacts with the stimulatory G-protein, Gαs. We explored if deletion of Gnas (the gene encoding Gαs) in cardiac tissue using a similar approach in the mouse led to an equivalent phenotype. The conditional deletion of the Gαs gene in the ventricle led to comparable effects on contractile function and cardiac histology. We conclude that ric-8b is essential to preserve cardiac contractile function likely through an interaction with the stimulatory G-protein and downstream phosphorylation of myosin light chain 2.


Assuntos
Contração Miocárdica , Animais , Camundongos , Contração Miocárdica/efeitos dos fármacos , Cadeias Leves de Miosina/metabolismo , Cadeias Leves de Miosina/genética , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo L/genética , Miosinas Cardíacas/metabolismo , Miosinas Cardíacas/genética , Miocárdio/metabolismo , Miocárdio/patologia , Camundongos Knockout , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Humanos , Inibidores da Colinesterase/farmacologia , Masculino , Apoptose/efeitos dos fármacos , Fatores de Troca do Nucleotídeo Guanina
2.
Physiol Genomics ; 53(4): 150-159, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33719582

RESUMO

We have assessed the role of ric-b8 in the control of heart rate after the gene was implicated in a recent genome-wide association study of resting heart rate. We developed a novel murine model in which it was possible to conditionally delete ric-8b in the sinoatrial (SA) node after the addition of tamoxifen. Despite this, we were unable to obtain homozygotes and thus studied heterozygotes. Haploinsufficiency of ric-8b in the sinoatrial node induced by the addition of tamoxifen in adult animals leads to mice with a reduced heart rate. However, other electrocardiographic intervals (e.g., PR and QRS) were normal, and there was no apparent arrhythmia such as heart block. The positive chronotropic response to isoprenaline was abrogated, whereas the response to carbachol was unchanged. The pacemaker current If (funny current) has an important role in regulating heart rate, and its function is modulated by both isoprenaline and carbachol. Using a heterologous system expressing HCN4, we show that ric-8b can modulate the HCN4 current. Overexpression of ric-8b led to larger HCN4 currents, whereas silencing ric-8b led to smaller currents. Ric-8b modulates heart rate responses in vivo likely via its actions on the stimulatory G-protein.


Assuntos
Fatores de Troca do Nucleotídeo Guanina , Frequência Cardíaca , Animais , Estudo de Associação Genômica Ampla , Fatores de Troca do Nucleotídeo Guanina/genética , Camundongos
3.
Physiol Genomics ; 51(8): 323-332, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31172864

RESUMO

Atrial fibrillation is a significant worldwide contributor to cardiovascular morbidity and mortality. Few studies have investigated the differences in gene expression between the left and right atrial appendages, leaving their characterization largely unexplored. In this study, differential gene expression was investigated in atrial fibrillation and sinus rhythm using left and right atrial appendages from the same patients. RNA sequencing was performed on the left and right atrial appendages from five sinus rhythm (SR) control patients and five permanent AF case patients. Differential gene expression in both the left and right atrial appendages was analyzed using the Bioconductor package edgeR. A selection of differentially expressed genes, with relevance to atrial fibrillation, were further validated using quantitative RT-PCR. The distribution of the samples assessed through principal component analysis showed distinct grouping between left and right atrial appendages and between SR controls and AF cases. Overall 157 differentially expressed genes were identified to be downregulated and 90 genes upregulated in AF. Pathway enrichment analysis indicated a greater involvement of left atrial genes in the Wnt signaling pathway whereas right atrial genes were involved in clathrin-coated vesicle and collagen formation. The differing expression of genes in both left and right atrial appendages indicate that there are different mechanisms for development, support and remodeling of AF within the left and right atria.


Assuntos
Apêndice Atrial/fisiopatologia , Fibrilação Atrial/genética , Análise de Sequência de RNA/métodos , Transcriptoma/genética , Idoso , Idoso de 80 Anos ou mais , Fibrilação Atrial/patologia , Vesículas Revestidas por Clatrina/metabolismo , Estudos de Coortes , Colágeno/metabolismo , Ponte de Artéria Coronária , Regulação para Baixo/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/genética , Regulação para Cima/genética , Via de Sinalização Wnt/genética
4.
Am J Physiol Cell Physiol ; 314(5): C616-C626, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29342363

RESUMO

G protein-gated inwardly rectifying K+ (GIRK) channels are the major inwardly rectifying K+ currents in cardiac atrial myocytes and an important determinant of atrial electrophysiology. Inhibitory G protein α-subunits can both mediate activation via acetylcholine but can also suppress basal currents in the absence of agonist. We studied this phenomenon using whole cell patch clamping in murine atria from mice with global genetic deletion of Gαi2, combined deletion of Gαi1/Gαi3, and littermate controls. We found that mice with deletion of Gαi2 had increased basal and agonist-activated currents, particularly in the right atria while in contrast those with Gαi1/Gαi3 deletion had reduced currents. Mice with global genetic deletion of Gαi2 had decreased action potential duration. Tissue preparations of the left atria studied with a multielectrode array from Gαi2 knockout mice showed a shorter effective refractory period, with no change in conduction velocity, than littermate controls. Transcriptional studies revealed increased expression of GIRK channel subunit genes in Gαi2 knockout mice. Thus different G protein isoforms have differential effects on GIRK channel behavior and paradoxically Gαi2 act to increase basal and agonist-activated GIRK currents. Deletion of Gαi2 is potentially proarrhythmic in the atria.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Átrios do Coração/metabolismo , Ativação do Canal Iônico , Potássio/metabolismo , Potenciais de Ação , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatologia , Função do Átrio Esquerdo , Função do Átrio Direito , Feminino , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/deficiência , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Subunidades alfa de Proteínas de Ligação ao GTP/deficiência , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Frequência Cardíaca , Cinética , Masculino , Camundongos da Linhagem 129 , Camundongos Knockout , Período Refratário Eletrofisiológico
5.
J Biol Chem ; 290(31): 19233-44, 2015 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-26088132

RESUMO

The description of potential molecular substrates for predisposition to atrial fibrillation (AF) is incomplete, and it is unknown what role regulators of G-protein signaling might play. We address whether the attenuation of RGS4 function may promote AF and the mechanism through which this occurs. For this purpose, we studied a mouse with global genetic deletion of RGS4 (RGS4(-/-)) and the normal littermate controls (RGS4(+/+)). In vivo electrophysiology using atrial burst pacing revealed that mice with global RGS4 deletion developed AF more frequently than control littermates. Isolated atrial cells from RGS4(-/-) mice show an increase in Ca(2+) spark frequency under basal conditions and after the addition of endothelin-1 and abnormal spontaneous Ca(2+) release events after field stimulation. Isolated left atria studied on a multielectrode array revealed modest changes in path length for re-entry but abnormal electrical events after a pacing train in RGS4(-/-) mice. RGS4 deletion results in a predisposition to atrial fibrillation from enhanced activity in the Gαq/11-IP3 pathway, resulting in abnormal Ca(2+) release and corresponding electrical events.


Assuntos
Fibrilação Atrial/genética , Cálcio/metabolismo , Proteínas RGS/genética , Potenciais de Ação , Animais , Fibrilação Atrial/metabolismo , Sinalização do Cálcio , Estimulação Elétrica , Predisposição Genética para Doença , Átrios do Coração/metabolismo , Átrios do Coração/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/fisiologia , Proteínas RGS/metabolismo
6.
Pharmacol Res ; 105: 54-61, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26773203

RESUMO

Atrial fibrillation is the commonest cardiac arrhythmia and leads to significant clinical morbidity and mortality. It has a complex pathophysiology but is often initiated by atrial ectopic beats and because of atrial remodelling once it occurs it can become established. Thus therapeutic interventions designed to prevent the initial occurrence of the arrhythmia are particularly needed. At the cellular level, these ectopic beats arise because of abnormal calcium release events from the sarcoplasmic reticulum leading to an inward current mediated by the sodium-calcium exchanger. There has been considerable interest in this over the last few years largely focused on the ryanodine receptor and related signalling pathways. However, atrial myocytes also possess a well-developed inositol trisphosphate (IP3) dependent calcium release system and this has been less studied. In this review we focus on pathways and molecules that couple via the Gq\11 family of G-proteins including regulators of G-protein signalling that may influence IP3 mediated calcium release and atrial fibrillation.


Assuntos
Fibrilação Atrial/metabolismo , Cálcio/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Fosfatos de Inositol/metabolismo , Transdução de Sinais , Animais , Fibrilação Atrial/tratamento farmacológico , Fibrilação Atrial/patologia , Descoberta de Drogas , Átrios do Coração/efeitos dos fármacos , Átrios do Coração/metabolismo , Átrios do Coração/patologia , Humanos , Terapia de Alvo Molecular , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Transdução de Sinais/efeitos dos fármacos
7.
Org Biomol Chem ; 14(2): 455-459, 2016 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-26603469

RESUMO

Described in this work is a novel method for photochemically manipulating peptides and proteins via the installation of cysteine-selective photoactive tags. Thiomaleimides, generated simply by the addition of bromomaleimides to reduced disulfide bonds, undergo [2 + 2] photocycloadditions to reconnect the crosslink between the two cysteine residues. This methodology is demonstrated to enable photoactivation of a peptide by macrocyclisation, and reconnection of the heavy and light chains in an antibody fragment to form thiol stable conjugates. Finally we report on an intriguing thiomaleimide mediated photochemical decarboxylation of C-terminal cysteines, discovered during this study.


Assuntos
Cisteína/química , Dissulfetos/química , Maleimidas/química , Ciclização , Descarboxilação , Maleimidas/síntese química , Estrutura Molecular , Processos Fotoquímicos
8.
Org Biomol Chem ; 13(37): 9559-63, 2015 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-26299430

RESUMO

We report a concise approach to a multimodal imaging reagent for peptide labelling via disulfide bridging. The reagent is constructed using a one pot, three component, [3 + 2] cycloaddition of a fluorescent azide with a dithiomaleimide-alkyne, with concomitant incorporation of (125)I. The dithiomaleimide handle then enables site selective conjugation to a disulfide bond of a peptide whilst retaining the key structural bridging functionality, as exemplified on the therapeutic peptide octreotide.


Assuntos
Dissulfetos/química , Medicina Nuclear/métodos , Imagem Óptica/métodos , Peptídeos/química , Reação de Cicloadição , Células HEK293 , Humanos , Imagem Multimodal , Coloração e Rotulagem
10.
Eur Heart J ; 33(15): 1942-53, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22240500

RESUMO

AIMS: Anecdotal observations suggest that sub-clinical electrophysiological manifestations of arrhythmogenic right ventricular cardiomyopathy (ARVC) develop before detectable structural changes ensue on cardiac imaging. To test this hypothesis, we investigated a murine model with conditional cardiac genetic deletion of one desmoplakin allele (DSP ±) and compared the findings to patients with non-diagnostic features of ARVC who carried mutations in desmoplakin. METHODS AND RESULTS: Murine: the DSP (±) mice underwent electrophysiological, echocardiographic, and immunohistochemical studies. They had normal echocardiograms but delayed conduction and inducible ventricular tachycardia associated with mislocalization and reduced intercalated disc expression of Cx43. Sodium current density and myocardial histology were normal at 2 months of age. Human: ten patients with heterozygous mutations in DSP without overt structural heart disease (DSP+) and 12 controls with supraventricular tachycardia were studied by high-density electrophysiological mapping of the right ventricle. Using a standard S(1)-S(2) protocol, restitution curves of local conduction and repolarization parameters were constructed. Significantly greater mean increases in delay were identified particularly in the outflow tract vs. controls (P< 0.01) coupled with more uniform wavefront progression. The odds of a segment with a maximal activation-repolarization interval restitution slope >1 was 99% higher (95% CI: 13%; 351%, P = 0.017) in DSP+ vs. controls. Immunostaining revealed Cx43 mislocalization and variable Na channel distribution. CONCLUSION: Desmoplakin disease causes connexin mislocalization in the mouse and man preceding any overt histological abnormalities resulting in significant alterations in conduction-repolarization kinetics prior to morphological changes detectable on conventional cardiac imaging. Haploinsufficiency of desmoplakin is sufficient to cause significant Cx43 mislocalization. Changes in sodium current density and histological abnormalities may contribute to a worsening phenotype or disease but are not necessary to generate an arrhythmogenic substrate. This has important implications for the earlier diagnosis of ARVC and risk stratification.


Assuntos
Displasia Arritmogênica Ventricular Direita/genética , Desmoplaquinas/genética , Mutação/genética , Adulto , Idoso , Animais , Estudos de Casos e Controles , Desmoplaquinas/deficiência , Eletrocardiografia , Feminino , Deleção de Genes , Sistema de Condução Cardíaco/fisiologia , Heterozigoto , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canais de Sódio/fisiologia , Adulto Jovem
11.
Nat Commun ; 14(1): 1722, 2023 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-37012244

RESUMO

Cardiogenesis relies on the precise spatiotemporal coordination of multiple progenitor populations. Understanding the specification and differentiation of these distinct progenitor pools during human embryonic development is crucial for advancing our knowledge of congenital cardiac malformations and designing new regenerative therapies. By combining genetic labelling, single-cell transcriptomics, and ex vivo human-mouse embryonic chimeras we uncovered that modulation of retinoic acid signaling instructs human pluripotent stem cells to form heart field-specific progenitors with distinct fate potentials. In addition to the classical first and second heart fields, we observed the appearance of juxta-cardiac field progenitors giving rise to both myocardial and epicardial cells. Applying these findings to stem-cell based disease modelling we identified specific transcriptional dysregulation in first and second heart field progenitors derived from stem cells of patients with hypoplastic left heart syndrome. This highlights the suitability of our in vitro differentiation platform for studying human cardiac development and disease.


Assuntos
Células-Tronco Pluripotentes , Tretinoína , Humanos , Animais , Camundongos , Tretinoína/farmacologia , Coração , Miocárdio , Diferenciação Celular , Miócitos Cardíacos
12.
Neuromuscul Disord ; 32(11-12): 908-922, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36418198

RESUMO

Mutations in the dystrophin gene cause the most common and currently incurable Duchenne muscular dystrophy (DMD) characterized by progressive muscle wasting. Although abnormal Ca2+ handling is a pathological feature of DMD, mechanisms underlying defective Ca2+ homeostasis remain unclear. Here we generate a novel DMD patient-derived pluripotent stem cell (PSC) model of skeletal muscle with an isogenic control using clustered regularly interspaced short palindromic repeat (CRISPR)-mediated precise gene correction. Transcriptome analysis identifies dysregulated gene sets in the absence of dystrophin, including genes involved in Ca2+ handling, excitation-contraction coupling and muscle contraction. Specifically, analysis of intracellular Ca2+ transients and mathematical modeling of Ca2+ dynamics reveal significantly reduced cytosolic Ca2+ clearance rates in DMD-PSC derived myotubes. Pharmacological assays demonstrate Ca2+ flux in myotubes is determined by both intracellular and extracellular sources. DMD-PSC derived myotubes display significantly reduced velocity of contractility. Compared with a non-isogenic wildtype PSC line, these pathophysiological defects could be rescued by CRISPR-mediated precise gene correction. Our study provides new insights into abnormal Ca2+ homeostasis in DMD and suggests that Ca2+ signaling pathways amenable to pharmacological modulation are potential therapeutic targets. Importantly, we have established a human physiology-relevant in vitro model enabling rapid pre-clinical testing of potential therapies for DMD.


Assuntos
Distrofia Muscular de Duchenne , Células-Tronco Pluripotentes , Humanos , Distrofina/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Sistemas CRISPR-Cas , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Distrofia Muscular de Duchenne/patologia , Músculo Esquelético/patologia , Fibras Musculares Esqueléticas/patologia , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/patologia
13.
J Biol Chem ; 285(36): 27817-26, 2010 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-20562107

RESUMO

In atrial and nodal cardiac myocytes, M2 muscarinic receptors activate inhibitory G-proteins (G(i/o)), which in turn stimulate G-protein-gated inwardly rectifying K(+) channels through direct binding of the Gbetagamma subunit. Despite also releasing Gbetagamma, G(s)-coupled receptors such as the beta-adrenergic receptor are not able to prominently activate this current. An appealing hypothesis would be if components were sequestered in membrane domains such as caveolae/rafts. Using biochemical fractionation followed by Western blotting and/or radioligand binding experiments, we examined the distribution of the components in stable HEK293 and HL-1 cells, which natively express the transduction cascade. The channel, M2 muscarinic, and A1 adenosine receptors were located in noncaveolar/nonraft fractions. G(i)alpha(1/2) was enriched in both caveolar/raft and noncaveolar/nonraft fractions. In contrast, G(s)alpha was only enriched in caveolar/raft fractions. We constructed YFP-tagged caveolin-2 (YFP-Cav2) and chimeras with the M2 (M2-YFP-Cav2) and A1 (A1-YFP-Cav2) receptors. Analysis of gradient fractions showed that these receptor chimeras were now localized to caveolae-enriched fractions. Microscopy showed that M2-YFP and A1-YFP had a diffuse homogenous membrane signal. YFP-Cav2, M2-YFP-Cav2, and A1-YFP-Cav2 revealed a more punctuate pattern. Finally, we looked at the consequences for signaling. Activation via M2-YFP-Cav2 or A1-YFP-Cav2 revealed substantially slower kinetics compared with M2-YFP or A1-YFP and was reversed by the addition of methyl-beta-cyclodextrin. Thus the localization of the channel signal transduction cascade in non-cholesterol rich domains substantially enhances the speed of signaling. The presence of G(s)alpha solely in caveolae may account for signaling selectivity between G(i/o) and G(s)-coupled receptors.


Assuntos
Cavéolas/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Ativação do Canal Iônico , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Caveolina 2/metabolismo , Linhagem Celular , Colesterol/metabolismo , Humanos , Microdomínios da Membrana/metabolismo , Camundongos , Subunidades Proteicas/metabolismo , Transdução de Sinais , Solubilidade , Frações Subcelulares/química , Frações Subcelulares/metabolismo
14.
J Biol Chem ; 285(6): 3664-3675, 2010 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-19940153

RESUMO

We have directly observed the trafficking and fusion of ion channel containing vesicles and monitored the release of individual ion channels at the plasma membrane of live mammalian cells using total internal reflection fluorescence microscopy. Proteins were fused in-frame with green or red fluorescent proteins and expressed at low level in HL-1 and HEK293 cells. Dual color imaging revealed that vesicle trafficking involved motorized movement along microtubules followed by stalling, fusion, and subsequent release of individual ion channels at the plasma membrane. We found that KCNQ1-KCNE1 complexes were released in batches of about 5 molecules per vesicle. To elucidate the properties of ion channel complexes at the cell membrane we tracked the movement of individual molecules and compared the diffusive behavior of two types of potassium channel complex (KCNQ1-KCNE1 and Kir6.2-SUR2A) to that of a G-protein coupled receptor, the A1 adenosine receptor. Plots of mean squared displacement against time intervals showed that mobility depended on channel type, cell type, and temperature. Analysis of the mobility of wild type KCNQ1-KCNE1 complexes showed the existence of a significant immobile subpopulation and also a significant number of molecules that demonstrated periodic stalling of diffusive movements. This behavior was enhanced in cells treated with jasplakinolide and was abrogated in a C-terminal truncated form (KCNQ1(R518X)-KCNE1) of the protein. This mutant has been identified in patients with the long QT syndrome. We propose that KCNQ1-KCNE1 complexes interact intermittently with the actin cytoskeleton via the C-terminal region and this interaction may have a functional role.


Assuntos
Membrana Celular/metabolismo , Canal de Potássio KCNQ1/fisiologia , Proteínas Recombinantes de Fusão/fisiologia , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/fisiologia , Animais , Linhagem Celular , Membrana Celular/fisiologia , Depsipeptídeos/farmacologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ1/metabolismo , Cinética , Potenciais da Membrana/fisiologia , Microscopia Confocal , Microscopia de Fluorescência , Mutação , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Técnicas de Patch-Clamp , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Ligação Proteica , Multimerização Proteica , Transporte Proteico/efeitos dos fármacos , Receptores de Droga/genética , Receptores de Droga/metabolismo , Receptores de Droga/fisiologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Receptores de Sulfonilureias , Transfecção
15.
Bioconjug Chem ; 22(2): 132-6, 2011 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-21271715

RESUMO

The introduction of non-natural entities into proteins by chemical modification has numerous applications in fundamental biological science and for the development and manipulation of peptide and protein therapeutics. The reduction of native disulfide bonds provides a convenient method to access two nucleophilic cysteine residues that can serve as ideal attachment points for such chemical modification. The optimum bioconjugation strategy utilizing these cysteine residues should include the reconstruction of a bridge to mimic the role of the disulfide bond, maintaining structure and stability of the protein. Furthermore, the bridging chemical modification should be as rapid as possible to prevent problems associated with protein unfolding, aggregation, or disulfide scrambling. This study reports on an in situ disulfide reduction-bridging strategy that ensures rapid sequestration of the free cysteine residues in a bridge, using dithiomaleimides. This approach is then used to PEGylate the peptide hormone somatostatin and retention of biological activity is demonstrated.


Assuntos
Dissulfetos/química , Maleimidas/química , Polietilenoglicóis/química , Somatostatina/química , Linhagem Celular , Humanos , Estrutura Molecular , Polietilenoglicóis/síntese química , Receptores de Somatostatina/química , Receptores de Somatostatina/metabolismo , Somatostatina/análogos & derivados , Somatostatina/síntese química
16.
Pflugers Arch ; 460(1): 99-108, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20186548

RESUMO

An inwardly rectifying K(+) current is present in atrial cardiac myocytes that is activated by acetylcholine (I(KACh)). Physiologically, activation of the current in the SA node is important in slowing the heart rate with increased parasympathetic tone. It is a paradigm for the direct regulation of signaling effectors by the Gbetagamma G-protein subunit. Many questions have been addressed in heterologous expression systems with less focus on the behaviour in native myocytes partly because of the technical difficulties in undertaking comparable studies in native cells. In this study, we characterise a potassium current in the atrial-derived cell line HL-1. Using an electrophysiological approach, we compare the characteristics of the potassium current with those in native atrial cells and in a HEK cell line expressing the cloned Kir3.1/3.4 channel. The potassium current recorded in HL-1 is inwardly rectifying and activated by the muscarinic agonist carbachol. Carbachol-activated currents were inhibited by pertussis toxin and tertiapin-Q. The basal current was time-dependently increased when GTP was substituted in the patch-clamp pipette by the non-hydrolysable analogue GTPgammaS. We compared the kinetics of current modulation in HL-1 with those of freshly isolated atrial mouse cardiomyocytes. The current activation and deactivation kinetics in HL-1 cells are comparable to those measured in atrial cardiomyocytes. Using immunofluorescence, we found GIRK4 at the membrane in HL-1 cells. Real-time RT-PCR confirms the presence of mRNA for the main G-protein subunits, as well as for M2 muscarinic and A1 adenosine receptors. The data suggest HL-1 cells are a good model to study IKAch.


Assuntos
Acetilcolina/metabolismo , Ativação do Canal Iônico , Miócitos Cardíacos/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Potássio/metabolismo , Receptor Muscarínico M2/metabolismo , Animais , Venenos de Abelha/farmacologia , Carbacol/farmacologia , Linhagem Celular , Agonistas Colinérgicos/farmacologia , Feminino , Imunofluorescência , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Átrios do Coração/metabolismo , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Cinética , Masculino , Camundongos , Miócitos Cardíacos/efeitos dos fármacos , Técnicas de Patch-Clamp , Toxina Pertussis/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização/antagonistas & inibidores , Canais de Potássio Corretores do Fluxo de Internalização/genética , RNA Mensageiro/metabolismo , Receptor Muscarínico M2/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
17.
Nature ; 427(6977): 853-8, 2004 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-14985765

RESUMO

Neutrophil leukocytes have a pivotal function in innate immunity. Dogma dictates that the lethal blow is delivered to microbes by reactive oxygen species (ROS) and halogens, products of the NADPH oxidase, whose impairment causes immunodeficiency. However, recent evidence indicates that the microbes might be killed by proteases, activated by the oxidase through the generation of a hypertonic, K+-rich and alkaline environment in the phagocytic vacuole. Here we show that K+ crosses the membrane through large-conductance Ca2+-activated K+ (BK(Ca)) channels. Specific inhibitors of these channels, iberiotoxin and paxilline, blocked oxidase-induced 86Rb+ fluxes and alkalinization of the phagocytic vacuole, whereas NS1619, a BK(Ca) channel opener, enhanced both. Characteristic outwardly rectifying K+ currents, reversibly inhibited by iberiotoxin, were demonstrated in neutrophils and eosinophils and the expression of the alpha-subunit of the BK channel was confirmed by western blotting. The channels were opened by the combination of membrane depolarization and elevated Ca2+ concentration, both consequences of oxidase activity. Remarkably, microbial killing and digestion were abolished when the BK(Ca) channel was blocked, revealing an essential and unexpected function for this K+ channel in the microbicidal process.


Assuntos
Imunidade Inata , Canais de Potássio Cálcio-Ativados/metabolismo , Cálcio/metabolismo , Condutividade Elétrica , Eosinófilos/citologia , Eosinófilos/efeitos dos fármacos , Eosinófilos/imunologia , Eosinófilos/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Imunidade Inata/efeitos dos fármacos , Transporte de Íons/efeitos dos fármacos , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta , Canais de Potássio Ativados por Cálcio de Condutância Alta , Potenciais da Membrana/efeitos dos fármacos , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Neutrófilos/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Fagocitose/efeitos dos fármacos , Potássio/metabolismo , Canais de Potássio Cálcio-Ativados/antagonistas & inibidores , Radioisótopos de Rubídio , Staphylococcus aureus/imunologia , Acetato de Tetradecanoilforbol/farmacologia , Vacúolos/efeitos dos fármacos , Vacúolos/metabolismo
18.
Bio Protoc ; 10(1): e3477, 2020 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-33654710

RESUMO

The cardiac conduction system allows the synchronized propagation of electrical activity through heart muscle. This is initiated by the spontaneous activity of the specialized pacemaker cells of the sino-atrial node (SAN). The SAN region underlies automaticity in mammals and therefore has a crucial role in the pathogenesis of cardiac disorders such as arrhythmia. Isolation of SAN tissue and SAN cells is critical to advance our understanding of SAN structure and function in health and disease. Initially, isolation of SAN tissue and SAN cells was carried out in the rabbit owing to its larger size and similar electrical properties to human. This protocol was optimized by Mangoni and Nargeot (2001) for use in mice to take advantage of advancements in transgenic models. Here, we provide a step-by-step guide to dissecting the SAN tissue and isolating pacemaker cardiomyocytes from mouse hearts using an enzyme digestion approach.

19.
Bio Protoc ; 10(1): e3478, 2020 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-33654711

RESUMO

Cardiac pacemaker cells of the sino-atrial node are responsible for the initiation of the heart beat and express an array of ion channels. The patch-clamp technique is the gold standard method for investigating the function of ion channels expressed in electrically active cells. Conventional whole-cell and perforated patch-clamp techniques can be used to investigate ionic currents in the voltage-clamp mode and changes in membrane potential (e.g., action potential) in the current-clamp mode. Here, we provide details of protocols used to measure spontaneous and triggered action potentials and whole-cell funny current If (HCN4) in single cardiomyocytes isolated from the mouse sino-atrial node (SAN).

20.
Cardiovasc Res ; 74(3): 515-25, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17391657

RESUMO

OBJECTIVE: C-type natriuretic peptide (CNP) has recently been suggested to represent an endothelium-derived hyperpolarising factor (EDHF) in the mammalian resistance vasculature and, as such, important in the regulation of local blood flow and systemic blood pressure. Additionally, this peptide has been shown to protect against ischaemia-reperfusion injury and inhibits leukocyte and platelet activation. Herein, we use a novel, selective natriuretic peptide receptor-C (NPR-C) antagonist (M372049) to highlight the pivotal contribution of CNP/NPR-C signalling in the EDHF-dependent regulation of vascular tone and investigate the mechanism(s) underlying the release and biological activity of CNP. METHODS: In vitro pharmacological investigation was conducted in rat (Sprague-Dawley) aorta and mesenteric resistance arteries. Relaxant responses to CNP, atrial natriuretic peptide (ANP), the nitric oxide donor spermine-NONOate (SPER-NO) and the endothelium-dependent vasodilator, acetylcholine (ACh) were examined in the absence and presence of M372049 or inhibitor cocktails shown previously to block endothelium-dependent dilatation in the resistance vasculature. RT-PCR was employed to characterize the expression of NPR subtypes in the vessels studied. RESULTS: M372049 produced concentration-dependent inhibition of the vasorelaxant activity of CNP in rat isolated mesenteric resistance arteries but not aorta; in contrast, M372049 did not affect relaxations to ANP or SPER-NO in either vessel. M372049 or ouabain alone produced small, significant inhibition of EDHF-dependent relaxations in mesenteric arteries and in combination acted synergistically to abolish such responses. A combination of M372049 with established inhibitors of EDHF-dependent relaxation revealed that multiple, distinct pathways coordinate the bioactivity of EDHF in the resistance vasculature, and that CNP/NPR-C signalling represents a major component. CONCLUSIONS: These data substantiate CNP/NPR-C signalling as a fundamental pathway underlying EDHF-dependent regulation of vascular tone in the rat mesenteric resistance vasculature. An increased understanding of the physiological roles of CNP/NPR-C signalling in the vasculature (now facilitated by the identification of a selective NPR-C antagonist) should aid determination of the (patho)physiological importance of EDHF and might provide the rationale for the design of novel therapeutics.


Assuntos
Fatores Biológicos/metabolismo , Peptídeo Natriurético Tipo C/farmacologia , Oligopeptídeos/farmacologia , Quinoxalinas/farmacologia , Receptores do Fator Natriurético Atrial/metabolismo , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Acetilcolina/farmacologia , Animais , Aorta , Fator Natriurético Atrial/farmacologia , Bário/farmacologia , Técnicas In Vitro , Masculino , Potenciais da Membrana/efeitos dos fármacos , Artérias Mesentéricas , Ouabaína/farmacologia , Técnicas de Patch-Clamp , Canais de Potássio/efeitos dos fármacos , Pirazóis/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores do Fator Natriurético Atrial/antagonistas & inibidores , Reação em Cadeia da Polimerase Via Transcriptase Reversa , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , Espermina/análogos & derivados , Espermina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA