Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Protein Sci ; 33(6): e4995, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38747377

RESUMO

Membrane proteins play critical physiological roles as receptors, channels, pumps, and transporters. Despite their importance, however, low expression levels often hamper the experimental characterization of membrane proteins. We present an automated and web-accessible design algorithm called mPROSS (https://mPROSS.weizmann.ac.il), which uses phylogenetic analysis and an atomistic potential, including an empirical lipophilicity scale, to improve native-state energy. As a stringent test, we apply mPROSS to the Kv1.2-Kv2.1 paddle chimera voltage-gated potassium channel. Four designs, encoding 9-26 mutations relative to the parental channel, were functional and maintained potassium-selective permeation and voltage dependence in Xenopus oocytes with up to 14-fold increase in whole-cell current densities. Additionally, single-channel recordings reveal no significant change in the channel-opening probability nor in unitary conductance, indicating that functional expression levels increase without impacting the activity profile of individual channels. Our results suggest that the expression levels of other dynamic channels and receptors may be enhanced through one-shot design calculations.


Assuntos
Xenopus laevis , Animais , Algoritmos , Canal de Potássio Kv1.2/genética , Canal de Potássio Kv1.2/metabolismo , Canal de Potássio Kv1.2/química , Oócitos/metabolismo , Filogenia , Canais de Potássio Shab/metabolismo , Canais de Potássio Shab/genética , Canais de Potássio Shab/química , Mutação , Xenopus
2.
PNAS Nexus ; 2(3): pgad068, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37007714

RESUMO

Store-operated calcium entry (SOCE) is a vital process aimed at refilling cellular internal Ca2+ stores and a primary cellular signaling driver for transcription factors' entry to the nucleus. SOCE-associated regulatory factor (SARAF)/TMEM66 is an endoplasmic reticulum (ER)-resident transmembrane protein that promotes SOCE inactivation and prevents Ca2+ overfilling of the cell. Here, we demonstrate that mice deficient in SARAF develop age-dependent sarcopenic obesity with decreased energy expenditure, lean mass, and locomotion without affecting food consumption. Moreover, SARAF ablation reduces hippocampal proliferation, modulates the activity of the hypothalamus-pituitary-adrenal (HPA) axis, and mediates changes in anxiety-related behaviors. Interestingly, selective SARAF ablation in the hypothalamus's paraventricular nucleus (PVN) neurons reduces old age-induced obesity and preserves locomotor activity, lean mass, and energy expenditure, suggesting a possible central control with a site-specific role for SARAF. At the cellular level, SARAF ablation in hepatocytes leads to elevated SOCE, elevated vasopressin-induced Ca2+ oscillations, and an increased mitochondrial spare respiratory capacity (SPC), thus providing insights into the cellular mechanisms that may affect the global phenotypes. These effects may be mediated via the liver X receptor (LXR) and IL-1 signaling metabolic regulators explicitly altered in SARAF ablated cells. In short, our work supports both central and peripheral roles of SARAF in regulating metabolic, behavioral, and cellular responses.

3.
Gigascience ; 112022 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-35640874

RESUMO

Venoms have evolved >100 times in all major animal groups, and their components, known as toxins, have been fine-tuned over millions of years into highly effective biochemical weapons. There are many outstanding questions on the evolution of toxin arsenals, such as how venom genes originate, how venom contributes to the fitness of venomous species, and which modifications at the genomic, transcriptomic, and protein level drive their evolution. These questions have received particularly little attention outside of snakes, cone snails, spiders, and scorpions. Venom compounds have further become a source of inspiration for translational research using their diverse bioactivities for various applications. We highlight here recent advances and new strategies in modern venomics and discuss how recent technological innovations and multi-omic methods dramatically improve research on venomous animals. The study of genomes and their modifications through CRISPR and knockdown technologies will increase our understanding of how toxins evolve and which functions they have in the different ontogenetic stages during the development of venomous animals. Mass spectrometry imaging combined with spatial transcriptomics, in situ hybridization techniques, and modern computer tomography gives us further insights into the spatial distribution of toxins in the venom system and the function of the venom apparatus. All these evolutionary and biological insights contribute to more efficiently identify venom compounds, which can then be synthesized or produced in adapted expression systems to test their bioactivity. Finally, we critically discuss recent agrochemical, pharmaceutical, therapeutic, and diagnostic (so-called translational) aspects of venoms from which humans benefit.


Assuntos
Proteômica , Peçonhas , Animais , Pesquisa , Serpentes/genética , Transcriptoma , Peçonhas/química , Peçonhas/genética
4.
Methods Enzymol ; 654: 169-201, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34120712

RESUMO

Animal venom is a rich source for peptide toxins that bind and modulate the function of ion channels. Owing to their ability to bind receptor sites on the channel protein with high affinity and specificity, peptide neurotoxins have become an indispensable tool for ion channel research. Recent breakthroughs in structural biology and advances in computer simulations of biomolecules have sparked a new interest in animal toxins as probes of channel protein structure and function. Here, we focus on methods used to produce animal toxins for research purposes using recombinant expression. The specific challenges associated with heterologous production of venom peptides are discussed, and several methods targeting these issues are presented with an emphasis on E. coli based systems. An efficient protocol for the bacterial expression, folding, and purification of recombinant venom peptides is described.


Assuntos
Escherichia coli , Peçonhas , Animais , Escherichia coli/genética , Escherichia coli/metabolismo , Canais Iônicos/genética , Peptídeos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
5.
J Gen Physiol ; 153(6)2021 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-34014250

RESUMO

It has been reported earlier that the slow (C-type) inactivated conformation in Kv channels is stabilized by a multipoint hydrogen-bond network behind the selectivity filter. Furthermore, MD simulations revealed that structural water molecules are also involved in the formation of this network locking the selectivity filter in its inactive conformation. We found that the application of an extracellular, but not intracellular, solution based on heavy water (D2O) dramatically slowed entry into the slow inactivated state in Shaker-IR mutants (T449A, T449A/I470A, and T449K/I470C, displaying a wide range of inactivation kinetics), consistent with the proposed effect of the dynamics of structural water molecules on the conformational stability of the selectivity filter. Alternative hypotheses capable of explaining the observed effects of D2O were examined. Increased viscosity of the external solution mimicked by the addition of glycerol had a negligible effect on the rate of inactivation. In addition, the inactivation time constants of K+ currents in the outward and the inward directions in asymmetric solutions were not affected by a H2O/D2O exchange, negating an indirect effect of D2O on the rate of K+ rehydration. The elimination of the nonspecific effects of D2O on our macroscopic current measurements supports the hypothesis that the rate of structural water exchange at the region behind the selectivity filter determines the rate of slow inactivation, as proposed by molecular modeling.


Assuntos
Ativação do Canal Iônico , Água , Óxido de Deutério , Ligação de Hidrogênio , Cinética
6.
J Mol Biol ; 433(17): 166957, 2021 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-33771569

RESUMO

Many venomous organisms carry in their arsenal short polypeptides that block K+ channels in a highly selective manner. These toxins may compete with the permeating ions directly via a "plug" mechanism or indirectly via a "pore-collapse" mechanism. An alternative "lid" mechanism was proposed but remained poorly defined. Here we study the Drosophila Shaker channel block by Conkunitzin-S1 and Conkunitzin-C3, two highly similar toxins derived from cone venom. Despite their similarity, the two peptides exhibited differences in their binding poses and biophysical assays, implying discrete action modes. We show that while Conkunitzin-S1 binds tightly to the channel turret and acts via a "pore-collapse" mechanism, Conkunitzin-C3 does not contact this region. Instead, Conk-C3 uses a non-conserved Arg to divert the permeant ions and trap them in off-axis cryptic sites above the SF, a mechanism we term a "molecular-lid". Our study provides an atomic description of the "lid" K+ blocking mode and offers valuable insights for the design of therapeutics based on venom peptides.


Assuntos
Ativação do Canal Iônico/efeitos dos fármacos , Peptídeos/farmacologia , Canais de Potássio/metabolismo , Potássio/metabolismo , Venenos de Escorpião/farmacologia , Sequência de Aminoácidos , Animais , Sítios de Ligação/efeitos dos fármacos , Biofísica/métodos , Xenopus laevis/metabolismo
7.
Cell ; 184(2): 299-301, 2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33482096

RESUMO

Ion channels can tailor their activity to the particular cellular context by incorporating auxiliary subunits that are channel-type specific. In this issue of Cell, Ávalos Prado et al. now find that a well-characterized voltage-gated K+ channel auxiliary subunit can also modulate the gating of Ca2+-activated Cl- channels.


Assuntos
Canais Iônicos , Humanos
8.
Chembiochem ; 22(5): 894-903, 2021 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-33105515

RESUMO

Quinone methide (QM) chemistry is widely applied including in enzyme inhibitors. Typically, enzyme-mediated bond breaking releases a phenol product that rearranges into an electrophilic QM that in turn covalently modifies protein side chains. However, the factors that govern the reactivity of QM-based inhibitors and their mode of inhibition have not been systematically explored. Foremost, enzyme inactivation might occur in cis, whereby a QM molecule inactivates the very same enzyme molecule that released it, or by trans if the released QMs diffuse away and inactivate other enzyme molecules. We examined QM-based inhibitors for enzymes exhibiting phosphoester hydrolase activity. We tested different phenolic substituents and benzylic leaving groups, thereby modulating the rates of enzymatic hydrolysis, phenolate-to-QM rearrangement, and the electrophilicity of the resulting QM. By developing assays that distinguish between cis and trans inhibition, we have identified certain combinations of leaving groups and phenyl substituents that lead to inhibition in the cis mode, while other combinations gave trans inhibition. Our results suggest that cis-acting QM-based substrates could be used as activity-based probes to identify various phospho- and phosphono-ester hydrolases, and potentially other hydrolases.


Assuntos
Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Indolquinonas/química , Indolquinonas/farmacologia , Monoéster Fosfórico Hidrolases/antagonistas & inibidores , Hidrólise , Organofosfatos/metabolismo
9.
J Physiol ; 599(2): 521-545, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33124684

RESUMO

KEY POINTS: G-protein inwardly rectifying K+ (GIRK) channels consist of four homologous subunits (GIRK1-4) and are essential regulators of electrical excitability in the nervous system. GIRK2-null mice have been widely investigated for their distinct behaviour and altered depotentiation following long-term potentiation (LTP), whereas GIRK1 mice are less well characterized. Here we utilize a novel knockin mouse strain in which the GIRK1 subunit is fluorescently tagged with yellow fluorescent protein (YFP-GIRK1) and the GIRK1-null mouse line to investigate the role of GIRK1 in neuronal processes such as spatial learning and memory, locomotion and depotentiation following LTP. Neurons dissected from YFP-GIRK1 mice had significantly reduced potassium currents and this mouse line phenotypically resembled GIRK1-null mice, making it a 'functional knockdown' model of GIRK1-containing channels. YFP-GIRK1 and GIRK1-null mice had increased locomotion, reduced spatial learning and memory and blunted depotentiation following LTP. ABSTRACT: GIRK channels are essential for the slow inhibition of electrical activity in the nervous system and heart rate regulation via the parasympathetic system. The implications of individual GIRK isoforms in specific physiological activities are based primarily on studies conducted with GIRK-null mouse lines. Here we utilize a novel knockin mouse line in which YFP was fused in-frame to the N-terminus of GIRK1 (YFP-GIRK1) to correlate GIRK1 spatial distribution with physiological activities. These mice, however, displayed spontaneous seizure-like activity and thus were investigated for the origin of such activity. We show that GIRK tetramers containing YFP-GIRK1 are correctly assembled and trafficked to the plasma membrane, but are functionally impaired. A battery of behavioural assays conducted on YFP-GIRK1 and GIRK1-null (GIRK1-/- ) mice revealed similar phenotypes, including impaired nociception, reduced anxiety and hyperactivity in an unfamiliar environment. However, YFP-GIRK1 mice exhibited increased home-cage locomotion while GIRK1-/- mice did not. In addition, we show that the GIRK1 subunit is essential for intact spatial learning and memory and synaptic plasticity in hippocampal brain slices. This study expands our knowledge regarding the role of GIRK1 in neuronal processes and underlines the importance of GIRK1-containing heterotetramers.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Aprendizagem Espacial , Animais , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Hipocampo/metabolismo , Camundongos , Plasticidade Neuronal , Neurônios/metabolismo
10.
Proc Natl Acad Sci U S A ; 116(37): 18700-18709, 2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31444298

RESUMO

Voltage-dependent potassium channels (Kvs) gate in response to changes in electrical membrane potential by coupling a voltage-sensing module with a K+-selective pore. Animal toxins targeting Kvs are classified as pore blockers, which physically plug the ion conduction pathway, or as gating modifiers, which disrupt voltage sensor movements. A third group of toxins blocks K+ conduction by an unknown mechanism via binding to the channel turrets. Here, we show that Conkunitzin-S1 (Cs1), a peptide toxin isolated from cone snail venom, binds at the turrets of Kv1.2 and targets a network of hydrogen bonds that govern water access to the peripheral cavities that surround the central pore. The resulting ectopic water flow triggers an asymmetric collapse of the pore by a process resembling that of inherent slow inactivation. Pore modulation by animal toxins exposes the peripheral cavity of K+ channels as a novel pharmacological target and provides a rational framework for drug design.


Assuntos
Membrana Celular/efeitos dos fármacos , Proteínas de Drosophila/antagonistas & inibidores , Ativação do Canal Iônico/efeitos dos fármacos , Canal de Potássio Kv1.2/antagonistas & inibidores , Venenos de Moluscos/toxicidade , Superfamília Shaker de Canais de Potássio/antagonistas & inibidores , Animais , Membrana Celular/metabolismo , Cristalografia por Raios X , Proteínas de Drosophila/genética , Proteínas de Drosophila/isolamento & purificação , Proteínas de Drosophila/metabolismo , Desenho de Fármacos , Feminino , Ligação de Hidrogênio/efeitos dos fármacos , Canal de Potássio Kv1.2/genética , Canal de Potássio Kv1.2/isolamento & purificação , Canal de Potássio Kv1.2/metabolismo , Dose Letal Mediana , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Venenos de Moluscos/química , Mutação , Oócitos , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Superfamília Shaker de Canais de Potássio/genética , Superfamília Shaker de Canais de Potássio/isolamento & purificação , Superfamília Shaker de Canais de Potássio/metabolismo , Água/química , Água/metabolismo , Xenopus laevis
11.
Cell ; 178(4): 776-778, 2019 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-31398335

RESUMO

Voltage sensing by ion channels is the key event enabling the generation and propagation of electrical activity in excitable cells. In this issue of Cell, Wisedchaisri et al. provide a structural view of a voltage-gated sodium channel in its resting closed conformation.


Assuntos
Canais Iônicos , Sódio , Conformação Molecular , Canais de Sódio Disparados por Voltagem
12.
Chemphyschem ; 20(14): 1860-1868, 2019 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-31054266

RESUMO

It is an open question whether the conformations of proteins sampled in dilute solutions are the same as in the cellular environment. Here we address this question by double electron-electron resonance (DEER) distance measurements with Gd(III) spin labels to probe the conformations of calmodulin (CaM) in vitro, in cell extract, and in human HeLa cells. Using the CaM mutants N53C/T110C and T34C/T117C labeled with maleimide-DOTA-Gd(III) in the N- and C-terminal domains, we observed broad and varied interdomain distance distributions. The in vitro distance distributions of apo-CaM and holo-CaM in the presence and absence of the IQ target peptide can be described by combinations of closed, open, and collapsed conformations. In cell extract, apo- and holo-CaM bind to target proteins in a similar way as apo- and holo-CaM bind to IQ peptide in vitro. In HeLa cells, however, in the presence or absence of elevated in-cell Ca2+ levels CaM unexpectedly produced more open conformations and very broad distance distributions indicative of many different interactions with in-cell components. These results show-case the importance of in-cell analyses of protein structures.


Assuntos
Calmodulina/química , Calmodulina/metabolismo , Calmodulina/genética , Extratos Celulares/química , Espectroscopia de Ressonância de Spin Eletrônica/métodos , Gadolínio/química , Células HeLa , Humanos , Mutação , Conformação Proteica , Marcadores de Spin
13.
J Mol Biol ; 431(15): 2869-2883, 2019 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-31082439

RESUMO

Store-Operated Calcium Entry (SOCE) plays key roles in cell proliferation, muscle contraction, immune responses, and memory formation. The coordinated interactions of a number of proteins from the plasma and endoplasmic reticulum membranes control SOCE to replenish internal Ca2+ stores and generate intracellular Ca2+ signals. SARAF, an endoplasmic reticulum resident component of the SOCE pathway having no homology to any characterized protein, serves as an important brake on SOCE. Here, we describe the X-ray crystal structure of the SARAF luminal domain, SARAFL. This domain forms a novel 10-stranded ß-sandwich fold that includes a set of three conserved disulfide bonds, denoted the "SARAF-fold." The structure reveals a domain-swapped dimer in which the last two ß-strands (ß9 and ß10) are exchanged forming a region denoted the "SARAF luminal switch" that is essential for dimerization. Sequence comparisons reveal that the SARAF-fold is highly conserved in vertebrates and in a variety of pathologic fungi. Förster resonance energy transfer experiments using full-length SARAF validate the formation of the domain-swapped dimer in cells and demonstrate that dimerization is reversible. A designed variant lacking the SARAF luminal switch shows that the domain swapping is essential to function and indicates that the SARAF dimer accelerates SOCE inactivation.


Assuntos
Cálcio/metabolismo , Proteínas Sensoras de Cálcio Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Sinalização do Cálcio , Cristalografia por Raios X , Células HEK293 , Humanos , Proteínas Sensoras de Cálcio Intracelular/química , Proteínas de Membrana/química , Modelos Moleculares , Conformação Proteica em Folha beta , Domínios Proteicos , Dobramento de Proteína , Multimerização Proteica
14.
Br J Pharmacol ; 176(17): 3161-3179, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31116876

RESUMO

BACKGROUND AND PURPOSE: A second-generation antihistamine, terfenadine, is known to induce arrhythmia by blocking hERG channels. In this study, we have shown that terfenadine also inhibits the activity of G-protein-gated inwardly rectifying K+ (GIRK) channels, which regulate the excitability of neurons and cardiomyocytes. To clarify the underlying mechanism(s), we examined the effects of several antihistamines on GIRK channels and identified the structural determinant for the inhibition. EXPERIMENTAL APPROACH: Electrophysiological recordings were made in Xenopus oocytes and rat atrial myocytes to analyse the effects of antihistamines on various GIRK subunits (Kir 3.x). Mutagenesis analyses identified the residues critical for inhibition by terfenadine and the regulation of ion selectivity. The potential docking site of terfenadine was analysed by molecular docking. KEY RESULTS: GIRK channels containing Kir 3.1 subunits heterologously expressed in oocytes and native GIRK channels in atrial myocytes were inhibited by terfenadine and other non-sedating antihistamines. In Kir 3.1 subunits, mutation of Phe137, located in the centre of the pore helix, to the corresponding Ser in Kir 3.2 subunits reduced the inhibition by terfenadine. Introduction of an amino acid with a large side chain in Kir 3.2 subunits at Ser148 increased the inhibition. When this residue was mutated to a non-polar amino acid, the channel became permeable to Na+ . Phosphoinositide-mediated activity was also decreased by terfenadine. CONCLUSION AND IMPLICATIONS: The Phe137 residue in Kir 3.1 subunits is critical for inhibition by terfenadine. This study provides novel insights into the regulation of GIRK channels by the pore helix and information for drug design.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/antagonistas & inibidores , Antagonistas dos Receptores Histamínicos/farmacologia , Animais , Relação Dose-Resposta a Droga , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Antagonistas dos Receptores Histamínicos/química , Masculino , Simulação de Acoplamento Molecular , Mutação , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Ratos , Ratos Wistar , Relação Estrutura-Atividade , Xenopus laevis
15.
Cell Rep ; 25(11): 3169-3179.e7, 2018 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-30540948

RESUMO

Importins mediate transport from synapse to soma and from cytoplasm to nucleus, suggesting that perturbation of importin-dependent pathways should have significant neuronal consequences. A behavioral screen on five importin α knockout lines revealed that reduced expression of importin α5 (KPNA1) in hippocampal neurons specifically decreases anxiety in mice. Re-expression of importin α5 in ventral hippocampus of knockout animals increased anxiety behaviors to wild-type levels. Hippocampal neurons lacking importin α5 reveal changes in presynaptic plasticity and modified expression of MeCP2-regulated genes, including sphingosine kinase 1 (Sphk1). Knockout of importin α5, but not importin α3 or α4, reduces MeCP2 nuclear localization in hippocampal neurons. A Sphk1 blocker reverses anxiolysis in the importin α5 knockout mouse, while pharmacological activation of sphingosine signaling has robust anxiolytic effects in wild-type animals. Thus, importin α5 influences sphingosine-sensitive anxiety pathways by regulating MeCP2 nuclear import in hippocampal neurons.


Assuntos
Ansiedade/metabolismo , Proteína 2 de Ligação a Metil-CpG/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , alfa Carioferinas/metabolismo , Animais , Ansiolíticos/farmacologia , Comportamento Animal , Carbolinas/farmacologia , Hipocampo/patologia , Camundongos Knockout , Neurônios/metabolismo , Fenótipo , Sinapses/metabolismo , Transcrição Gênica , alfa Carioferinas/deficiência
16.
ACS Chem Neurosci ; 5(9): 812-22, 2014 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-25028803

RESUMO

Use of randomized peptide libraries to evolve molecules with new functions provides a means for developing novel regulators of protein activity. Despite the demonstrated power of such approaches for soluble targets, application of this strategy to membrane systems, such as ion channels, remains challenging. Here, we have combined libraries of a tethered protein scaffold with functional selection in yeast to develop a novel activator of the G-protein-coupled mammalian inwardly rectifying potassium channel Kir3.2 (GIRK2). We show that the novel regulator, denoted N5, increases Kir3.2 (GIRK2) basal activity by inhibiting clearance of the channel from the cellular surface rather than affecting the core biophysical properties of the channel. These studies establish the tethered protein display strategy as a means to create new channel modulators and highlight the power of approaches that couple randomized libraries with direct selections for functional effects. Our results further underscore the possibility for the development of modulators that influence channel function by altering cell surface expression densities rather than by direct action on channel biophysical parameters. The use of tethered library selection strategies coupled with functional selection bypasses the need for a purified target and is likely to be applicable to a range of membrane protein systems.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Animais , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética , Regulação da Expressão Gênica , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/genética , Camundongos , Microinjeções , Oócitos , Técnicas de Patch-Clamp , Biblioteca de Peptídeos , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Transporte Proteico , Xenopus laevis
18.
Cell ; 149(2): 425-38, 2012 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-22464749

RESUMO

Store operated calcium entry (SOCE) is a principal cellular process by which cells regulate basal calcium, refill intracellular Ca(2+) stores, and execute a wide range of specialized activities. STIM and Orai proteins have been identified as the essential components enabling the reconstitution of Ca(2+) release-activated Ca(2+) (CRAC) channels that mediate SOCE. Here, we report the molecular identification of SARAF as a negative regulator of SOCE. Using heterologous expression, RNAi-mediated silencing and site directed mutagenesis combined with electrophysiological, biochemical and imaging techniques we show that SARAF is an endoplasmic reticulum membrane resident protein that associates with STIM to facilitate slow Ca(2+)-dependent inactivation of SOCE. SARAF plays a key role in shaping cytosolic Ca(2+) signals and determining the content of the major intracellular Ca(2+) stores, a role that is likely to be important in protecting cells from Ca(2+) overfilling.


Assuntos
Cálcio/metabolismo , Proteínas de Membrana/metabolismo , Sinalização do Cálcio , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Citosol/metabolismo , Citometria de Fluxo , Humanos , Proteínas Sensoras de Cálcio Intracelular , Proteínas de Membrana/genética , Dados de Sequência Molecular , Proteínas de Neoplasias/metabolismo , Molécula 1 de Interação Estromal , Molécula 2 de Interação Estromal
19.
Proc Natl Acad Sci U S A ; 109(7): 2642-7, 2012 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-22308328

RESUMO

G protein-activated inwardly rectifying K+ channels (GIRK) generate slow inhibitory postsynaptic potentials in the brain via G(i/o) protein-coupled receptors. GIRK2, a GIRK subunit, is widely abundant in the brain and has been implicated in various functions and pathologies, such as learning and memory, reward, motor coordination, and Down syndrome. Down syndrome, the most prevalent cause of mental retardation, results from the presence of an extra maternal chromosome 21 (trisomy 21), which comprises the Kcnj6 gene (GIRK2). The present study examined the behaviors and cellular physiology properties in mice harboring a single trisomy of the Kcnj6 gene. Kcnj6 triploid mice exhibit deficits in hippocampal-dependent learning and memory, altered responses to rewards, hampered depotentiation, a form of excitatory synaptic plasticity, and have accentuated long-term synaptic depression. Collectively the findings suggest that triplication of Kcnj6 gene may play an active role in some of the abnormal neurological phenotypes found in Down syndrome.


Assuntos
Cognição , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Plasticidade Neuronal , Recompensa , Trissomia , Animais , Ritmo Circadiano , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética , Hipocampo/fisiologia , Camundongos
20.
Cell ; 143(5): 750-60, 2010 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-21111235

RESUMO

G protein-coupled receptors (GPCRs) respond to agonists to activate downstream enzymatic pathways or to gate ion channel function. Turning off GPCR signaling is known to involve phosphorylation of the GPCR by GPCR kinases (GRKs) to initiate their internalization. The process, however, is relatively slow and cannot account for the faster desensitization responses required to regulate channel gating. Here, we show that GRKs enable rapid desensitization of the G protein-coupled potassium channel (GIRK/Kir3.x) through a mechanism independent of their kinase activity. On GPCR activation, GRKs translocate to the membrane and quench channel activation by competitively binding and titrating G protein ßγ subunits away from the channel. Of interest, the ability of GRKs to effect this rapid desensitization depends on the receptor type. The findings thus reveal a stimulus-specific, phosphorylation-independent mechanism for rapidly downregulating GPCR activity at the effector level.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Quinases de Receptores Acoplados a Proteína G/metabolismo , Animais , Fenômenos Fisiológicos Celulares , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/química , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Fosforilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA