Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 968
Filtrar
1.
BMC Biol ; 22(1): 29, 2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38317233

RESUMO

BACKGROUND: Cyclic Nucleotide-Binding Domain (CNBD)-family channels display distinct voltage-sensing properties despite sharing sequence and structural similarity. For example, the human Ether-a-go-go Related Gene (hERG) channel and the Hyperpolarization-activated Cyclic Nucleotide-gated (HCN) channel share high amino acid sequence similarity and identical domain structures. hERG conducts outward current and is activated by positive membrane potentials (depolarization), whereas HCN conducts inward current and is activated by negative membrane potentials (hyperpolarization). The structural basis for the "opposite" voltage-sensing properties of hERG and HCN remains unknown. RESULTS: We found the voltage-sensing domain (VSD) involves in modulating the gating polarity of hERG. We identified that a long-QT syndrome type 2-related mutation within the VSD, K525N, mediated an inwardly rectifying non-deactivating current, perturbing the channel closure, but sparing the open state and inactivated state. K525N rescued the current of a non-functional mutation in the pore helix region (F627Y) of hERG. K525N&F627Y switched hERG into a hyperpolarization-activated channel. The reactivated inward current induced by hyperpolarization mediated by K525N&F627Y can be inhibited by E-4031 and dofetilide quite well. Moreover, we report an extracellular interaction between the S1 helix and the S5-P region is crucial for modulating the gating polarity. The alanine substitution of several residues in this region (F431A, C566A, I607A, and Y611A) impaired the inward current of K525N&F627Y. CONCLUSIONS: Our data provide evidence that a potential cooperation mechanism in the extracellular vestibule of the VSD and the PD would determine the gating polarity in hERG.


Assuntos
Canal de Potássio ERG1 , Ativação do Canal Iônico , Humanos , Sequência de Aminoácidos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Ativação do Canal Iônico/genética , Mutação , Nucleotídeos Cíclicos , Canal de Potássio ERG1/genética
2.
Nature ; 622(7982): 410-417, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37758949

RESUMO

The Kv2.1 voltage-activated potassium (Kv) channel is a prominent delayed-rectifier Kv channel in the mammalian central nervous system, where its mechanisms of activation and inactivation are critical for regulating intrinsic neuronal excitability1,2. Here we present structures of the Kv2.1 channel in a lipid environment using cryo-electron microscopy to provide a framework for exploring its functional mechanisms and how mutations causing epileptic encephalopathies3-7 alter channel activity. By studying a series of disease-causing mutations, we identified one that illuminates a hydrophobic coupling nexus near the internal end of the pore that is critical for inactivation. Both functional and structural studies reveal that inactivation in Kv2.1 results from dynamic alterations in electromechanical coupling to reposition pore-lining S6 helices and close the internal pore. Consideration of these findings along with available structures for other Kv channels, as well as voltage-activated sodium and calcium channels, suggests that related mechanisms of inactivation are conserved in voltage-activated cation channels and likely to be engaged by widely used therapeutics to achieve state-dependent regulation of channel activity.


Assuntos
Ativação do Canal Iônico , Mutação , Canais de Potássio Shab , Animais , Humanos , Microscopia Crioeletrônica , Interações Hidrofóbicas e Hidrofílicas , Ativação do Canal Iônico/genética , Canais de Potássio Shab/genética , Canais de Potássio Shab/metabolismo , Canais de Potássio Shab/ultraestrutura , Espasmos Infantis/genética
3.
FEBS J ; 290(4): 970-973, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36315610

RESUMO

So far one gene for Hv1 has been detected in studied species. The work presented by Chaves et al. in The FEBS Journal reported an 'Unexpected expansion of the voltage-gated proton channel family'. They searched for proton channel candidates and found three sequences in the genome of Aplysia californica (Ac), which were named AcHv1, AcHv2 and AcHv3. Based on electrophysiological experiments, AcHv1 and AcHv2 are voltage-gated channels. While AcHv1 behaves like Hv1 in other species, that is, it is voltage and pH-dependent, it can be inhibited by zinc and conducts protons outwardly, AcHv2 conducts protons inwards at symmetrical pH. AcHv3 constantly leaks protons, and its C-terminal part contains several cytoplasmic retention motifs. Through carefully designed and carried out electrophysiological experiments, Chaves et al. determined the biophysical parameters of all three proton channels, such as the voltage and the pH dependence, the threshold-voltage, the gating charge and the time constants of activation and inactivation. Comment on: https://doi.org/10.1111/febs.16617.


Assuntos
Ativação do Canal Iônico , Prótons , Ativação do Canal Iônico/genética , Canais Iônicos/metabolismo , Zinco/metabolismo
4.
Life Sci ; 308: 120943, 2022 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-36096246

RESUMO

AIMS: GABAA receptors belong to Cys-loop ion channel family and mediate inhibition in the brain. Despite the abundance of structural data on receptor structure, the molecular scenarios of activation are unknown. In this study we investigated the role of a ß2P273 residue in channel gating transitions. This residue is located in a central position of the M2-M3 linker of the interdomain interface, expected to be predisposed to interact with another interfacial element, the ß1-ß2 loop of the extracellular side. The interactions occurring on this interface have been reported to couple agonist binding to channel gating. MAIN METHODS: We recorded micro- and macroscopic current responses of recombinant GABAA receptors mutated at the ß2P273 residue (to A, K, E) to saturating GABA. Electrophysiological data served as basis to kinetic modeling, used to decipher which gating transition were affected by mutations. KEY FINDINGS: Mutations of this residue impaired macroscopic desensitization and accelerated current deactivation with P273E mutant showing greatest deviation from wild-type. Single-channel analysis revealed alterations mainly in short-lived shut times and shortening of openings, resulting in dramatic changes in intraburst open probability. Kinetic modeling indicated that ß2P273 mutants show diminished entry into desensitized and open states as well as faster channel closing transitions. SIGNIFICANCE: In conclusion, we demonstrate that ß2P273 of the M2-M3 linker is a crucial element of the ECD-TMD interface regulating the receptor's ability to undergo late gating transitions. Henceforth, this region could be an important target for new pharmacological tools affecting GABAAR-mediated inhibition.


Assuntos
Receptores de GABA-A , Ácido gama-Aminobutírico , Ativação do Canal Iônico/genética , Técnicas de Patch-Clamp , Prolina , Receptores de GABA-A/metabolismo , Ácido gama-Aminobutírico/metabolismo
5.
Elife ; 112022 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-35816168

RESUMO

Deep mutational scanning provides new insights into how mutations alter the expression and activity of the potassium ion channel Kir2.1, which is associated with many diseases.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização , Ativação do Canal Iônico/genética , Mutação , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo
6.
Proc Natl Acad Sci U S A ; 119(25): e2204620119, 2022 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-35704760

RESUMO

In neurosecretion, allosteric communication between voltage sensors and Ca2+ binding in BK channels is crucially involved in damping excitatory stimuli. Nevertheless, the voltage-sensing mechanism of BK channels is still under debate. Here, based on gating current measurements, we demonstrate that two arginines in the transmembrane segment S4 (R210 and R213) function as the BK gating charges. Significantly, the energy landscape of the gating particles is electrostatically tuned by a network of salt bridges contained in the voltage sensor domain (VSD). Molecular dynamics simulations and proton transport experiments in the hyperpolarization-activated R210H mutant suggest that the electric field drops off within a narrow septum whose boundaries are defined by the gating charges. Unlike Kv channels, the charge movement in BK appears to be limited to a small displacement of the guanidinium moieties of R210 and R213, without significant movement of the S4.


Assuntos
Ativação do Canal Iônico , Canais de Potássio Ativados por Cálcio de Condutância Alta , Arginina/metabolismo , Ativação do Canal Iônico/genética , Simulação de Dinâmica Molecular , Mutação
7.
Nat Commun ; 12(1): 7114, 2021 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-34880224

RESUMO

Protein domains are the basic units of protein structure and function. Comparative analysis of genomes and proteomes showed that domain recombination is a main driver of multidomain protein functional diversification and some of the constraining genomic mechanisms are known. Much less is known about biophysical mechanisms that determine whether protein domains can be combined into viable protein folds. Here, we use massively parallel insertional mutagenesis to determine compatibility of over 300,000 domain recombination variants of the Inward Rectifier K+ channel Kir2.1 with channel surface expression. Our data suggest that genomic and biophysical mechanisms acted in concert to favor gain of large, structured domain at protein termini during ion channel evolution. We use machine learning to build a quantitative biophysical model of domain compatibility in Kir2.1 that allows us to derive rudimentary rules for designing domain insertion variants that fold and traffic to the cell surface. Positional Kir2.1 responses to motif insertion clusters into distinct groups that correspond to contiguous structural regions of the channel with distinct biophysical properties tuned towards providing either folding stability or gating transitions. This suggests that insertional profiling is a high-throughput method to annotate function of ion channel structural regions.


Assuntos
Biofísica , Canais de Potássio/química , Canais de Potássio/genética , Recombinação Genética , Linhagem Celular , Membrana Celular , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G , Perfilação da Expressão Gênica , Células HEK293 , Humanos , Ativação do Canal Iônico/genética , Ativação do Canal Iônico/fisiologia , Aprendizado de Máquina , Mutagênese Insercional , Potássio/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/química , Canais de Potássio Corretores do Fluxo de Internalização/genética , Domínios Proteicos/genética , Transcriptoma
8.
Nat Commun ; 12(1): 5457, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34526505

RESUMO

GABAA receptors are vital for controlling neuronal excitability and can display significant levels of constitutive activity that contributes to tonic inhibition. However, the mechanisms underlying spontaneity are poorly understood. Here we demonstrate a strict requirement for ß3 subunit incorporation into receptors for spontaneous gating, facilitated by α4, α6 and δ subunits. The crucial molecular determinant involves four amino acids (GKER) in the ß3 subunit's extracellular domain, which interacts with adjacent receptor subunits to promote transition to activated, open channel conformations. Spontaneous activity is further regulated by ß3 subunit phosphorylation and by allosteric modulators including neurosteroids and benzodiazepines. Promoting spontaneous activity reduced neuronal excitability, indicating that spontaneous currents will alter neural network activity. This study demonstrates how regional diversity in GABAA receptor isoform, protein kinase activity, and neurosteroid levels, can impact on tonic inhibition through the modulation of spontaneous GABAA receptor gating.


Assuntos
Hipocampo/fisiologia , Ativação do Canal Iônico/fisiologia , Neurônios/fisiologia , Receptores de GABA-A/fisiologia , Algoritmos , Sequência de Aminoácidos , Animais , Células Cultivadas , Células HEK293 , Hipocampo/citologia , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/genética , Camundongos , Modelos Moleculares , Modelos Neurológicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Técnicas de Patch-Clamp/métodos , Conformação Proteica , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/fisiologia , Ratos Sprague-Dawley , Receptores de GABA-A/química , Receptores de GABA-A/genética , Homologia de Sequência de Aminoácidos , Ácido gama-Aminobutírico/metabolismo , Ácido gama-Aminobutírico/farmacologia
9.
J Cell Mol Med ; 25(20): 9685-9696, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34514691

RESUMO

Hepatocellular carcinoma (HCC) is a leading cause of cancer-related death worldwide. Its high metastasis rate is significantly correlated with poor patient prognosis. Elucidating the molecular mechanism underlying HCC metastasis is essential for HCC treatment. Owing to their high conductance, large-conductance calcium-activated potassium channels (BK channels) play a critical role in the control of membrane potential and have repeatedly been proposed as potential targets for cancer therapy. Emerging evidence suggests that BK channels are involved in the progression of cancer malignancies. The present study investigated the role of BK channels in mediating the hypoxia-stimulated migration of HCC cells both in vitro and in vivo in the absence and presence of various BK channels modulators. We found that BK channels were functionally expressed on the membranes of the SMMC-7721 and Huh7 HCC cell lines. Furthermore, blockage or activation of BK channels on the surface of HCC cells correspondingly inhibited or promoted HCC cell proliferation, migration and invasion in hypoxia conditions, with altered expression and distribution of cell-cell adhesion molecule E-cadherin and typical marker of mesenchymal cells, Vimentin, but not N-cadherin. Hypoxia conditions did not alter BK channels expression but increased its open probability. Moreover, BK channels blocker IbTX significantly inhibited HCC cell remote colonization in HCC cell xenografted mice. In conclusion, the results of this study suggest that blocking BK channels offers an attractive strategy for treating HCC.


Assuntos
Movimento Celular/genética , Canais de Potássio Cálcio-Ativados/genética , Canais de Potássio Cálcio-Ativados/metabolismo , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Fenômenos Eletrofisiológicos , Xenoenxertos , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Camundongos , Bloqueadores dos Canais de Potássio/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Mol Brain ; 14(1): 126, 2021 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-34399820

RESUMO

Developmental and epileptic encephalopathies (DEEs) are a group of severe epilepsies that are characterized by seizures and developmental delay. DEEs are primarily attributed to genetic causes and an increasing number of cases have been correlated with variants in ion channel genes. In this study, we report a child with an early severe DEE. Whole exome sequencing showed a de novo heterozygous variant (c.4873-4881 duplication) in the SCN8A gene and an inherited heterozygous variant (c.952G > A) in the CACNA1H gene encoding for Nav1.6 voltage-gated sodium and Cav3.2 voltage-gated calcium channels, respectively. In vitro functional analysis of human Nav1.6 and Cav3.2 channel variants revealed mild but significant alterations of their gating properties that were in general consistent with a gain- and loss-of-channel function, respectively. Although additional studies will be required to confirm the actual pathogenic involvement of SCN8A and CACNA1H, these findings add to the notion that rare ion channel variants may contribute to the etiology of DEEs.


Assuntos
Deficiências do Desenvolvimento/genética , Epilepsia Resistente a Medicamentos/genética , Epilepsia Tônico-Clônica/genética , Canal de Sódio Disparado por Voltagem NAV1.6/genética , Anormalidades Múltiplas/genética , Canais de Cálcio Tipo T/genética , Canais de Cálcio Tipo T/fisiologia , Feminino , Mutação com Ganho de Função , Duplicação Gênica , Predisposição Genética para Doença , Humanos , Recém-Nascido , Ativação do Canal Iônico/genética , Ativação do Canal Iônico/fisiologia , Mutação de Sentido Incorreto , Canal de Sódio Disparado por Voltagem NAV1.6/fisiologia , Linhagem , Mutação Puntual , Escoliose/genética
11.
Biochem Biophys Res Commun ; 569: 112-117, 2021 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-34243066

RESUMO

P2X7 receptors are trimeric ion channels activated by extracellular ATP. Upon activation, they trigger cytolysis and apoptosis but also control cell proliferation. To shed more light on channel gating and the underlying function of the individual subunits, receptors of concatenated subunits were built containing a defined number of functional binding sites. The currents evoked by ATP were obtained in the outside-out configuration of the patch-clamp technique, and steady-state activation, as well as time courses, were analyzed. Our results show that each occupied binding site contributes to channel activation. While the occupation of a single binding site can already activate the channels, three bound ligands maximally stabilize the open state. Hence, P2X7 receptors can be described by a stepwise activation process.


Assuntos
Trifosfato de Adenosina/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Mutação de Sentido Incorreto , Oócitos/fisiologia , Receptores Purinérgicos P2X7/genética , Trifosfato de Adenosina/metabolismo , Algoritmos , Animais , Sítios de Ligação/genética , Feminino , Ativação do Canal Iônico/genética , Cinética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Potenciais da Membrana/fisiologia , Oócitos/metabolismo , Técnicas de Patch-Clamp/métodos , Ratos , Receptores Purinérgicos P2X7/química , Receptores Purinérgicos P2X7/metabolismo , Fatores de Tempo , Xenopus laevis
12.
Biochem J ; 478(14): 2843-2869, 2021 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-34195804

RESUMO

The interaction of insect-selective scorpion depressant ß-toxins (LqhIT2 and Lqh-dprIT3 from Leiurus quinquestriatus hebraeus) with the Blattella germanica sodium channel, BgNav1-1a, was investigated using site-directed mutagenesis, electrophysiological analyses, and structural modeling. Focusing on the pharmacologically defined binding site-4 of scorpion ß-toxins at the voltage-sensing domain II (VSD-II), we found that charge neutralization of D802 in VSD-II greatly enhanced the channel sensitivity to Lqh-dprIT3. This was consistent with the high sensitivity of the splice variant BgNav2-1, bearing G802, to Lqh-dprIT3, and low sensitivity of BgNav2-1 mutant, G802D, to the toxin. Further mutational and electrophysiological analyses revealed that the sensitivity of the WT = D802E < D802G < D802A < D802K channel mutants to Lqh-dprIT3 correlated with the depolarizing shifts of activation in toxin-free channels. However, the sensitivity of single mutants involving IIS4 basic residues (K4E = WT << R1E < R2E < R3E) or double mutants (D802K = K4E/D802K = R3E/D802K > R2E/D802K > R1E/D802K > WT) did not correlate with the activation shifts. Using the cryo-EM structure of the Periplaneta americana channel, NavPaS, as a template and the crystal structure of LqhIT2, we constructed structural models of LqhIT2 and Lqh-dprIT3-c in complex with BgNav1-1a. These models along with the mutational analysis suggest that depressant toxins approach the salt-bridge between R1 and D802 at VSD-II to form contacts with linkers IIS1-S2, IIS3-S4, IIIP5-P1 and IIIP2-S6. Elimination of this salt-bridge enables deeper penetration of the toxin into a VSD-II gorge to form new contacts with the channel, leading to increased channel sensitivity to Lqh-dprIT3.


Assuntos
Neópteros/metabolismo , Venenos de Escorpião/metabolismo , Escorpiões/metabolismo , Canais de Sódio/metabolismo , Animais , Sítios de Ligação/genética , Ativação do Canal Iônico/genética , Ativação do Canal Iônico/fisiologia , Potenciais da Membrana/genética , Potenciais da Membrana/fisiologia , Modelos Moleculares , Mutação , Neópteros/genética , Oócitos/metabolismo , Oócitos/fisiologia , Técnicas de Patch-Clamp/métodos , Ligação Proteica , Domínios Proteicos , Mapeamento de Interação de Proteínas , Venenos de Escorpião/química , Venenos de Escorpião/genética , Escorpiões/genética , Canais de Sódio/química , Canais de Sódio/genética , Xenopus
13.
Proc Natl Acad Sci U S A ; 118(19)2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33941706

RESUMO

The dissipation of acute acid loads by the voltage-gated proton channel (Hv1) relies on regulating the channel's open probability by the voltage and the ΔpH across the membrane (ΔpH = pHex - pHin). Using monomeric Ciona-Hv1, we asked whether ΔpH-dependent gating is produced during the voltage sensor activation or permeation pathway opening. A leftward shift of the conductance-voltage (G-V) curve was produced at higher ΔpH values in the monomeric channel. Next, we measured the voltage sensor pH dependence in the absence of a functional permeation pathway by recording gating currents in the monomeric nonconducting D160N mutant. Increasing the ΔpH leftward shifted the gating charge-voltage (Q-V) curve, demonstrating that the ΔpH-dependent gating in Hv1 arises by modulating its voltage sensor. We fitted our data to a model that explicitly supposes the Hv1 voltage sensor free energy is a function of both the proton chemical and the electrical potential. The parameters obtained showed that around 60% of the free energy stored in the ΔpH is coupled to the Hv1 voltage sensor activation. Our results suggest that the molecular mechanism underlying the Hv1 ΔpH dependence is produced by protons, which alter the free-energy landscape around the voltage sensor domain. We propose that this alteration is produced by accessibility changes of the protons in the Hv1 voltage sensor during activation.


Assuntos
Algoritmos , Ativação do Canal Iônico/fisiologia , Canais Iônicos/fisiologia , Modelos Biológicos , Prótons , Sequência de Aminoácidos , Animais , Feminino , Humanos , Concentração de Íons de Hidrogênio , Ativação do Canal Iônico/genética , Canais Iônicos/genética , Canais Iônicos/metabolismo , Potenciais da Membrana/fisiologia , Camundongos , Simulação de Dinâmica Molecular , Mutação , Oócitos/metabolismo , Oócitos/fisiologia , Homologia de Sequência de Aminoácidos , Xenopus laevis
14.
Brain ; 144(7): 2092-2106, 2021 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-33704440

RESUMO

T-type calcium channels (Cav3.1 to Cav3.3) regulate low-threshold calcium spikes, burst firing and rhythmic oscillations of neurons and are involved in sensory processing, sleep, and hormone and neurotransmitter release. Here, we examined four heterozygous missense variants in CACNA1I, encoding the Cav3.3 channel, in patients with variable neurodevelopmental phenotypes. The p.(Ile860Met) variant, affecting a residue in the putative channel gate at the cytoplasmic end of the IIS6 segment, was identified in three family members with variable cognitive impairment. The de novo p.(Ile860Asn) variant, changing the same amino acid residue, was detected in a patient with severe developmental delay and seizures. In two additional individuals with global developmental delay, hypotonia, and epilepsy, the variants p.(Ile1306Thr) and p.(Met1425Ile), substituting residues at the cytoplasmic ends of IIIS5 and IIIS6, respectively, were found. Because structure modelling indicated that the amino acid substitutions differentially affect the mobility of the channel gate, we analysed possible effects on Cav3.3 channel function using patch-clamp analysis in HEK293T cells. The mutations resulted in slowed kinetics of current activation, inactivation, and deactivation, and in hyperpolarizing shifts of the voltage-dependence of activation and inactivation, with Cav3.3-I860N showing the strongest and Cav3.3-I860M the weakest effect. Structure modelling suggests that by introducing stabilizing hydrogen bonds the mutations slow the kinetics of the channel gate and cause the gain-of-function effect in Cav3.3 channels. The gating defects left-shifted and increased the window currents, resulting in increased calcium influx during repetitive action potentials and even at resting membrane potentials. Thus, calcium toxicity in neurons expressing the Cav3.3 variants is one likely cause of the neurodevelopmental phenotype. Computer modelling of thalamic reticular nuclei neurons indicated that the altered gating properties of the Cav3.3 disease variants lower the threshold and increase the duration and frequency of action potential firing. Expressing the Cav3.3-I860N/M mutants in mouse chromaffin cells shifted the mode of firing from low-threshold spikes and rebound burst firing with wild-type Cav3.3 to slow oscillations with Cav3.3-I860N and an intermediate firing mode with Cav3.3-I860M, respectively. Such neuronal hyper-excitability could explain seizures in the patient with the p.(Ile860Asn) mutation. Thus, our study implicates CACNA1I gain-of-function mutations in neurodevelopmental disorders, with a phenotypic spectrum ranging from borderline intellectual functioning to a severe neurodevelopmental disorder with epilepsy.


Assuntos
Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Ativação do Canal Iônico/genética , Transtornos do Neurodesenvolvimento/genética , Adulto , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Criança , Simulação por Computador , Feminino , Mutação com Ganho de Função , Predisposição Genética para Doença/genética , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Modelos Moleculares , Modelos Neurológicos , Mutação de Sentido Incorreto , Neurônios/metabolismo , Linhagem , Conformação Proteica
15.
Commun Biol ; 4(1): 84, 2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33469156

RESUMO

Mechanosensitive Piezo1 channels are essential mechanotransduction proteins in eukaryotes. Their curved transmembrane domains, called arms, create a convex membrane deformation, or footprint, which is predicted to flatten in response to increased membrane tension. Here, using a hyperbolic tangent model, we show that, due to the intrinsic bending rigidity of the membrane, the overlap of neighboring Piezo1 footprints produces a flattening of the Piezo1 footprints and arms. Multiple all-atom molecular dynamics simulations of Piezo1 further reveal that this tension-independent flattening is accompanied by gating motions that open an activation gate in the pore. This open state recapitulates experimentally obtained ionic selectivity, unitary conductance, and mutant phenotypes. Tracking ion permeation along the open pore reveals the presence of intracellular and extracellular fenestrations acting as cation-selective sites. Simulations also reveal multiple potential binding sites for phosphatidylinositol 4,5-bisphosphate. We propose that the overlap of Piezo channel footprints may act as a cooperative mechanism to regulate channel activity.


Assuntos
Canais Iônicos/metabolismo , Células HEK293 , Humanos , Ativação do Canal Iônico/genética , Ativação do Canal Iônico/fisiologia , Canais Iônicos/genética , Canais Iônicos/fisiologia , Íons/metabolismo , Mecanotransdução Celular/genética , Mecanotransdução Celular/fisiologia , Modelos Moleculares , Modelos Teóricos , Simulação de Dinâmica Molecular , Domínios Proteicos/genética
16.
Mol Brain ; 14(1): 4, 2021 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-33413531

RESUMO

A novel missense mutation in the CACNA1A gene that encodes the pore forming α1 subunit of the CaV2.1 voltage-gated calcium channel was identified in a patient with trigeminal neuralgia. This mutation leads to a substitution of proline 2455 by histidine (P2455H) in the distal C-terminus region of the channel. Due to the well characterized role of this channel in neurotransmitter release, our aim was to characterize the biophysical properties of the P2455H variant in heterologously expressed CaV2.1 channels. Whole-cell patch clamp recordings of wild type and mutant CaV2.1 channels expressed in tsA-201 cells reveal that the mutation mediates a depolarizing shift in the voltage-dependence of activation and inactivation. Moreover, the P2455H mutant strongly reduced calcium-dependent inactivation of the channel that is consistent with an overall gain of function. Hence, the P2455H CaV2.1 missense mutation alters the gating properties of the channel, suggesting that associated changes in CaV2.1-dependent synaptic communication in the trigeminal system may contribute to the development of trigeminal neuralgia.


Assuntos
Canais de Cálcio/genética , Estudos de Associação Genética , Predisposição Genética para Doença , Variação Genética , Ativação do Canal Iônico/genética , Neuralgia do Trigêmeo/genética , Linhagem Celular , Humanos , Mutação/genética
17.
J Biol Chem ; 296: 100225, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33361157

RESUMO

Mechanotransduction is the process by which cells convert physical forces into electrochemical responses. On a molecular scale, these forces are detected by mechanically activated ion channels, which constitute the basis for hearing, touch, pain, cold, and heat sensation, among other physiological processes. Exciting high-resolution structural details of these channels are currently emerging that will eventually allow us to delineate the molecular determinants of gating and ion permeation. However, our structural-functional understanding across the family remains limited. Piezo1 is one of the largest and least understood of these channels, with various structurally identified features within its trimeric assembly. This study seeks to determine the modularity and function of Piezo1 channels by constructing deletion proteins guided by cryo EM structural knowledge. Our comprehensive functional study identified, for the first time, the minimal amino acid sequence of the full-length Piezo1 that can fold and function as the channel's pore domain between E2172 and the last residue E2547. While the addition of an anchor region has no effect on permeation properties. The Piezo1 pore domain is not pressure-sensitive and the appending of Piezo Repeat-A did not restore pressure-dependent gating, hence the sensing module must exist between residues 1 to 1952. Our efforts delineating the permeation and gating regions within this complex ion channel have implications in identifying small molecules that exclusively regulate the activity of the channel's pore module to influence mechanotransduction and downstream processes.


Assuntos
Ativação do Canal Iônico/genética , Canais Iônicos/química , Magnésio/química , Mecanotransdução Celular/genética , Potássio/química , Substituição de Aminoácidos , Animais , Sítios de Ligação , Cálcio/química , Cálcio/metabolismo , Expressão Gênica , Células HEK293 , Humanos , Canais Iônicos/genética , Canais Iônicos/metabolismo , Transporte de Íons , Cinética , Magnésio/metabolismo , Camundongos , Modelos Moleculares , Mutação , Técnicas de Patch-Clamp , Potássio/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Sódio/química , Sódio/metabolismo
18.
J Biol Chem ; 296: 100224, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33361160

RESUMO

The initial activation step in the gating of ubiquitously expressed Orai1 calcium (Ca2+) ion channels represents the activation of the Ca2+-sensor protein STIM1 upon Ca2+ store depletion of the endoplasmic reticulum. Previous studies using constitutively active Orai1 mutants gave rise to, but did not directly test, the hypothesis that STIM1-mediated Orai1 pore opening is accompanied by a global conformational change of all Orai transmembrane domain (TM) helices within the channel complex. We prove that a local conformational change spreads omnidirectionally within the Orai1 complex. Our results demonstrate that these locally induced global, opening-permissive TM motions are indispensable for pore opening and require clearance of a series of Orai1 gating checkpoints. We discovered these gating checkpoints in the middle and cytosolic extended TM domain regions. Our findings are based on a library of double point mutants that contain each one loss-of-function with one gain-of-function point mutation in a series of possible combinations. We demonstrated that an array of loss-of-function mutations are dominant over most gain-of-function mutations within the same as well as of an adjacent Orai subunit. We further identified inter- and intramolecular salt-bridge interactions of Orai subunits as a core element of an opening-permissive Orai channel architecture. Collectively, clearance and synergistic action of all these gating checkpoints are required to allow STIM1 coupling and Orai1 pore opening. Our results unravel novel insights in the preconditions of the unique fingerprint of CRAC channel activation, provide a valuable source for future structural resolutions, and help to understand the molecular basis of disease-causing mutations.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Ativação do Canal Iônico/genética , Proteínas de Neoplasias/química , Proteína ORAI1/química , Molécula 1 de Interação Estromal/química , Substituição de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Regulação da Expressão Gênica , Genes Reporter , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Lipossomos/química , Lipossomos/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Simulação de Dinâmica Molecular , Mutação , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Técnicas de Patch-Clamp , Fosfatidilcolinas/química , Fosfatidilcolinas/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Molécula 1 de Interação Estromal/genética , Molécula 1 de Interação Estromal/metabolismo
19.
Prog Biophys Mol Biol ; 159: 146-156, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33011190

RESUMO

Atrial fibrillation (AF) is a multifactorial disease, which often occurs in the presence of underlying cardiac abnormalities and is supported by electrophysiological and structural alterations, generally referred to as atrial remodeling. Abnormal substrates are commonly encountered in various conditions that predispose to AF, such as hypertension, heart failure, obesity, and sleep apnea, in which atrial stretch plays a key mechanistic role. Emerging evidence suggests a role for microRNAs (small non-coding RNAs) in the pathogenesis of AF, where they can act as post-transcriptional regulators of the genes involved in atrial remodeling. This review summarizes the experimental and clinical evidence that supports the role of microRNAs in the modulation of atrial electrical and structural remodeling with a focus on overload-induced atrial alterations, and discusses the potential contribution of microRNAs to mechano-electrical coupling and AF.


Assuntos
Fibrilação Atrial/metabolismo , Fenômenos Biomecânicos/fisiologia , Átrios do Coração/metabolismo , Ativação do Canal Iônico/fisiologia , MicroRNAs/metabolismo , Animais , Função Atrial , Remodelamento Atrial/fisiologia , Biomarcadores/metabolismo , Cálcio/metabolismo , Fenômenos Eletrofisiológicos , Retroalimentação Fisiológica , Regulação da Expressão Gênica , Humanos , Ativação do Canal Iônico/genética
20.
Am J Respir Crit Care Med ; 203(5): 585-593, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33023304

RESUMO

Rationale: We previously reported that ivacaftor was safe and well tolerated in cohorts aged 12 to <24 months with cystic fibrosis and gating mutations in the ARRIVAL study; here, we report results for cohorts aged 4 to <12 months.Objectives: To evaluate the safety, pharmacokinetics, and pharmacodynamics of ivacaftor in infants aged 4 to <12 months with one or more gating mutations.Methods: ARRIVAL is a single-arm phase 3 study. Infants received 25 mg or 50 mg ivacaftor every 12 hours on the basis of age and weight for 4 days in part A and 24 weeks in part B.Measurements and Main Results: Primary endpoints were safety (parts A and B) and pharmacokinetics (part A). Secondary/tertiary endpoints (part B) included pharmacokinetics and changes in sweat chloride levels, growth, and markers of pancreatic function. Twenty-five infants received ivacaftor, 12 in part A and 17 in part B (four infants participated in both parts). Pharmacokinetics was consistent with that in older groups. Most adverse events were mild or moderate. In part B, cough was the most common adverse event (n = 10 [58.8%]). Five infants (part A, n = 1 [8.3%]; part B, n = 4 [23.5%]) had serious adverse events, all of which were considered to be not or unlikely related to ivacaftor. No deaths or treatment discontinuations occurred. One infant (5.9%) experienced an alanine transaminase elevation >3 to ≤5× the upper limit of normal at Week 24. No other adverse trends in laboratory tests, vital signs, or ECG parameters were reported. Sweat chloride concentrations and measures of pancreatic obstruction improved.Conclusions: This study of ivacaftor in the first year of life supports treating the underlying cause of cystic fibrosis in children aged ≥4 months with one or more gating mutations.Clinical trial registered with clinicaltrials.gov (NCT02725567).


Assuntos
Aminofenóis/uso terapêutico , Agonistas dos Canais de Cloreto/uso terapêutico , Fibrose Cística/tratamento farmacológico , Quinolonas/uso terapêutico , Aminofenóis/farmacocinética , Agonistas dos Canais de Cloreto/farmacocinética , Cloretos/metabolismo , Tosse/epidemiologia , Fibrose Cística/genética , Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Insuficiência Pancreática Exócrina/metabolismo , Feminino , Febre/epidemiologia , Genótipo , Humanos , Lactente , Ativação do Canal Iônico/genética , Masculino , Mutação , Otite Média/epidemiologia , Elastase Pancreática/metabolismo , Quinolonas/farmacocinética , Infecções Respiratórias/epidemiologia , Rinorreia/epidemiologia , Suor/metabolismo , Resultado do Tratamento , Vômito/epidemiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...