Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Mol Cell ; 83(24): 4555-4569.e4, 2023 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-38035882

RESUMO

Modulation of large conductance intracellular ligand-activated potassium (BK) channel family (Slo1-3) by auxiliary subunits allows diverse physiological functions in excitable and non-excitable cells. Cryoelectron microscopy (cryo-EM) structures of voltage-gated potassium (Kv) channel complexes have provided insights into how voltage sensitivity is modulated by auxiliary subunits. However, the modulation mechanisms of BK channels, particularly as ligand-activated ion channels, remain unknown. Slo1 is a Ca2+-activated and voltage-gated BK channel and is expressed in neurons, muscle cells, and epithelial cells. Using cryo-EM and electrophysiology, we show that the LRRC26-γ1 subunit modulates not only voltage but also Ca2+ sensitivity of Homo sapiens Slo1. LRRC26 stabilizes the active conformation of voltage-senor domains of Slo1 by an extracellularly S4-locking mechanism. Furthermore, it also stabilizes the active conformation of Ca2+-sensor domains of Slo1 intracellularly, which is functionally equivalent to intracellular Ca2+ in the activation of Slo1. Such a dual allosteric modulatory mechanism may be general in regulating the intracellular ligand-activated BK channel complexes.


Assuntos
Cálcio , Canais de Potássio Ativados por Cálcio de Condutância Alta , Humanos , Cálcio/metabolismo , Microscopia Crioeletrônica , Ativação do Canal Iônico/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/genética , Canais de Potássio Ativados por Cálcio de Condutância Alta/química , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Ligantes , Potássio , Regulação Alostérica
2.
Biomolecules ; 11(11)2021 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-34827626

RESUMO

Potassium ion concentrations, controlled by ion pumps and potassium channels, predominantly govern a cell's membrane potential and the tone in the vessels. Calcium-activated potassium channels respond to two different stimuli-changes in voltage and/or changes in intracellular free calcium. Large conductance calcium-activated potassium (BKCa) channels assemble from pore forming and various modulatory and auxiliary subunits. They are of vital significance due to their very high unitary conductance and hence their ability to rapidly cause extreme changes in the membrane potential. The pathophysiology of lung diseases in general and pulmonary hypertension, in particular, show the implication of either decreased expression and partial inactivation of BKCa channel and its subunits or mutations in the genes encoding different subunits of the channel. Signaling molecules, circulating humoral molecules, vasorelaxant agents, etc., have an influence on the open probability of the channel in pulmonary arterial vascular cells. BKCa channel is a possible therapeutic target, aimed to cause vasodilation in constricted or chronically stiffened vessels, as shown in various animal models. This review is a comprehensive collation of studies on BKCa channels in the pulmonary circulation under hypoxia (hypoxic pulmonary vasoconstriction; HPV), lung pathology, and fetal to neonatal transition, emphasising pharmacological interventions as viable therapeutic options.


Assuntos
Canais de Potássio Ativados por Cálcio de Condutância Alta , Cálcio , Circulação Pulmonar
3.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33431687

RESUMO

Goblet cells (GCs) are specialized cells of the intestinal epithelium contributing critically to mucosal homeostasis. One of the functions of GCs is to produce and secrete MUC2, the mucin that forms the scaffold of the intestinal mucus layer coating the epithelium and separates the luminal pathogens and commensal microbiota from the host tissues. Although a variety of ion channels and transporters are thought to impact on MUC2 secretion, the specific cellular mechanisms that regulate GC function remain incompletely understood. Previously, we demonstrated that leucine-rich repeat-containing protein 26 (LRRC26), a known regulatory subunit of the Ca2+-and voltage-activated K+ channel (BK channel), localizes specifically to secretory cells within the intestinal tract. Here, utilizing a mouse model in which MUC2 is fluorescently tagged, thereby allowing visualization of single GCs in intact colonic crypts, we show that murine colonic GCs have functional LRRC26-associated BK channels. In the absence of LRRC26, BK channels are present in GCs, but are not activated at physiological conditions. In contrast, all tested MUC2- cells completely lacked BK channels. Moreover, LRRC26-associated BK channels underlie the BK channel contribution to the resting transepithelial current across mouse distal colonic mucosa. Genetic ablation of either LRRC26 or BK pore-forming α-subunit in mice results in a dramatically enhanced susceptibility to colitis induced by dextran sodium sulfate. These results demonstrate that normal potassium flux through LRRC26-associated BK channels in GCs has protective effects against colitis in mice.


Assuntos
Colite/genética , Canais de Potássio Ativados por Cálcio de Condutância Alta/genética , Mucina-2/genética , Animais , Colite/patologia , Colite/prevenção & controle , Colite/terapia , Colo/metabolismo , Colo/patologia , Modelos Animais de Doenças , Células Caliciformes/metabolismo , Células Caliciformes/patologia , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Potenciais da Membrana/genética , Camundongos , Técnicas de Patch-Clamp
4.
Epigenomics ; 12(18): 1633-1650, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32573269

RESUMO

Aim: We aim to identify driving genes of colorectal cancer (CRC) through multi-omics analysis. Materials & methods: We downloaded multi-omics data of CRC from The Cancer Genome Atlas dataset. Integrative analysis of single-nucleotide variants, copy number variations, DNA methylation and differentially expressed genes identified candidate genes that carry CRC risk. Kernal genes were extracted from the weighted gene co-expression network analysis. A competing endogenous RNA network composed of CRC-related genes was constructed. Biological roles of genes were further investigated in vitro. Results: We identified LRRC26 and REP15 as novel prognosis-related driving genes for CRC. LRRC26 hindered tumorigenesis of CRC in vitro. Conclusion: Our study identified novel driving genes and may provide new insights into the molecular mechanisms of CRC.


Assuntos
Neoplasias Colorretais/genética , Genes Neoplásicos , Proteínas Adaptadoras de Transdução de Sinal/genética , Idoso , Linhagem Celular Tumoral , Neoplasias Colorretais/classificação , Neoplasias Colorretais/patologia , Variações do Número de Cópias de DNA , Metilação de DNA , Epigenômica , Feminino , Regulação Neoplásica da Expressão Gênica , Variação Genética , Genômica , Genótipo , Humanos , Masculino , Proteínas de Membrana/genética , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Prognóstico
5.
Am J Physiol Lung Cell Mol Physiol ; 318(2): L366-L375, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31800260

RESUMO

In visceral smooth muscle cells (SMCs), the large-conductance Ca2+-activated K+ (BK) channel is one of the key elements underlying a negative feedback mechanism that is essential for the regulation of intracellular Ca2+ concentration. Although leucine-rich repeat-containing (LRRC) proteins have been identified as novel auxiliary γ-subunits of the BK channel (BKγ) in several cell types, its physiological roles in SMCs are unclear. The BKγ expression patterns in selected SM tissues were examined using real-time PCR analyses and Western blotting. The functional contribution of BKγ1 to BK channel activity was examined by whole cell patch-clamp in SMCs and heterologous expression systems. BKγ1 expression in mouse bronchial SMCs (mBSMCs) was higher than in other several SMC types. Coimmunoprecipitation and total internal reflection fluorescence imaging analyses revealed molecular interaction between BKα and BKγ1 in mBSMCs. Under voltage-clamp, steady-state activation of BK channel currents at pCa 8.0 in mBSMCs occurred in a voltage range comparable to that of reconstituted BKα/BKγ1 complex. However, this range was much more negative than in mouse aortic SMCs (mASMCs) or in HEK293 cells expressing BKα alone and ß-subunit (BKß1). Mallotoxin, a selective activator of BK channel that lacks BKγ1, dose-dependently activated BK currents in mASMCs but not in mBSMCs. The abundant expression of BKγ1 in mBSMCs extensively facilitates BK channel activity to keep the resting membrane potential at negative values and prevents contraction under physiological conditions.


Assuntos
Brônquios/citologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas de Neoplasias/metabolismo , Subunidades Proteicas/metabolismo , Acetofenonas/farmacologia , Animais , Benzopiranos/farmacologia , Cálcio/metabolismo , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Miócitos de Músculo Liso/efeitos dos fármacos , Ratos Wistar
6.
Proc Natl Acad Sci U S A ; 114(18): E3739-E3747, 2017 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-28416688

RESUMO

Leucine-rich-repeat-containing protein 26 (LRRC26) is the regulatory γ1 subunit of Ca2+- and voltage-dependent BK-type K+ channels. BK channels that contain LRRC26 subunits are active near normal resting potentials even without Ca2+, suggesting they play unique physiological roles, likely limited to very specific cell types and cellular functions. By using Lrrc26 KO mice with a ß-gal reporter, Lrrc26 promoter activity is found in secretory epithelial cells, especially acinar epithelial cells in lacrimal and salivary glands, and also goblet and Paneth cells in intestine and colon, although absent from neurons. We establish the presence of LRRC26 protein in eight secretory tissues or tissues with significant secretory epithelium and show that LRRC26 protein coassembles with the pore-forming BK α-subunit in at least three tissues: lacrimal gland, parotid gland, and colon. In lacrimal, parotid, and submandibular gland acinar cells, LRRC26 KO shifts BK gating to be like α-subunit-only BK channels. Finally, LRRC26 KO mimics the effect of SLO1/BK KO in reducing [K+] in saliva. LRRC26-containing BK channels are competent to contribute to resting K+ efflux at normal cell membrane potentials with resting cytosolic Ca2+ concentrations and likely play a critical physiological role in supporting normal secretory function in all secretory epithelial cells.


Assuntos
Colo/metabolismo , Células Epiteliais/metabolismo , Aparelho Lacrimal/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Potenciais da Membrana , Glândula Parótida/metabolismo , Animais , Cálcio/metabolismo , Colo/citologia , Células Epiteliais/citologia , Aparelho Lacrimal/citologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/genética , Camundongos , Camundongos Knockout , Glândula Parótida/citologia , Potássio/metabolismo
7.
Int Rev Neurobiol ; 128: 369-99, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27238269

RESUMO

The perception of complex acoustic stimuli begins with the deconstruction of sound into its frequency components. This spectral processing occurs first and foremost in the inner ear. In vertebrates, two very different strategies of frequency analysis have evolved. In nonmammalian vertebrates, the sensory hair cells of the inner ear are intrinsically electrically tuned to a narrow band of acoustic frequencies. This electrical tuning relies on the interplay between BK channels and voltage-gated calcium channels. Systematic variations in BK channel density and kinetics establish a gradient in electrical resonance that enables the coding of a broad range of acoustic frequencies. In contrast, mammalian hair cells are extrinsically tuned by mechanical properties of the cochlear duct. Even so, mammalian hair cells also express BK channels. These BK channels play critical roles in various aspects of mammalian auditory signaling, from developmental maturation to protection against acoustic trauma. This review summarizes the anatomical localization, biophysical properties, and functional contributions of BK channels in vertebrate inner ears. Areas of future research, based on an updated understanding of the biology of both BK channels and the inner ear, are also highlighted. Investigation of BK channels in the inner ear continues to provide fertile research grounds for examining both BK channel biophysics and the molecular mechanisms underlying signal processing in the auditory periphery.


Assuntos
Orelha Interna/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Potenciais da Membrana/fisiologia , Animais , Vias Auditivas/fisiologia , Humanos , Vertebrados
8.
J Biol Chem ; 290(42): 25710-6, 2015 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-26338706

RESUMO

Transforming growth factor ß1 (TGF-ß1) is not only elevated in airways of cystic fibrosis (CF) patients, whose airways are characterized by abnormal ion transport and mucociliary clearance, but TGF-ß1 is also associated with worse clinical outcomes. Effective mucociliary clearance depends on adequate airway hydration, governed by ion transport. Apically expressed, large-conductance, Ca(2+)- and voltage-dependent K(+) (BK) channels play an important role in this process. In this study, TGF-ß1 decreased airway surface liquid volume, ciliary beat frequency, and BK activity in fully differentiated CF bronchial epithelial cells by reducing mRNA expression of the BK γ subunit leucine-rich repeat-containing protein 26 (LRRC26) and its function. Although LRRC26 knockdown itself reduced BK activity, LRRC26 overexpression partially reversed TGF-ß1-induced BK dysfunction. TGF-ß1-induced airway surface liquid volume hyper-absorption was reversed by the BK opener mallotoxin and the clinically useful TGF-ß signaling inhibitor pirfenidone. The latter increased BK activity via rescue of LRRC26. Therefore, we propose that TGF-ß1-induced mucociliary dysfunction in CF airways is associated with BK inactivation related to a LRRC26 decrease and is amenable to treatment with clinically useful TGF-ß1 inhibitors.


Assuntos
Brônquios/patologia , Fibrose Cística/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Piridonas/farmacologia , Fator de Crescimento Transformador beta1/fisiologia , Trifosfato de Adenosina/metabolismo , Brônquios/efeitos dos fármacos , Fibrose Cística/tratamento farmacológico , Fibrose Cística/patologia , Técnicas de Silenciamento de Genes , Humanos , Depuração Mucociliar/efeitos dos fármacos , Proteínas de Neoplasias/genética , Piridonas/uso terapêutico , Fator de Crescimento Transformador beta1/antagonistas & inibidores
9.
Circ Res ; 115(4): 423-31, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24906643

RESUMO

RATIONALE: Smooth muscle cell (myocyte) large-conductance calcium (Ca)(2+)-activated potassium (BK) channels are functionally significant modulators of arterial contractility. Arterial myocytes express both pore-forming BKα and auxiliary ß1 subunits, which increase channel Ca(2+) sensitivity. Recently, several leucine-rich repeat containing (LRRC) proteins have been identified as auxiliary γ subunits that elevate the voltage sensitivity of recombinant and prostate adenocarcinoma BK channels. LRRC expression and physiological functions in native cell types are unclear. OBJECTIVE: Investigate the expression and physiological functions of leucine-rich repeat containing protein 26 (LRRC26) in arterial myocytes. METHODS AND RESULTS: Reverse transcription polymerase chain reaction and Western blotting detected LRRC26 mRNA and protein in cerebral artery myocytes. Biotinylation, immunofluorescence resonance energy transfer microscopy, and coimmunoprecipitation indicated that LRRC26 was located in close spatial proximity to, and associated with, plasma membrane BKα subunits. LRRC26 knockdown (RNAi) reduced total and surface LRRC26, but did not alter BKα or ß1, proteins in arteries. LRRC26 knockdown did not alter Ca(2+) sparks but reduced BK channel voltage sensitivity, which decreased channel apparent Ca(2+) sensitivity and transient BK current frequency and amplitude in myocytes. LRRC26 knockdown also increased myogenic tone over a range (40-100 mm Hg) of intravascular pressures, and reduced vasoconstriction to iberiotoxin and vasodilation to NS1619, BK channel inhibitors and activators, respectively. In contrast, LRRC26 knockdown did not alter depolarization (60 mmol/L K(+))-induced vasoconstriction. CONCLUSIONS: LRRC26 is expressed, associates with BKα subunits, and elevates channel voltage- and apparent Ca(2+) sensitivity in arterial myocytes to induce vasodilation. This study indicates that arterial myocytes express a functional BK channel γ subunit.


Assuntos
Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas de Neoplasias/metabolismo , Animais , Western Blotting , Sinalização do Cálcio , Artérias Cerebrais/metabolismo , Regulação da Expressão Gênica , Imunoprecipitação , Canais de Potássio Ativados por Cálcio de Condutância Alta/genética , Masculino , Potenciais da Membrana , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Proteínas de Neoplasias/genética , Subunidades Proteicas , Interferência de RNA , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Técnicas de Cultura de Tecidos , Transfecção , Vasoconstrição , Vasoconstritores/farmacologia , Vasodilatação , Vasodilatadores/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA