Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 293(10): 3675-3684, 2018 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-29358325

RESUMO

The epithelial sodium channel (ENaC) mediates sodium absorption in lung, kidney, and colon epithelia. Channels in the ENaC/degenerin family possess an extracellular region that senses physicochemical changes in the extracellular milieu and allosterically regulates the channel opening. Proteolytic cleavage activates the ENaC opening, by the removal of specific segments in the finger domains of the α- and γ ENaC-subunits. Cleavage causes perturbations in the extracellular region that propagate to the channel gate. However, it is not known how the channel structure mediates the propagation of activation signals through the extracellular sensing domains. Here, to identify the structure-function determinants that mediate allosteric ENaC activation, we performed MD simulations, thiol modification of residues substituted by cysteine, and voltage-clamp electrophysiology recordings. Our simulations of an ENaC heterotetramer, α1ßα2γ, in the proteolytically cleaved and uncleaved states revealed structural pathways in the α-subunit that are responsible for ENaC proteolytic activation. To validate these findings, we performed site-directed mutagenesis to introduce cysteine substitutions in the extracellular domains of the α-, ß-, and γ ENaC-subunits. Insertion of a cysteine at the α-subunit Glu557 site, predicted to stabilize a closed state of ENaC, inhibited ENaC basal activity and retarded the kinetics of proteolytic activation by 2-fold. Our results suggest that the lower palm domain of αENaC is essential for ENaC activation. In conclusion, our integrated computational and experimental approach suggests key structure-function determinants for ENaC proteolytic activation and points toward a mechanistic model for the allosteric communication in the extracellular domains of the ENaC/degenerin family channels.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Modelos Moleculares , Regulação Alostérica , Animais , Células Cultivadas , Ativação Enzimática , Canais Epiteliais de Sódio/química , Canais Epiteliais de Sódio/genética , Humanos , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Mutação , Oócitos/citologia , Oócitos/fisiologia , Técnicas de Patch-Clamp , Conformação Proteica , Dobramento de Proteína , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Multimerização Proteica , Estabilidade Proteica , Proteólise , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Xenopus laevis
2.
J Biol Chem ; 291(8): 3682-92, 2016 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-26668308

RESUMO

Sodium absorption in epithelial cells is rate-limited by the epithelial sodium channel (ENaC) activity in lung, kidney, and the distal colon. Pathophysiological conditions, such as cystic fibrosis and Liddle syndrome, result from water-electrolyte imbalance partly due to malfunction of ENaC regulation. Because the quaternary structure of ENaC is yet undetermined, the bases of pathologically linked mutations in ENaC subunits α, ß, and γ are largely unknown. Here, we present a structural model of heterotetrameric ENaC α1ßα2γ that is consistent with previous cross-linking results and site-directed mutagenesis experiments. By using this model, we show that the disease-causing mutation αW493R rewires structural dynamics of the intersubunit interfaces α1ß and α2γ. Changes in dynamics can allosterically propagate to the channel gate. We demonstrate that cleavage of the γ-subunit, which is critical for full channel activation, does not mediate activation of ENaC by αW493R. Our molecular dynamics simulations led us to identify a channel-activating electrostatic interaction between α2Arg-493 and γGlu-348 at the α2γ interface. By neutralizing a sodium-binding acidic patch at the α1ß interface, we reduced ENaC activation of αW493R by more than 2-fold. By combining homology modeling, molecular dynamics, cysteine cross-linking, and voltage clamp experiments, we propose a dynamics-driven model for the gain-of-function in ENaC by αW493R. Our integrated computational and experimental approach advances our understanding of structure, dynamics, and function of ENaC in its disease-causing state.


Assuntos
Canais Epiteliais de Sódio/química , Modelos Moleculares , Mutação de Sentido Incorreto , Sódio/química , Regulação Alostérica , Substituição de Aminoácidos , Animais , Sítios de Ligação , Canais Epiteliais de Sódio/genética , Canais Epiteliais de Sódio/metabolismo , Humanos , Mutagênese Sítio-Dirigida , Estrutura Quaternária de Proteína , Ratos , Sódio/metabolismo , Homologia Estrutural de Proteína , Relação Estrutura-Atividade
3.
Am J Respir Crit Care Med ; 194(6): 701-10, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27014936

RESUMO

RATIONALE: In cystic fibrosis (CF) a reduction in airway surface liquid (ASL) height compromises mucociliary clearance, favoring mucus plugging and chronic bacterial infection. Inhibitors of the epithelial sodium channel (ENaC) have therapeutic potential in CF airways to reduce hyperstimulated sodium and fluid absorption to levels that can restore airway hydration. OBJECTIVES: To determine whether a novel compound (QUB-TL1) designed to inhibit protease/ENaC signaling in CF airways restores ASL volume and mucociliary function. METHODS: Protease activity was measured using fluorogenic activity assays. Differentiated primary airway epithelial cell cultures (F508del homozygotes) were used to determined ENaC activity (Ussing chamber recordings), ASL height (confocal microscopy), and mucociliary function (by tracking the surface flow of apically applied microbeads). Cell toxicity was measured using a lactate dehydrogenase assay. MEASUREMENTS AND MAIN RESULTS: QUB-TL1 inhibits extracellularly located channel activating proteases (CAPs), including prostasin, matriptase, and furin, the activities of which are observed at excessive levels at the apical surface of CF airway epithelial cells. QUB-TL1-mediated CAP inhibition results in diminished ENaC-mediated Na(+) absorption in CF airway epithelial cells caused by internalization of a prominent pool of cleaved (active) ENaCγ from the cell surface. Importantly, diminished ENaC activity correlates with improved airway hydration status and mucociliary clearance. We further demonstrate QUB-TL1-mediated furin inhibition, which is in contrast to other serine protease inhibitors (camostat mesylate and aprotinin), affords protection against neutrophil elastase-mediated ENaC activation and Pseudomonas aeruginosa exotoxin A-induced cell death. CONCLUSIONS: QUB-TL1 corrects aberrant CAP activities, providing a mechanism to delay or prevent the development of CF lung disease in a manner independent of CF transmembrane conductance regulator mutation.


Assuntos
Arginina/análogos & derivados , Fibrose Cística/tratamento farmacológico , Depuração Mucociliar/efeitos dos fármacos , Organofosfonatos/farmacologia , Mucosa Respiratória/efeitos dos fármacos , Serina Endopeptidases/efeitos dos fármacos , Bloqueadores dos Canais de Sódio/uso terapêutico , Canais de Sódio/efeitos dos fármacos , Arginina/farmacologia , Células Cultivadas , Humanos , Depuração Mucociliar/fisiologia , Mucosa Respiratória/citologia , Mucosa Respiratória/fisiologia , Canais de Sódio/fisiologia
4.
Proc Natl Acad Sci U S A ; 110(40): 15973-8, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24043776

RESUMO

The ability to maintain proper airway surface liquid (ASL) volume homeostasis is vital for mucus hydration and clearance, which are essential aspects of the mammalian lung's innate defense system. In cystic fibrosis (CF), one of the most common life-threatening genetic disorders, ASL dehydration leads to mucus accumulation and chronic infection. In normal airways, the secreted protein short palate lung and nasal epithelial clone 1 (SPLUNC1) effectively inhibits epithelial Na(+) channel (ENaC)-dependent Na(+) absorption and preserves ASL volume. In CF airways, it has been hypothesized that increased ENaC-dependent Na(+) absorption contributes to ASL depletion, and hence increased disease. However, this theory is controversial, and the mechanism for abnormal ENaC regulation in CF airways has remained elusive. Here, we show that SPLUNC1 is a pH-sensitive regulator of ENaC and is unable to inhibit ENaC in the acidic CF airway environment. Alkalinization of CF airway cultures prevented CF ASL hyperabsorption, and this effect was abolished when SPLUNC1 was stably knocked down. Accordingly, we resolved the crystal structure of SPLUNC1 to 2.8 Å. Notably, this structure revealed two pH-sensitive salt bridges that, when removed, rendered SPLUNC1 pH-insensitive and able to regulate ASL volume in acidic ASL. Thus, we conclude that ENaC hyperactivity is secondary to reduced CF ASL pH. Together, these data provide molecular insights into the mucosal dehydration associated with a range of pulmonary diseases, including CF, and suggest that future therapy be directed toward alkalinizing the pH of CF airways.


Assuntos
Fibrose Cística/patologia , Desidratação/metabolismo , Canais Epiteliais de Sódio/metabolismo , Glicoproteínas/química , Modelos Moleculares , Muco/química , Fosfoproteínas/química , Mucosa Respiratória/química , Adulto , Análise de Variância , Células Cultivadas , Cristalização , Fibrose Cística/complicações , Desidratação/etiologia , Desidratação/patologia , Técnicas de Silenciamento de Genes , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Concentração de Íons de Hidrogênio , North Carolina , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia
5.
J Biol Chem ; 289(33): 23029-23042, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24973914

RESUMO

The epithelial sodium channel (ENaC) is activated upon endoproteolytic cleavage of specific segments in the extracellular domains of the α- and γ-subunits. Cleavage is accomplished by intracellular proteases prior to membrane insertion and by surface-expressed or extracellular soluble proteases once ENaC resides at the cell surface. These cleavage events are partially regulated by intracellular signaling through an unknown allosteric mechanism. Here, using a combination of computational and experimental techniques, we show that the intracellular N terminus of γ-ENaC undergoes secondary structural transitions upon interaction with phosphoinositides. From ab initio folding simulations of the N termini in the presence and absence of phosphatidylinositol 4,5-bisphosphate (PIP2), we found that PIP2 increases α-helical propensity in the N terminus of γ-ENaC. Electrophysiology and mutation experiments revealed that a highly conserved cluster of lysines in the γ-ENaC N terminus regulates accessibility of extracellular cleavage sites in γ-ENaC. We also show that conditions that decrease PIP2 or enhance ubiquitination sharply limit access of the γ-ENaC extracellular domain to proteases. Further, the efficiency of allosteric control of ENaC proteolysis is dependent on Tyr(370) in γ-ENaC. Our findings provide an allosteric mechanism for ENaC activation regulated by the N termini and sheds light on a potential general mechanism of channel and receptor activation.


Assuntos
Canais Epiteliais de Sódio/química , Simulação de Dinâmica Molecular , Regulação Alostérica/fisiologia , Animais , Canais Epiteliais de Sódio/genética , Mutação , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteólise , Ratos
6.
J Physiol ; 592(23): 5251-68, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25260629

RESUMO

In cystic fibrosis (CF) lung disease, the absence of functional CF transmembrane conductance regulator results in Cl(-)/HCO3 (-) hyposecretion and triggers Na(+) hyperabsorption through the epithelial Na(+) channel (ENaC), which contribute to reduced airway surface liquid (ASL) pH and volume. Prostasin, a membrane-anchored serine protease with trypsin-like substrate specificity has previously been shown to activate ENaC in CF airways. However, prostasin is typically inactive below pH 7.0, suggesting that it may be less relevant in acidic CF airways. Cathepsin B (CTSB) is present in both normal and CF epithelia and is secreted into ASL, but little is known about its function in the airways. We hypothesized that the acidic ASL seen in CF airways may stimulate CTSB to activate ENaC, contributing to Na(+) hyperabsorption and depletion of CF ASL volume. In Xenopus laevis oocytes, CTSB triggered α- and γENaC cleavage and induced an increase in ENaC activity. In bronchial epithelia from both normal and CF donor lungs, CTSB localized to the apical membrane. In normal and CF human bronchial epithelial cultures, CTSB was detected at the apical plasma membrane and in the ASL. CTSB activity was significantly elevated in acidic ASL, which correlated with increased abundance of ENaC in the plasma membrane and a reduction in ASL volume. This acid/CTSB-dependent activation of ENaC was ameliorated with the cell impermeable, CTSB-selective inhibitor CA074, suggesting that CTSB inhibition may have therapeutic relevance. Taken together, our data suggest that CTSB is a pathophysiologically relevant protease that activates ENaC in CF airways.


Assuntos
Catepsina B/metabolismo , Fibrose Cística/metabolismo , Sódio/metabolismo , Animais , Catepsina B/antagonistas & inibidores , Membrana Celular/metabolismo , Células Cultivadas , Quimotripsina/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Fibrose Cística/tratamento farmacológico , Dipeptídeos/farmacologia , Canais Epiteliais de Sódio/química , Canais Epiteliais de Sódio/genética , Canais Epiteliais de Sódio/metabolismo , Feminino , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Oócitos/metabolismo , Subunidades Proteicas , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Mucosa Respiratória/metabolismo , Xenopus laevis
7.
Am J Physiol Lung Cell Mol Physiol ; 304(11): L746-56, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23542952

RESUMO

Mucus clearance is an important component of the lung's innate defense system. A failure of this system brought on by mucus dehydration is common to both cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD). Mucus clearance rates are regulated by the volume of airway surface liquid (ASL) and by ciliary beat frequency (CBF). Chronic treatment with macrolide antibiotics is known to be beneficial to both CF and COPD patients. However, chronic macrolide usage may induce bacterial resistance. We have developed a novel macrolide, 2'-desoxy-9-(S)-erythromycylamine (GS-459755), that has significantly diminished antibiotic activity against Staphylococcus aureus, Streptococcus pneumonia, Moraxella catarrhalis, and Haemophilus influenzae. Since neutrophilia frequently occurs in chronic lung disease and human neutrophil elastase (HNE) induces mucus stasis by activating the epithelial sodium channel (ENaC), we tested the ability of GS-459755 to protect against HNE-induced mucus stasis. GS-459755 had no effect on HNE activity. However, GS-459755 pretreatment protected against HNE-induced ASL volume depletion in human bronchial epithelial cells (HBECs). The effect of GS-459755 on ASL volume was dose dependent (IC50 ~3.9 µM) and comparable to the antibacterial macrolide azithromycin (IC50 ~2.4 µM). Macrolides had no significant effect on CBF or on transepithelial water permeability. However, the amiloride-sensitive transepithelial voltage, a marker of ENaC activity, was diminished by macrolide pretreatment. We conclude that GS-459755 may limit HNE-induced activation of ENaC and may be useful for the treatment of mucus dehydration in CF and COPD without inducing bacterial resistance.


Assuntos
Canais Epiteliais de Sódio/efeitos dos fármacos , Eritromicina/análogos & derivados , Elastase de Leucócito/antagonistas & inibidores , Macrolídeos/farmacologia , Muco/fisiologia , Azitromicina/farmacologia , Eritromicina/farmacologia , Humanos , Elastase de Leucócito/metabolismo , Muco/efeitos dos fármacos , Mucosa Respiratória/efeitos dos fármacos , Sistema Respiratório/metabolismo
8.
Am J Physiol Lung Cell Mol Physiol ; 305(12): L990-L1001, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24124190

RESUMO

The epithelial sodium channel (ENaC) is responsible for Na(+) and fluid absorption across colon, kidney, and airway epithelia. Short palate lung and nasal epithelial clone 1 (SPLUNC1) is a secreted, innate defense protein and an autocrine inhibitor of ENaC that is highly expressed in airway epithelia. While SPLUNC1 has a bactericidal permeability-increasing protein (BPI)-type structure, its NH2-terminal region lacks structure. Here we found that an 18 amino acid peptide, S18, which corresponded to residues G22-A39 of the SPLUNC1 NH2 terminus inhibited ENaC activity to a similar degree as full-length SPLUNC1 (∼2.5 fold), while SPLUNC1 protein lacking this region was without effect. S18 did not inhibit the structurally related acid-sensing ion channels, indicating specificity for ENaC. However, S18 preferentially bound to the ßENaC subunit in a glycosylation-dependent manner. ENaC hyperactivity is contributory to cystic fibrosis (CF) lung disease. Unlike control, CF human bronchial epithelial cultures (HBECs) where airway surface liquid (ASL) height was abnormally low (4.2 ± 0.6 µm), addition of S18 prevented ENaC-led ASL hyperabsorption and maintained CF ASL height at 7.9 ± 0.6 µm, even in the presence of neutrophil elastase, which is comparable to heights seen in normal HBECs. Our data also indicate that the ENaC inhibitory domain of SPLUNC1 may be cleaved away from the main molecule by neutrophil elastase, suggesting that it may still be active during inflammation or neutrophilia. Furthermore, the robust inhibition of ENaC by the S18 peptide suggests that this peptide may be suitable for treating CF lung disease.


Assuntos
Absorção/fisiologia , Fibrose Cística/metabolismo , Células Epiteliais/metabolismo , Glicoproteínas/metabolismo , Fosfoproteínas/metabolismo , Sódio/metabolismo , Células Cultivadas , Canais Epiteliais de Sódio/metabolismo , Glicoproteínas/genética , Humanos , Transporte de Íons/fisiologia , Elastase de Leucócito/metabolismo , Pulmão/metabolismo , Fosfoproteínas/genética , Mucosa Respiratória/metabolismo
9.
FASEB J ; 26(10): 4348-59, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22798424

RESUMO

The epithelial sodium channel (ENaC) is responsible for Na+ and fluid absorption across colon, kidney, and airway epithelia. We have previously identified SPLUNC1 as an autocrine inhibitor of ENaC. We have now located the ENaC inhibitory domain of SPLUNC1 to SPLUNC1's N terminus, and a peptide corresponding to this domain, G22-A39, inhibited ENaC activity to a similar degree as full-length SPLUNC1 (∼2.5 fold). However, G22-A39 had no effect on the structurally related acid-sensing ion channels, indicating specificity for ENaC. G22-A39 preferentially bound to the ß-ENaC subunit in a glycosylation-dependent manner. ENaC hyperactivity is contributory to cystic fibrosis (CF) lung disease. Addition of G22-A39 to CF human bronchial epithelial cultures (HBECs) resulted in an increase in airway surface liquid height from 4.2±0.6 to 7.9±0.6 µm, comparable to heights seen in normal HBECs, even in the presence of neutrophil elastase. Our data also indicate that the ENaC inhibitory domain of SPLUNC1 may be cleaved away from the main molecule by neutrophil elastase, which suggests that it may still be active during inflammation or neutrophilia. Furthermore, the robust inhibition of ENaC by the G22-A39 peptide suggests that this peptide may be suitable for treating CF lung disease.


Assuntos
Canais Iônicos Sensíveis a Ácido/metabolismo , Fibrose Cística/metabolismo , Canais Epiteliais de Sódio/metabolismo , Sódio/metabolismo , Absorção/efeitos dos fármacos , Animais , Western Blotting , Linhagem Celular , Dicroísmo Circular , Eletrofisiologia , Glicoproteínas/metabolismo , Humanos , Oócitos , Peptídeos/farmacologia , Fosfoproteínas/metabolismo , Estrutura Terciária de Proteína , Xenopus
10.
Annu Rev Physiol ; 71: 361-79, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-18928407

RESUMO

The study of human monogenic diseases [pseudohypoaldosteronism type 1 (PHA-1) and Liddle's syndrome] as well as mouse models mimicking the salt-losing syndrome (PHA-1) or salt-sensitive hypertension (Liddle's syndrome) have established the epithelial sodium channel ENaC as a limiting factor in vivo in the control of ionic composition of the extracellular fluid, regulation of blood volume and blood pressure, lung alveolar clearance, and airway mucociliary clearance. In this review, we discuss more specifically the activation of ENaC by serine proteases. Recent in vitro and in vivo experiments indicate that membrane-bound serine proteases are of critical importance in the activation of ENaC in different organs, such as the kidney, the lung, or the cochlea. Progress in understanding the basic mechanism of proteolytic activation of ENaC is accelerating, but uncertainty about the most fundamental aspects persists, leaving numerous still-unanswered questions.


Assuntos
Canais Epiteliais de Sódio/fisiologia , Serina Endopeptidases/fisiologia , Animais , Cóclea/citologia , Cóclea/fisiologia , Líquido Extracelular/fisiologia , Humanos , Rim/citologia , Rim/fisiologia , Pulmão/citologia , Pulmão/fisiologia
11.
Sci Transl Med ; 15(699): eabo7728, 2023 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-37285404

RESUMO

Unlike solid organs, human airway epithelia derive their oxygen from inspired air rather than the vasculature. Many pulmonary diseases are associated with intraluminal airway obstruction caused by aspirated foreign bodies, virus infection, tumors, or mucus plugs intrinsic to airway disease, including cystic fibrosis (CF). Consistent with requirements for luminal O2, airway epithelia surrounding mucus plugs in chronic obstructive pulmonary disease (COPD) lungs are hypoxic. Despite these observations, the effects of chronic hypoxia (CH) on airway epithelial host defense functions relevant to pulmonary disease have not been investigated. Molecular characterization of resected human lungs from individuals with a spectrum of muco-obstructive lung diseases (MOLDs) or COVID-19 identified molecular features of chronic hypoxia, including increased EGLN3 expression, in epithelia lining mucus-obstructed airways. In vitro experiments using cultured chronically hypoxic airway epithelia revealed conversion to a glycolytic metabolic state with maintenance of cellular architecture. Chronically hypoxic airway epithelia unexpectedly exhibited increased MUC5B mucin production and increased transepithelial Na+ and fluid absorption mediated by HIF1α/HIF2α-dependent up-regulation of ß and γENaC (epithelial Na+ channel) subunit expression. The combination of increased Na+ absorption and MUC5B production generated hyperconcentrated mucus predicted to perpetuate obstruction. Single-cell and bulk RNA sequencing analyses of chronically hypoxic cultured airway epithelia revealed transcriptional changes involved in airway wall remodeling, destruction, and angiogenesis. These results were confirmed by RNA-in situ hybridization studies of lungs from individuals with MOLD. Our data suggest that chronic airway epithelial hypoxia may be central to the pathogenesis of persistent mucus accumulation in MOLDs and associated airway wall damage.


Assuntos
COVID-19 , Fibrose Cística , Doença Pulmonar Obstrutiva Crônica , Humanos , Doença Pulmonar Obstrutiva Crônica/metabolismo , Pulmão/metabolismo , Muco/metabolismo , Hipóxia/metabolismo
12.
Am J Physiol Cell Physiol ; 303(7): C767-80, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22814399

RESUMO

Active ion transport and coupled osmotic water flow are essential to maintain ocular surface health. We investigated regional differences in the ion transport activities of the rat conjunctivas and compared these activities with those of cornea and lacrimal gland. The epithelial sodium channel (ENaC), sodium/glucose cotransporter 1 (Slc5a1), transmembrane protein 16 (Tmem16a, b, f, and g), cystic fibrosis transmembrane conductance regulator (Cftr), and mucin (Muc4, 5ac, and 5b) mRNA expression was characterized by RT-PCR. ENaC proteins were measured by Western blot. Prespecified regions (palpebral, fornical, and bulbar) of freshly isolated conjunctival tissues and cell cultures were studied electrophysiologically with Ussing chambers. The transepithelial electrical potential difference (PD) of the ocular surface was also measured in vivo. The effect of amiloride and UTP on the tear volume was evaluated in lacrimal gland excised rats. All selected genes were detected but with different expression patterns. We detected αENaC protein in all tissues, ßENaC in palpebral and fornical conjunctiva, and γENaC in all tissues except lacrimal glands. Electrophysiological studies of conjunctival tissues and cell cultures identified functional ENaC, SLC5A1, CFTR, and TMEM16. Fornical conjunctiva exhibited the most active ion transport under basal conditions amongst conjunctival regions. PD measurements confirmed functional ENaC-mediated Na(+) transport on the ocular surface. Amiloride and UTP increased tear volume in lacrimal gland excised rats. This study demonstrated that the different regions of the conjunctiva exhibited a spectrum of ion transport activities. Understanding the specific functions of distinct regions of the conjunctiva may foster a better understanding of the physiology maintaining hydration of the ocular surface.


Assuntos
Túnica Conjuntiva/metabolismo , Canais Iônicos/metabolismo , Transporte de Íons/fisiologia , Animais , Células Cultivadas , Feminino , Humanos , Masculino , Técnicas de Cultura de Órgãos , Ratos , Ratos Sprague-Dawley , Xenopus laevis
13.
Biochemistry ; 51(16): 3460-9, 2012 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-22471557

RESUMO

Limited proteolysis, accomplished by endopeptidases, is a ubiquitous phenomenon underlying the regulation and activation of many enzymes, receptors, and other proteins synthesized as inactive precursors. Serine proteases make up one of the largest and most conserved families of endopeptidases involved in diverse cellular activities, including wound healing, blood coagulation, and immune responses. Heteromeric α,ß,γ-epithelial sodium channels (ENaC) associated with diseases like cystic fibrosis and Liddle's syndrome are irreversibly stimulated by membrane-anchored proteases (MAPs) and furin-like convertases. Matriptase/channel activating protease-3 (CAP3) is one of the several MAPs that potently activate ENaC. Despite identification of protease cleavage sites, the basis for the enhanced susceptibility of α- and γ-ENaC to proteases remains elusive. Here, we elucidate the energetic and structural bases for activation of ENaC by CAP3. We find a region near the γ-ENaC furin site that has previously not been identified as a critical cleavage site for CAP3-mediated stimulation. We also report that CAP3 mediates cleavage of ENaC at basic residues downstream of the furin site. Our results indicate that surface proteases alone are sufficient to fully activate uncleaved ENaC and explain how ENaC in epithelia expressing surface-active proteases can appear refractory to soluble proteases. Our results support a model in which proteases prime ENaC for activation by cleaving at the furin site, and cleavage at downstream sites is accomplished by membrane surface proteases or extracellular soluble proteases. On the basis of our results, we propose a dynamics-driven "anglerfish" mechanism that explains less stringent sequence requirements for substrate recognition and cleavage by matriptase than by furin.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Serina Endopeptidases/metabolismo , Animais , Canais Epiteliais de Sódio/química , Furina/metabolismo , Humanos , Transporte de Íons , Oócitos/metabolismo , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Ratos , Serpinas/química , Serpinas/genética , Serpinas/metabolismo , Relação Estrutura-Atividade , Xenopus laevis/metabolismo
14.
Proc Natl Acad Sci U S A ; 106(27): 11412-7, 2009 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-19541605

RESUMO

Many epithelia, including the superficial epithelia of the airways, are thought to secrete "volume sensors," which regulate the volume of the mucosal lining fluid. The epithelial Na(+) channel (ENaC) is often the rate limiting factor in fluid absorption, and must be cleaved by extracellular and/or intracellular proteases before it can conduct Na(+) and absorb excess mucosal liquid, a process that can be blocked by proteases inhibitors. In the airways, airway surface liquid dilution or removal activates ENaC. Therefore, we hypothesized that endogenous proteases are membrane-anchored, whereas endogenous proteolysis inhibitors are soluble and can function as airway surface liquid volume sensors to inhibit ENaC activity. Using a proteomic approach, we identified short palate, lung, and nasal epithelial clone (SPLUNC)1 as a candidate volume sensor. Recombinant SPLUNC1 inhibited ENaC activity in both human bronchial epithelial cultures and Xenopus oocytes. Knockdown of SPLUNC1 by shRNA resulted in a failure of bronchial epithelial cultures to regulate ENaC activity and airway surface liquid volume, which was restored by adding recombinant SPLUNC1 to the airway surface liquid. Despite being able to inhibit ENaC, recombinant SPLUNC1 had little effect on extracellular serine protease activity. However, SPLUNC1 specifically bound to ENaC, preventing its cleavage and activation by serine proteases. SPLUNC1 is highly expressed in the airways, as well as in colon and kidney. Thus, we propose that SPLUNC1 is secreted onto mucosal surfaces as a soluble volume sensor whose concentration and dilution can regulate ENaC activity and mucosal volumes, including that of airway surface liquid.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Glicoproteínas/metabolismo , Fosfoproteínas/metabolismo , Processamento de Proteína Pós-Traducional , Mucosa Respiratória/fisiologia , Proteínas de Xenopus/metabolismo , Animais , Polaridade Celular , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Homeostase , Humanos , Ativação do Canal Iônico , Transporte de Íons , Oócitos/metabolismo , Ligação Proteica , Propriedades de Superfície , Tripsina/metabolismo , Xenopus
15.
J Biol Chem ; 285(42): 32227-32, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20709758

RESUMO

Cystic fibrosis (CF) is caused by mutations in the CF transmembrane conductance regulator (CFTR) that prevent its proper folding and trafficking to the apical membrane of epithelial cells. Absence of cAMP-mediated Cl(-) secretion in CF airways causes poorly hydrated airway surfaces in CF patients, and this condition is exacerbated by excessive Na(+) absorption. The mechanistic link between missing CFTR and increased Na(+) absorption in airway epithelia has remained elusive, although substantial evidence implicates hyperactivity of the epithelial Na(+) channel (ENaC). ENaC is known to be activated by selective endoproteolysis of the extracellular domains of its α- and γ-subunits, and it was recently reported that ENaC and CFTR physically associate in mammalian cells. We confirmed this interaction in oocytes by co-immunoprecipitation and found that ENaC associated with wild-type CFTR was protected from proteolytic cleavage and stimulation of open probability. In contrast, ΔF508 CFTR, the most common mutant protein in CF patients, failed to protect ENaC from proteolytic cleavage and stimulation. In normal airway epithelial cells, ENaC was contained in the anti-CFTR immunoprecipitate. In CF airway epithelial cultures, the proportion of full-length to total α-ENaC protein signal was consistently reduced compared with normal cultures. Our results identify limiting proteolytic cleavage of ENaC as a mechanism by which CFTR down-regulates Na(+) absorption.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Canais Epiteliais de Sódio/metabolismo , Animais , Células Cultivadas , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Canais Epiteliais de Sódio/genética , Humanos , Oócitos/metabolismo , Técnicas de Patch-Clamp , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Ratos , Sódio/metabolismo
16.
Pflugers Arch ; 460(1): 1-17, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20401730

RESUMO

Mammalian airways are protected from infection by a thin film of airway surface liquid (ASL) which covers airway epithelial surfaces and acts as a lubricant to keep mucus from adhering to the epithelial surface. Precise regulation of ASL volume is essential for efficient mucus clearance and too great a reduction in ASL volume causes mucus dehydration and mucus stasis which contributes to chronic airway infection. The epithelial Na(+) channel (ENaC) is the rate-limiting step that governs Na(+) absorption in the airways. Recent in vitro and in vivo data have demonstrated that ENaC is a critical determinant of ASL volume and hence mucus clearance. ENaC must be cleaved by either intracellular furin-type proteases or extracellular serine proteases to be active and conduct Na(+), and this process can be inhibited by protease inhibitors. ENaC can be regulated by multiple pathways, and once proteolytically cleaved ENaC may then be inhibited by intracellular second messengers such as cAMP and PIP(2). In the airways, however, regulation of ENaC by proteases seems to be the predominant mode of regulation since knockdown of either endogenous serine proteases such as prostasin, or inhibitors of ENaC proteolysis such as SPLUNC1, has large effects on ENaC activity in airway epithelia. In this review, we shall discuss how ENaC is proteolytically cleaved, how this process can regulate ASL volume, and how its failure to operate correctly may contribute to chronic airway disease.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Ativação do Canal Iônico , Depuração Mucociliar , Mucosa Respiratória/enzimologia , Sistema Respiratório/enzimologia , Serina Proteases/metabolismo , Animais , Canais Epiteliais de Sódio/química , Humanos , Conformação Proteica , Doenças Respiratórias/metabolismo , Sistemas do Segundo Mensageiro , Inibidores de Serina Proteinase/metabolismo , Relação Estrutura-Atividade
17.
J Clin Invest ; 117(2): 364-74, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17235394

RESUMO

The role of the cystic fibrosis transmembrane conductance regulator (CFTR) as a cAMP-dependent chloride channel on the apical membrane of epithelia is well established. However, the processes by which CFTR is regulated on the cell surface are not clear. Here we report the identification of a protein-protein interaction between CFTR and the cytoskeletal filamin proteins. Using proteomic approaches, we identified filamins as proteins that associate with the extreme CFTR N terminus. Furthermore, we identified a disease-causing missense mutation in CFTR, serine 13 to phenylalanine (S13F), which disrupted this interaction. In cells, filamins tethered plasma membrane CFTR to the underlying actin network. This interaction stabilized CFTR at the cell surface and regulated the plasma membrane dynamics and confinement of the channel. In the absence of filamin binding, CFTR was internalized from the cell surface, where it prematurely accumulated in lysosomes and was ultimately degraded. Our data demonstrate what we believe to be a previously unrecognized role for the CFTR N terminus in the regulation of the plasma membrane stability and metabolic stability of CFTR. In addition, we elucidate the molecular defect associated with the S13F mutation.


Assuntos
Proteínas Contráteis/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Proteínas dos Microfilamentos/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sítios de Ligação/genética , Linhagem Celular , Membrana Celular/metabolismo , Cricetinae , Regulador de Condutância Transmembrana em Fibrose Cística/química , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Estabilidade de Medicamentos , Filaminas , Células HeLa , Humanos , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Ligação Proteica , Conformação Proteica , Proteômica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos
18.
Mol Brain ; 12(1): 12, 2019 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-30736831

RESUMO

This study describes the functional interaction between Cav3.2 calcium channels and the Epithelial Sodium Channel (ENaC). ß-ENaC subunits showed overlapping expression with endogenous Cav3.2 calcium channels in the thalamus and hypothalamus as detected by immunostaining. Moreover, ß- and γ-ENaC subunits could be co-immunoprecipitated with Cav3.2 calcium channels from brain lysates, dorsal horn and lumbar dorsal root ganglia. Mutation of a cluster of lysines present in the intracellular N-terminus region of ß-ENaC (K4R/ K5R/ K9R/ K16R/ K23R) reduced interactions with Cav3.2 calcium channels. Αßγ-ENaC channels enhanced Cav3.2 calcium channel trafficking to the plasma membrane in tsA-201 cells. This effect was reciprocal such that Cav3.2 channel expression also enhanced ß-ENaC trafficking to the cell surface. T-type current density was increased when fully assembled αßγ-ENaC channels were transiently expressed in CAD cells, a neuronal derived cell line. Altogether, these findings reveal ENaC as an interactor and potential regulator of Cav3.2 calcium channels expressed in neuronal tissues.


Assuntos
Canais de Cálcio Tipo T/metabolismo , Canais Epiteliais de Sódio/metabolismo , Animais , Encéfalo/metabolismo , Membrana Celular/metabolismo , Canais Epiteliais de Sódio/química , Ativação do Canal Iônico , Camundongos Endogâmicos C57BL , Ligação Proteica , Subunidades Proteicas/metabolismo , Transporte Proteico , Ratos
19.
J Gen Physiol ; 150(8): 1179-1187, 2018 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-29980634

RESUMO

Epithelial Na+ channels comprise three homologous subunits (α, ß, and γ) that are regulated by alternative splicing and proteolytic cleavage. Here, we determine the basis of the reduced Na+ current (INa) that results from expression of a previously identified, naturally occurring splice variant of the α subunit (α-ENaC), in which residues 34-82 are deleted (αΔ34-82). αΔ34-82-ENaC expression with WT ß and γ subunits in Xenopus oocytes produces reduced basal INa, which can largely be recovered by exogenous trypsin. With this αΔ34-82-containing ENaC, both α and γ subunits display decreased cleavage fragments, consistent with reduced processing by furin or furin-like convertases. Data using MTSET modification of a cysteine, introduced into the degenerin locus in ß-ENaC, suggest that the reduced INa of αΔ34-82-ENaC arises from an increased population of uncleaved, near-silent ENaC, rather than from a reduced open probability spread uniformly across all channels. After treatment with brefeldin A to disrupt anterograde trafficking of channel subunits, INa in oocytes expressing αΔ34-82-ENaC is reestablished more slowly than that in oocytes expressing WT ENaC. Overnight or acute incubation of oocytes expressing WT ENaC in the pore blocker amiloride increases basal ENaC proteolytic stimulation, consistent with relief of Na+ feedback inhibition. These responses are reduced in oocytes expressing αΔ34-82-ENaC. We conclude that the α-ENaC N terminus mediates interactions that govern the delivery of cleaved and uncleaved ENaC populations to the oocyte membrane.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Furina/metabolismo , Animais , Feminino , Oócitos , Xenopus
20.
Arch Otolaryngol Head Neck Surg ; 132(12): 1352-62, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17178948

RESUMO

OBJECTIVES: To create well-differentiated cultures of normal and chronic sinusitis paranasal sinus epithelial cells and to compare their electrophysiologic properties. DESIGN: In vitro investigation using primary sinus epithelial cells, initially cultured on plastic tissue culture dishes. Cells were characterized by means of immunocytochemical analysis and then passaged to air-liquid interface culture conditions. The morphologic features of air-liquid interface cultures were assessed using light and electron microscopy. Epithelial Na(+) channel, Na(+)-K(+)-2Cl(-) cotransporter, cystic fibrosis transmembrane conductance regulator, and Ca(2+)-activated Cl(-) channel function were investigated in Ussing chambers. SUBJECTS: Specimens were obtained from 15 patients undergoing transsphenoidal pituitary procedures, tumor removal, or trauma repair and from 9 patients with chronic sinusitis. RESULTS: After culture at an air-liquid interface for 21 days, the epithelium was pseudostratified and contained basal, mucous secretory, and ciliated cells. There were no detectable morphologic differences between normal and chronic sinusitis cells. In cultures of normal cells, median basal short circuit current was 4.7 microA/cm(2), and Na(+) transport, defined as the amiloride hydrochloride-sensitive component, was approximately 20% of the total. Basal and amiloride-sensitive short circuit currents were greater in cultures of chronic sinusitis cells. Basal short circuit currents in both types of cultures were insensitive to the Cl(-) transport inhibitor bumetanide, but all responded to forskolin or uridine triphosphate. After amiloride pretreatment, forskolin and uridine triphosphate responses were greater in chronic sinusitis cells. CONCLUSIONS: We established methods for well-differentiated sinus epithelial cultures. The cells exhibited Na(+) absorption and Cl(-) secretion, and elevated rates of ion transport may be pathophysiologically relevant in chronic sinusitis.


Assuntos
Potenciais da Membrana/fisiologia , Sinusite/metabolismo , Células Cultivadas , Canais de Cloreto/metabolismo , Doença Crônica , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Humanos , Imuno-Histoquímica , Transporte de Íons/fisiologia , Microscopia Eletrônica de Transmissão , Mucosa Nasal/metabolismo , Mucosa Nasal/ultraestrutura , Sinusite/patologia , Canais de Sódio/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA