Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Am J Physiol Lung Cell Mol Physiol ; 318(5): L931-L942, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32130033

RESUMO

The human airway is protected by an efficient innate defense mechanism that requires healthy secretion of airway surface liquid (ASL) to clear pathogens from the lungs. Most of the ASL in the upper airway is secreted by submucosal glands. In cystic fibrosis (CF), the function of airway submucosal glands is abnormal, and these abnormalities are attributed to anomalies in ion transport across the epithelia lining the different sections of the glands that function coordinately to produce the ASL. However, the ion transport properties of most of the anatomical regions of the gland have never been measured, and there is controversy regarding which segments express CFTR. This makes it difficult to determine the glandular abnormalities that may contribute to CF lung disease. Using a noninvasive, extracellular self-referencing ion-selective electrode technique, we characterized ion transport properties in all four segments of submucosal glands from wild-type and CFTR-/- swine. In wild-type airways, the serous acini, mucus tubules, and collecting ducts secrete Cl- and Na+ into the lumen in response to carbachol and forskolin stimulation. The ciliated duct also transports Cl- and Na+ but in the opposite direction, i.e., reabsorption from the ASL, which may contribute to lowering Na+ and Cl- activities in the secreted fluid. In CFTR-/- airways, the serous acini, collecting ducts, and ciliated ducts fail to transport ions after forskolin stimulation, resulting in the production of smaller volumes of ASL with normal Cl-, Na+, and K+ concentration.


Assuntos
Células Acinares/metabolismo , Cílios/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/metabolismo , Pulmão/metabolismo , Células Acinares/efeitos dos fármacos , Células Acinares/patologia , Animais , Carbacol/farmacologia , Cátions Monovalentes , Cloretos/metabolismo , Cílios/efeitos dos fármacos , Cílios/patologia , Colforsina/farmacologia , Fibrose Cística/genética , Fibrose Cística/patologia , Regulador de Condutância Transmembrana em Fibrose Cística/deficiência , Modelos Animais de Doenças , Técnicas Eletroquímicas , Eletrodos , Deleção de Genes , Expressão Gênica , Humanos , Transporte de Íons , Pulmão/efeitos dos fármacos , Pulmão/patologia , Potássio/metabolismo , Sódio/metabolismo , Suínos
2.
Cell Microbiol ; 20(1)2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28876505

RESUMO

N-(3-Oxododecanoyl)-l-homoserine lactone (C12) is produced by Pseudomonas aeruginosa to function as a quorum-sensing molecule for bacteria-bacteria communication. C12 is also known to influence many aspects of human host cell physiology, including induction of cell death. However, the signalling pathway(s) leading to C12-triggered cell death is (are) still not completely known. To clarify cell death signalling induced by C12, we examined mouse embryonic fibroblasts deficient in "initiator" caspases or "effector" caspases. Our data indicate that C12 selectively induces the mitochondria-dependent intrinsic apoptotic pathway by quickly triggering mitochondrial outer membrane permeabilisation. Importantly, the activities of C12 to permeabilise mitochondria are independent of activation of both "initiator" and "effector" caspases. Furthermore, C12 directly induces mitochondrial outer membrane permeabilisation in vitro. Overall, our study suggests a mitochondrial apoptotic signalling pathway triggered by C12, in which C12 or its metabolite(s) acts on mitochondria to permeabilise mitochondria, leading to activation of apoptosis.


Assuntos
4-Butirolactona/análogos & derivados , Apoptose/fisiologia , Homosserina/análogos & derivados , Membranas Mitocondriais/metabolismo , Percepção de Quorum/fisiologia , 4-Butirolactona/metabolismo , Animais , Caspase 3/genética , Caspase 7/genética , Caspase 9/genética , Caspase 9/metabolismo , Linhagem Celular Tumoral , Citocromos c/metabolismo , Fibroblastos/metabolismo , Células HCT116 , Homosserina/metabolismo , Humanos , Camundongos , Camundongos Knockout , Interações Microbianas/fisiologia , Mitocôndrias/metabolismo , Pseudomonas aeruginosa/metabolismo
3.
Proc Natl Acad Sci U S A ; 111(35): 12930-5, 2014 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-25136096

RESUMO

Cystic fibrosis (CF) is an autosomal recessive genetic disorder caused by mutations in the gene encoding for the anion channel cystic fibrosis transmembrane conductance regulator (CFTR). Several organs are affected in CF, but most of the morbidity and mortality comes from lung disease. Recent data show that the initial consequence of CFTR mutation is the failure to eradicate bacteria before the development of inflammation and airway remodeling. Bacterial clearance depends on a layer of airway surface liquid (ASL) consisting of both a mucus layer that traps, kills, and inactivates bacteria and a periciliary liquid layer that keeps the mucus at an optimum distance from the underlying epithelia, to maximize ciliary motility and clearance of bacteria. The airways in CF patients and animal models of CF demonstrate abnormal ASL secretion and reduced antimicrobial properties. Thus, it has been proposed that abnormal ASL secretion in response to bacteria may facilitate the development of the infection and inflammation that characterize CF airway disease. Whether the inhalation of bacteria triggers ASL secretion, and the role of CFTR, have never been tested, however. We developed a synchrotron-based imaging technique to visualize the ASL layer and measure the effect of bacteria on ASL secretion. We show that the introduction of Pseudomonas aeruginosa and other bacteria into the lumen of intact isolated swine tracheas triggers CFTR-dependent ASL secretion by the submucosal glands. This response requires expression of the bacterial protein flagellin. In patients with CF, the inhalation of bacteria would fail to trigger ASL secretion, leading to infection and inflammation.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/microbiologia , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa/metabolismo , Traqueia/metabolismo , Traqueia/microbiologia , Animais , Fibrose Cística/imunologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/imunologia , Feminino , Haemophilus influenzae/metabolismo , Imunidade Inata/fisiologia , Masculino , Mucinas/metabolismo , Infecções por Pseudomonas/imunologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/microbiologia , Staphylococcus aureus/metabolismo , Suínos , Síncrotrons , Traqueia/imunologia
4.
J Biol Chem ; 290(11): 7247-58, 2015 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-25627690

RESUMO

Pseudomonas aeruginosa use quorum-sensing molecules, including N-(3-oxododecanoyl)-homoserine lactone (C12), for intercellular communication. C12 activated apoptosis in mouse embryo fibroblasts (MEF) from both wild type (WT) and Bax/Bak double knock-out mice (WT MEF and DKO MEF that were responsive to C12, DKOR MEF): nuclei fragmented; mitochondrial membrane potential (Δψmito) depolarized; Ca(2+) was released from the endoplasmic reticulum (ER), increasing cytosolic [Ca(2+)] (Cacyto); and caspase 3/7 was activated. DKOR MEF had been isolated from a nonclonal pool of DKO MEF that were non-responsive to C12 (DKONR MEF). RNAseq analysis, quantitative PCR, and Western blots showed that WT and DKOR MEF both expressed genes associated with cancer, including paraoxonase 2 (PON2), whereas DKONR MEF expressed little PON2. Adenovirus-mediated expression of human PON2 in DKONR MEF rendered them responsive to C12: Δψmito depolarized, Cacyto increased, and caspase 3/7 activated. Human embryonic kidney 293T (HEK293T) cells expressed low levels of endogenous PON2, and these cells were also less responsive to C12. Overexpression of PON2, but not PON2-H114Q (no lactonase activity) in HEK293T cells caused them to become sensitive to C12. Because [C12] may reach high levels in biofilms in lungs of cystic fibrosis (CF) patients, PON2 lactonase activity may control Δψmito, Ca(2+) release from the ER, and apoptosis in CF airway epithelia. Coupled with previous data, these results also indicate that PON2 uses its lactonase activity to prevent Bax- and Bak-dependent apoptosis in response to common proapoptotic drugs like doxorubicin and staurosporine, but activates Bax- and Bak-independent apoptosis in response to C12.


Assuntos
4-Butirolactona/análogos & derivados , Apoptose , Arildialquilfosfatase/metabolismo , Homosserina/análogos & derivados , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa/fisiologia , Percepção de Quorum , 4-Butirolactona/metabolismo , Animais , Células Cultivadas , Células HEK293 , Homosserina/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Camundongos , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/patologia
5.
Am J Physiol Gastrointest Liver Physiol ; 310(9): G671-81, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26847387

RESUMO

A computer model, constructed for evaluation of integrated functioning of cellular components involved in acid secretion by the gastric parietal cell, has provided new interpretations of older experimental evidence, showing the functional significance of a canalicular space separated from a mucosal bath by a gland lumen and also shedding light on basolateral Cl(-) transport. The model shows 1) changes in levels of parietal cell secretion (with stimulation or H-K-ATPase inhibitors) result mainly from changes in electrochemical driving forces for apical K(+) and Cl(-) efflux, as canalicular [K(+)] ([K(+)]can) increases or decreases with changes in apical H(+)/K(+) exchange rate; 2) H-K-ATPase inhibition in frog gastric mucosa would increase [K(+)]can similarly with low or high mucosal [K(+)], depolarizing apical membrane voltage similarly, so electrogenic H(+) pumping is not indicated by inhibition causing similar increase in transepithelial potential difference (Vt) with 4 and 80 mM mucosal K(+); 3) decreased H(+) secretion during strongly mucosal-positive voltage clamping is consistent with an electroneutral H-K-ATPase being inhibited by greatly decreased [K(+)]can (Michaelis-Menten mechanism); 4) slow initial change ("long time-constant transient") in current or Vt with clamping of Vt or current involves slow change in [K(+)]can; 5) the Na(+)-K(+)-2Cl(-) symporter (NKCC) is likely to have a significant role in Cl(-) influx, despite evidence that it is not necessary for acid secretion; and 6) relative contributions of Cl(-)/HCO3 (-) exchanger (AE2) and NKCC to Cl(-) influx would differ greatly between resting and stimulated states, possibly explaining reported differences in physiological characteristics of stimulated open-circuit Cl(-) secretion (≈H(+)) and resting short-circuit Cl(-) secretion (>>H(+)).


Assuntos
Simulação por Computador , Células Parietais Gástricas/metabolismo , Potássio/metabolismo , Animais , Bicarbonatos/metabolismo , Cloretos/metabolismo , Humanos , Bombas de Íon/metabolismo
6.
J Immunol ; 193(3): 1459-67, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24990083

RESUMO

Pseudomonas aeruginosa secrete N-(3-oxododecanoyl)-homoserine lactone (HSL-C12) as a quorum-sensing molecule to regulate bacterial gene expression. Because HSL-C12 is membrane permeant, multiple cell types in P. aeruginosa-infected airways may be exposed to HSL-C12, especially adjacent to biofilms where local (HSL-C12) may be high. Previous reports showed that HSL-C12 causes both pro- and anti-inflammatory effects. To characterize HSL-C12's pro- and anti-inflammatory effects in host cells, we measured protein synthesis, NF-κB activation, and KC (mouse IL-8) and IL-6 mRNA and protein secretion in wild-type mouse embryonic fibroblasts (MEF). To test the role of the endoplasmic reticulum stress inducer, PERK we compared these responses in PERK(-/-) and PERK-corrected PERK(-/-) MEF. During 4-h treatments of wild-type MEF, HSL-C12 potentially activated NF-κB p65 by preventing the resynthesis of IκB and increased transcription of KC and IL-6 genes (quantitative PCR). HSL-C12 also inhibited secretion of KC and/or IL-6 into the media (ELISA) both in control conditions and also during stimulation by TNF-α. HSL-C12 also activated PERK (as shown by increased phosphorylation of eI-F2α) and inhibited protein synthesis (as measured by incorporation of [(35)S]methionine by MEF). Comparisons of PERK(-/-) and PERK-corrected MEF showed that HSL-C12's effects were explained in part by activation of PERK→phosphorylation of eI-F2α→inhibition of protein synthesis→reduced IκBα production→activation of NF-κB→increased transcription of the KC gene but reduced translation and secretion of KC. HSL-C12 may be an important modulator of early (up to 4 h) inflammatory signaling in P. aeruginosa infections.


Assuntos
4-Butirolactona/análogos & derivados , Fator de Iniciação 2 em Eucariotos/fisiologia , Mediadores da Inflamação/fisiologia , Pseudomonas aeruginosa/imunologia , Percepção de Quorum/imunologia , Transdução de Sinais/imunologia , eIF-2 Quinase/fisiologia , 4-Butirolactona/fisiologia , Animais , Linhagem Celular , Estresse do Retículo Endoplasmático/imunologia , Camundongos , eIF-2 Quinase/deficiência
7.
Cell Microbiol ; 16(7): 1094-104, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24438098

RESUMO

Pseudomonas aeruginosa use N-(3-oxododecanoyl)-homoserine lactone (C12) as a quorum-sensing molecule to regulate gene expression in the bacteria. It is expected that in patients with chronic infections with P. aeruginosa, especially as biofilms, local [C12] will be high and, since C12 is lipid soluble, diffuse from the airways into the epithelium and underlying fibroblasts, capillary endothelia and white blood cells. Previous work showed that C12 has multiple effects in human host cells, including activation of apoptosis. The present work tested the involvement of Bak and Bax in C12-triggered apoptosis in mouse embryo fibroblasts (MEF) by comparing MEF isolated from embryos of wild-type (WT) and Bax(-/-) /Bak(-/-) (DKO) mice. In WT MEF C12 rapidly triggered (minutes to 2 h): activation of caspases 3/7 and 8, depolarization of mitochondrial membrane potential (Δψmito ), release of cytochrome C from mitochondria into the cytosol, blebbing of plasma membranes, shrinkage/condensation of cells and nuclei and, subsequently, cell killing. A DKO MEF line that was relatively unaffected by the Bak/Bax-dependent proapoptotic stimulants staurosporine and etoposide responded to C12 similarly to WT MEF: activation of caspase 3/7, depolarization of Δψmito and release of cytochrome C and cell death. Re-expression of Bax or Bak in DKO MEF did not alter the WT-like responses to C12 in DKO MEF. These data showed that C12 triggers novel, rapid proapoptotic Bak/Bax-independent responses that include events commonly associated with activation of both the intrinsic pathway (depolarization of Δψmito and release of cytochrome C from mitochondria into the cytosol) and the extrinsic pathway (activation of caspase 8). Unlike the proapoptotic agonists staurosporine and etoposide that release cytochrome C from mitochondria, C12's effects do not require participation of either Bak or Bax.


Assuntos
4-Butirolactona/análogos & derivados , Apoptose , Citocromos c/metabolismo , Fibroblastos/fisiologia , Mitocôndrias/metabolismo , Pseudomonas aeruginosa/fisiologia , 4-Butirolactona/fisiologia , Animais , Caspase 3/metabolismo , Caspase 7/metabolismo , Forma Celular , Fibroblastos/microbiologia , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Potencial da Membrana Mitocondrial , Camundongos , Células NIH 3T3 , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo
8.
Am J Physiol Cell Physiol ; 306(9): C844-55, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24598360

RESUMO

Pseudomonas aeruginosa secretes N-(3-oxododecanoyl)-homoserine lactone (C12) as a quorum-sensing molecule to regulate gene expression. Micromolar concentrations are found in the airway surface liquid of infected lungs. Exposure of the airway surface to C12 caused a loss of transepithelial resistance within 1 h that was accompanied by disassembly of tight junctions, as indicated by relocation of the tight junction protein zonula occludens 1 from the apical to the basolateral pole and into the cytosol of polarized human airway epithelial cell cultures (Calu-3 and primary tracheal epithelial cells). These effects were blocked by carbobenzoxy-valyl-alanyl-aspartyl-[O-methyl]-fluoromethylketone, a pan-caspase blocker, indicating that tight junction disassembly was an early event in C12-triggered apoptosis. Short-duration (10 min) pretreatment of airway epithelial (Calu-3 and JME) cells with 1 µM thapsigargin (Tg), an inhibitor of Ca(2+) uptake into the endoplasmic reticulum (ER), was found to be protective against the C12-induced airway epithelial barrier breakdown and also against other apoptosis-related effects, including shrinkage and fragmentation of nuclei, activation of caspase 3/7 (the executioner caspase in apoptosis), release of ER-targeted redox-sensitive green fluorescent protein into the cytosol, and depolarization of mitochondrial membrane potential. Pretreatment of Calu-3 airway cell monolayers with BAPTA-AM [to buffer cytosolic Ca(2+) concentration (Cacyto)] or Ca(2+)-free solution + BAPTA-AM reduced C12 activation of apoptotic events, suggesting that C12-triggered apoptosis may involve Ca(2+). Because C12 and Tg reduced Ca(2+) concentration in the ER and increased Cacyto, while Tg increased mitochondrial Ca(2+) concentration (Camito) and C12 reduced Camito, it is proposed that Tg may reduce C12-induced apoptosis in host cells not by raising Cacyto, but by preventing C12-induced decreases in Camito.


Assuntos
4-Butirolactona/análogos & derivados , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Pulmão/efeitos dos fármacos , Pseudomonas aeruginosa/metabolismo , Tapsigargina/farmacologia , Traqueia/efeitos dos fármacos , 4-Butirolactona/metabolismo , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular , Quelantes/farmacologia , Citoproteção , Impedância Elétrica , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/patologia , Ativação Enzimática , Humanos , Pulmão/metabolismo , Pulmão/microbiologia , Pulmão/patologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Cultura Primária de Células , Transporte Proteico , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo , Junções Íntimas/patologia , Fatores de Tempo , Traqueia/metabolismo , Traqueia/microbiologia , Traqueia/patologia , Proteína da Zônula de Oclusão-1/metabolismo
9.
Am J Physiol Gastrointest Liver Physiol ; 306(8): G699-710, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24578340

RESUMO

Selective inhibitors of myosin or actin function and confocal microscopy were used to test the role of an actomyosin complex in controlling morphology, trafficking, and fusion of tubulovesicles (TV) containing H-K-ATPase with the apical secretory canaliculus (ASC) of primary-cultured rabbit gastric parietal cells. In resting cells, myosin IIB and IIC, ezrin, and F-actin were associated with ASC, whereas H-K-ATPase localized to intracellular TV. Histamine caused fusion of TV with ASC and subsequent expansion resulting from HCl and water secretion; F-actin and ezrin remained associated with ASC whereas myosin IIB and IIC appeared to dissociate from ASC and relocalize to the cytoplasm. ML-7 (inhibits myosin light chain kinase) caused ASC of resting cells to collapse and most myosin IIB, F-actin, and ezrin to dissociate from ASC. TV were unaffected by ML-7. Jasplakinolide (stabilizes F-actin) caused ASC to develop large blebs to which actin, myosin II, and ezrin, as well as tubulin, were prominently localized. When added prior to stimulation, ML-7 and jasplakinolide prevented normal histamine-stimulated transformations of ASC/TV and the cytoskeleton, but they did not affect cells that had been previously stimulated with histamine. These results indicate that dynamic pools of actomyosin are required for maintenance of ASC structure in resting cells and for trafficking of TV to ASC during histamine stimulation. However, the dynamic pools of actomyosin are not required once the histamine-stimulated transformation of TV/ASC and cytoskeleton has occurred. These results also show that vesicle trafficking in parietal cells shares mechanisms with similar processes in renal collecting duct cells, neuronal synapses, and skeletal muscle.


Assuntos
Actinas/metabolismo , Miosina não Muscular Tipo IIB/metabolismo , Células Parietais Gástricas , Vesículas Transportadoras , Animais , Azepinas/farmacologia , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Fenômenos Fisiológicos Celulares/efeitos dos fármacos , Células Cultivadas , Inibidores Enzimáticos/farmacologia , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Naftalenos/farmacologia , Células Parietais Gástricas/metabolismo , Células Parietais Gástricas/patologia , Coelhos , Vesículas Transportadoras/efeitos dos fármacos , Vesículas Transportadoras/fisiologia
10.
Cell Microbiol ; 14(5): 698-709, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22233488

RESUMO

Pseudomonas aeruginosa (PA) forms biofilms in lungs of cystic fibrosis (CF) patients, a process regulated by quorum-sensing molecules including N-(3-oxododecanoyl)-l-homoserine lactone (C12). C12 (10-100 µM) rapidly triggered events commonly associated with the intrinsic apoptotic pathway in JME (CF ΔF508CFTR, nasal surface) epithelial cells: depolarization of mitochondrial (mito) membrane potential (Δψ(mito)) and release of cytochrome C (cytoC) from mitos into cytosol and activation of caspases 3/7, 8 and 9. C12 also had novel effects on the endoplasmic reticulum (release of both Ca(2+) and ER-targeted GFP and oxidized contents into the cytosol). Effects began within 5 min and were complete in 1-2 h. C12 caused similar activation of caspases and release of cytoC from mitos in Calu-3 (wtCFTR, bronchial gland) cells, showing that C12-triggered responses occurred similarly in different airway epithelial types. C12 had nearly identical effects on three key aspects of the apoptosis response (caspase 3/7, depolarization of Δψ(mito) and reduction of redox potential in the ER) in JME and CFTR-corrected JME cells (adenoviral expression), showing that CFTR was likely not an important regulator of C12-triggered apoptosis in airway epithelia. Exposure of airway cultures to biofilms from PAO1wt caused depolarization of Δψ(mito) and increases in Ca(cyto) like 10-50 µM C12. In contrast, biofilms from PAO1ΔlasI (C12 deficient) had no effect, suggesting that C12 from P. aeruginosa biofilms may contribute to accumulation of apoptotic cells that cannot be cleared from CF lungs. A model to explain the effects of C12 is proposed.


Assuntos
4-Butirolactona/análogos & derivados , Apoptose , Biofilmes/crescimento & desenvolvimento , Células Epiteliais/efeitos dos fármacos , Homosserina/análogos & derivados , Pseudomonas aeruginosa/fisiologia , 4-Butirolactona/metabolismo , 4-Butirolactona/toxicidade , Linhagem Celular , Retículo Endoplasmático/efeitos dos fármacos , Homosserina/metabolismo , Homosserina/toxicidade , Humanos , Membranas Mitocondriais/efeitos dos fármacos , Pseudomonas aeruginosa/crescimento & desenvolvimento , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/patogenicidade , Fatores de Tempo
11.
J Pharmacol Sci ; 122(3): 213-22, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23877017

RESUMO

The proposed mechanism for proton pump inhibitors (PPIs) is that PPIs are activated at low pH to the sulfenamide form, which reacts with the sulfhydryl group of cysteine(s) at the active site of the proton pump, to produce reducible disulfide-bonded PPI-proton pump conjugates. However, this mechanism cannot explain the observations that some PPI-protein conjugates are irreducible. This study was designed to investigate the chemistry of the irreducible conjugates by mass spectrometry, using three PPIs and 17 cysteine-containing peptides. While some peptides favored the formation of reducible PPI-peptide adduct, the other peptides mainly produced irreducible adducts. Characterization of the irreducible adduct revealed that the irreducible bonding required the participation of both a sulfhydryl group and a nearby primary amino group. High resolution mass spectrometry suggested a molecular structure of the irreducible adduct. These results suggested a reaction mechanism in which the PPI pyridone form reacted with an amino group and a sulfhydryl group to form an irreducible adduct. The irreducible adduct becomes the dominant product over time because of the irreversible nature of the pyridone-mediated reaction. These findings may explain the irreducible inhibition of H/K-ATPase by PPIs and their relatively slow biological turnover in vivo. [Supplementary materials: available only at http://dx.doi.org/10.1254/jphs.13058FP].


Assuntos
Cisteína/química , Peptídeos/química , Inibidores da Bomba de Prótons/química , Inibidores da Bomba de Prótons/farmacologia , 2-Piridinilmetilsulfinilbenzimidazóis/química , 2-Piridinilmetilsulfinilbenzimidazóis/farmacologia , Domínio Catalítico , Inibidores Enzimáticos , ATPase Trocadora de Hidrogênio-Potássio , Concentração de Íons de Hidrogênio , Lansoprazol/química , Lansoprazol/farmacologia , Espectrometria de Massas , Estrutura Molecular , Omeprazol/química , Omeprazol/farmacologia , Pantoprazol , Piridonas , Sulfamerazina
12.
Front Cell Infect Microbiol ; 13: 1196581, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37680748

RESUMO

Lung infection with the fungus Aspergillus fumigatus (Af) is a common complication in cystic fibrosis (CF) and is associated with loss of pulmonary function. We established a fungal epithelial co-culture model to examine the impact of Af infection on CF bronchial epithelial barrier function using Af strains 10AF and AF293-GFP, and the CFBE41o- cell line homozygous for the F508del mutation with (CF+CFTR) and without (CF) normal CFTR expression. Following exposure of the epithelial surface to Af conidia, formation of germlings (early stages of fungal growth) was detected after 9-12 hours and hyphae (mature fungal growth) after 12-24 hours. During fungal morphogenesis, bronchial epithelial cells showed signs of damage including rounding, and partial detachment after 24 hours. Fluorescently labeled conidia were internalized after 6 hours and more internalized conidia were observed in CF compared to CF+CFTR cells. Infection of the apical surface with 10AF conidia, germlings, or hyphae was performed to determine growth stage-specific effects on tight junction protein zona occludens protein 1 (ZO-1) expression and transepithelial electrical resistance (TER). In response to infection with conidia or germlings, epithelial barrier function degraded time-dependently (based on ZO-1 immunofluorescence and TER) with a delayed onset in CF+CFTR cell monolayers and required viable fungi and apical application. Infection with hyphae caused an earlier onset and faster rate of decline in TER compared to conidia and germlings. Gliotoxin, a major Af virulence factor, caused a rapid decline in TER and induced a transient chloride secretory response in CF+CFTR but not CF cells. Our findings suggest growth and internalization of Af result in deleterious effects on bronchial epithelial barrier function that occurred more rapidly in the absence of CFTR. Bronchial epithelial barrier breakdown was time-dependent and morphotype-specific and mimicked by acute administration of gliotoxin. Our study also suggests a protective role for CFTR by turning on CFTR-dependent chloride transport in response to gliotoxin, a mechanism that will support mucociliary clearance, and could delay the loss of epithelial integrity during fungal development in vivo.


Assuntos
Fibrose Cística , Gliotoxina , Micoses , Aspergillus fumigatus , Fibrose Cística/complicações , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Cloretos , Células Epiteliais
13.
Am J Physiol Cell Physiol ; 303(12): C1301-11, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23099641

RESUMO

In primary culture, the gastric parietal cell's deeply invaginated apical membrane, seen in microscopy by phalloidin binding to F-actin (concentrated in microvilli and a subapical web), is engulfed into the cell, separated from the basolateral membrane (which then becomes the complete plasma membrane), and converted, from a lacy interconnected system of canaliculi, into several separate vacuoles. In this study, vacuolar morphology was achieved by 71% of parietal cells 8 h after typical collagenase digestion of rabbit gastric mucosa, but the tight-junctional protein zonula occludens-1 (ZO-1) was completely delocalized after ∼2 h, when cells were ready for culturing. Use of low-Ca(2+) medium (4 mM EGTA) to release cells quickly from gastric glands yielded parietal cells in which ZO-1 was seen in a small spot or ring, a localization quickly lost if these cells were then cultured in normal Ca(2+) but remaining up to 20 h if they were cultured in low Ca(2+). The cells in low Ca(2+) mostly retained, at 20 h, an intermediate morphology of many bulbous canalicular expansions ("prevacuoles"), seemingly with narrow interconnections. Histamine stimulation of 20-h cells with intermediate morphology caused colocalization of proton-pumping H-K-ATPase with canaliculi and prevacuoles but little swelling of those structures, consistent with a remaining apical pore through which secreted acid could escape. Apparent canalicular interconnections, lack of stimulated swelling, and lingering ZO-1 staining indicate inhibition of membrane fission processes that separate apical from basolateral membrane and vacuoles from each other, suggesting an important role for extracellular Ca(2+) in these, and possibly other, endocytotic processes.


Assuntos
Cálcio/farmacologia , Células Parietais Gástricas/citologia , Vacúolos/metabolismo , Animais , Células Cultivadas , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Histamina/farmacologia , Microvilosidades/metabolismo , Células Parietais Gástricas/efeitos dos fármacos , Células Parietais Gástricas/metabolismo , Coelhos , Vacúolos/efeitos dos fármacos , Proteína da Zônula de Oclusão-1/metabolismo
14.
Am J Physiol Lung Cell Mol Physiol ; 303(4): L327-33, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22683572

RESUMO

The airway is kept sterile by an efficient innate defense mechanism. The cornerstone of airway defense is mucus containing diverse antimicrobial factors that kill or inactivate pathogens. Most of the mucus in the upper airways is secreted by airway submucosal glands. In patients with cystic fibrosis (CF), airway defense fails and the lungs are colonized by bacteria, usually Pseudomonas aeruginosa. Accumulating evidence suggests that airway submucosal glands contribute to CF pathogenesis by failing to respond appropriately to inhalation of bacteria. However, the regulation of submucosal glands by the innate immune system remains poorly understood. We studied the response of submucosal glands to the proinflammatory cytokines interleukin-1ß and tumor necrosis factor-α. These are released into the airway submucosa in response to infection with the bacterium P. aeruginosa and are elevated in CF airways. Stimulation with IL-1ß and TNF-α increased submucosal gland secretion in a concentration-dependent manner with a maximal secretion rate of 240 ± 20 and 190 ± 40 pl/min, respectively. The half maximal effective concentrations were 11 and 20 ng/ml, respectively. The cytokine effect was dependent on cAMP but was independent of cGMP, nitric oxide, Ca(2+), or p38 MAP kinase. Most importantly, IL-1ß- and TNF-α-stimulated secretion was blocked by the CF transmembrane conductance regulator (CFTR) blocker, CFTRinh172 (100 µmol/l) but was not affected by the Ca(2+)-activated Cl(-) channel blocker, niflumic acid (1 µmol/l). The data suggest, that during bacterial infections and resulting release of proinflammatory cytokines, the glands are stimulated to secrete fluid, and this response is mediated by cAMP-activated CFTR, a process that would fail in patients with CF.


Assuntos
Líquidos Corporais/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Interleucina-1beta/farmacologia , Muco/metabolismo , Sistema Respiratório/anatomia & histologia , Sistema Respiratório/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Líquidos Corporais/efeitos dos fármacos , Cálcio/metabolismo , AMP Cíclico/farmacologia , GMP Cíclico/farmacologia , Feminino , Técnicas In Vitro , Masculino , Muco/efeitos dos fármacos , Ácido Niflúmico/farmacologia , Óxido Nítrico/metabolismo , Sistema Respiratório/efeitos dos fármacos , Sistema Respiratório/enzimologia , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Suínos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
J Biol Chem ; 285(45): 34850-63, 2010 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-20739289

RESUMO

The ubiquitous bacterium Pseudomonas aeruginosa frequently causes hospital-acquired infections. P. aeruginosa also infects the lungs of cystic fibrosis (CF) patients and secretes N-(3-oxo-dodecanoyl)-S-homoserine lactone (3O-C12) to regulate bacterial gene expression critical for P. aeruginosa persistence. In addition to its effects as a quorum-sensing gene regulator in P. aeruginosa, 3O-C12 elicits cross-kingdom effects on host cell signaling leading to both pro- or anti-inflammatory effects. We find that in addition to these slow effects mediated through changes in gene expression, 3O-C12 also rapidly increases Cl(-) and fluid secretion in the cystic fibrosis transmembrane regulator (CFTR)-expressing airway epithelia. 3O-C12 does not stimulate Cl(-) secretion in CF cells, suggesting that lactone activates the CFTR. 3O-C12 also appears to directly activate the inositol trisphosphate receptor and release Ca(2+) from the endoplasmic reticulum (ER), lowering [Ca(2+)] in the ER and thereby activating the Ca(2+)-sensitive ER signaling protein STIM1. 3O-C12 increases cytosolic [Ca(2+)] and, strikingly, also cytosolic [cAMP], the known activator of CFTR. Activation of Cl(-) current by 3O-C12 was inhibited by a cAMP antagonist and increased by a phosphodiesterase inhibitor. Finally, a Ca(2+) buffer that lowers [Ca(2+)] in the ER similar to the effect of 3O-C12 also increased cAMP and I(Cl). The results suggest that 3O-C12 stimulates CFTR-dependent Cl(-) and fluid secretion in airway epithelial cells by activating the inositol trisphosphate receptor, thus lowering [Ca(2+)] in the ER and activating STIM1 and store-operated cAMP production. In CF airways, where CFTR is absent, the adaptive ability to rapidly flush the bacteria away is compromised because the lactone cannot affect Cl(-) and fluid secretion.


Assuntos
4-Butirolactona/análogos & derivados , Cloretos/metabolismo , AMP Cíclico/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa/metabolismo , Mucosa Respiratória/metabolismo , 4-Butirolactona/metabolismo , Ânions/metabolismo , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/genética , Linhagem Celular Transformada , AMP Cíclico/antagonistas & inibidores , AMP Cíclico/genética , Fibrose Cística/genética , Fibrose Cística/metabolismo , Fibrose Cística/microbiologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Retículo Endoplasmático/genética , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/genética , Humanos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Proteínas de Membrana/genética , Proteínas de Neoplasias/genética , Infecções por Pseudomonas/genética , Percepção de Quorum/efeitos dos fármacos , Mucosa Respiratória/microbiologia , Molécula 1 de Interação Estromal
16.
Cell Rep ; 37(1): 109795, 2021 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-34610318

RESUMO

A controversial hypothesis pertaining to cystic fibrosis (CF) lung disease is that the CF transmembrane conductance regulator (CFTR) channel fails to inhibit the epithelial Na+ channel (ENaC), yielding increased Na+ reabsorption and airway dehydration. We use a non-invasive self-referencing Na+-selective microelectrode technique to measure Na+ transport across individual folds of distal airway surface epithelium preparations from CFTR-/- (CF) and wild-type (WT) swine. We show that, under unstimulated control conditions, WT and CF epithelia exhibit similar, low rates of Na+ transport that are unaffected by the ENaC blocker amiloride. However, in the presence of the cyclic AMP (cAMP)-elevating agents forskolin+IBMX (isobutylmethylxanthine), folds of WT tissues secrete large amounts of Na+, while CFTR-/- tissues absorb small, but potentially important, amounts of Na+. In cAMP-stimulated conditions, amiloride inhibits Na+ absorption in CFTR-/- tissues but does not affect secretion in WT tissues. Our results are consistent with the hypothesis that ENaC-mediated Na+ absorption may contribute to dehydration of CF distal airways.


Assuntos
AMP Cíclico/metabolismo , Canais Epiteliais de Sódio/metabolismo , Epitélio/metabolismo , Sódio/metabolismo , 1-Metil-3-Isobutilxantina/farmacologia , Amilorida/farmacologia , Animais , Animais Geneticamente Modificados/metabolismo , Colforsina/farmacologia , Fibrose Cística , Regulador de Condutância Transmembrana em Fibrose Cística/deficiência , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Modelos Animais de Doenças , Bloqueadores do Canal de Sódio Epitelial/farmacologia , Canais Epiteliais de Sódio/química , Transporte de Íons/efeitos dos fármacos , Masculino , Suínos
17.
Infect Immun ; 77(7): 2857-65, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19451246

RESUMO

Pseudomonas aeruginosa-induced activation of NF-kappaB and secretion of proinflammatory cytokines by airway epithelial cells require that the bacteria express flagellin. We tested whether P. aeruginosa and human airway epithelial cells secrete factors that modulated this response. Experiments were performed with both the Calu-3 cell line and primary cultures of tracheal epithelial cells. P. aeruginosa strain PAK DeltafliC (flagellin knockout) did not activate NF-kappaB or interleukin-8 (IL-8) but inhibited flagellin-activated NF-kappaB by 40 to 50% and IL-8 secretion by 20 to 25%. PAK DeltafliC also inhibited NF-kappaB induced by IL-1beta and Toll-like receptor 2 agonist Pam3CSK4. Similar inhibitions were observed with strains PAK, PAO1, and PA14. The inhibitory factor was present in conditioned medium isolated from PAK DeltafliC or Calu-3 plus PAK DeltafliC, but it was not present in conditioned medium isolated from Calu-3 cells alone or from PAK DeltafliC that had been heat treated. Inhibition by PAK DeltafliC-conditioned medium was exerted from either the apical or the basolateral side of the epithelium, was enhanced in simple Ringer's solution over that in tissue culture medium, and did not result from altered pH or depletion of glucose. The inhibitory effect of conditioned medium was abolished by boiling and appeared from filtration studies to result from effects of a factor with a molecular mass of <3 kDa. These and further studies with isogenic mutants led to the conclusion that the NF-kappaB and IL-8 response of airway epithelial cells to P. aeruginosa results from a balance of proinflammatory effects of flagellin and antiinflammatory effects of a small (<3-kDa), heat-sensitive factor(s) that is not lipopolysaccharide, C12 homoserine lactone, alginate, CIF, or exotoxin A, S, T, U, or Y.


Assuntos
Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Flagelina/imunologia , Interleucina-8/antagonistas & inibidores , NF-kappa B/antagonistas & inibidores , Pseudomonas aeruginosa/imunologia , Linhagem Celular , Células Cultivadas , Meios de Cultivo Condicionados/química , Flagelina/genética , Deleção de Genes , Humanos , Interleucina-1beta/antagonistas & inibidores , Lipopeptídeos/antagonistas & inibidores , Mucosa Respiratória
18.
Sci Rep ; 9(1): 540, 2019 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-30679487

RESUMO

Inhaled hypertonic saline (HTS) treatment is used to improve lung health in patients with cystic fibrosis (CF). The current consensus is that the treatment generates an osmotic gradient that draws water into the airways and increases airway surface liquid (ASL) volume. However, there is evidence that HTS may also stimulate active secretion of ASL by airway epithelia through the activation of sensory neurons. We tested the contribution of the nervous system and airway epithelia on HTS-stimulated ASL height increase in CF and wild-type swine airway. We used synchrotron-based imaging to investigate whether airway neurons and epithelia are involved in HTS treatment-triggered ASL secretion in CFTR-/- and wild-type swine. We showed that blocking parasympathetic and sensory neurons in airway resulted in ~50% reduction of the effect of HTS treatment on ASL volume in vivo. Incubating tracheal preparations with inhibitors of epithelial ion transport across airway decreased secretory responses to HTS treatment. CFTR-/- swine ex-vivo tracheal preparations showed substantially decreased secretory response to HTS treatment after blockage of neuronal activity. Our results indicated that HTS-triggered ASL secretion is partially mediated by the stimulation of airway neurons and the subsequent activation of active epithelia secretion; osmosis accounts for only ~50% of the effect.


Assuntos
Fibrose Cística/tratamento farmacológico , Fibrose Cística/metabolismo , Cisto Mediastínico/tratamento farmacológico , Cisto Mediastínico/metabolismo , Solução Salina Hipertônica/uso terapêutico , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/metabolismo , Administração por Inalação , Animais , Animais Geneticamente Modificados , Secreções Corporais/efeitos dos fármacos , Secreções Corporais/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Feminino , Técnicas de Inativação de Genes , Transporte de Íons/efeitos dos fármacos , Masculino , Osmose/efeitos dos fármacos , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/metabolismo , Solução Salina Hipertônica/administração & dosagem , Solução Salina Hipertônica/farmacologia , Suínos
19.
Free Radic Biol Med ; 45(12): 1653-62, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18845244

RESUMO

Pyocyanin (N-methyl-1-hydroxyphenazine), a redox-active virulence factor produced by the human pathogen Pseudomonas aeruginosa, is known to compromise mucociliary clearance. Exposure of human bronchial epithelial cells to pyocyanin increased the rate of cellular release of H(2)O(2) threefold above the endogenous H(2)O(2) production. Real-time measurements of the redox potential of the cytosolic compartment using the redox sensor roGFP1 showed that pyocyanin (100 microM) oxidized the cytosol from a resting value of -318+/-5 mV by 48.0+/-4.6 mV within 2 h; a comparable oxidation was induced by 100 microM H(2)O(2). Whereas resting Cl(-) secretion was slightly activated by pyocyanin (to 10% of maximal currents), forskolin-stimulated Cl(-) secretion was inhibited by 86%. The decline was linearly related to the cytosolic redox potential (1.8% inhibition/mV oxidation). Cystic fibrosis bronchial epithelial cells homozygous for DeltaF508 CFTR failed to secrete Cl(-) in response to pyocyanin or H(2)O(2), indicating that these oxidants specifically target the CFTR and not other Cl(-) conductances. Treatment with pyocyanin also decreased total cellular glutathione levels to 62% and cellular ATP levels to 46% after 24 h. We conclude that pyocyanin is a key factor that redox cycles in the cytosol, generates H(2)O(2), depletes glutathione and ATP, and impairs CFTR function in Pseudomonas-infected lungs.


Assuntos
Cloretos/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Transporte de Íons/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Piocianina/farmacologia , Trifosfato de Adenosina/metabolismo , Brônquios/citologia , Brônquios/efeitos dos fármacos , Brônquios/metabolismo , Células Cultivadas , Fibrose Cística/metabolismo , Fibrose Cística/patologia , Citosol/metabolismo , Células Epiteliais , Glutationa/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Mutação/genética , Oxirredução
20.
Free Radic Biol Med ; 43(2): 300-16, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17603939

RESUMO

In cystic fibrosis reduced CFTR function may alter redox properties of airway epithelial cells. Redox-sensitive GFP (roGFP1) and imaging microscopy were used to measure the redox potentials of the cytosol, endoplasmic reticulum (ER), mitochondria, and cell surface of cystic fibrosis nasal epithelial cells and CFTR-corrected cells. We also measured glutathione and cysteine thiol redox states in cell lysates and apical fluids to provide coverage over a range of redox potentials and environments that might be affected by CFTR. As measured with roGFP1, redox potentials at the cell surface (approx -207+/-8 mV) and in the ER (approx -217+/-1 mV) and rates of regulation of the apical fluid and ER lumen after DTT treatment were similar for CF and CFTR-corrected cells. CF and CFTR-corrected cells had similar redox potentials in mitochondria (-344+/-9 mV) and cytosol (-322+/-7 mV). Oxidation of carboxydichlorodihydrofluorescein diacetate and of apical Amplex red occurred at equal rates in CF and CFTR-corrected cells. Glutathione and cysteine redox couples in cell lysates and apical fluid were equal in CF and CFTR-corrected cells. These quantitative estimates of organelle redox potentials combined with apical and cell measurements using small-molecule couples confirmed there were no differences in the redox properties of CF and CFTR-corrected cells.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/fisiopatologia , Mucosa Respiratória/fisiopatologia , Fibrose Cística/genética , Cistina/metabolismo , Genes Reporter , Glutationa/metabolismo , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Humanos , Microscopia Confocal , Mucosa Nasal/patologia , Mucosa Nasal/fisiopatologia , Organelas/patologia , Organelas/fisiologia , Oxirredução , Plasmídeos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA