Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
1.
Nature ; 493(7431): 255-8, 2013 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-23222544

RESUMEN

Half the world's population is chronically infected with Helicobacter pylori, causing gastritis, gastric ulcers and an increased incidence of gastric adenocarcinoma. Its proton-gated inner-membrane urea channel, HpUreI, is essential for survival in the acidic environment of the stomach. The channel is closed at neutral pH and opens at acidic pH to allow the rapid access of urea to cytoplasmic urease. Urease produces NH(3) and CO(2), neutralizing entering protons and thus buffering the periplasm to a pH of roughly 6.1 even in gastric juice at a pH below 2.0. Here we report the structure of HpUreI, revealing six protomers assembled in a hexameric ring surrounding a central bilayer plug of ordered lipids. Each protomer encloses a channel formed by a twisted bundle of six transmembrane helices. The bundle defines a previously unobserved fold comprising a two-helix hairpin motif repeated three times around the central axis of the channel, without the inverted repeat of mammalian-type urea transporters. Both the channel and the protomer interface contain residues conserved in the AmiS/UreI superfamily, suggesting the preservation of channel architecture and oligomeric state in this superfamily. Predominantly aromatic or aliphatic side chains line the entire channel and define two consecutive constriction sites in the middle of the channel. Mutation of Trp 153 in the cytoplasmic constriction site to Ala or Phe decreases the selectivity for urea in comparison with thiourea, suggesting that solute interaction with Trp 153 contributes specificity. The previously unobserved hexameric channel structure described here provides a new model for the permeation of urea and other small amide solutes in prokaryotes and archaea.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Helicobacter pylori/química , Protones , Urea/metabolismo , Secuencias de Aminoácidos , Cristalografía por Rayos X , Concentración de Iones de Hidrógeno , Modelos Moleculares , Multimerización de Proteína , Estructura Secundaria de Proteína , Homología Estructural de Proteína
2.
J Bacteriol ; 200(14)2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29735759

RESUMEN

Helicobacter pylori is an organism known to colonize the normal human stomach. Previous studies have shown that the bacterium does this by elevating its periplasmic pH via the hydrolysis of urea. However, the value of the periplasmic pH was calculated indirectly from the proton motive force equation. To measure the periplasmic pH directly in H. pylori, we fused enhanced green fluorescent protein (EGFP) to the predicted twin-arginine signal peptides of HydA and KapA from H. pylori and TorA from Escherichia coli The fusion proteins were expressed in the H. pylori genome under the control of the cagA promoter. Confocal microscopic and cell fractionation/immunoblotting analyses detected TorA-EGFP in the periplasm and KapA-EGFP in both the periplasm and cytoplasm, while the mature form of HydA-EGFP was seen at low levels in the periplasm, with major cytoplasmic retention of the precursor form. With H. pylori expressing TorA-EGFP, we established a system to directly measure periplasmic pH based on the pH-sensitive fluorimetry of EGFP. These measurements demonstrated that the addition of 5 mM urea has little effect on the periplasmic pH at a medium pH higher than pH 6.5 but rapidly increases the periplasmic pH to pH 6.1 at an acidic medium pH (pH 5.0), corresponding to the opening of the proton-gated channel, UreI, and confirming the basis of gastric colonization. Measurements of the periplasmic pH in an HP0244 (FlgS)-deficient mutant of H. pylori expressing TorA-EGFP revealed a significant loss of the urea-dependent increase in the periplasmic pH at an acidic medium pH, providing additional evidence that FlgS is responsible for recruitment of urease to the inner membrane in association with UreI.IMPORTANCEHelicobacter pylori has been identified as the major cause of chronic superficial gastritis and peptic ulcer disease. In addition, persistent infection with H. pylori, which, if untreated, lasts for the lifetime of an infected individual, predisposes one to gastric malignancies, such as adenocarcinoma and mucosa-associated lymphoid tissue (MALT) lymphoma. A unique feature of the neutralophilic bacterium H. pylori is its ability to survive in the extremely acidic environment of the stomach through its acid acclimation mechanism. The presented results on measurements of periplasmic pH in H. pylori based on fluorimetry of fully active green fluorescent protein fusion proteins exported with the twin-arginine translocase system provide a reliable and rapid tool for the investigation of acid acclimation in H. pylori.


Asunto(s)
Fluorometría/métodos , Proteínas Fluorescentes Verdes/metabolismo , Helicobacter pylori/metabolismo , Antígenos Bacterianos , Proteínas Bacterianas , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Regulación Bacteriana de la Expresión Génica/fisiología , Helicobacter pylori/genética , Concentración de Iones de Hidrógeno , Mutación , Regiones Promotoras Genéticas , Urea/metabolismo , Urea/farmacología
3.
J Cell Sci ; 129(12): 2394-406, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27142834

RESUMEN

FXYD5 (also known as dysadherin), a regulatory subunit of the Na,K-ATPase, impairs intercellular adhesion by a poorly understood mechanism. Here, we determined whether FXYD5 disrupts the trans-dimerization of Na,K-ATPase molecules located in neighboring cells. Mutagenesis of the Na,K-ATPase ß1 subunit identified four conserved residues, including Y199, that are crucial for the intercellular Na,K-ATPase trans-dimerization and adhesion. Modulation of expression of FXYD5 or of the ß1 subunit with intact or mutated ß1-ß1 binding sites demonstrated that the anti-adhesive effect of FXYD5 depends on the presence of Y199 in the ß1 subunit. Immunodetection of the plasma membrane FXYD5 was prevented by the presence of O-glycans. Partial FXYD5 deglycosylation enabled antibody binding and showed that the protein level and the degree of O-glycosylation were greater in cancer than in normal cells. FXYD5-induced impairment of adhesion was abolished by both genetic and pharmacological inhibition of FXYD5 O-glycosylation. Therefore, the extracellular O-glycosylated domain of FXYD5 impairs adhesion by interfering with intercellular ß1-ß1 interactions, suggesting that the ratio between FXYD5 and α1-ß1 heterodimer determines whether the Na,K-ATPase acts as a positive or negative regulator of intercellular adhesion.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Multimerización de Proteína , Subunidades de Proteína/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Células A549 , Aminoácidos/metabolismo , Animales , Especificidad de Anticuerpos , Adhesión Celular , Línea Celular Tumoral , Membrana Celular/metabolismo , Perros , Células Epiteliales/metabolismo , Técnicas de Silenciamiento del Gen , Glicosilación , Células HEK293 , Humanos , Canales Iónicos , Células de Riñón Canino Madin Darby , Ratones , Proteínas de Microfilamentos , Unión Proteica , Subunidades de Proteína/química , Ratas , ATPasa Intercambiadora de Sodio-Potasio/química
4.
Helicobacter ; 23(3): e12490, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29696729

RESUMEN

BACKGROUND: The pathogen Helicobacter pylori encounters many stressors as it transits to and infects the gastric epithelium. Gastric acidity is the predominate stressor encountered by the bacterium during initial infection and establishment of persistent infection. H. pylori initiates a rapid response to acid to maintain intracellular pH and proton motive force appropriate for a neutralophile. However, acid sensing by H. pylori may also serve as a transcriptional trigger to increase the levels of other pathogenic factors needed to subvert host defenses such as acid acclimation, antioxidants, flagellar synthesis and assembly, and CagA secretion. MATERIALS AND METHODS: Helicobacter pylori were acid challenged at pH 3.0, 4.5, 6.0 vs nonacidic pH for 4 hours in the presence of urea, followed by RNA-seq analysis and qPCR. Cytoplasmic pH was monitored under the same conditions. RESULTS: About 250 genes were induced, and an equal number were repressed at acidic pHs. Genes encoding for antioxidant proteins, flagellar structural proteins, particularly class 2 genes, T4SS/Cag-PAI, Fo F1 -ATPase, and proteins involved in acid acclimation were highly expressed at acidic pH. Cytoplasmic pH decreased from 7.8 at pHout of 8.0 to 6.0 at pHout of 3.0. CONCLUSIONS: These results suggest that increasing extracellular or intracellular acidity or both are detected by the bacterium and serve as a signal to initiate increased production of protective and pathogenic factors needed to counter host defenses for persistent infection. These changes are dependent on degree of acidity and time of acid exposure, triggering a coordinated response to the environment required for colonization.


Asunto(s)
Ácidos/metabolismo , Regulación Bacteriana de la Expresión Génica , Helicobacter pylori/genética , Estómago/microbiología , Transcriptoma/genética , Adaptación Fisiológica , Proteínas Bacterianas/genética , Medios de Cultivo , Helicobacter pylori/fisiología , Concentración de Iones de Hidrógeno , Familia de Multigenes , Proteoma/genética , ARN Bacteriano/genética , Ureasa/genética , Factores de Virulencia/genética
5.
J Biol Chem ; 291(44): 23159-23174, 2016 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-27624940

RESUMEN

The Na,K-ATPase α2 subunit plays a key role in cardiac muscle contraction by regulating intracellular Ca2+, whereas α1 has a more conventional role of maintaining ion homeostasis. The ß subunit differentially regulates maturation, trafficking, and activity of α-ß heterodimers. It is not known whether the distinct role of α2 in the heart is related to selective assembly with a particular one of the three ß isoforms. We show here by immunofluorescence and co-immunoprecipitation that α2 is preferentially expressed with ß2 in T-tubules of cardiac myocytes, forming α2ß2 heterodimers. We have expressed human α1ß1, α2ß1, α2ß2, and α2ß3 in Pichia pastoris, purified the complexes, and compared their functional properties. α2ß2 and α2ß3 differ significantly from both α2ß1 and α1ß1 in having a higher K0.5K+ and lower K0.5Na+ for activating Na,K-ATPase. These features are the result of a large reduction in binding affinity for extracellular K+ and shift of the E1P-E2P conformational equilibrium toward E1P. A screen of perhydro-1,4-oxazepine derivatives of digoxin identified several derivatives (e.g. cyclobutyl) with strongly increased selectivity for inhibition of α2ß2 and α2ß3 over α1ß1 (range 22-33-fold). Molecular modeling suggests a possible basis for isoform selectivity. The preferential assembly, specific T-tubular localization, and low K+ affinity of α2ß2 could allow an acute response to raised ambient K+ concentrations in physiological conditions and explain the importance of α2ß2 for cardiac muscle contractility. The high sensitivity of α2ß2 to digoxin derivatives explains beneficial effects of cardiac glycosides for treatment of heart failure and potential of α2ß2-selective digoxin derivatives for reducing cardiotoxicity.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Transporte de Catión/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Inhibidores Enzimáticos/química , Miocardio/enzimología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Adenosina Trifosfatasas/antagonistas & inhibidores , Adenosina Trifosfatasas/química , Animales , Proteínas de Transporte de Catión/antagonistas & inhibidores , Proteínas de Transporte de Catión/química , Moléculas de Adhesión Celular Neuronal/antagonistas & inhibidores , Moléculas de Adhesión Celular Neuronal/química , Dimerización , Inhibidores Enzimáticos/metabolismo , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Ratones , Miocardio/química , Potasio/química , Potasio/metabolismo , Sodio/química , Sodio/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores , ATPasa Intercambiadora de Sodio-Potasio/química , ATPasa Intercambiadora de Sodio-Potasio/genética
6.
Biochem J ; 473(12): 1703-18, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27048593

RESUMEN

Septins are a family of cytoskeletal GTP-binding proteins that assemble into membrane-associated hetero-oligomers and organize scaffolds for recruitment of cytosolic proteins or stabilization of membrane proteins. Septins have been implicated in a diverse range of cancers, including gastric cancer, but the underlying mechanisms remain unclear. The hypothesis tested here is that septins contribute to cancer by stabilizing the receptor tyrosine kinase ErbB2, an important target for cancer treatment. Septins and ErbB2 were highly over-expressed in gastric cancer cells. Immunoprecipitation followed by MS analysis identified ErbB2 as a septin-interacting protein. Knockdown of septin-2 or cell exposure to forchlorfenuron (FCF), a well-established inhibitor of septin oligomerization, decreased surface and total levels of ErbB2. These treatments had no effect on epidermal growth factor receptor (EGFR), emphasizing the specificity and functionality of the septin-ErbB2 interaction. The level of ubiquitylated ErbB2 at the plasma membrane was elevated in cells treated with FCF, which was accompanied by a decrease in co-localization of ErbB2 with septins at the membrane. Cathepsin B inhibitor, but not bafilomycin or lactacystin, prevented FCF-induced decrease in total ErbB2 by increasing accumulation of ubiquitylated ErbB2 in lysosomes. Therefore, septins protect ErbB2 from ubiquitylation, endocytosis and lysosomal degradation. The FCF-induced degradation pathway is distinct from and additive with the degradation induced by inhibiting ErbB2 chaperone Hsp90. These results identify septins as novel regulators of ErbB2 expression that contribute to the remarkable stabilization of the receptor at the plasma membrane of cancer cells and may provide a basis for the development of new ErbB2-targeting anti-cancer therapies.


Asunto(s)
Receptor ErbB-2/metabolismo , Septinas/metabolismo , Neoplasias Gástricas/metabolismo , Western Blotting , Línea Celular Tumoral , Cromatografía Liquida , Citoesqueleto/metabolismo , Humanos , Inmunoprecipitación , Compuestos de Fenilurea/farmacología , Unión Proteica/efectos de los fármacos , Piridinas/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/fisiología , Receptor ErbB-2/genética , Septinas/antagonistas & inhibidores , Septinas/genética , Transducción de Señal/fisiología , Espectrometría de Masas en Tándem , Ubiquitinación/efectos de los fármacos
7.
J Biol Chem ; 290(9): 5280-97, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25575596

RESUMEN

Septins are a family of 14 cytoskeletal proteins that dynamically form hetero-oligomers and organize membrane microdomains for protein complexes. The previously reported interactions with SNARE proteins suggested the involvement of septins in exocytosis. However, the contradictory results of up- or down-regulation of septin-5 in various cells and mouse models or septin-4 in mice suggested either an inhibitory or a stimulatory role for these septins in exocytosis. The involvement of the ubiquitously expressed septin-2 or general septin polymerization in exocytosis has not been explored to date. Here, by nano-LC with tandem MS and immunoblot analyses of the septin-2 interactome in mouse brain, we identified not only SNARE proteins but also Munc-18-1 (stabilizes assembled SNARE complexes), N-ethylmaleimide-sensitive factor (NSF) (disassembles SNARE complexes after each membrane fusion event), and the chaperones Hsc70 and synucleins (maintain functional conformation of SNARE proteins after complex disassembly). Importantly, α-soluble NSF attachment protein (SNAP), the adaptor protein that mediates NSF binding to the SNARE complex, did not interact with septin-2, indicating that septins undergo reorganization during each exocytosis cycle. Partial depletion of septin-2 by siRNA or impairment of septin dynamics by forchlorfenuron inhibited constitutive and stimulated exocytosis of secreted and transmembrane proteins in various cell types. Forchlorfenuron impaired the interaction between SNAP-25 and its chaperone Hsc70, decreasing SNAP-25 levels in cultured neuroendocrine cells, and inhibited both spontaneous and stimulated acetylcholine secretion in mouse motor neurons. The results demonstrate a stimulatory role of septin-2 and the dynamic reorganization of septin oligomers in exocytosis.


Asunto(s)
Encéfalo/metabolismo , Exocitosis , Proteoma/metabolismo , Septinas/metabolismo , Animales , Western Blotting , Encéfalo/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Perros , Femenino , Células HEK293 , Humanos , Células de Riñón Canino Madin Darby , Masculino , Ratones Endogámicos BALB C , Microscopía Confocal , Células PC12 , Compuestos de Fenilurea/farmacología , Unión Proteica/efectos de los fármacos , Multimerización de Proteína , Proteómica , Piridinas/farmacología , Interferencia de ARN , Ratas , Septinas/química , Septinas/genética , Proteína 25 Asociada a Sinaptosomas/metabolismo
8.
J Cell Sci ; 127(Pt 15): 3294-308, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24928902

RESUMEN

Proteolytic cleavage of synaptosomal-associated protein 25 by the light chain of botulinum neurotoxin type A (LCA) results in a blockade of neurotransmitter release that persists for several months in motor neurons. The L428A/L429A mutation in LCA is known to significantly shorten both the proteolytic and neuroparalytic effects of the neurotoxin in mice. To elucidate the cellular mechanism for LCA longevity, we studied the effects of L428A/L429A mutation on the interactome, localization and stability of LCA expressed in cultured neuronal cells. Mass spectrometry analysis of the LCA interactome showed that the mutation prevented the interaction of LCA with septins. The wild-type LCA was concentrated in plasma-membrane-associated clusters, colocalizing with septins-2 and septin-7, which accumulated in these clusters only in the presence of LCA. The L428A/L429A mutation decreased co-clustering of LCA and septins and accelerated proteasomal and non-proteasomal degradation of LCA. Similarly, the impairment of septin oligomerization by forchlorfenuron or silencing of septin-2 prevented LCA interaction and clustering with septins and increased LCA degradation. Therefore, the dileucine-mediated LCA-septin co-clustering is crucial for the long-lasting stabilization of LCA-related proteolytic and presumably neuroparalytic activity.


Asunto(s)
Toxinas Botulínicas Tipo A/metabolismo , Membrana Celular/metabolismo , Neuronas/fisiología , Síndromes de Neurotoxicidad/metabolismo , Septinas/metabolismo , Animales , Toxinas Botulínicas Tipo A/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Humanos , Ratones , Mutación/genética , Neuronas/microbiología , Síndromes de Neurotoxicidad/microbiología , Compuestos de Fenilurea/farmacología , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Multimerización de Proteína/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , Piridinas/farmacología , ARN Interferente Pequeño/genética , Septinas/genética
9.
Helicobacter ; 21(1): 69-81, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25997502

RESUMEN

BACKGROUND: The pH-sensitive Helicobacter pylori ArsRS two-component system (TCS) aids survival of this neutralophile in the gastric environment by directly sensing and responding to environmental acidity. ArsS is required for acid-induced trafficking of urease and its accessory proteins to the inner membrane, allowing rapid, urea-dependent cytoplasmic and periplasmic buffering. Expression of ArsR, but not its phosphorylation, is essential for bacterial viability. The aim of this study was to characterize the roles of ArsS and ArsR in the response of H. pylori to acid. MATERIALS AND METHODS: Wild-type H. pylori and an arsR(D52N) phosphorylation-deficient strain were incubated at acidic or neutral pH. Gene and protein expression, survival, membrane trafficking of urease proteins, urease activity, and internal pH were studied. RESULTS: Phosphorylation of ArsR is not required for acid survival. ArsS-driven trafficking of urease proteins to the membrane in acid, required for recovery of internal pH, is independent of ArsR phosphorylation. ArsR phosphorylation increases expression of the urease gene cluster, and the loss of negative feedback in a phosphorylation-deficient mutant leads to an increase in total urease activity. CONCLUSIONS: ArsRS has a dual function in acid acclimation: regulation of urease trafficking to UreI at the cytoplasmic membrane, driven by ArsS, and regulation of urease gene cluster expression, driven by phosphorylation of ArsR. ArsS and ArsR work through phosphorylation-dependent and phosphorylation-independent regulatory mechanisms to impact acid acclimation and allow gastric colonization. Furthering understanding of the intricacies of acid acclimation will impact the future development of targeted, nonantibiotic treatment regimens.


Asunto(s)
Ácidos/metabolismo , Proteínas Bacterianas/metabolismo , Infecciones por Helicobacter/microbiología , Helicobacter pylori/metabolismo , Proteínas Bacterianas/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Regulación Bacteriana de la Expresión Génica , Helicobacter pylori/genética , Humanos , Fosforilación , Transporte de Proteínas , Ureasa/genética , Ureasa/metabolismo
10.
Curr Gastroenterol Rep ; 18(7): 33, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27177639

RESUMEN

Helicobacter pylori infects about 50 % of the world's population, causing at a minimum chronic gastritis. A subset of infected patients will ultimately develop gastric or duodenal ulcer disease, gastric adenocarcinoma, or MALT (mucosa-associated lymphoid tissue) lymphoma. Eradication of H. pylori requires complex regimens that include acid suppression and multiple antibiotics. The efficacy of treatment using what were once considered standard regimens have declined in recent years, mainly due to widespread development of antibiotic resistance. Addition of bismuth to standard triple therapy regimens, use of alternate antibiotics, or development of alternative regimens using known therapies in novel combinations have improved treatment efficacy in specific populations, but overall success of eradication remains less than ideal. Novel regimens under investigation either in vivo or in vitro, involving increased acid suppression ideally with fewer antibiotics or development of non-antibiotic treatment targets, show promise for future therapy.


Asunto(s)
Gastritis/tratamiento farmacológico , Gastritis/microbiología , Infecciones por Helicobacter/tratamiento farmacológico , Helicobacter pylori , Antibacterianos/uso terapéutico , Bismuto/uso terapéutico , Enfermedad Crónica , Farmacorresistencia Bacteriana , Quimioterapia Combinada , Humanos , Inhibidores de la Bomba de Protones/uso terapéutico
11.
J Cell Sci ; 125(Pt 6): 1605-16, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22328500

RESUMEN

Epithelial junctions depend on intercellular interactions between ß(1) subunits of the Na(+)/K(+)-ATPase molecules of neighboring cells. The interaction between dog and rat subunits is less effective than the interaction between two dog ß(1) subunits, indicating the importance of species-specific regions for ß(1)-ß(1) binding. To identify these regions, the species-specific amino acid residues were mapped on a high-resolution structure of the Na(+)/K(+)-ATPase ß(1) subunit to select those exposed towards the ß(1) subunit of the neighboring cell. These exposed residues were mutated in both dog and rat YFP-linked ß(1) subunits (YFP-ß(1)) and also in the secreted extracellular domain of the dog ß(1) subunit. Five rat-like mutations in the amino acid region spanning residues 198-207 of the dog YFP-ß(1) expressed in Madin-Darby canine kidney (MDCK) cells decreased co-precipitation of the endogenous dog ß(1) subunit with YFP-ß(1) to the level observed between dog ß(1) and rat YFP-ß(1). In parallel, these mutations impaired the recognition of YFP-ß(1) by the dog-specific antibody that inhibits cell adhesion between MDCK cells. Accordingly, dog-like mutations in rat YFP-ß(1) increased both the (YFP-ß(1))-ß(1) interaction in MDCK cells and recognition by the antibody. Conversely, rat-like mutations in the secreted extracellular domain of the dog ß(1) subunit increased its interaction with rat YFP-ß(1) in vitro. In addition, these mutations resulted in a reduction of intercellular adhesion between rat lung epithelial cells following addition of the secreted extracellular domain of the dog ß(1) subunit to a cell suspension. Therefore, the amino acid region 198-207 is crucial for both trans-dimerization of the Na(+)/K(+)-ATPase ß(1) subunits and cell-cell adhesion.


Asunto(s)
Comunicación Celular/fisiología , Células Epiteliales/enzimología , ATPasa Intercambiadora de Sodio-Potasio/química , ATPasa Intercambiadora de Sodio-Potasio/fisiología , Secuencia de Aminoácidos , Aminoácidos/química , Aminoácidos/genética , Aminoácidos/fisiología , Animales , Línea Celular , Perros , Células Epiteliales/citología , Datos de Secuencia Molecular , Multimerización de Proteína , Estructura Terciaria de Proteína/genética , Ratas , ATPasa Intercambiadora de Sodio-Potasio/genética , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
12.
J Bacteriol ; 195(3): 444-52, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23104809

RESUMEN

Urease is an essential component of gastric acid acclimation by Helicobacter pylori. The increased level of urease in gastric acidity is due, in part, to acid activation of the two-component system consisting of the membrane sensor HP0165 (ArsS) and its response regulator HP0166 (ArsR), which regulates transcription of the seven genes in two separate operons (ureAB and ureIEFGH) of the urease gene cluster. Recently, we identified a novel cis-encoded antisense small RNA, 5'ureB-sRNA, targeted at the 5' end of ureB, which downregulates ureAB expression by truncation of the ureAB transcript at neutral pH. It is not known whether the truncated transcript is due to transcription termination or processing of the full-length mRNA by codegradation of a ureAB mRNA-sRNA hybrid complex. S1 nuclease mapping assays show that the truncated transcript is due to transcription termination. Further studies using an in vitro transcription assay found that 5'ureB-sRNA promotes premature termination of transcription of ureAB mRNA. These results suggest that the antisense small RNA 5'ureB-sRNA downregulates ureAB expression by enhancing transcription termination 5' of ureB. With this mechanism, a limited amount of 5'ureB-sRNA is sufficient to regulate the relatively high level of ureAB transcript.


Asunto(s)
Proteínas Bacterianas/metabolismo , Helicobacter pylori/metabolismo , ARN Bacteriano/metabolismo , Terminación de la Transcripción Genética/fisiología , Ureasa/metabolismo , Proteínas Bacterianas/genética , Secuencia de Bases , Regulación hacia Abajo , Regulación Bacteriana de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica , Helicobacter pylori/genética , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , ARN Bacteriano/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ureasa/genética
13.
J Biol Chem ; 287(31): 26115-25, 2012 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-22696220

RESUMEN

To catalyze ion transport, the Na,K-ATPase must contain one α and one ß subunit. When expressed by transfection in various expression systems, each of the four α subunit isoforms can assemble with each of the three ß subunit isoforms and form an active enzyme, suggesting the absence of selective α-ß isoform assembly. However, it is unknown whether in vivo conditions the α-ß assembly is random or isoform-specific. The α(2)-ß(2) complex was selectively immunoprecipitated by both anti-α(2) and anti-ß(2) antibodies from extracts of mouse brain, which contains cells co-expressing multiple Na,K-ATPase isoforms. Neither α(1)-ß(2) nor α(2)-ß(1) complexes were detected in the immunoprecipitates. Furthermore, in MDCK cells co-expressing α(1), ß(1), and ß(2) isoforms, a greater fraction of the ß(2) subunits was unassembled with α(1) as compared with that of the ß(1) subunits, indicating preferential association of the α(1) isoform with the ß(1) isoform. In addition, the α(1)-ß(2) complex was less resistant to various detergents than the α(1)-ß(1) complex isolated from MDCK cells or the α(2)-ß(2) complex isolated from mouse brain. Therefore, the diversity of the α-ß Na,K-ATPase heterodimers in vivo is determined not only by cell-specific co-expression of particular isoforms, but also by selective association of the α and ß subunit isoforms.


Asunto(s)
Multimerización de Proteína , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Unión Competitiva , Encéfalo/enzimología , Membrana Celular/enzimología , Células Cultivadas , Perros , Estabilidad de Enzimas , Humanos , Inmunoprecipitación , Isoenzimas/metabolismo , Riñón , Proteínas Luminiscentes/metabolismo , Ratones , Modelos Moleculares , Especificidad de Órganos , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Cuaternaria de Proteína , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Nervio Ciático/enzimología , ATPasa Intercambiadora de Sodio-Potasio/química , ATPasa Intercambiadora de Sodio-Potasio/aislamiento & purificación
14.
Am J Physiol Renal Physiol ; 304(1): F41-8, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23077097

RESUMEN

Metabolic acidosis is thought to exacerbate chronic kidney disease in part by stimulating the release of potentially injurious substances. To define the genes whose expression is affected by exposure to an acidic milieus, we examined the effect of exposure of MDCK cells to pH 7.4 and pH 7.0 for 24 h on gene expression using a canine derived microarray. Exposure to this pH stress for 24 h led to increased expression of 278 genes (2.2% of the transcriptome) by at least 2-fold and 60 of these (21%) were upregulated by >3-fold. On the other hand, 186 genes (1.5% of the transcriptome) were downregulated by at least 2-fold and 16 of these (9%) were downregulated by 3-fold or more. Ten percent of the genes upregulated by at least threefold encode proinflammatory cytokine proteins, including colony stimulating factor 2, chemokine ligand 7, chemokine ligand 20, chemokine ligand 8, and interleukin-1α. Two others encode metallopeptidases. The most highly upregulated gene encodes a protein, lubricin, shown to be important in preventing cartilage damage and in tissue injury or repair. Upregulation of four genes was confirmed by quantitative PCR. Housekeeping genes were not increased. To examine the effect of decreasing medium pH, we measured intracellular pH (pH(i)) using 2,7-bis (2-carboxyethyl)5-carboxyfluorescein. With extracellular pH (pH(o)) of 7.0, pH(i) fell and remained depressed. These findings suggest that a pH stress alone can increase renal expression of proinflammatory and other genes that contribute to renal injury.


Asunto(s)
Acidosis/fisiopatología , Citocinas/biosíntesis , Riñón/metabolismo , Animales , Perros , Regulación hacia Abajo , Fluoresceínas , Concentración de Iones de Hidrógeno , Células de Riñón Canino Madin Darby , Proteínas de Microfilamentos/biosíntesis , Quinasa de Cadena Ligera de Miosina/biosíntesis , Análisis por Matrices de Proteínas , Factor de Transcripción CHOP/biosíntesis , Transcriptoma/efectos de los fármacos , Regulación hacia Arriba
15.
Am J Physiol Gastrointest Liver Physiol ; 305(10): G731-9, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23989011

RESUMEN

Gastric infection by Helicobacter pylori is the most common cause of ulcer disease and gastric cancer. The mechanism of progression from gastritis and inflammation to ulcers and cancer in a fraction of those infected is not definitively known. Significant acidity is unique to the gastric environment and is required for ulcer development. The interplay between gastric acidity and H. pylori pathogenesis is important in progression to advanced disease. The aim of this study was to characterize the impact of acid on gastric epithelial integrity and cytokine release and how H. pylori infection alters these responses. Human gastric epithelial (HGE-20) cells were grown on porous inserts, and survival, barrier function, and cytokine release were studied at various apical pH levels in the presence and absence of H. pylori. With apical acidity, gastric epithelial cells demonstrate increased barrier function, as evidenced by increased transepithelial electrical resistance (TEER) and decreased paracellular permeability. This effect is reduced in the presence of wild-type, but not urease knockout, H. pylori. The epithelial inflammatory response is also modulated by acidity and H. pylori infection. Without H. pylori, epithelial IL-8 release decreases in acid, while IL-6 release increases. In the presence of H. pylori, acidic pH diminishes the magnitude of the previously reported increase in IL-8 and IL-6 release. H. pylori interferes with the gastric epithelial response to acid, contributing to altered barrier function and inflammatory response. H. pylori diminishes acid-induced tightening of cell junctions in a urease-dependent manner, suggesting that local pH elevation promotes barrier compromise and progression to mucosal damage.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Células Epiteliales/microbiología , Helicobacter pylori/metabolismo , Línea Celular Tumoral , Medios de Cultivo/química , Impedancia Eléctrica , Fenómenos Electrofisiológicos , Regulación Bacteriana de la Expresión Génica , Helicobacter pylori/genética , Humanos , Concentración de Iones de Hidrógeno
16.
Helicobacter ; 18(5): 363-72, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23600974

RESUMEN

BACKGROUND: Helicobacter pylori, a neutralophile, colonizes the acidic environment of the human stomach by employing acid acclimation mechanisms that regulate periplasmic and cytoplasmic pH. The regulation of urease activity is central to acid acclimation. Inactive urease apoenzyme, UreA/B, requires nickel for activation. Accessory proteins UreE, F, G, and H are required for nickel insertion into apoenzyme. The ExbB/ExbD/TonB complex transfers energy from the inner to outer membrane, providing the driving force for nickel uptake. Therefore, the aim of this study was to determine the contribution of ExbD to pH homeostasis. MATERIALS AND METHODS: A nonpolar exbD knockout was constructed and survival, growth, urease activity, and membrane potential were determined in comparison with wildtype. RESULTS: Survival of the ΔexbD strain was significantly reduced at pH 3.0. Urease activity as a function of pH and UreI activation was similar to the wildtype strain, showing normal function of the proton-gated urea channel, UreI. The increase in total urease activity over time in acid seen in the wildtype strain was abolished in the ΔexbD strain, but recovered in the presence of supraphysiologic nickel concentrations, demonstrating that the effect of the ΔexbD mutant is due to loss of a necessary constant supply of nickel. In acid, ΔexbD also decreased its ability to maintain membrane potential and periplasmic buffering in the presence of urea. CONCLUSIONS: ExbD is essential for maintenance of periplasmic buffering and membrane potential by transferring energy required for nickel uptake, making it a potential nonantibiotic target for H. pylori eradication.


Asunto(s)
Adaptación Fisiológica , Proteínas Bacterianas/metabolismo , Helicobacter pylori/fisiología , Homeostasis , Proteínas Bacterianas/genética , Membrana Celular/fisiología , Técnicas de Inactivación de Genes , Helicobacter pylori/efectos de los fármacos , Helicobacter pylori/genética , Helicobacter pylori/crecimiento & desarrollo , Humanos , Concentración de Iones de Hidrógeno , Potenciales de la Membrana , Viabilidad Microbiana/efectos de los fármacos , Ureasa/metabolismo
17.
J Bacteriol ; 194(20): 5545-51, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22865848

RESUMEN

Helicobacter pylori survives and grows at low pHs via acid acclimation mechanisms that enable periplasmic pH homeostasis. Important components include a cytoplasmic urease; a pH-gated urea channel, UreI; and periplasmic α-carbonic anhydrase. To allow the rapid adjustment of periplasmic pH, acid acclimation components are recruited to the inner membrane in acid. The ArsRS two-component system, in an acid-responsive manner, controls the transcription of the urease gene cluster and α-carbonic anhydrase. The aim of this study is to determine the role of ArsS in protein trafficking as a component of acid acclimation. H. pylori wild-type and ΔarsS bacteria were incubated at acidic and neutral pHs. Intact bacteria, purified membranes, and total protein were analyzed by Western blotting and urease activity measurements. The total urease activity level was decreased in the ΔarsS strain, but the acid activation of UreI was unaffected. A 30-min acid exposure increased the level and activity of urease proteins at the membrane in the wild type but not in the ΔarsS strain. The urease levels and activity of the ΔarsS strain after a 90-min acid exposure were similar to those of the wild type. ArsS, in addition to its role in urease gene transcription, is also involved in the recruitment of urease proteins to the inner membrane to augment acid acclimation during acute acid exposure. Urease membrane recruitment following prolonged acid exposure in the absence of ArsS was similar to that of the wild type, suggesting a compensatory mechanism, possibly regulated by FlgS, underscoring the importance of urease membrane recruitment and activation in periplasmic pH homeostasis.


Asunto(s)
Ácidos/toxicidad , Proteínas Bacterianas/metabolismo , Helicobacter pylori/fisiología , Proteínas Quinasas/metabolismo , Estrés Fisiológico , Western Blotting , Membrana Celular/química , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica , Helicobacter pylori/química , Helicobacter pylori/genética , Helicobacter pylori/metabolismo , Proteínas Quinasas/genética , Transporte de Proteínas , Ureasa/análisis
18.
Am J Physiol Cell Physiol ; 302(9): C1271-81, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22277755

RESUMEN

The ion gradients generated by the Na-K-ATPase play a critical role in epithelia by driving transepithelial transport of various solutes. The efficiency of this Na-K-ATPase-driven vectorial transport depends on the integrity of epithelial junctions that maintain polar distribution of membrane transporters, including the basolateral sodium pump, and restrict paracellular diffusion of solutes. The review summarizes the data showing that, in addition to pumping ions, the Na-K-ATPase located at the sites of cell-cell junction acts as a cell adhesion molecule by interacting with the Na-K-ATPase of the adjacent cell in the intercellular space accompanied by anchoring to the cytoskeleton in the cytoplasm. The review also discusses the experimental evidence on the importance of a specific amino acid region in the extracellular domain of the Na-K-ATPase ß(1) subunit for the Na-K-ATPase trans-dimerization and intercellular adhesion. Furthermore, a possible role of N-glycans linked to the Na-K-ATPase ß(1) subunit in regulation of epithelial junctions by modulating ß(1)-ß(1) interactions is discussed.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Células Epiteliales/metabolismo , Multimerización de Proteína , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Animales , Moléculas de Adhesión Celular/química , Células Epiteliales/química , Humanos , Uniones Intercelulares/química , Uniones Intercelulares/metabolismo , Estructura Cuaternaria de Proteína , ATPasa Intercambiadora de Sodio-Potasio/química
19.
Physiol Genomics ; 44(2): 152-61, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22108211

RESUMEN

cAMP mediates diverse cellular signals including prostaglandin (PG) E(2)-mediated intraocular pressure (IOP)-lowering activity in human ocular ciliary smooth muscle cells (hCSM). We have identified gene regulatory networks and key genes upon activation of the cAMP pathway in hCSM, using novel agonists highly selective for PGE(2) receptor subtypes EP2 or EP4, which are G protein-coupled receptors well known to activate cAMP signaling. Here we describe a novel, EP2/EP4-induced, primate-specific gene of hitherto unknown function, also known as C6orf176 (chromosome 6 open reading frame 176) and recently reclassified as noncoding RNA in NCBI's database. Its expression, as determined by quantitative real-time RT-PCR (qRT-PCR), is dramatically upregulated (>2,000-fold) subsequent to transduction of EP2/EP4/Gs/cAMP signaling not only in hCSM, but also in HEK cells overexpressing the recombinant receptors. Moreover, activation of other IOP lowering, Gs-coupled prostanoid receptors, such as DP1 and IP, as well as a direct activator of adenylyl cyclase, forskolin, also substantially upregulated C6orf176 in hCSM, while FP and TP, which are Gq-coupled prostanoid receptor subtypes, did not. Novel transcript variants carrying open reading frames, derived from an at least 67 kb genomic locus on chromosome 6q27 with putative alternative transcription start sites, were identified. Transcriptional upregulation of transcript variants as well as of two genes expressed in antisense orientation that partially overlap the transcribed C6orf176 region was observed, to varying degrees, subsequent to induction of cAMP signaling using various agonists. Small interfering RNA-mediated C6orf176 gene silencing experiments showed modulation of several cAMP-responsive genes. These transcriptional activities identify C6orf176 as a potential biomarker and/or therapeutic target in context with diseases linked to deregulated cAMP signaling. Also, the cAMP-inducible C6orf176 gene locus could be useful as a model system for studying transcriptional regulation by chromatin and RNA polymerase II.


Asunto(s)
AMP Cíclico/metabolismo , Expresión Génica , ARN no Traducido/genética , Cromatina/metabolismo , Dinoprostona/genética , Dinoprostona/metabolismo , Células HEK293 , Humanos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , ARN no Traducido/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Transcripción Genética , Regulación hacia Arriba
20.
J Biol Chem ; 286(29): 25801-12, 2011 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-21642423

RESUMEN

N-Glycans of the Na,K-ATPase ß1 subunit are important for intercellular adhesion in epithelia, suggesting that epithelial junctions depend on N-glycan-mediated interactions between the ß1 subunits of neighboring cells. The level of co-immunoprecipitation of the endogenous ß1 subunit with various YFP-linked ß1 subunits expressed in Madin-Darby canine kidney cells was used to assess ß1-ß1 interactions. The amount of co-precipitated endogenous dog ß1 was greater with dog YFP-ß1 than with rat YFP-ß1, showing that amino acid-mediated interactions are important for ß1-ß1 binding. Co-precipitation of ß1 was also less with the unglycosylated YFP-ß1 than with glycosylated YFP-ß1, indicating a role for N-glycans. Mixing cells expressing dog YFP-ß1 with non-transfected cells increased the amount of co-precipitated ß1, confirming the presence of intercellular (YFP-ß1)-ß1 complexes. Accordingly, disruption of intercellular junctions decreased the amount of co-precipitated ß1 subunits. The decrease in ß1 co-precipitation both with rat YFP-ß1 and unglycosylated YFP-ß1 was associated with decreased detergent stability of junctional proteins and increased paracellular permeability. Reducing N-glycan branching by specific inhibitors increased (YFP-ß1)-ß1 co-precipitation and strengthened intercellular junctions. Therefore, interactions between the ß1 subunits of neighboring cells maintain integrity of intercellular junctions, and alterations in the ß1 subunit N-glycan structure can regulate stability and tightness of intercellular junctions.


Asunto(s)
Células Epiteliales/citología , Uniones Intercelulares/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Secuencia de Aminoácidos , Animales , Adhesión Celular , Línea Celular , Perros , Humanos , Permeabilidad , Polisacáridos/metabolismo , Unión Proteica , Ratas , ATPasa Intercambiadora de Sodio-Potasio/química
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda