Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Adv Mater ; 35(15): e2207255, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36779454

RESUMO

The intestinal muscle layers execute various gut wall movements to achieve controlled propulsion and mixing of intestinal content. Engineering intestinal muscle layers with complex contractile function is critical for developing bioartificial intestinal tissue to treat patients with short bowel syndrome. Here, the first demonstration of a living intestinal muscle patch capable of generating three distinct motility patterns and displaying multiple digesta manipulations is reported. Assessment of contractility, cellular morphology, and transcriptome profile reveals that successful generation of the contracting muscle patch relies on both biological factors in a serum-free medium and environmental cues from an elastic electrospun gelatin scaffold. By comparing gene-expression patterns among samples, it is shown that biological factors from the medium strongly affect ion-transport activities, while the scaffold unexpectedly regulates cell-cell communication. Analysis of ligandreceptor interactome identifies scaffold-driven changes in intercellular communication, and 78% of the upregulated ligand-receptor interactions are involved in the development and function of enteric neurons. The discoveries highlight the importance of combining biomolecular and biomaterial approaches for tissue engineering. The living intestinal muscle patch represents a pivotal advancement for building functional replacement intestinal tissue. It offers a more physiological model for studying GI motility and for preclinical drug discovery.


Assuntos
Conteúdo Gastrointestinal , Músculo Liso , Humanos , Músculo Liso/fisiologia , Intestinos , Engenharia Tecidual , Contração Muscular , Fatores Biológicos
2.
Am J Physiol Gastrointest Liver Physiol ; 318(5): G931-G945, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32174134

RESUMO

Helicobacter pylori infection always induces gastritis, which may progress to ulcer disease or cancer. The mechanisms underlying mucosal injury by the bacteria are incompletely understood. Here, we identify a novel pathway for H. pylori-induced gastric injury, the impairment of maturation of the essential transport enzyme and cell adhesion molecule, Na-K-ATPase. Na-K-ATPase comprises α- and ß-subunits that assemble in the endoplasmic reticulum (ER) before trafficking to the plasma membrane. Attachment of H. pylori to gastric epithelial cells increased Na-K-ATPase ubiquitylation, decreased its surface and total levels, and impaired ion balance. H. pylori did not alter degradation of plasmalemma-resident Na-K-ATPase subunits or their mRNA levels. Infection decreased association of α- and ß-subunits with ER chaperone BiP and impaired assembly of α/ß-heterodimers, as was revealed by quantitative mass spectrometry and immunoblotting of immunoprecipitated complexes. The total level of BiP was not altered, and the decrease in interaction with BiP was not observed for other BiP client proteins. The H. pylori-induced decrease in Na-K-ATPase was prevented by BiP overexpression, stopping protein synthesis, or inhibiting proteasomal, but not lysosomal, protein degradation. The results indicate that H. pylori impairs chaperone-assisted maturation of newly made Na-K-ATPase subunits in the ER independently of a generalized ER stress and induces their ubiquitylation and proteasomal degradation. The decrease in Na-K-ATPase levels is also seen in vivo in the stomachs of gerbils and chronically infected children. Further understanding of H. pylori-induced Na-K-ATPase degradation will provide insights for protection against advanced disease.NEW & NOTEWORTHY This work provides evidence that Helicobacter pylori decreases levels of Na-K-ATPase, a vital transport enzyme, in gastric epithelia, both in acutely infected cultured cells and in chronically infected patients and animals. The bacteria interfere with BiP-assisted folding of newly-made Na-K-ATPase subunits in the endoplasmic reticulum, accelerating their ubiquitylation and proteasomal degradation and decreasing efficiency of the assembly of native enzyme. Decreased Na-K-ATPase expression contributes to H. pylori-induced gastric injury.


Assuntos
Retículo Endoplasmático/enzimologia , Células Epiteliais/enzimologia , Mucosa Gástrica/enzimologia , Gastrite/enzimologia , Proteínas de Choque Térmico/metabolismo , Infecções por Helicobacter/enzimologia , Helicobacter pylori/patogenicidade , ATPase Trocadora de Sódio-Potássio/metabolismo , Células Cultivadas , Retículo Endoplasmático/microbiologia , Chaperona BiP do Retículo Endoplasmático , Estabilidade Enzimática , Células Epiteliais/microbiologia , Mucosa Gástrica/microbiologia , Gastrite/genética , Gastrite/microbiologia , Infecções por Helicobacter/genética , Infecções por Helicobacter/microbiologia , Interações Hospedeiro-Patógeno , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Dobramento de Proteína , Proteólise , ATPase Trocadora de Sódio-Potássio/genética , Ubiquitinação
3.
Mol Neurobiol ; 56(11): 7557-7571, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31062247

RESUMO

The Na,K-ATPase, consisting of a catalytic α-subunit and a regulatory ß-subunit, is a ubiquitously expressed ion pump that carries out the transport of Na+ and K+ across the plasma membranes of most animal cells. In addition to its pump function, Na,K-ATPase serves as a signaling scaffold and a cell adhesion molecule. Of the three ß-subunit isoforms, ß1 is found in almost all tissues, while ß2 expression is mostly restricted to brain and muscle. In cerebellar granule cells, the ß2-subunit, also known as adhesion molecule on glia (AMOG), has been linked to neuron-astrocyte adhesion and granule cell migration, suggesting its role in cerebellar development. Nevertheless, little is known about molecular pathways that link the ß2-subunit to its cellular functions. Using cerebellar granule precursor cells, we found that the ß2-subunit, but not the ß1-subunit, negatively regulates the expression of a key activator of the Hippo/YAP signaling pathway, Merlin/neurofibromin-2 (NF2). The knockdown of the ß2-subunit resulted in increased Merlin/NF2 expression and affected downstream targets of Hippo signaling, i.e., increased YAP phosphorylation and decreased expression of N-Ras. Further, the ß2-subunit knockdown altered the kinetics of epidermal growth factor receptor (EGFR) signaling in a Merlin-dependent mode and impaired EGF-induced reorganization of the actin cytoskeleton. Therefore, our studies for the first time provide a functional link between the Na,K-ATPase ß2-subunit and Merlin/NF2 and suggest a role for the ß2-subunit in regulating cytoskeletal dynamics and Hippo/YAP signaling during neuronal differentiation.


Assuntos
Cerebelo/citologia , Grânulos Citoplasmáticos/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Neurofibromina 2/metabolismo , Subunidades Proteicas/metabolismo , Transdução de Sinais , ATPase Trocadora de Sódio-Potássio/metabolismo , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Adesão Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/metabolismo , Transporte de Íons/efeitos dos fármacos , Isoenzimas/metabolismo , Camundongos Endogâmicos C57BL , Modelos Biológicos , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Proteínas de Sinalização YAP
4.
J Bacteriol ; 200(14)2018 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-29735759

RESUMO

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.


Assuntos
Fluorometria/métodos , Proteínas de Fluorescência Verde/metabolismo , Helicobacter pylori/metabolismo , Antígenos de Bactérias , Proteínas de Bactérias , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Regulação Bacteriana da Expressão Gênica/fisiologia , Helicobacter pylori/genética , Concentração de Íons de Hidrogênio , Mutação , Regiões Promotoras Genéticas , Ureia/metabolismo , Ureia/farmacologia
5.
Front Immunol ; 8: 623, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28620381

RESUMO

The alveolar epithelium secretes cytokines and chemokines that recruit immune cells to the lungs, which is essential for fighting infections but in excess can promote lung injury. Overexpression of FXYD5, a tissue-specific regulator of the Na,K-ATPase, in mice, impairs the alveolo-epithelial barrier, and FXYD5 overexpression in renal cells increases C-C chemokine ligand-2 (CCL2) secretion in response to lipopolysaccharide (LPS). The aim of this study was to determine whether FXYD5 contributes to the lung inflammation and injury. Exposure of alveolar epithelial cells (AEC) to LPS increased FXYD5 levels at the plasma membrane, and FXYD5 silencing prevented both the activation of NF-κB and the secretion of cytokines in response to LPS. Intratracheal instillation of LPS into mice increased FXYD5 levels in the lung. FXYD5 overexpression increased the recruitment of interstitial macrophages and classical monocytes to the lung in response to LPS. FXYD5 silencing decreased CCL2 levels, number of cells, and protein concentration in bronchoalveolar lavage fluid (BALF) after LPS treatment, indicating that FXYD5 is required for the NF-κB-stimulated epithelial production of CCL2, the influx of immune cells, and the increase in alveolo-epithelial permeability in response to LPS. Silencing of FXYD5 also prevented the activation of NF-κB and cytokine secretion in response to interferon α and TNF-α, suggesting that pro-inflammatory effects of FXYD5 are not limited to the LPS-induced pathway. Furthermore, in the absence of other stimuli, FXYD5 overexpression in AEC activated NF-κB and increased cytokine production, while FXYD5 overexpression in mice increased cytokine levels in BALF, indicating that FXYD5 is sufficient to induce the NF-κB-stimulated cytokine secretion by the alveolar epithelium. The FXYD5 overexpression also increased cell counts in BALF, which was prevented by silencing the CCL2 receptor (CCR2), or by treating mice with a CCR2-blocking antibody, confirming that FXYD5-induced CCL2 production leads to the recruitment of monocytes to the lung. Taken together, the data demonstrate that FXYD5 is a key contributor to inflammatory lung injury.

6.
Methods Mol Biol ; 1594: 35-42, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28456975

RESUMO

The vast majority of lysosomal proteins are heavily glycosylated. The present protocol describes the method of analyzing N- and O-linked glycans in lysosomal proteins of interest. The method is based on using deglycosylating enzymes, endoglycosidases, and exoglycosidases. Endoglycosidases catalyze the cleavage of an internal bond in an oligosaccharide, while exoglycosidases remove terminal carbohydrates from glycans. Different types of carbohydrate residues or chains can be removed by specific glycosidases. Removing oligosaccharides with glycosidases increases the electrophoretic mobility of a protein. This increase in mobility depends on the size and number of removed carbohydrate chains. Therefore, the treatment of lysosomal proteins with specific glycosidases followed by a western blot analysis of a protein of interest provides a way to determine which types of glycans are present in the protein by comparing the gel mobility before and after treatment.


Assuntos
Glicoproteínas/análise , Polissacarídeos/análise , Proteínas/análise , Western Blotting , Glicoproteínas/metabolismo , Glicosilação , Oligossacarídeos/análise , Oligossacarídeos/metabolismo , Polissacarídeos/metabolismo , Proteínas/metabolismo
7.
J Biol Chem ; 291(44): 23159-23174, 2016 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-27624940

RESUMO

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.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Inibidores Enzimáticos/química , Miocárdio/enzimologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Adenosina Trifosfatases/antagonistas & inibidores , Adenosina Trifosfatases/química , Animais , Proteínas de Transporte de Cátions/antagonistas & inibidores , Proteínas de Transporte de Cátions/química , Moléculas de Adesão Celular Neuronais/antagonistas & inibidores , Moléculas de Adesão Celular Neuronais/química , Dimerização , Inibidores Enzimáticos/metabolismo , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Camundongos , Miocárdio/química , Potássio/química , Potássio/metabolismo , Sódio/química , Sódio/metabolismo , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , ATPase Trocadora de Sódio-Potássio/química , ATPase Trocadora de Sódio-Potássio/genética
8.
J Cell Sci ; 129(12): 2394-406, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27142834

RESUMO

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.


Assuntos
Glicoproteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Multimerização Proteica , Subunidades Proteicas/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Células A549 , Aminoácidos/metabolismo , Animais , Especificidade de Anticorpos , Adesão Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Cães , Células Epiteliais/metabolismo , Técnicas de Silenciamento de Genes , Glicosilação , Células HEK293 , Humanos , Canais Iônicos , Células Madin Darby de Rim Canino , Camundongos , Proteínas dos Microfilamentos , Ligação Proteica , Subunidades Proteicas/química , Ratos , ATPase Trocadora de Sódio-Potássio/química
9.
Biochem J ; 473(12): 1703-18, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27048593

RESUMO

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.


Assuntos
Receptor ErbB-2/metabolismo , Septinas/metabolismo , Neoplasias Gástricas/metabolismo , Western Blotting , Linhagem Celular Tumoral , Cromatografia Líquida , Citoesqueleto/metabolismo , Humanos , Imunoprecipitação , Compostos de Fenilureia/farmacologia , Ligação Proteica/efeitos dos fármacos , Piridinas/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/fisiologia , Receptor ErbB-2/genética , Septinas/antagonistas & inibidores , Septinas/genética , Transdução de Sinais/fisiologia , Espectrometria de Massas em Tandem , Ubiquitinação/efeitos dos fármacos
10.
Cell Mol Gastroenterol Hepatol ; 1(6): 678-694, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26693174

RESUMO

BACKGROUND & AIMS: The pathogenic mechanism of pancreatitis is poorly understood. Recent evidence implicates defective autophagy in pancreatitis responses; however, the pathways mediating impaired autophagy in pancreas remain largely unknown. Here, we investigate the role of lysosome associated membrane proteins (LAMPs) in pancreatitis. METHODS: We analyzed changes in LAMPs in experimental models and human pancreatitis, and the underlying mechanisms: LAMP de-glycosylation and degradation. LAMP cleavage by cathepsin B (CatB) was analyzed by mass spectrometry. We used mice deficient in LAMP-2 to assess its role in pancreatitis. RESULTS: Pancreatic levels of LAMP-1 and LAMP-2 greatly decrease across various pancreatitis models and in human disease. Pancreatitis does not trigger LAMPs' bulk de-glycosylation, but induces their degradation via CatB-mediated cleavage of LAMP molecule close to the boundary between luminal and transmembrane domains. LAMP-2 null mice spontaneously develop pancreatitis that begins with acinar cell vacuolization due to impaired autophagic flux, and progresses to severe pancreas damage characterized by trypsinogen activation, macrophage-driven inflammation, and acinar cell death. LAMP-2 deficiency causes a decrease in pancreatic digestive enzymes content, stimulates the basal and inhibits CCK-induced amylase secretion by acinar cells. The effects of LAMP-2 knockout and acute cerulein pancreatitis overlap, which corroborates the pathogenic role of LAMP decrease in experimental pancreatitis models. CONCLUSIONS: The results indicate a critical role for LAMPs, particularly LAMP-2, in maintaining pancreatic acinar cell homeostasis, and provide evidence that defective lysosomal function, resulting in impaired autophagy, leads to pancreatitis. Mice with LAMP-2 deficiency present a novel genetic model of human pancreatitis caused by lysosomal/autophagic dysfunction.

11.
J Biol Chem ; 290(9): 5280-97, 2015 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-25575596

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Exocitose , Proteoma/metabolismo , Septinas/metabolismo , Animais , Western Blotting , Encéfalo/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Cães , Feminino , Células HEK293 , Humanos , Células Madin Darby de Rim Canino , Masculino , Camundongos Endogâmicos BALB C , Microscopia Confocal , Células PC12 , Compostos de Fenilureia/farmacologia , Ligação Proteica/efeitos dos fármacos , Multimerização Proteica , Proteômica , Piridinas/farmacologia , Interferência de RNA , Ratos , Septinas/química , Septinas/genética , Proteína 25 Associada a Sinaptossoma/metabolismo
12.
J Cell Sci ; 127(Pt 15): 3294-308, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24928902

RESUMO

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.


Assuntos
Toxinas Botulínicas Tipo A/metabolismo , Membrana Celular/metabolismo , Neurônios/fisiologia , Síndromes Neurotóxicas/metabolismo , Septinas/metabolismo , Animais , Toxinas Botulínicas Tipo A/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Humanos , Camundongos , Mutação/genética , Neurônios/microbiologia , Síndromes Neurotóxicas/microbiologia , Compostos de Fenilureia/farmacologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Multimerização Proteica/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , Piridinas/farmacologia , RNA Interferente Pequeno/genética , Septinas/genética
13.
Am J Physiol Gastrointest Liver Physiol ; 305(10): G731-9, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23989011

RESUMO

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.


Assuntos
Células Epiteliais/efeitos dos fármacos , Células Epiteliais/microbiologia , Helicobacter pylori/metabolismo , Linhagem Celular Tumoral , Meios de Cultura/química , Impedância Elétrica , Fenômenos Eletrofisiológicos , Regulação Bacteriana da Expressão Gênica , Helicobacter pylori/genética , Humanos , Concentração de Íons de Hidrogênio
14.
Int J Biol Macromol ; 51(5): 908-14, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22910577

RESUMO

The objective of this study is analysis of stability and antioxidant and antiradical activities of the gossypol derivative - megosin conjugated with N-polyvinylpyrrolidone (PVP). The results of study have shown the greater stability of megosin+PVP than megosin in aqueous solution of wide range of pH. Here we also demonstrated that megosin+PVP, named rometin, possess high antioxidant activity in the same range as well known antioxidant trolox as determined by its ability to scavenge free ABTS(+) and DPPH radicals in vitro. In addition, megosin+PVP was able to prevent accumulation of products of lipid peroxidation (thiobarbituric acid reactive substances and diene conjugates) and lysophospholipids formation in mitochondria membranes caused by CCl(4)-induced oxidative stress in rat liver in vivo. Furthermore, megosin+PVP rescued mitochondrial functions, such as respiration and oxidative phosphorylation, which declined after CCl(4) administration. Thus we present that the conjugation of megosin to PVP increase its stability and remain antioxidant activity in vivo and in vitro.


Assuntos
Sequestradores de Radicais Livres/química , Sequestradores de Radicais Livres/farmacologia , Gossipol/análogos & derivados , Povidona/química , Animais , Antipaína , Benzotiazóis/química , Compostos de Bifenilo/química , Tetracloreto de Carbono/toxicidade , Estabilidade de Medicamentos , Gossipol/química , Gossipol/farmacologia , Fígado/citologia , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Picratos/química , Ratos , Ratos Wistar , Ácidos Sulfônicos/química , Água/química
15.
J Biol Chem ; 287(31): 26115-25, 2012 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-22696220

RESUMO

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.


Assuntos
Multimerização Proteica , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Ligação Competitiva , Encéfalo/enzimologia , Membrana Celular/enzimologia , Células Cultivadas , Cães , Estabilidade Enzimática , Humanos , Imunoprecipitação , Isoenzimas/metabolismo , Rim , Proteínas Luminescentes/metabolismo , Camundongos , Modelos Moleculares , Especificidade de Órgãos , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Quaternária de Proteína , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Nervo Isquiático/enzimologia , ATPase Trocadora de Sódio-Potássio/química , ATPase Trocadora de Sódio-Potássio/isolamento & purificação
16.
J Cell Sci ; 125(Pt 6): 1605-16, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22328500

RESUMO

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.


Assuntos
Comunicação Celular/fisiologia , Células Epiteliais/enzimologia , ATPase Trocadora de Sódio-Potássio/química , ATPase Trocadora de Sódio-Potássio/fisiologia , Sequência de Aminoácidos , Aminoácidos/química , Aminoácidos/genética , Aminoácidos/fisiologia , Animais , Linhagem Celular , Cães , Células Epiteliais/citologia , Dados de Sequência Molecular , Multimerização Proteica , Estrutura Terciária de Proteína/genética , Ratos , ATPase Trocadora de Sódio-Potássio/genética , ATPase Trocadora de Sódio-Potássio/metabolismo
17.
Am J Physiol Cell Physiol ; 302(9): C1271-81, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22277755

RESUMO

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.


Assuntos
Moléculas de Adesão Celular/metabolismo , Células Epiteliais/metabolismo , Multimerização Proteica , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Moléculas de Adesão Celular/química , Células Epiteliais/química , Humanos , Junções Intercelulares/química , Junções Intercelulares/metabolismo , Estrutura Quaternária de Proteína , ATPase Trocadora de Sódio-Potássio/química
18.
J Pharmacol Exp Ther ; 339(2): 412-20, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21828261

RESUMO

Inhibition of the gastric H,K-ATPase by the potassium-competitive acid blocker (P-CAB) 1-[5-(2-fluorophenyl)-1-(pyridin-3-ylsulfonyl)-1H-pyrrol-3-yl]-N-methylmethanamine (TAK-438), is strictly K(+)-competitive with a K(i) of 10 nM at pH 7. In contrast to previous P-CABs, this structure has a point positive charge (pK(a) 9.06) allowing for greater accumulation in parietal cells compared with previous P-CABs [e.g., (8-benzyloxy-2-methyl-imidazo(1,2-a)pyridin-3-yl)acetonitrile (SCH28080), pK(a) 5.6]. The dissociation rate of the compound from the isolated ATPase is slower than other P-CABs, with the t(1/2) being 7.5 h in 20 mM KCl at pH 7. The stoichiometry of binding of TAK-438 to the H,K-ATPase is 2.2 nmol/mg in the presence of Mg-ATP, vanadate, or MgP(i). However, TAK-438 also binds enzyme at 1.3 nmol/mg in the absence of Mg(2+). Modeling of the H,K-ATPase to the homologous Na,K-ATPase predicts a close approach and hydrogen bonding between the positively charged N-methylamino group and the negatively charged Glu795 in the K(+)-binding site in contrast to the planar diffuse positive charge of previous P-CABs. This probably accounts for the slow dissociation and high affinity. The model also predicts hydrogen bonding between the hydroxyl of Tyr799 and the oxygens of the sulfonyl group of TAK-438. A Tyr799Phe mutation resulted in a 3-fold increase of the dissociation rate, showing that this hydrogen bonding also contributes to the slow dissociation rate. Hence, this K(+)-competitive inhibitor of the gastric H,K-ATPase should provide longer-lasting inhibition of gastric acid secretion compared with previous drugs of this class.


Assuntos
Inibidores da Bomba de Prótons , Inibidores da Bomba de Prótons/farmacologia , Pirróis/farmacologia , Estômago/efeitos dos fármacos , Sulfonamidas/farmacologia , Laranja de Acridina/metabolismo , Animais , Corantes Fluorescentes/metabolismo , Refluxo Gastroesofágico/tratamento farmacológico , ATPase Trocadora de Hidrogênio-Potássio/química , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Terapia de Alvo Molecular , Fosforilação/efeitos dos fármacos , Ligação Proteica , Inibidores da Bomba de Prótons/análise , Inibidores da Bomba de Prótons/química , Pirróis/química , Pirróis/metabolismo , Software , Estereoisomerismo , Estômago/enzimologia , Sulfonamidas/química , Sulfonamidas/metabolismo , Suínos
19.
J Biol Chem ; 286(29): 25801-12, 2011 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-21642423

RESUMO

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.


Assuntos
Células Epiteliais/citologia , Junções Intercelulares/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Sequência de Aminoácidos , Animais , Adesão Celular , Linhagem Celular , Cães , Humanos , Permeabilidade , Polissacarídeos/metabolismo , Ligação Proteica , Ratos , ATPase Trocadora de Sódio-Potássio/química
20.
J Biol Chem ; 285(50): 39289-302, 2010 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-20937802

RESUMO

Proper folding of the Na,K-ATPase ß subunits followed by assembly with the α subunits is necessary for their export from the endoplasmic reticulum (ER). Here we examine roles of the ER lectin chaperone, calnexin, and non-lectin chaperone, BiP, in folding and quality control of the ß(1) and ß(2) subunits in Madin-Darby canine kidney cells. Short term prevention of glycan-calnexin interactions by castanospermine slightly increases ER retention of ß(1), suggesting minor involvement of calnexin in subunit folding. However, both prolonged incubation with castanospermine and removal of N-glycosylation sites do not affect the α(1)-assembly or trafficking of ß(1) but increase the amount of the ß(1)-bound BiP, showing that BiP can compensate for calnexin in assisting ß(1) folding. In contrast to ß(1), prevention of either N-glycosylation or glycan-calnexin interactions abolishes the α(1)-assembly and export of ß(2) from the ER despite increased ß(2)-BiP binding. Mutations in the α(1)-interacting regions of ß(1) and ß(2) subunits impair α(1) assembly but do not affect folding of the ß subunits tested by their sensitivity to trypsin. At the same time, these mutations increase the amount of ß-bound BiP but not of ß-bound calnexin and increase ER retention of both ß-isoforms. BiP, therefore, prevents the ER export of folded but α(1)-unassembled ß subunits. These α(1)-unassembled ß subunits are degraded faster than α(1)-bound ß subunits, preventing ER overload. In conclusion, folding of the ß(1) and ß(2) subunits is assisted predominantly by BiP and calnexin, respectively. Folded ß(1) and ß(2) either assemble with α(1) or bind BiP. The α(1)-bound ß subunits traffic to the Golgi, whereas BiP-bound ß subunits are retained and degraded in the ER.


Assuntos
Adenosina Trifosfatases/fisiologia , Proteínas de Transporte de Cátions/fisiologia , Moléculas de Adesão Celular Neuronais/fisiologia , Retículo Endoplasmático/metabolismo , Regulação da Expressão Gênica , ATPase Trocadora de Sódio-Potássio/fisiologia , Adenosina Trifosfatases/genética , Animais , Calnexina/química , Proteínas de Transporte de Cátions/genética , Moléculas de Adesão Celular Neuronais/genética , Linhagem Celular , Cães , Glicosilação , Complexo de Golgi/metabolismo , Modelos Biológicos , Polissacarídeos/química , Dobramento de Proteína , Isoformas de Proteínas , Estrutura Terciária de Proteína , Ratos , ATPase Trocadora de Sódio-Potássio/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...