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

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Gastroenterology ; 156(8): 2158-2173, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30831083

RESUMEN

Acidification of the gastric lumen poses a barrier to transit of potentially pathogenic bacteria and enables activation of pepsin to complement nutrient proteolysis initiated by salivary proteases. Histamine-induced activation of the PKA signaling pathway in gastric corpus parietal cells causes insertion of proton pumps into their apical plasma membranes. Parietal cell secretion and homeostasis are regulated by signaling pathways that control cytoskeletal changes required for apical membrane remodeling and organelle and proton pump activities. Helicobacter pylori colonization of human gastric mucosa affects gastric epithelial cell plasticity and homeostasis, promoting epithelial progression to neoplasia. By intervening in proton pump expression, H pylori regulates the abundance and diversity of microbiota that populate the intestinal lumen. We review stimulation-secretion coupling and renewal mechanisms in parietal cells and the mechanisms by which H pylori toxins and effectors alter cell secretory pathways (constitutive and regulated) and organelles to establish and maintain their inter- and intracellular niches. Studies of bacterial toxins and their effector proteins have provided insights into parietal cell physiology and the mechanisms by which pathogens gain control of cell activities, increasing our understanding of gastrointestinal physiology, microbial infectious disease, and immunology.


Asunto(s)
Microbioma Gastrointestinal/efectos de los fármacos , Infecciones por Helicobacter/patología , Helicobacter pylori/patogenicidad , Células Parietales Gástricas/patología , Lesiones Precancerosas/patología , Neoplasias Gástricas/patología , Antibacterianos/uso terapéutico , Femenino , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Microbioma Gastrointestinal/fisiología , Infecciones por Helicobacter/diagnóstico , Infecciones por Helicobacter/tratamiento farmacológico , Humanos , Masculino , Lesiones Precancerosas/tratamiento farmacológico , Pronóstico , Medición de Riesgo , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/cirugía , Resultado del Tratamiento
2.
Curr Top Microbiol Immunol ; 400: 227-252, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28124156

RESUMEN

Appropriate management of Helicobacter pylori infection of the human stomach is evolving and remains a significant clinical challenge. Acute infection results in hypochlorhydria, whereas chronic infection results in either hypo- or hyperchlorhydria, depending upon the anatomic site of infection. Acute hypochlorhydria facilitates survival of the bacterium and its infection of the stomach. Interestingly, most patients chronically infected with H. pylori manifest a pangastritis with reduced acid secretion due to bacterial virulence factors, inflammatory cytokines, and various degrees of gastric atrophy. While these patients are predisposed to develop gastric adenocarcinoma (~1%), there is increasing evidence from population studies that they are also protected from gastroesophageal reflux disease (GERD), Barrett's esophagus (BE), and esophageal adenocarcinoma (EAC). Eradication of H. pylori, in these patients, may provoke GERD in predisposed individuals and may be a contributory factor for the rising incidence of refractory GERD, BE, and EAC observed in Westernized societies. Only ~10% of chronically infected patients, mainly the young, manifest an antral predominant gastritis with increased acid secretion due to a decrease in somatostatin and increase in gastrin secretion; these patients are predisposed to develop peptic ulcer disease. H. pylori-induced changes in acid secretion, in particular hypochlorhydria, may allow ingested microorganisms to survive transit through the stomach and colonize the distal intestine and colon. Such perturbation of gut microbiota, i.e. dysbiosis, may influence human health and disease.


Asunto(s)
Ácido Gástrico/metabolismo , Mucosa Gástrica/metabolismo , Infecciones por Helicobacter/microbiología , Helicobacter pylori/fisiología , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/patología , Helicobacter pylori/genética , Humanos , Estómago/microbiología , Estómago/patología , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
3.
Am J Physiol Gastrointest Liver Physiol ; 309(3): G193-201, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26045613

RESUMEN

Acute Helicobacter pylori infection of gastric epithelial cells and human gastric biopsies represses H,K-ATPase α subunit (HKα) gene expression and inhibits acid secretion, causing transient hypochlorhydria and supporting gastric H. pylori colonization. Infection by H. pylori strains deficient in the cag pathogenicity island (cag PAI) genes cagL, cagE, or cagM, which do not transfer CagA into host cells or induce interleukin-8 secretion, does not inhibit HKα expression, nor does a cagA-deficient strain that induces IL-8. To test the hypothesis that virulence factors other than those mediating CagA translocation or IL-8 induction participate in HKα repression by activating NF-κB, AGS cells transfected with HKα promoter-Luc reporter constructs containing an intact or mutated NF-κB binding site were infected with wild-type H. pylori strain 7.13, isogenic mutants lacking cag PAI genes responsible for CagA translocation and/or IL-8 induction (cagA, cagζ, cagε, cagZ, and cagß), or deficient in genes encoding two peptidoglycan hydrolases (slt and cagγ). H. pylori-induced AGS cell HKα promoter activities, translocated CagA, and IL-8 secretion were measured by luminometry, immunoblotting, and ELISA, respectively. Human gastric biopsy acid secretion was measured by microphysiometry. Taken together, the data showed that HKα repression is independent of IL-8 expression, and that CagA translocation together with H. pylori transglycosylases encoded by slt and cagγ participate in NF-κB-dependent HKα repression and acid inhibition. The findings are significant because H. pylori factors other than CagA and IL-8 secretion are now implicated in transient hypochlorhydria which facilitates gastric colonization and potential triggering of epithelial progression to neoplasia.


Asunto(s)
Mucosa Gástrica/metabolismo , Helicobacter pylori , FN-kappa B/metabolismo , Bombas de Protones/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Aclorhidria/etiología , Aclorhidria/metabolismo , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Células Cultivadas , Células Epiteliales/metabolismo , Ácido Gástrico/metabolismo , Mucosa Gástrica/microbiología , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/patogenicidad , Helicobacter pylori/fisiología , Humanos , Interleucina-8/metabolismo , Regiones Promotoras Genéticas , Transducción de Señal , Factores de Virulencia/metabolismo
4.
Dig Dis Sci ; 60(6): 1645-54, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25577268

RESUMEN

BACKGROUND: Most patients presenting with symptoms of esophageal cancer (EC) have advanced disease. Even with resection, the cure rate is extremely low due to local recurrence and metastatic disease. Early detection and effective therapeutic intervention are essential to improve survival. AIMS: This study tested the hypothesis that the presence of EC modulates concentrations of specific plasma proteins and peptides, potentially allowing discrimination between EC and controls based on mass spectrometric analysis of the respective plasma proteomes. METHODS: Blood samples from 79 esophageal cancer patients and 40 age-matched normal subjects were processed to plasma, and protein/peptide sub-fractions were isolated using HIC8 or WAX-derivatized superparamagnetic beads. Triplicate matrix-assisted laser desorption time-of-flight mass spectra were acquired for specific plasma fractions from each subject. RESULTS: HIC8 and WAX-derivatized plasma eluates yielded 79 and 77 candidate features, respectively, and a Random Forest algorithm identified a subset of features whose peak intensities allowed discrimination between cancer patients and controls. Areas under the curve in receiver operating characteristic curves for HIC8 spectra were 0.88 and 0.83 for WAX spectra. The combined feature set discriminated EC from control plasma with 79 % sensitivity and 79 % specificity, with positive and negative test likelihood ratios of >14 and 0.17, respectively. CONCLUSIONS: These data lay the foundation for the development of a clinically useful test for esophageal cancer based on statistical analysis of proteomic spectra of patient plasma samples. This approach will be validated by analysis of larger patient cohorts, development of cancer-specific classifiers, and assessment of racial origin imbalances.


Asunto(s)
Adenocarcinoma/sangre , Biomarcadores de Tumor/sangre , Carcinoma de Células Escamosas/sangre , Neoplasias Esofágicas/sangre , Proteómica/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Anciano , Anciano de 80 o más Años , Algoritmos , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , Sensibilidad y Especificidad
5.
Am J Physiol Gastrointest Liver Physiol ; 306(7): G606-13, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24503769

RESUMEN

Acute Helicobacter pylori infection of gastric epithelial cells induces CagA oncoprotein- and peptidoglycan (SLT)-dependent mobilization of NF-κB p50 homodimers that bind to H-K-ATPase α-subunit (HKα) promoter and repress HKα gene transcription. This process may facilitate gastric H. pylori colonization by induction of transient hypochlorhydria. We hypothesized that H. pylori also regulates HKα expression posttranscriptionally by miRNA interaction with HKα mRNA. In silico analysis of the HKα 3' untranslated region (UTR) identified miR-1289 as a highly conserved putative HKα-regulatory miRNA. H. pylori infection of AGS cells transfected with HKα 3' UTR-Luc reporter construct repressed luciferase activity by 70%, whereas ΔcagA or Δslt H. pylori infections partially abrogated repression. Transfection of AGS cells expressing HKα 3' UTR-Luc construct with an oligoribonucleotide mimetic of miR-1289 induced maximal repression (54%) of UTR activity within 30 min; UTR activity was unchanged by nontargeting siRNA transfection. Gastric biopsies from patients infected with cagA(+) H. pylori showed a significant increase in miR-1289 expression compared with uninfected patients or those infected with cagA(-) H. pylori. Finally, miR-1289 expression was necessary and sufficient to attenuate biopsy HKα protein expression in the absence of infection. Taken together, these data indicate that miR-1289 is upregulated by H. pylori in a CagA- and SLT-dependent manner and targets HKα 3' UTR, affecting HKα mRNA translation. The sensitivity of HKα mRNA 3' UTR to binding of miR-1289 identifies a novel regulatory mechanism of gastric acid secretion and offers new insights into mechanisms underlying transient H. pylori-induced hypochlorhydria.


Asunto(s)
Mucosa Gástrica/enzimología , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Infecciones por Helicobacter/enzimología , Helicobacter pylori/metabolismo , MicroARNs/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Células Parietales Gástricas/enzimología , Procesamiento Postranscripcional del ARN , Regiones no Traducidas 3' , Aclorhidria/enzimología , Aclorhidria/microbiología , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Línea Celular , Mucosa Gástrica/microbiología , Regulación Enzimológica de la Expresión Génica , Genes Reporteros , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/microbiología , Helicobacter pylori/genética , Helicobacter pylori/patogenicidad , Interacciones Huésped-Patógeno , Humanos , Subunidad p50 de NF-kappa B/genética , Células Parietales Gástricas/microbiología , Peptidoglicano/metabolismo , Interferencia de ARN , ARN Mensajero/metabolismo , Factores de Tiempo , Transfección , Virulencia
6.
Am J Physiol Gastrointest Liver Physiol ; 304(2): G157-66, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23154976

RESUMEN

Gastric acid secretion by the H(+)-K(+)-ATPase at the apical surface of activated parietal cells requires luminal K(+) provided by the KCNQ1/KCNE2 K(+) channel. However, little is known about the trafficking and relative spatial distribution of KCNQ1 and H(+)-K(+)-ATPase in resting and activated parietal cells and the capacity of KCNQ1 to control acid secretion. Here we show that inhibition of KCNQ1 activity quickly curtails gastric acid secretion in vivo, even when the H(+)-K(+)-ATPase is permanently anchored in the apical membrane, demonstrating a key role of the K(+) channel in controlling acid secretion. Three-dimensional imaging analysis of isolated mouse gastric units revealed that the majority of KCNQ1 resides in an intracytoplasmic, Rab11-positive compartment in resting parietal cells, distinct from H(+)-K(+)-ATPase-enriched tubulovesicles. Upon activation, there was a significant redistribution of H(+)-K(+)-ATPase and KCNQ1 from intracytoplasmic compartments to the apical secretory canaliculi. Significantly, high Förster resonance energy transfer was detected between H(+)-K(+)-ATPase and KCNQ1 in activated, but not resting, parietal cells. These findings demonstrate that H(+)-K(+)-ATPase and KCNQ1 reside in independent intracytoplasmic membrane compartments, or membrane domains, and upon activation of parietal cells, both membrane proteins are transported, possibly via Rab11-positive recycling endosomes, to apical membranes, where the two molecules are closely physically opposed. In addition, these studies indicate that acid secretion is regulated by independent trafficking of KCNQ1 and H(+)-K(+)-ATPase.


Asunto(s)
Ácido Gástrico/metabolismo , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Canal de Potasio KCNQ1/metabolismo , Células Parietales Gástricas/enzimología , Animales , Membrana Celular/enzimología , Cromanos/farmacología , Citoplasma/enzimología , Endosomas/enzimología , Transferencia Resonante de Energía de Fluorescencia , Histamina/metabolismo , Canal de Potasio KCNQ1/efectos de los fármacos , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Células Parietales Gástricas/efectos de los fármacos , Células Parietales Gástricas/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Transporte de Proteínas , Sulfonamidas/farmacología , Factores de Tiempo , Proteínas de Unión al GTP rab/metabolismo
7.
J Biol Chem ; 285(30): 23515-26, 2010 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-20507990

RESUMEN

Fibronectin, a 250-kDa eukaryotic extracellular matrix protein containing an RGD motif plays crucial roles in cell-cell communication, development, tissue homeostasis, and disease development. The highly complex fibrillar fibronectin meshwork orchestrates the functions of other extracellular matrix proteins, promoting cell adhesion, migration, and intracellular signaling. Here, we demonstrate that CagL, a 26-kDa protein of the gastric pathogen and type I carcinogen Helicobacter pylori, mimics fibronectin in various cellular functions. Like fibronectin, CagL contains a RGD motif and is located on the surface of the bacterial type IV secretion pili as previously shown. CagL binds to the integrin receptor alpha(5)beta(1) and mediates the injection of virulence factors into host target cells. We show that purified CagL alone can directly trigger intracellular signaling pathways upon contact with mammalian cells and can complement the spreading defect of fibronectin(-/-) knock-out cells in vitro. During interaction with various human and mouse cell lines, CagL mimics fibronectin in triggering cell spreading, focal adhesion formation, and activation of several tyrosine kinases in an RGD-dependent manner. Among the activated factors are the nonreceptor tyrosine kinases focal adhesion kinase and Src but also the epidermal growth factor receptor and epidermal growth factor receptor family member Her3/ErbB3. Interestingly, fibronectin activates a similar range of tyrosine kinases but not Her3/ErbB3. These findings suggest that the bacterial protein CagL not only exhibits functional mimicry with fibronectin but is also capable of activating fibronectin-independent signaling events. We thus postulate that CagL may contribute directly to H. pylori pathogenesis by promoting aberrant signaling cross-talk within host cells.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/farmacología , Forma de la Célula/efectos de los fármacos , Fibronectinas/metabolismo , Helicobacter pylori , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/química , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibronectinas/química , Humanos , Proteínas Inmovilizadas/química , Proteínas Inmovilizadas/metabolismo , Proteínas Inmovilizadas/farmacología , Integrinas/metabolismo , Ratones , Oligopéptidos/metabolismo , Fosfotirosina/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Gastroenterology ; 139(1): 239-48, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20303353

RESUMEN

BACKGROUND & AIMS: Infection with Helicobacter pylori represses expression of the gastric H, K-adenosine triphosphatase alpha-subunit (HKalpha), which could contribute to transient hypochlorhydria. CagL, a pilus protein component of the H pylori type IV secretion system, binds to the integrin alpha(5)beta1 to mediate translocation of virulence factors into the host cell and initiate signaling. alpha(5)beta1 binds a disintegrin and metalloprotease (ADAM) 17, a metalloenzyme that catalyzes ectodomain shedding of receptor tyrosine kinase ligands. We investigated whether H pylori-induced repression of HKalpha is mediated by CagL activation of ADAM17 and release of heparin-binding epidermal growth factor (HB-EGF). METHODS: HKalpha promoter and ADAM17 activity were measured in AGS gastric epithelial cells transfected with HKalpha promoter-reporter constructs or ADAM17-specific small interfering RNAs and infected with H pylori. HB-EGF secretion was measured by enzyme-linked immunosorbent assay analysis, and ADAM17 interaction with integrins was investigated by coimmunoprecipitation analyses. RESULTS: Infection of AGS cells with wild-type H pylori or an H pylori cagL-deficient isogenic mutant that also contained a wild-type version of cagL (P12DeltacagL/cagL) repressed HKalpha promoter-Luc reporter activity and stimulated ADAM17 activity. Both responses were inhibited by point mutations in the nuclear factor-kappaB binding site of HKalpha or by infection with P12DeltacagL. Small interfering RNA-mediated silencing of ADAM17 in AGS cells inhibited the repression of wild-type HKalpha promoter and reduced ADAM17 activity and HB-EGF production, compared to controls. Coimmunoprecipitation studies of AGS lysates showed that wild-type H pylori disrupted ADAM17-alpha5beta1 complexes. CONCLUSIONS: During acute H pylori infection, CagL dissociates ADAM17 from the integrin alpha(5)beta1 and activates ADAM17-dependent, nuclear factor-kappaB-mediated repression of HKalpha. This might contribute to transient hypochlorhydria in patients with H pylori infection.


Asunto(s)
Proteínas ADAM/fisiología , Helicobacter pylori/fisiología , ATPasa Intercambiadora de Sodio-Potasio/genética , Estómago/enzimología , Proteína ADAM17 , Represión Enzimática , Factor de Crecimiento Similar a EGF de Unión a Heparina , Humanos , Integrina beta1/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Oligopéptidos/fisiología , Regiones Promotoras Genéticas , ATPasa Intercambiadora de Sodio-Potasio/biosíntesis
9.
Gut ; 59(7): 874-81, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20581234

RESUMEN

BACKGROUND AND AIMS: Helicobacter pylori infection of gastric mucosa causes gastritis and transient hypochlorhydria, which may provoke emergence of a mucosal precancer phenotype; H pylori strains containing a cag pathogenicity island (PAI) augment cancer risk. Acid secretion is mediated by the catalytic alpha subunit of parietal cell H,K-ATPase (HKalpha). In AGS gastric epithelial cells, H pylori induces nuclear factor-kappaB (NF-kappaB) binding to and repression of transfected HKalpha promoter activity. This study sought to identify bacterial genes involved in HKalpha repression and to assess their impact on acid secretion. METHODS AND RESULTS: AGS cells transfected with an HKalpha promoter construct or human gastric body biopsies were infected with wild-type (wt) or isogenic mutant (IM) H pylori strains. AGS cell HKalpha promoter activity, and biopsy HKalpha mRNA, protein and H(+) secretory activity were measured by luminometry, reverse transcription-PCR, immunoblotting and extracellular acidification, respectively. Wt H pylori and DeltavacA, DeltaureA, Deltaslt and DeltaflaA IM strains repressed HKalpha promoter activity by approximately 50%, a DeltacagA IM strain repressed HKalpha by approximately 33%, and DeltacagE, DeltacagM and DeltacagL IM strains elicited no HKalpha repression. Wt H pylori-infected biopsies had markedly reduced HKalpha mRNA and protein compared with IM strain infections or mock-infected controls. Histamine-stimulated, SCH28080-sensitive biopsy acid secretion was significantly inhibited by wt but not by DeltacagL IM H pylori infection compared with vehicle-only controls. CONCLUSIONS: It is concluded that H pylori cag PAI gene products CagE, CagM, CagL and, possibly, CagA are mechanistically involved in repression of HKalpha transcription. Further, acute H pylori infection of human gastric mucosa downregulates parietal cell H,K-ATPase expression, significantly inhibiting acid secretion.


Asunto(s)
Ácido Gástrico/metabolismo , Mucosa Gástrica/metabolismo , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/patogenicidad , Bombas de Protones/metabolismo , Adulto , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Mucosa Gástrica/microbiología , Genes Bacterianos , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Infecciones por Helicobacter/microbiología , Helicobacter pylori/genética , Humanos , Persona de Mediana Edad , Regiones Promotoras Genéticas , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Técnicas de Cultivo de Tejidos , Células Tumorales Cultivadas , Virulencia/genética , Adulto Joven
10.
Bioinformatics ; 23(2): 264-5, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17121773

RESUMEN

UNLABELLED: We introduce a simple-to-use graphical tool that enables researchers to easily prepare time-of-flight mass spectrometry data for analysis. For ease of use, the graphical executable provides default parameter settings, experimentally determined to work well in most situations. These values, if desired, can be changed by the user. PrepMS is a stand-alone application made freely available (open source), and is under the General Public License (GPL). Its graphical user interface, default parameter settings, and display plots allow PrepMS to be used effectively for data preprocessing, peak detection and visual data quality assessment. AVAILABILITY: Stand-alone executable files and Matlab toolbox are available for download at: http://sourceforge.net/projects/prepms


Asunto(s)
Gráficos por Computador , Almacenamiento y Recuperación de la Información/métodos , Mapeo Peptídico/métodos , Proteínas/química , Programas Informáticos , Interfaz Usuario-Computador , Algoritmos , Sistemas de Administración de Bases de Datos , Bases de Datos de Proteínas , Espectrometría de Masas/métodos
11.
PLoS One ; 11(10): e0163696, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27783698

RESUMEN

The stomach, which is characterized by acid peptic digestion in vertebrates, has been lost secondarily multiple times in the evolution of the teleost fishes. The Cypriniformes are largely seen as an agastric order; however, within the superfamily Cobitoidea, the closely related sister groups Nemacheilidae and Balitoridae have been identified as gastric families. The presence of these most recently diverged gastric families in an otherwise agastric clade indicates that either multiple (>2-3) loss events occurred with the Cyprinidae, Catostomidae and Cobitidae, or that gastric reinvention arose in a recent ancestor of the Nemacheilidae/Balitoridae sister clade. In the present study, the foregut regions of Cobitidae, Nemacheilidae/Balitoridae and the ancestral Botiidae family members were examined for the presence of gastric glands and gastric proton pump (Atp4a) α subunit expression by histology and immunohistochemistry respectively. Atp4a gene expression was assessed by reverse transcriptase-polymerase chain reaction (RT-PCR). Gastric glands expressing apical H+/K+-ATPase α subunit and isolated partial sequences of atp4a, identified using degenerate primers showing clear orthology to other vertebrate atp4a sequences, were detected in representative species from Nemacheilidae/ Balitoridae and Botiidae, but not Cobitidae (Misgurnus anguillicaudatus). In summary, we provide evidence for an uninterrupted gastric evolutionary lineage in the Cobitoidea, making it highly improbable that the stomach was reinvented in the Nemacheilidae/Balitoridae clade consistent with Dollo's principle. These results also indicate that the gastric trait may be present elsewhere in the Cobitoidea.


Asunto(s)
Cipriniformes/metabolismo , Mucosa Gástrica/metabolismo , Secuencia de Aminoácidos , Animales , Cipriniformes/clasificación , Evolución Molecular , Expresión Génica , ATPasa Intercambiadora de Hidrógeno-Potásio/química , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Inmunohistoquímica , Mucosa Intestinal/metabolismo , Intestinos/patología , Datos de Secuencia Molecular , Fenotipo , Filogenia , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN/aislamiento & purificación , ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Alineación de Secuencia , Estómago/patología
12.
CBE Life Sci Educ ; 14(1): ar3, 2015 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-25673355

RESUMEN

Academic medical centers nationwide face numerous fiscal challenges resulting from implementation of restructured healthcare delivery models, contracting state support for higher education, and increased competition for federal and other sources of biomedical research funding. In pursuing greater accountability and transparency in its fiscal operations, the Medical University of South Carolina (MUSC) has implemented a responsibility centers management budgetary model, which requires all MUSC colleges to be eventually self-sustaining financially. Graduate schools in the biomedical sciences are particularly vulnerable in the face of these challenges, depending traditionally as they do on financial support from training grant tuition, occasional medical school tuition and medical practice plan revenues, graduate college-based revenue-generating programs, and faculty payment of PhD tuition. The revenue streams are often insufficient to support PhD training programs, and supplemental financial support is required from the institution. In the context of a college of graduate studies, estimates of the cost of educating a graduate student become a significant necessity. This study presents a readily applicable model of empirically estimating the faculty salary costs that may provide a basis for budgetary planning that will help to sustain a biomedical sciences graduate school's commitment to its teaching, research, and service mission goals.


Asunto(s)
Biología/economía , Biología/educación , Educación de Postgrado/economía , Docentes , Facultades de Medicina/economía , Centros Médicos Académicos/economía , Investigación Biomédica/economía , Investigación Biomédica/educación , Recolección de Datos , Docentes Médicos , Humanos , Salarios y Beneficios , Ciencia/economía , South Carolina , Estudiantes , Apoyo a la Formación Profesional , Universidades/economía
13.
Life Sci ; 74(17): 2111-28, 2004 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-14969716

RESUMEN

This study tested the hypothesis that rat adrenocortical secretion of endogenous ouabain-like factor (OLF) is regulated by nicotinic mechanisms. OLF secreted by dispersed cell suspensions of zona glomerulosa (ZG) and fasciculata/reticularis (ZFR) cells was found to co-elute with authentic ouabain by reverse phase HPLC; OLF concentrations in cell supernatants were measured by radioimmunoassay. Nicotine (10(-6) - 10(-3) M) stimulated significant OLF secretion in rat adrenocortical cells. Acetylcholine (10(-7) - 10(-4) M) and eserine (10(-7) - 10(-3) M) stimulated OLF secretion in ZG cells at lower concentrations and stimulated at higher concentrations. Acetylcholine had no effect on ZFR secretion of OLF, but eserine stimulated OLF secretion. ACTH (10(-8) M) strongly potentiated the OLF stimulatory effect of nicotine in ZG cells; however significant interactions between nicotine and ACTH or angiotensin II on OLF secretion in ZFR cells were not apparent. The ganglionic blockers hexamethonium and mecamylamine further potentiated the effect of nicotine, implicating nicotinic acetylcholine receptors (nAChRs) in regulation of OLF secretion. The alpha7-receptor antagonist methyllycaconitine (MLA) dose-dependently inhibited the effect of nicotine in the ZG cells, and in ZFR cells MLA potentiated nicotine-induced OLF secretion. These data suggest that nicotinic regulation may underlie OLF secretion by rat adrenocortical cells, and strongly suggest presence of functional nicotinic acetylcholine receptors on these cells.


Asunto(s)
Aconitina/análogos & derivados , Corteza Suprarrenal/metabolismo , Receptores Nicotínicos/metabolismo , Saponinas/metabolismo , Acetilcolina/farmacología , Aconitina/farmacología , Corteza Suprarrenal/efectos de los fármacos , Animales , Cardenólidos , Digoxina , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Hexametonio/farmacología , Masculino , Mecamilamina/farmacología , Nicotina/farmacología , Antagonistas Nicotínicos/farmacología , Fisostigmina/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Nicotínicos/efectos de los fármacos
14.
BMC Gastroenterol ; 4: 4, 2004 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-15028114

RESUMEN

BACKGROUND: ML 3000 ([2,2-dimethyl-6-(4-chlorophenyl)-7-phenyl-2,3-dihydro-1H-pyrrolizine-5-yl]-acetic acid) is an inhibitor of both cyclooxygenase and 5-lipoxygenase in vitro, and shows promise as a novel non-steroidal anti-inflammatory drug (NSAID). Unlike conventional NSAIDs which are associated with gastric ulcerogenic effects, ML 3000 causes little or no damage to the gastric mucosa, even though it significantly depresses gastric prostaglandin synthesis. METHODS: As part of an effort to clarify mechanisms underlying the gastric sparing properties of ML 3000, we studied the effects of ML 3000 on H,K-ATPase activity in vitro, on acid accumulation in isolated gastric parietal cells, and on IL-8 secretion by gastric epithelial cells in culture. RESULTS: SCH28080-sensitive H,K-ATPase activity in highly-purified pig gastric microsomes was dose-dependently inhibited by ML 3000 (IC50 = 16.4 microM). Inhibition was reversible, and insensitive to ML 3000 acidification in the pH range 2.0-8.0. In rabbit gastric parietal cells,ML 3000 dose-dependently inhibited histamine-stimulated acid accumulation (IC50 = 40 microM) and forskolin-stimulated acid accumulation (IC50 = 45 microM). Lastly, in human gastric adenocarcinoma (AGS) cells, ML 3000 dose-dependently inhibited both baseline and IL-1beta-stimulated (20 ng/ml) IL-8 secretion with IC50s of 0.46 microM and 1.1 microM respectively. CONCLUSION: The data indicate that ML 3000 affects acid-secretory mechanisms downstream of cAMP mobilization induced by histamine H2 receptor activation, that it directly inhibits H,K-ATPase specific activity, and that baseline gastric epithelial cell IL-8 secretory inhibition may be mediated by ML 3000 inhibition of 5-lipoxygenase activity. We conclude that these gastric function inhibitory data may underlie the gastric sparing properties of ML 3000.


Asunto(s)
Acetatos/farmacología , Antiinflamatorios no Esteroideos/farmacología , Mucosa Gástrica/efectos de los fármacos , Interleucina-8/metabolismo , Células Parietales Gástricas/efectos de los fármacos , Inhibidores de la Bomba de Protones , Pirroles/farmacología , Animales , Línea Celular Tumoral/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Mucosa Gástrica/enzimología , Mucosa Gástrica/metabolismo , Humanos , Ácido Clorhídrico/metabolismo , Interleucina-1/farmacología , Omeprazol/farmacología , Células Parietales Gástricas/metabolismo , Conejos
15.
J Gastroenterol ; 47(6): 609-18, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22565637

RESUMEN

Infection of the human stomach mucosa by Helicobacter pylori induces strong inflammatory responses and a transitory hypochlorhydria which can progress in ~2 % of patients to atrophic gastritis, dysplasia, or gastric adenocarcinoma. H. pylori infection of gastric biopsies or cultured gastric epithelial cells in vitro represses the activity of endogenous or transfected promoter of the alpha-subunit (HKα) of gastric H,K-adenosine triphosphatase (H,K-ATPase), the parietal cell enzyme mediating acid secretion. Some mechanistic details of H. pylori-mediated repression of HKα and ensuing hypochlorhydria have been recently elucidated. H. pylori strains expressing a type IV secretion system (T4SS) encoded by the cag pathogenicity island are known to upregulate the transcription factor nuclear factor (NF)-κB. The NF-κB-binding regions in the HKα promoter were identified and shown to repress its transcriptional activity. Interaction studies have indicated that although active phosphorylated NF-κB p65 is present in infected cells, an NF-κB p50/p65 heterodimeric complex fails to bind to the HKα promoter. Point mutations at -159 and -161 bp in the HKα promoter NF-κB binding sequence prevent the binding of NF-κB p50 and prevent H. pylori repression of point-mutated HKα promoter activity. The T4SS factors CagL, CagE, CagM, and possibly CagA and the lytic transglycosylase Slt, are mechanistically involved in NF-κB activation and repression of HKα transcription. CagL, a T4SS pilus component, binds to the integrin α(5)ß(1) to mediate translocation of virulence factors into the host cell and initiate signaling. During acute H. pylori infection, CagL dissociates ADAM 17 (a disintegrin and a metalloprotease 17) from the integrin α(5)ß(1) complex and stimulates ADAM17-dependent release of heparin-binding epidermal growth factor (HB-EGF), EGF receptor (EGFR) stimulation, ERK1/2 kinase activation, and NF-κB-mediated repression of HKα. These studies suggest that H. pylori inhibits HKα gene expression by an integrin α(5)ß(1) → ADAM17 → HB-EGF → EGFR → ERK1/2 → NF-κB pathway mediating NF-κB p50 homodimer binding to the HKα promoter. Here we review the molecular basis and recent progress of this novel pathogen-dependent mechanism of H,K-ATPase inhibition, which contributes significantly to our current understanding of H. pylori pathophysiology.


Asunto(s)
Ácido Gástrico/metabolismo , Infecciones por Helicobacter/metabolismo , Helicobacter pylori , Aclorhidria/microbiología , Animales , Modelos Animales de Enfermedad , Medicina Basada en la Evidencia/métodos , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/genética , Humanos , Bombas de Protones/fisiología , Transcripción Genética
16.
Am J Physiol Gastrointest Liver Physiol ; 295(5): G977-86, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18772363

RESUMEN

Helicobacter pylori infection of the gastric body induces transient hypochlorhydria and contributes to mucosal progression toward gastric carcinoma. Acid secretion is mediated by parietal cell H,K-ATPase, in which the catalytic alpha-subunit (HKalpha) promoter activity in transfected gastric epithelial [gastric adenocarcinoma (AGS)] cells is repressed by H. pylori through NF-kappaB p50 homodimer binding to the promoter. IL-1beta, an acid secretory inhibitor whose mucosal level is increased by H. pylori, upregulates HKalpha promoter activity in AGS cells. Because IL-1beta also activates NF-kappaB signaling, we investigated disparate HKalpha regulation by H. pylori and IL-1beta, testing the hypothesis that IL-1beta-induced HKalpha promoter activation is mediated by the transcription factor Sp1. DNase I footprinting revealed Sp1 binding to the HKalpha promoter at -56 to -39 bp. IL-1beta stimulated the activity of three HKalpha promoter constructs containing NF-kappaB and Sp1 sites transfected into AGS cells and also stimulated a construct containing only an Sp1 site. This stimulation was abrogated by mutating the HKalpha promoter Sp1 binding site. Gelshift assays showed that IL-1beta increased Sp1 but not p50 binding to cognate HKalpha probes and that Sp1 also interacts with an HKalpha NF-kappaB site when bound to its cognate HKalpha cis-response element. H. pylori did not augment Sp1 binding to an HKalpha Sp1 probe, and small interfering RNA-mediated knockdown of Sp1 expression abrogated IL-1beta-induced HKalpha promoter stimulation. We conclude that IL-1beta upregulates HKalpha gene transcription by inducing Sp1 binding to HKalpha Sp1 and NF-kappaB sites and that the H. pylori perturbation of HKalpha gene expression is independent of Sp1-mediated basal HKalpha transcription.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/fisiología , ATPasa Intercambiadora de Hidrógeno-Potásio/metabolismo , Helicobacter pylori/fisiología , Interleucina-1beta/metabolismo , Factor de Transcripción Sp1/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , Humanos , FN-kappa B/metabolismo , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/fisiología , Unión Proteica , Transducción de Señal
17.
Am J Physiol Gastrointest Liver Physiol ; 294(3): G795-807, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18202112

RESUMEN

Infection of human gastric body mucosa by the gram-negative, microaerophilic bacterium Helicobacter pylori induces an inflammatory response and a transitory hypochlorhydria that progresses in approximately 2% of patients to atrophic gastritis, dysplasia, and gastric adenocarcinoma. We have previously shown that H. pylori infection of cultured gastric epithelial cells (AGS) represses the activity of the transfected alpha-subunit (HKalpha) promoter of H,K-ATPase, the parietal cell enzyme mediating acid secretion. However, the mechanistic details of H. pylori-mediated repression of HKalpha and ensuing hypochlorhydria are unknown. H. pylori is known to upregulate the transcription factor NF-kappaB through the ERK1/2 MAPK pathway. We identified NF-kappaB-binding regions in the HKalpha promoter and found that H. pylori inoculation of AGS cells increased NF-kappaB p50 binding to the transfected HKalpha promoter and repressed its transcriptional activity. Immunoblot and DNA-protein interaction studies showed that although active phosphorylated NF-kappaB p65 is present in H. pylori-infected AGS cells, an NF-kappaB p50/p65 heterodimeric complex fails to bind to the HKalpha promoter. Point mutations at -159 and -161 bp in the HKalpha promoter NF-kappaB binding sequence prevented binding of NF-kappaB p50 and prevented H. pylori repression of point-mutated HKalpha promoter activity in transfected AGS cells. Small interfering RNA-mediated knockdown of NF-kappaB p50 in H. pylori-infected AGS cells also abrogated H. pylori-induced HKalpha repression, whereas NF-kappaB p65 knockdown did not. We conclude that H. pylori inhibits HKalpha gene expression by ERK1/2-mediated NF-kappaB p50 homodimer binding to the HKalpha promoter. This study identifies a novel pathogen-dependent mechanism of H,K-ATPase inhibition and contributes to understanding of H. pylori pathophysiology.


Asunto(s)
Infecciones por Helicobacter/enzimología , Infecciones por Helicobacter/genética , Helicobacter pylori , Subunidad p50 de NF-kappa B/genética , ATPasa Intercambiadora de Sodio-Potasio/genética , Secuencia de Bases , Western Blotting , Línea Celular Tumoral , Núcleo Celular/metabolismo , Desoxirribonucleasa I/genética , Desoxirribonucleasa I/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Humanos , Indicadores y Reactivos , Proteínas Quinasas Activadas por Mitógenos/fisiología , Datos de Secuencia Molecular , Plásmidos/genética , Regiones Promotoras Genéticas/genética , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología , Neoplasias Gástricas/patología , Transcripción Genética , Transfección
18.
Am J Physiol Gastrointest Liver Physiol ; 292(4): G1055-61, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17204545

RESUMEN

Helicobacter pylori infection of the human gastric body induces hypochlorhydria by perturbing acid secretion. H. pylori inhibits parietal cell H,K-ATPase alpha-subunit (HKalpha) gene and protein expression, providing a mechanistic basis for clinical hypochlorhydria. Given that H. pylori infection increases gastric mucosal IL-1beta, an acid secretory inhibitor, we investigated the role of IL-1beta in H. pylori-mediated inhibition of HKalpha transcription. Human gastric adenocarcinoma (AGS) cells were transfected with promoter-reporter constructs containing human HKalpha 5'-flanking sequence deletions. IL-1beta (10 ng/ml) had no effect on the transcriptional activity of six progressively shorter deletion constructs of the HKalpha promoter (HKalpha2179-HKalpha340) and significantly stimulated the activity of HKalpha206, HKalpha177, HKalpha165, and HKalpha102 deletion constructs (80%, 100%, 46%, and 35%, respectively). H. pylori inhibited the transcriptional activity of HKalpha2179, HKalpha206, HKalpha177, and HKalpha165; IL-1beta relieved the H. pylori inhibition of HKalpha2179 and HKalpha206 activity but not HKalpha177 and HKalpha165 activity. AGS cell pretreatment with a MEK1/2 inhibitor prevented the IL-1beta-mediated stimulation, but p38 and JNK pathway inhibitors did not. IL-1beta mRNA levels in AGS cells were low and unaffected by H. pylori, and ELISAs of H. pylori-conditioned AGS culture media showed no measurable IL-1beta secretion. These data indicate that an IL-1beta-dependent cis-response element lies downstream of -206 nt in the HKalpha promoter and that IL-1beta-mediated upregulation of HKalpha transcription is affected by an ERK1/2 kinase signal pathway. We conclude that an IL-1beta-responsive HKalpha cis element positively regulates HKalpha gene transcription in shortened deletion constructs and that H. pylori-induced inhibition of HKalpha transcription is not mediated by IL-1beta.


Asunto(s)
Células Epiteliales/metabolismo , Mucosa Gástrica/metabolismo , ATPasa Intercambiadora de Hidrógeno-Potásio/biosíntesis , Infecciones por Helicobacter/metabolismo , Helicobacter pylori , Interleucina-1beta/metabolismo , Transcripción Genética , Región de Flanqueo 5' , Adenocarcinoma/patología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Células Epiteliales/efectos de los fármacos , Células Epiteliales/microbiología , Células Epiteliales/patología , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Genes Reporteros , ATPasa Intercambiadora de Hidrógeno-Potásio/genética , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/microbiología , Humanos , Interleucina-1beta/farmacología , Luciferasas , Sistema de Señalización de MAP Quinasas , Inhibidores de Proteínas Quinasas/farmacología , Subunidades de Proteína , ARN Mensajero/biosíntesis , Neoplasias Gástricas/patología , Factores de Tiempo , Transfección , Regulación hacia Arriba
19.
Am J Physiol Gastrointest Liver Physiol ; 292(5): G1249-62, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17255364

RESUMEN

Gastric parietal cells possess an amplified apical membrane recycling system dedicated to regulated apical recycling of H-K-ATPase. While amplified in parietal cells, apical recycling is critical to polarized secretory processes in most epithelial cells. To clarify putative regulators of apical recycling, we prepared immunoisolated parietal cell H-K-ATPase-containing recycling membranes from human stomachs and analyzed protein contents by tryptic digestion and mass spectrometry. We identified and validated by Western blots many of the proteins previously identified on immunoisolated rabbit tubulovesicles, including Rab11, Rab25, syntaxin 3, secretory carrier membrane proteins (SCAMPs), and vesicle-associated membrane protein (VAMP)2. In addition, we detected several previously unrecognized proteins, including Rab10, VAMP8, syntaxin 7, and syntaxin 12/13. We also identified the K(+) channel component KCNQ1. Immunostaining of human gastric mucosal sections confirmed the presence of each of these proteins in parietal cells and their colocalization with H-K-ATPase on tubulovesicles. To investigate the role of the identified soluble N-ethylmaleimide-sensitive factor attachment receptor (SNARE) proteins in apical recycling, we transfected them as DsRed2 fusions into an enhanced green fluorescent protein (EGFP)-Rab11a-expressing Madin-Darby canine kidney (MDCK) cell line. Syntaxin 12/13 and VAMP8 caused a collapse of the EGFP-Rab11a compartment, whereas a less dramatic effect was observed in cells transfected with syntaxin 3, syntaxin 7, or VAMP2. The five DsRed2-SNARE chimeras were also transfected into a MDCK cell line overexpressing Rab11-FIP2(129-512). All five of the chimeras were drawn into the collapsed apical recycling system. This study, which represents the first proteomic analysis of an immunoisolated vesicle population from native human tissue, demonstrates the diversity of putative regulators of the apical recycling system.


Asunto(s)
ATPasa Intercambiadora de Hidrógeno-Potásio/fisiología , Células Parietales Gástricas/fisiología , Adulto , Animales , Western Blotting , Células Cultivadas , Cromatografía Liquida , Perros , Femenino , ATPasa Intercambiadora de Hidrógeno-Potásio/análisis , Humanos , Masculino , Persona de Mediana Edad , Células Parietales Gástricas/ultraestructura , Proteoma/análisis , Proteínas SNARE/fisiología , Sistemas de Mensajero Secundario , Espectrometría de Masas en Tándem , Proteínas de Unión al GTP rab/análisis
20.
Ultrastruct Pathol ; 27(2): 87-94, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12746199

RESUMEN

The case is reported of a clinically aggressive parietal cell carcinoma of the gastric cardia in a 67-year-old man. Histologically, the tumor was a poorly differentiated adenocarcinoma with a predominantly solid growth pattern, though with areas exhibiting glandular morphology and with extensive lymphatic invasion. The tumor cells had eosinophilic, finely granular cytoplasm, with focal Alcian blue-positive mucin in the gland lumens. Ultrastructural examination of the pleural metastasis and gastrectomy specimen demonstrated many mitochondria, tubulovesicular profiles of endoplasmic reticulum, and intracytoplasmic lumens, which resembled intracellular canaliculi of parietal cells. Immunohistochemically, there was positive staining of tumor cells for the parietal cell specific antibodies to H/K-ATPase and human milk fat globule-2 (HMFG-2).


Asunto(s)
Adenocarcinoma/secundario , Cardias/patología , Células Parietales Gástricas/patología , Neoplasias Gástricas/patología , Adenocarcinoma/metabolismo , Adenocarcinoma/ultraestructura , Anciano , Diferenciación Celular , Diagnóstico Diferencial , Resultado Fatal , Humanos , Inmunofenotipificación , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/ultraestructura , Masculino , Células Parietales Gástricas/metabolismo , Células Parietales Gástricas/ultraestructura , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA