Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Am J Ther ; 15(3): 225-30, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18496260

RESUMO

The objective of this study was to determine if coagulation is different between 6% hetastarch in normal saline (NS) and 6% hetastarch in lactated Ringer's solution (LR), with use of an ex vivo thromboelastography (TEG) model with healthy donated volunteer blood. We simulated hemodilution that occurs during clinical resuscitation of hemorrhagic or hypovolemic shock, using healthy human donor whole blood (WB) ex vivo. Coagulopathy related to low, medium, high, or very high dilution of WB with NS or a high-molecular-weight hetastarch-based plasma expander, 6% hetastarch in NS (HSNS) or 6% hetastarch in lactated Ringer's [Hextend (HSLR)], was analyzed by thromboelastography (TEG). No changes were noted in the TEG profile of undiluted WB controls during the 6-hour period of use (P > 0.95). Dilution with HSNS and HSLR significantly impaired coagulation compared to both WB control and NS. Progressive dilution with NS impaired coagulation but to a lesser extent than colloids (P < 0.01). Low dilution of blood with NS increased clot strength by 12% (not significant; P = 0.097). We conclude that WB containing citrate obtained from healthy donors for TEG analysis yields reproducible data over a minimum of 6 hours. Either hetastarch, when present at concentrations comparable to the manufacturer's maximum recommended dose of 20 mL/kg (equivalent to the high dilution used in these experiments), decreases clot tensile strength to levels associated with an increased risk of bleeding. Substitution of lactated Ringer's for NS in 6% hetastarch appears to offer no advantage in avoiding hemostatic compromise in an in vitro model.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Derivados de Hidroxietil Amido/farmacologia , Substitutos do Plasma/farmacologia , Tromboelastografia/métodos , Relação Dose-Resposta a Droga , Hemodiluição , Humanos , Derivados de Hidroxietil Amido/administração & dosagem , Técnicas In Vitro , Soluções Isotônicas/química , Veículos Farmacêuticos/química , Substitutos do Plasma/administração & dosagem , Lactato de Ringer , Cloreto de Sódio/química , Resistência à Tração , Fatores de Tempo
2.
J Cardiothorac Vasc Anesth ; 22(1): 34-9, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18249328

RESUMO

OBJECTIVES: Because hetastarches have deleterious effects on coagulation that increase with molecular weight (MWt), risk of coagulopathy associated with a high MWt hemoglobin-based oxygen carrier (HBOC) was studied. DESIGN: Preliminary laboratory study of donor blood using thromboelastography (TEG). SETTING: University laboratory. PARTICIPANTS: Volunteer donor blood. INTERVENTIONS: Experiments simulated hemodilution during clinical resuscitation of hemorrhagic shock with varying doses of HBOCs. Coagulopathy related to 1:11, 1:5, 1:2, or 1:1 dilution of whole blood with normal saline, 6% hetastarch (670 kilodaltons [kD]), hemoglobin glutamer-200 (HBOC-200, 200 kD), or OxyVita (OXYVITA Inc, New Windsor, NY) (a new-generation, zero-link polymerized bovine hemoglobin-based oxygen carrier, 33 megadaltons) were analyzed. MEASUREMENTS AND MAIN RESULTS: At 2 lower levels of hemodilution, hetastarch, HBOC-200, and OxyVita produced equivalent reductions in maximum clot strength (TEG-MA and TEG-G) that reached statistical significance compared with whole blood and normal saline. At 2 higher dilutions, OxyVita and HBOC-200 impaired maximum clot strength compared with whole blood, normal saline, and hetastarch. Dilution with hetastarch had a greater effect on clot propagation (K and alpha) than either HBOC. CONCLUSIONS: OxyVita and HBOC-200, HBOCs with different MWt, had similar effects on coagulation as measured by TEG. The impairment of coagulation by HBOCs and hetastarch occurred at doses corresponding to 12 mL/kg or a blood volume replacement of 17%. The use of HBOCs at doses corresponding to 23 mL/kg or a blood volume replacement of 33% significantly decreased coagulation to levels associated with increased clinical bleeding in this preliminary study. Minimal coagulopathic effects are expected with use of OxyVita at the manufacturer's anticipated effective dose of 10 g or 2 to 3 mL/kg.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Substitutos Sanguíneos/farmacologia , Hemoglobinas/farmacologia , Tromboelastografia/efeitos dos fármacos , Substitutos Sanguíneos/administração & dosagem , Relação Dose-Resposta a Droga , Hemoglobinas/administração & dosagem , Humanos , Derivados de Hidroxietil Amido/administração & dosagem , Derivados de Hidroxietil Amido/farmacologia , Choque Hemorrágico/sangue , Choque Hemorrágico/terapia , Cloreto de Sódio/farmacologia
3.
J Med Microbiol ; 52(Pt 3): 211-216, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12621085

RESUMO

Helicobacter pylori is an aetiological agent of gastric disease. Although the role of urease in gastric colonization of H. pylori has been shown, it remains unclear as to where urease is located in this bacterial cell. The purpose of this study was to define the urease-associated apparatus in the H. pylori cytoplasm. H. pylori was incubated at both a neutral and an acidic pH in the presence or absence of urea and examined by double indirect immunoelectron microscopy. The density of gold particles for UreA was greatest in the inner portion of the wild-type H. pylori cytoplasm at neutral pH but was greatest in the outer portion at acidic pH. This difference was independent of the presence of urea and was not observed in the ureI-deletion mutant. Also, the eccentric shift of urease in acidic pH was not observed in UreI. After a 2 day incubation period at acidic pH, it was observed that the urease gold particles in H. pylori assembled and were associated with UreI gold particles. Urease immunoreactivity shifted from the inner to the outer portion of H. pylori as a result of an extracellular decrease in pH. This shift was urea-independent and UreI-dependent, suggesting an additional role of UreI in urease-dependent acid resistance. This is the first report of the intracellular transport of molecules in bacteria in response to changes in the extracellular environment.


Assuntos
Helicobacter pylori/enzimologia , Proteínas de Membrana Transportadoras , Urease/metabolismo , Proteínas de Bactérias/genética , Citoplasma/enzimologia , Relação Dose-Resposta a Droga , Deleção de Genes , Genes Bacterianos/genética , Helicobacter pylori/efeitos dos fármacos , Helicobacter pylori/genética , Helicobacter pylori/ultraestrutura , Concentração de Íons de Hidrogênio , Ureia/metabolismo , Ureia/farmacologia
6.
Cancer Immunol Immunother ; 55(6): 663-71, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16133107

RESUMO

Dendritic cell (DC) administration to CD8alpha knock-out (CD8alphaKO) mice results in a strong antigen-non-specific protection to a B16 murine melanoma tumor challenge. This response is mediated by lytic NK cells and cytokine-producing CD4 cells. We aimed to determine the signals that guide tumor targeting of this response. CD8alphaKO mice in the C57BL/6 background received subcutaneous (s.c.) injections of immature DC. Mice were challenged in vivo or assayed for lytic activity in vitro to a panel of syngeneic tumors with different levels of MHC class I expression. These studies support the following conclusions: (1) DC administration to CD8alphaKO mice results in protective in vivo responses to syngeneic tumors from epithelial, neuroectodermal and hematopoietic origin; in vivo protection is independent of the level of MHC classes I and II expression. (2) The in vitro lytic activity of DC-activated NK cells from CD8alphaKO mice has sensitive and insensitive targets, which is independent of the cell lineage or the level of inhibitory self-MHC surface molecules. (3) In sensitive targets a putative activating NK ligand in DC-stimulated NK cells from CD8alphaKO mice signals directly to PI3-K, but is distinct from NKG2D.


Assuntos
Antígenos CD8/imunologia , Células Dendríticas/imunologia , Células Matadoras Naturais/imunologia , Ativação Linfocitária/imunologia , Neoplasias Experimentais/imunologia , Animais , Antígenos CD8/genética , Linhagem Celular Tumoral , Antígenos de Histocompatibilidade/imunologia , Camundongos , Camundongos Knockout , Fosfatidilinositol 3-Quinases/imunologia
7.
Physiology (Bethesda) ; 20: 429-38, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16287992

RESUMO

Helicobacter pylori is a Gram-negative neutralophile associated with peptic ulcers and gastric cancer. It has a unique ability to colonize the human stomach by acid acclimation. It uses the pH-gated urea channel, UreI, to enhance urea access to intrabacterial urease and a membrane-anchored periplasmic carbonic anhydrase to regulate periplasmic pH to approximately 6.1 in acidic media, whereas other neutralophiles cannot regulate periplasmic pH and thus only transit the stomach.


Assuntos
Adaptação Fisiológica , Ácido Gástrico/metabolismo , Helicobacter pylori/fisiologia , Animais , Helicobacter pylori/metabolismo , Humanos , Urease/metabolismo
8.
J Biol Chem ; 279(11): 9944-50, 2004 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-14701805

RESUMO

The size and complexity of many pH-gated channels have frustrated the development of specific structural models. The small acid-activated six-membrane segment urea channel of Helicobacter hepaticus (HhUreI), homologous to the essential UreI of the gastric pathogen Helicobacter pylori, enables identification of all the periplasmic sites of proton gating by site-directed mutagenesis. Exposure to external acidity enhances [(14)C]urea uptake by Xenopus oocytes expressing HhUreI, with half-maximal activity (pH(0.5)) at pH 6.8. A downward shift of pH(0.5) in single site mutants identified four of six protonatable periplasmic residues (His-50 at the boundary of the second transmembrane segment TM2, Glu-56 in the first periplasmic loop, Asp-59 at the boundary of TM3, and His-170 at the boundary of TM6) that affect proton gating. Asp-59 was the only site at which a protonatable residue appeared to be essential for pH gating. Mutation of Glu-110 or Glu-114 in PL2 did not affect the pH(0.5) of gating. A chimera, where the entire periplasmic domain of HhUreI was fused to the membrane domain of Streptococcus salivarius UreI (SsUreI), retained the pH-independent properties of SsUreI. Hence, proton gating of HhUreI likely depends upon the formation of hydrogen bonds by periplasmic residues that in turn produce conformational changes of the transmembrane domain. Further studies on HhUreI may facilitate understanding of other physiologically important pH-responsive channels.


Assuntos
Prótons , Ureia/química , Sequência de Aminoácidos , Animais , Ácido Aspártico/química , Proteínas de Bactérias/metabolismo , Transporte Biológico , Membrana Celular/metabolismo , Ácido Glutâmico/química , Helicobacter hepaticus/metabolismo , Helicobacter pylori/metabolismo , Histidina/química , Concentração de Íons de Hidrogênio , Proteínas de Membrana Transportadoras/metabolismo , Modelos Biológicos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Conformação Proteica , Estrutura Terciária de Proteína , RNA/metabolismo , Homologia de Sequência de Aminoácidos , Streptococcus/metabolismo , Ureia/metabolismo , Urease/metabolismo , Xenopus , Xenopus laevis
9.
Am J Physiol Gastrointest Liver Physiol ; 284(1): G96-G106, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12388207

RESUMO

Survival of Helicobacter pylori in acid depends on intrabacterial urease. This urease is a Ni(2+)-containing oligomeric heterodimer. Regulation of its activity and assembly is important for gastric habitation by this neutralophile. The gene complex encodes catalytic subunits (ureA/B), an acid-gated urea channel (ureI), and accessory assembly proteins (ureE-H). With the use of yeast two-hybrid analysis for determining protein-protein interactions, UreF as bait identified four interacting sequences encoding UreH, whereas UreG as bait detected five UreE sequences. These results were confirmed by coimmunoprecipitation and beta-galactosidase assays. Native PAGE immunoblotting of H. pylori inner membranes showed interaction of UreA/B with UreI, whereas UreI deletion mutants lacked this protein interaction. Deletion of ureE-H did not affect this interaction with UreI. Hence, the accessory proteins UreE/G and UreF/H form dimeric complexes and UreA/B form a membrane complex with UreI, perhaps enabling assembly of the urease apoenzyme at the membrane surface and immediate urea access to intrabacterial urease to allow rapid periplasmic neutralization.


Assuntos
Helicobacter pylori/enzimologia , Proteínas de Membrana Transportadoras , Urease/genética , Urease/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Membrana Celular/enzimologia , Eletroforese , Ácido Gástrico , Técnicas In Vitro , Mutagênese/fisiologia , Proteínas de Ligação a Fosfato , Testes de Precipitina , Técnicas do Sistema de Duplo-Híbrido
10.
Gastroenterology ; 123(1): 187-95, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12105847

RESUMO

BACKGROUND & AIMS: Helicobacter pylori, a neutralophile, uses acid neutralization by urease to combat gastric acidity, allowing gastric colonization. Both acute and chronic acid resistance mechanisms are present. Acute mechanisms of acid adaptation could be due to surface urease, increased inner-membrane urea permeability via UreI, or both. Slower mechanisms may involve increased nickel insertion into apoenzyme, posttranscriptional regulation, or increased enzyme synthesis. The aim of this study was to further define regulation of urease under acidic conditions. METHODS: Surface-bound urease was analyzed by measurement of free and bound urease after centrifugation through a step gradient and by quantitative urease immunostaining of intact and fixed bacteria. Changes in urease synthesis or assembly were determined by incubation of the organisms at pH 5.5 or 7.0 in the absence and presence of chloramphenicol, urea, or nickel chelator and in ureI-positive and -negative organisms. RESULTS: The amount of surface urease was below detection limits with either centrifugation washing or immunostaining. Total bacterial urease activity was increased 3-5-fold by incubation at pH 5.5 in the presence of chloramphenicol but not in nickel-free medium or in ureI knockout organisms. There was also a 3-fold increase in survival of acid shock in acid-adapted organisms. CONCLUSIONS: Surface-bound urease is too low to contribute to acid resistance. Acidic medium pH induces UreI-dependent nickel incorporation into apoenzyme. This augmentation of urease activity increases survival in acid and is part of the gastric colonization strategy of the organism.


Assuntos
Ácidos/farmacologia , Helicobacter pylori/efeitos dos fármacos , Urease/metabolismo , Western Blotting , Membrana Celular/enzimologia , Resistência Microbiana a Medicamentos , Ativação Enzimática , Helicobacter pylori/metabolismo , Helicobacter pylori/fisiologia , Concentração de Íons de Hidrogênio , Distribuição Tecidual
11.
Annu Rev Physiol ; 65: 349-69, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12471160

RESUMO

Helicobacter pylori is a neutralophilic, gram-negative, ureolytic organism that is able to colonize the human stomach but does not survive in a defined medium with a pH <4.0 unless urea is present. In order to live in the gastric environment, it has developed a repertoire of acid resistance mechanisms that can be classified into time-independent, acute, and chronic responses. Time-independent acid resistance depends on the structure of the organism's inner and outer membrane proteins that have a high isoelectric point, thereby reducing their proton permeability. Acute acid resistance depends on the constitutive synthesis of a neutral pH optimum urease that is an oligomeric Ni(2+)-containing heterodimer of UreA and UreB subunits. Gastric juice urea is able to rapidly access intrabacterial urease when the periplasmic pH falls below approximately 6.2 owing to pH-gating of a urea channel, UreI. This results in the formation of NH3, which then neutralizes the bacterial periplasm to provide a pH of approximately 6.2 and an inner membrane potential of -101 mV, giving a proton motive force of approximately -200 mV. UreI is a six-transmembrane segment protein, with homology to the amiS genes of the amidase gene cluster and to UreI of Helicobacter hepaticus and Streptococcus salivarius. Expression of these UreI proteins in Xenopus oocytes has shown that UreI of H. pylori and H. hepaticus can transport urea only at acidic pH, whereas that of S. salivarius is open at both neutral and acidic pH. Site-directed mutagenesis and chimeric analysis have identified amino acids implicated in maintaining the closed state of the channel at neutral pH and other amino acids that play a structural role in channel function. Deletion of ureI abolishes the ability of the organism to survive in acid and also to colonize the mouse or gerbil stomach. However, if acid secretion is inhibited in gerbils, the deletion mutants do colonize but are eradicated when acid secretion is allowed to return, showing that UreI is essential for gastric survival and that the habitat of H. pylori at the gastric surface must fall to pH 3.5 or below. The chronic response is from increased Ni(2+) insertion into the apo-enzyme, which results in a threefold increase in urease, which is also dependent on expression of UreI. This allows the organism to live in either gastric fundus or gastric antrum depending on the level of acidity at the gastric surface. There are other effects of acid on transcript stability that may alter levels of protein synthesis in acid. Incubation of the organism at acidic pH also results in regulation of expression of a variety of genes, such as some outer membrane proteins, that constitutes an acid tolerance response. Understanding of these acid resistance and tolerance responses should provide novel eradication therapies for this carcinogenic gastric pathogen.


Assuntos
Gastrite/microbiologia , Gastrite/fisiopatologia , Infecções por Helicobacter/fisiopatologia , Helicobacter pylori/fisiologia , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/fisiologia , Helicobacter pylori/patogenicidade , Humanos , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/fisiologia , Dados de Sequência Molecular , Estômago/microbiologia , Estômago/fisiologia , Virulência
12.
J Bacteriol ; 184(22): 6155-62, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12399485

RESUMO

Secretion of proteins by Helicobacter pylori may contribute to gastric inflammation and epithelial damage. An in vitro analysis was designed to identify proteins released by mechanisms other than nonspecific lysis. The radioactivity of proteins in the supernatant was compared with that of the intact organism by two-dimensional gel phosphorimaging following a 4-h pulse-chase. The ratio of the amount of UreB, a known cytoplasmic protein, in the supernatant to that in the pellet was found to be 0.25, and this was taken as an index of lysis during the experiments (n = 6). Ratios greater than that of UreB were used to distinguish proteins that were selectively released into the medium. Thus, proteins enriched more than 10-fold in the supernatant compared to UreB were identified by mass spectrometry. Sixteen such proteins were present in the supernatant: VacA; a conserved secreted protein (HP1286); putative peptidyl cis-trans isomerase (HP0175); six proteins encoded by HP0305, HP0231, HP0973, HP0721, HP0129, and HP0902; thioredoxin (HP1458); single-stranded-DNA-binding 12RNP2 precursor (HP0827); histone-like DNA-binding protein HU (HP0835); ribosomal protein L11 (HP1202); a putative outer membrane protein (HP1564); and outer membrane proteins Omp21 (HP0913) and Omp20 (HP0912). All except HP0902, thioredoxin, HP0827, HP0835, and HP1202 had a signal peptide. When nalidixic acid, a DNA synthesis inhibitor, was added to inhibit cell division but not protein synthesis, to decrease possible contamination due to outer membrane shedding, two outer membrane proteins (Omp21 and Omp20) disappeared from the supernatant, and the amount of VacA also decreased. Thus, 13 proteins were still enriched greater than 10-fold in the medium after nalidixic acid treatment, suggesting these were released specifically, possibly by secretion. These proteins may be implicated in H. pylori-induced effects on the gastric epithelium.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Helicobacter pylori/crescimento & desenvolvimento , Helicobacter pylori/metabolismo , Técnicas Bacteriológicas , Meios de Cultivo Condicionados/química , Eletroforese em Gel Bidimensional , Espectrometria de Massas , Metionina/metabolismo , Ácido Nalidíxico/farmacologia , Radioisótopos de Enxofre/metabolismo
13.
Infect Immun ; 72(4): 2358-68, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15039361

RESUMO

Reciprocal interactions between Helicobacter pylori and cells of the gastric epithelium to which it adheres may affect colonization. Changes in gene expression of H. pylori induced by adhesion to AGS gastric cancer cells by coculture were compared to changes in gene expression of H. pylori cultured without AGS cells by using cDNA filter macroarrays. Adhesion was quantitatively verified by confocal microscopy of green fluorescent protein-expressing bacteria. Four experiments showed that 22 and 21 H. pylori genes were consistently up- and down-regulated, respectively. The up-regulated genes included pathogenicity island, motility, outer membrane protein, and translational genes. The sigma(28) factor antagonist flgM, flgG, the stress response gene, flaA, omp11, and the superoxide dismutase gene (sodB) were down-regulated. The up-regulation of cag3, flgB, tonB, rho, and deaD was confirmed by quantitative PCR, and the up-regulation of lpxD, omp6, secG, fabH, HP1285, HP0222, and HP0836 was confirmed by reverse transcription (RT)-PCR. The down-regulation of flaA, sodB, and HP0874 was confirmed by quantitative PCR, and the down-regulation of omp11 was confirmed by RT-PCR. The alteration of gene expression in H. pylori after adhesion to gastric cells in vitro suggests that changes in motility, outer membrane composition, and stress responses, among other changes, may be involved in gastric colonization.


Assuntos
Aderência Bacteriana , Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Helicobacter pylori/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas de Bactérias/genética , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Helicobacter pylori/genética , Helicobacter pylori/metabolismo , Humanos , Reação em Cadeia da Polimerase , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Gástricas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA