Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biomed Sci ; 26(1): 23, 2019 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-30841890

RESUMEN

BACKGROUND: Helicobacter pylori bacteria colonize human gastric mucosa, cause chronic inflammation, peptic ulcers and gastric cancer. Colonization is mediated by H. pylori adhesins, which preferentially bind mucin 5 (MUC5AC) and Lewis (Le) determinants. The aim of this study was to evaluate the influence of H. pylori and their components on MUC5AC production and deposition of LeX/LeY in gastric epithelial cells in relation to bacterial adhesion using Caviae porcellus primary gastric epithelial cells and an in vivo model of experimental H. pylori infection in these animals. METHODS: MUCA5C and LeX/LeY were induced in vitro by live H. pylori reference strain CCUG 17874 (2 × 107 CFU/ml), H. pylori glycine acid extract (GE), 10 µg/ml; cytotoxin associated gene A (CagA) protein, 1 µl/ml; UreA urease subunit, 5 µg/ml; lipopolysaccharide (LPS) 25 ng/ml and imaged by fluorescence microscopy after anti-MUC5AC or anti-LeX/LeY FITC antibody staining. Bacterial adhesion was imaged by using anti-H. pylori FITC antibodies. The animals were inoculated per os with H. pylori (3 times in 2 days intervals, 1 × 1010 CFU/ml). After 7 or 28 days an infection and inflammation were assessed by histological, serological and molecular methods. Gastric tissue sections of infected and control animals were screend for MUCA5C and LeX, and H. pylori adhesion as above. RESULTS: MUC5AC production and deposition of Lewis determinants, especially LeX were upregulated in the milieu of live H. pylori as well as GE, CagA, UreA or LPS in vitro and in vivo during infection, more effectively in the acute (7 days) than in the chronic (28 days) phase of infection. This was related to enhanced adhesion of H. pylori, which was abrogated by anti-MUC5AC and anti-LeX or anti-LeY antibody treatment. CONCLUSIONS: Modulation of MUCA5C production and LeX/LeY deposition in the gastric mucosa by H. pylori can significantly increase gastric tissue colonization during H. pylori infection.


Asunto(s)
Infecciones por Helicobacter/inmunología , Helicobacter pylori/fisiología , Antígenos del Grupo Sanguíneo de Lewis/inmunología , Antígeno Lewis X/inmunología , Mucina 5AC/genética , Gastropatías/inmunología , Animales , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Cobayas , Infecciones por Helicobacter/microbiología , Masculino , Mucina 5AC/metabolismo , Estómago , Gastropatías/microbiología , Regulación hacia Arriba
2.
Biomacromolecules ; 14(1): 153-9, 2013 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-23215344

RESUMEN

Antibody-derived fragments have enormous potential application in solid-phase assays such as biomarker detection and protein purification. Controlled orientation of the immobilized antibody molecules is a critical requirement for the sensitivity and efficacy of such assays. We present an approach for covalent, correctly oriented attachment of scFv antibody fragments on solid supports. Glycosylated scFvs were expressed in Escherichia coli and the C-terminal, binding pocket-distal glycan tag was oxidized for covalent attachment to amine-functionalized beads. The glycosylated scFvs could be immobilized at salt concentrations that precluded nonspecific adsorption of unglycosylated molecules and the covalently attached antibody fragments exhibited 4-fold higher functional activity than ionically adsorbed scFvs. The glyco-tethered scFvs were stable in NaCl concentrations that removed greater than 90% of adsorbed scFvs and they exhibited improved stability of antigen binding over both adsorbed scFvs and soluble, nonimmobilized scFvs in accelerated degradation tests. The simple expression and immobilization approach reported is likely to find broad application in in vitro antibody tests.


Asunto(s)
Anticuerpos Inmovilizados/metabolismo , Polisacáridos/metabolismo , Ingeniería de Proteínas/métodos , Anticuerpos de Cadena Única/metabolismo , Anticuerpos Inmovilizados/química , Sitios de Unión/fisiología , Campylobacter jejuni/metabolismo , Polisacáridos/química , Estructura Secundaria de Proteína , Anticuerpos de Cadena Única/química
3.
J Immunol ; 186(4): 2462-71, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21220698

RESUMEN

Helicobacter pylori causes chronic gastritis, peptic ulcers, and gastric carcinoma. Gastric epithelial cells provide the first point of contact between H. pylori and the host. TLRs present on these cells recognize various microbial products, resulting in the initiation of innate immunity. Although previous reports investigated TLR signaling in response to intact H. pylori, the specific contribution of H. pylori LPS with regard to functional genomics and cell-signaling events has not been defined. This study set out to define downstream signaling components and altered gene expression triggered by H. pylori LPS and to investigate the role of the signaling protein tribbles 3 (TRIB3) during the TLR-mediated response to H. pylori LPS. Cotransfections using small interfering RNA and dominant-negative constructs demonstrated that H. pylori LPS functions as a classic TLR2 ligand by signaling through pathways involving the key TLR signaling components MyD88 adaptor-like, MyD88, IRAK1, IRAK4, TNFR-associated factor 6, IκB kinase ß, and IκBα. Microarray analysis, real-time PCR, and ELISA revealed the induction of a discrete pattern of chemokines as a direct effect of LPS:TLR2 signaling. H. pylori infection was associated with decreased expression of TRIB3 in human gastric epithelial cell lines and tissue samples. Additionally, H. pylori decreased expression of C/EBP homologous protein and activating transcription factor 4, the transcription factors involved in the induction of TRIB3 expression. Furthermore, knockdown of TRIB3 and C/EBP homologous protein enhanced TLR2-mediated NF-κB activation and chemokine induction in response to H. pylori LPS. Thus, modulation of TRIB3 by H. pylori and/or its products may be an important mechanism during H. pylori-associated pathogenesis.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Helicobacter pylori/inmunología , Lipopolisacáridos/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Represoras/fisiología , Transducción de Señal/inmunología , Receptor Toll-Like 2/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/biosíntesis , Línea Celular , Línea Celular Tumoral , Quimiocinas/biosíntesis , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/metabolismo , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Células HEK293 , Humanos , Inmunidad Innata/genética , Interleucina-8/biosíntesis , Interleucina-8/genética , Lipopolisacáridos/aislamiento & purificación , FN-kappa B/metabolismo , Regiones Promotoras Genéticas/inmunología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/biosíntesis , Transducción de Señal/genética , Receptor Toll-Like 2/fisiología , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
4.
J Biomed Biotechnol ; 2012: 206463, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22550396

RESUMEN

In this study we asked whether Helicobacter pylori whole cells and lipopolysaccharide (LPS) utilize sugar moieties of Lewis (Le) antigenic determinants to interact with DC-SIGN (dendritic cell specific ICAM grabbing nonintegrin) receptor on dendritic cells (DCs). For this purpose the soluble DC-SIGN/Fc adhesion assay and the THP-1 leukemia cells with induced expression of DC-SIGN were used. We showed that the binding specificity of DC-SIGN with H. pylori Le(X/Y) positive whole cells and H. pylori LPS of Le(X/Y) type was fucose dependent, whereas in Le(XY) negative H. pylori strains and LPS preparations without Lewis determinants, this binding was galactose dependent. The binding of soluble synthetic Le(X) and Le(Y) to the DC-SIGN-like receptor on THP-1 cells was also observed. In conclusion, the Le(XY) dependent as well as independent binding of H. pylori whole cells and H. pylori LPS to DC-SIGN was described. Moreover, we demonstrated that THP-1 cells may serve as an in vitro model for the assessment of H. pylori-DC-SIGN interactions mediated by Le(X) and Le(Y) determinants.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Helicobacter pylori/metabolismo , Lectinas Tipo C/metabolismo , Receptores de Superficie Celular/metabolismo , Moléculas de Adhesión Celular/química , Línea Celular Tumoral , Fucosa/química , Fucosa/metabolismo , Galactosa/química , Galactosa/metabolismo , Helicobacter pylori/química , Humanos , Lectinas Tipo C/química , Antígenos del Grupo Sanguíneo de Lewis/química , Antígenos del Grupo Sanguíneo de Lewis/metabolismo , Antígeno Lewis X/química , Antígeno Lewis X/metabolismo , Lipopolisacáridos/química , Lipopolisacáridos/metabolismo , Microscopía Fluorescente , Monocitos/química , Monocitos/metabolismo , Receptores de Superficie Celular/química
5.
Microbiol Immunol ; 56(1): 62-75, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22040089

RESUMEN

Helicobacter pylori (H.p) colonizes human gastric mucosa and causes gastric and duodenal ulcer disease or gastric cancer. Various H.p compounds may modulate the host immune response in regards to tolerance of the infection or disease development. The aim of this study was to determine whether H.p lipopolysaccharide (LPS) and glycine acid extract antigens (GE) or E. coli LPS influence the cytotoxic activity of peripheral blood lymphocytes from H.p infected - H.p (+) or uninfected - H.p (-) individuals, in the presence or absence of exogenous interleukin (IL)12. Individual H.p status was defined by the urea breath test. Lymphocytes, stimulated or not with H.p, and control antigens, with or without IL-12, were used as effector cells and epithelial HeLa cells as targets. The cytotoxicity of lymphocytes was expressed as the percentage of dead target cells unable to reduce tetrazolium salt. The supernatants from HeLa/lymphocyte cultures were used for detection of the cellular cytotoxicity markers granzyme B and caspase 8. The natural cytotoxic activity of lymphocytes from H.p (+) was less than that of H.p (-) donors. This may have been due to fewer natural killer cells of CD3(-) CD56(+) Nkp46(+) phenotype in H.p (+) in comparison to H.p (-) subjects. H.p GE and standard E. coli LPS enhanced the cytotoxicity of lymphocytes towards target cells whereas H.p LPS downregulated this activity. The decrease in lymphocyte cytotoxicity in response to H.p LPS correlated with a lack of IL-2 and IL-12 production, inhibition of interferon-γ production, and low IL-10 secretion by mononuclear leukocytes. IL-12 significantly enhanced the natural as well as H.p LPS and H.p GE driven cytotoxic capacity of lymphocytes. In conclusion, H.p LPS may negatively modulate natural cytotoxic activity and cytokine secretion by immunocompetent cells and thus be involved in the maintenance of infection and development of gastric pathologies.


Asunto(s)
Antígenos Bacterianos/inmunología , Infecciones por Helicobacter/inmunología , Helicobacter pylori/inmunología , Linfocitos T Citotóxicos/inmunología , Adulto , Pruebas Respiratorias/métodos , Caspasa 8/metabolismo , Medios de Cultivo/metabolismo , Escherichia coli/inmunología , Glicina/farmacología , Granzimas/metabolismo , Células HeLa , Infecciones por Helicobacter/microbiología , Helicobacter pylori/metabolismo , Humanos , Inmunoensayo , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Interleucina-12/inmunología , Interleucina-12/farmacología , Interleucina-2/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Activación de Linfocitos , Persona de Mediana Edad , Fenotipo , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/metabolismo , Sales de Tetrazolio/metabolismo
6.
Subcell Biochem ; 53: 209-40, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20593269

RESUMEN

Both Helicobacter pylori and Campylobacter jejuni are highly prevalent Gram-negative microaerophilic bacteria which are gastrointestinal pathogens of humans; H. pylori colonizes the gastroduodenal compartment and C. jejuni the intestinal mucosa. Although H. pylori causes chronic gastric infection leading to gastritis, peptic ulcers and eventually gastric cancer while C. jejuni causes acute infection inducing diarrhoeal disease, the endotoxin molecules of both bacterial species contrastingly contribute to their pathogenesis and the autoimmune sequelae each induces. Compared with enterobacterial endotoxin, that of H. pylori has significantly lower endotoxic and immuno-activities, the molecular basis for which is the underphosphorylation and underacylation of the lipid A component that interacts with immune receptors. This induction of low immunological responsiveness by endotoxin may aid the prolongation of H. pylori infection and therefore infection chronicity. On the other hand, this contrasts with acute infection-causing C. jejuni where overt inflammation contributes to pathology and diarrhoea production, and whose endotoxin is immunologically and endotoxically active. Futhermore, both H. pylori and C. jejuni exhibit molecular mimicry in the saccharide components of their endotoxins which can induce autoreactive antibodies; H. pylori expresses mimicry of Lewis and some ABO blood group antigens, C. jejuni mimicry of gangliosides. The former has been implicated in influencing the development of inflammation and gastric atrophy (a precursor of gastic cancer), the latter is central to the development of the neurological disorder Guillain-Barré syndrome. Both diseases raise important questions concerning infection-induced autoimmunity awaiting to be addressed.


Asunto(s)
Infecciones por Campylobacter/inmunología , Campylobacter jejuni/patogenicidad , Endotoxinas/inmunología , Infecciones por Helicobacter/inmunología , Helicobacter pylori/patogenicidad , Animales , Enfermedades Autoinmunes/inmunología , Campylobacter jejuni/inmunología , Conformación de Carbohidratos , Secuencia de Carbohidratos , Endotoxinas/química , Gangliósidos/química , Gangliósidos/inmunología , Síndrome de Guillain-Barré/inmunología , Helicobacter pylori/inmunología , Humanos , Antígenos del Grupo Sanguíneo de Lewis/química , Antígenos del Grupo Sanguíneo de Lewis/inmunología , Lípido A/química , Lípido A/inmunología , Imitación Molecular , Datos de Secuencia Molecular , Estructura Molecular
7.
Cells ; 10(6)2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-34199843

RESUMEN

BACKGROUND: Lipopolysaccharide (LPS) of Helicobacter pylori (Hp) bacteria causes disintegration of gastric tissue cells in vitro. It has been suggested that interleukin (IL)-33 is involved in healing gastric injury. AIM: To elucidate whether Hp LPS affects regeneration of gastric barrier initiated by IL-33. METHODS: Primary gastric epithelial cells or fibroblasts from Caviae porcellus were transfected with siRNA IL-33. Such cells, not exposed or treated with LPS Hp, were sub-cultured in the medium with or without exogenous IL-33. Then cell migration was assessed in conjunction with oxidative stress and apoptosis, activation of extracellular signal-regulated kinase (Erk), production of collagen I and soluble ST2 (IL-33 decoy). RESULTS: Control cells not treated with LPS Hp migrated in the presence of IL-33. The pro-regenerative activity of IL-33 was related to stimulation of cells to collagen I production. Wound healing by cells exposed to LPS Hp was inhibited even in the presence of IL-33. This could be due to increased oxidative stress and apoptosis in conjunction with Erk activation, sST2 elevation and modulation of collagen I production. CONCLUSIONS: The recovery of gastric barrier cells during Hp infection potentially can be affected due to downregulation of pro-regenerative activity of IL-33 by LPS Hp.


Asunto(s)
Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Mucosa Gástrica/fisiología , Helicobacter pylori/química , Interleucina-33/metabolismo , Lipopolisacáridos/farmacología , Regeneración/efectos de los fármacos , Animales , Cobayas , Lipopolisacáridos/química
8.
J Lipid Res ; 51(6): 1394-406, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20154333

RESUMEN

Fast migrating cerebrosides (FMC) are derivatives of galactosylceramide (GalCer). The structures of the most hydrophobic FMC-5, FMC-6, and FMC-7 were determined by electrospray ionization linear ion-trap mass spectrometry (MS) and nuclear magnetic resonance (NMR) spectroscopy complementing previous NMR spectroscopy and gas chromatography-mass spectrometry to be 3-O-acetyl-sphingosine-GalCer derivatives with galactose O-acetyl modifications. FMC-5 and FMC-6 are 3-O-acetyl-sphingosine-2,3,4,6-tetra-O-acetyl-GalCer with nonhydroxy and hydroxy-N-fatty-acids, while FMC-7 has an additional O-acetylation of the 2-hydroxy-fatty acid. The immuno-reactivity in human cerebrospinal fluid (CSF) to these acetylated glycolipids was examined in central nervous system (CNS) infectious disease, noninflammatory disorders, and multiple sclerosis (MS). Screening for lipid binding in MS and other neurological disease groups revealed that the greatest anti-hydrophobic FMC reactivity was observed in the inflammatory CNS diseases (meningitis, meningo-encephalitis, and subacute sclerosing panencephalitis). Some MS patients had increased reactivity with the hydrophobic FMCs and with glycoglycerophospholipid MfGL-II from Mycoplasma fermentans. The cross-reactivity of highly acetylated GalCer with microbial acyl-glycolipid raises the possibility that myelin-O-acetyl-cerebrosides, bacterial infection, and neurological disease are linked.


Asunto(s)
Líquido Cefalorraquídeo/inmunología , Galactosilceramidas/química , Galactosilceramidas/inmunología , Vaina de Mielina/química , Acetilación , Animales , Anticuerpos/inmunología , Encéfalo/citología , Bovinos , Reacciones Cruzadas , Ensayo de Inmunoadsorción Enzimática , Escherichia coli , Femenino , Galactosilceramidas/análisis , Glucolípidos/inmunología , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Lipopolisacáridos/inmunología , Espectroscopía de Resonancia Magnética , Masculino , Espectrometría de Masas , Mycoplasma fermentans , Ratas , Esfingosina/química
9.
Infect Immun ; 78(4): 1750-9, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20086085

RESUMEN

A human Campylobacter jejuni infection model provided controlled exposure to assess vaccine efficacy and investigate protective immunity for this important diarrheal pathogen. A well-characterized outbreak strain, C. jejuni 81-176, was investigated using a volunteer experimental infection model to evaluate the dose range and duration of protection. Healthy Campylobacter-seronegative adults received C. jejuni strain 81-176 via oral inoculation of 10(5), 10(7), or 10(9) CFU (5 adults/dose), which was followed by clinical and immunological monitoring. Based on dose range clinical outcomes, the 10(9)-CFU dose (n = 31) was used to assess homologous protection at 28 to 49 days (short-term veterans [STV]; n = 8) or 1 year (long-term veterans [LTV]; n = 7) after primary infection. An illness dose effect was observed for naïve subjects (with lower doses, 40 to 60% of the subjects were ill; with the 10(9)-CFU dose, 92% of the subjects were ill) along with complete protection for the STV group and attenuated illness for the LTV group (57%). Partial resistance to colonization was seen in STV (25% of the subjects were not infected; 3-log-lower maximum excretion level). Systemic and mucosal immune responses were robust in naïve subjects irrespective of the dose or the severity of illness. In contrast, in STV there was a lack of circulating antibody-secreting cells (ASC), reflecting the local mucosal effector responses. LTV exhibited comparable ASC responses to primary infection, and anamnestic fecal IgA responses likely contributed to self-resolving illness prior to antibiotic treatment. Campylobacter antigen-dependent production of gamma interferon by peripheral blood mononuclear cells was strongly associated with protection from illness, supporting the hypothesis that TH1 polarization has a primary role in acquired immunity to C. jejuni. This study revealed a C. jejuni dose-related increase in campylobacteriosis rates, evidence of complete short-term protection that waned with time, and immune response patterns associated with protection.


Asunto(s)
Vacunas Bacterianas/inmunología , Infecciones por Campylobacter/prevención & control , Campylobacter jejuni/inmunología , Administración Oral , Adulto , Anticuerpos Antibacterianos/sangre , Vacunas Bacterianas/administración & dosificación , Infecciones por Campylobacter/inmunología , Infecciones por Campylobacter/patología , Diarrea/inmunología , Diarrea/patología , Diarrea/prevención & control , Heces/química , Femenino , Experimentación Humana , Humanos , Inmunidad Mucosa , Inmunoglobulina A/análisis , Memoria Inmunológica , Interferón gamma/metabolismo , Leucocitos Mononucleares/inmunología , Masculino , Índice de Severidad de la Enfermedad , Factores de Tiempo , Adulto Joven
10.
Glycobiology ; 20(8): 1046-57, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20466654

RESUMEN

Salivary agglutinin plays a vital biological role modulating the protective effect in the oral cavity by interacting with a broad range of oral pathogens. Here, we describe the first characterization of the O-linked oligosaccharides of salivary agglutinin identified by negative ion liquid chromatography-mass spectrometry. The dominating structures were neutral or monosialylated core 1 (Galbeta1-3GalNAcalpha1-Ser/Thr) and core 2 (Galbeta1-3(GlcNAcbeta1-6)GalNAcalpha1-Ser/Thr) structures extended by fucosylated oligo-N-acetyllactosamine units. Oligosaccharides detected as [M-H](-) or [M-2H](2)(-) ions ranged from the disaccharide Galbeta1-3GalNAcol up to structures of almost 4000 Da, corresponding to core 1/2 structures with five N-acetyllactosamine units and 11 fucoses. Fucose was found either as terminal or internal blood group H structures in type 1 (Galbeta1-3GlcNAcbeta1-R), type 2 (Galbeta1-4GlcNAcbeta1-R) and type 3 (Galbeta1-3GalNAcalpha1-Ser/Thr) units, where the chains also could be fucosylated on GlcNAc yielding repeated Lewis a/b or Lewis x/y structures. Sialylation was located either at the non-reducing end of the N-acetyllactosamine chains as sialyl-Lewis x or as sialyl-T (NeuAcalpha2-3Galbeta1-3GalNAcalpha1-Ser/Thr) type structures with or without further extension of the C-6 branch of GalNAc with neutral fucosylated N-acetyllactosamine chains. The data indicated that sialylation, fucosylation and type 1 N-acetyllactosamine termination are important regulatory elements for controlling the oligosaccharide chain length. Furthermore, it was shown that these regulatory oligosaccharide elements could be utilized by the pathogen Helicobacter pylori to colonize the oral cavity, reside in dental plaque and serve as a reservoir for reinfection after successful clearance of H. pylori gastric infection.


Asunto(s)
Acetilglucosamina/química , Aglutininas/química , Helicobacter pylori/metabolismo , Antígenos del Grupo Sanguíneo de Lewis/química , Antígenos del Grupo Sanguíneo de Lewis/metabolismo , Oligosacáridos/química , Saliva/química , Acetilglucosamina/análogos & derivados , Sitios de Unión , Humanos , Oligosacáridos/metabolismo
11.
BMC Microbiol ; 10: 305, 2010 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-21118497

RESUMEN

BACKGROUND: Campylobacter jejuni is a major bacterial cause of food-borne enteritis, and its lipooligosaccharide (LOS) plays an initiating role in the development of the autoimmune neuropathy, Guillain-Barré syndrome, by induction of anti-neural cross-reactive antibodies through ganglioside molecular mimicry. RESULTS: Herein we describe the existence and heterogeneity of multiple LOS forms in C. jejuni strains of human and chicken origin grown at 37 °C and 42 °C, respectively, as determined on sodium dodecyl sulphate-polyacrylamide electrophoresis gels with carbohydrate-specific silver staining and blotting with anti-ganglioside ligands, and confirmed by nuclear magnetic resonance (NMR) spectroscopy. The C. jejuni NCTC 11168 original isolate (11168-O) was compared to its genome-sequenced variant (11168-GS), and both were found to have a lower-M(r) LOS form, which was different in size and structure to the previously characterized higher-M(r) form bearing GM1 mimicry. The lower-M(r) form production was found to be dependent on the growth temperature as the production of this form increased from ~5%, observed at 37 °C to ~35% at 42 °C. The structure of the lower-M(r) form contained a ß-D-Gal-(1→3)-ß-D-GalNAc disaccharide moiety which is consistent with the termini of the GM1, asialo-GM1, GD1, GT1 and GQ1 gangliosides, however, it did not display GM1 mimicry as assessed in blotting studies but was shown in NMR to resemble asialo-GM1. The production of multiple LOS forms and lack of GM1 mimicry was not a result of phase variation in the genes tested of NCTC 11168 and was also observed in most of the human and chicken isolates of C. jejuni tested. CONCLUSION: The presence of differing amounts of LOS forms at 37 and 42 °C, and the variety of forms observed in different strains, indicate that LOS form variation may play a role in an adaptive mechanism or a stress response of the bacterium during the colonization of different hosts.


Asunto(s)
Infecciones por Campylobacter/microbiología , Infecciones por Campylobacter/veterinaria , Campylobacter jejuni/metabolismo , Lipopolisacáridos/metabolismo , Enfermedades de las Aves de Corral/microbiología , Animales , Campylobacter jejuni/química , Campylobacter jejuni/genética , Campylobacter jejuni/aislamiento & purificación , Pollos , Humanos , Lipopolisacáridos/análisis , Fenotipo , Temperatura
12.
PLoS One ; 14(8): e0220636, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31390383

RESUMEN

BACKGROUND: Helicobacter pylori colonizes the human gastric mucosa, causing chronic inflammation, peptic ulcers and gastric cancer. A cascade of harmful processes results from the interaction of these bacteria with the gastric epithelium. AIM: To investigate these processes in terms of upregulation of oxidative stress and cell apoptosis and downregulation of the pro-regenerative activity of cells. METHODS: We employed an in vivo guinea pig model at 7 or 28 days postinoculation with H. pylori, corresponding to an acute or chronic stage of infection, respectively, and an in vitro model of guinea pig primary gastric epithelial cells and fibroblasts treated with bacterial components: glycine acid extract (GE), urease subunit A (UreA), cytotoxin-associated gene A protein (CagA) and lipopolysaccharide (LPS). Cells were evaluated for metabolic activity (MTT reduction), myeloperoxidase (MPO) and metalloproteinase (MMP-9) secretion, lipid peroxidation (4-hydroxynonenal (4HNE)), migration (wound healing), proliferation (Ki-67 antigen) and cell apoptosis (TUNEL assay; Bcl-xL, Bax, Bcl-2 expression; caspase 3 cleavage). RESULTS: Significant infiltration of the gastric mucosa by inflammatory cells in vivo in response to H. pylori was accompanied by oxidative stress and cell apoptosis, which were more intense 7 than 28 days after inoculation. The increase in cell proliferation was more intense in chronic than acute infection. H. pylori components GE, CagA, UreA, and LPS upregulated oxidative stress and apoptosis. Only H. pylori LPS inhibited cell migration and proliferation, which was accompanied by the upregulation of MMP-9. CONCLUSIONS: H. pylori infection induces cell apoptosis in conjunction with increased oxidative stress. Elevated apoptosis protects against deleterious inflammation and neoplasia; however, it reduces cell integrity. Upregulation of cell migration and proliferation in response to injury in the milieu of GE, CagA or UreA facilitates tissue regeneration but increases the risk of neoplasia. By comparison, downregulation of cell regeneration by H. pylori LPS may promote chronic inflammation.


Asunto(s)
Apoptosis , Proliferación Celular , Células Epiteliales/patología , Fibroblastos/patología , Mucosa Gástrica/patología , Infecciones por Helicobacter/patología , Helicobacter pylori/patogenicidad , Animales , Movimiento Celular , Células Epiteliales/microbiología , Fibroblastos/microbiología , Mucosa Gástrica/microbiología , Cobayas , Infecciones por Helicobacter/complicaciones , Humanos , Inflamación , Neoplasias/etiología , Estrés Oxidativo
13.
J Neurol Sci ; 275(1-2): 22-8, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18706662

RESUMEN

Multiple sclerosis (MS) is an autoimmune disorder characterised by clinical relapse and remission and pathological demyelination with varying inflammation. Because it is suggested that T-cells expressing natural killer cell receptors (NKR) play important roles in regulating human autoimmune diseases, we have quantified populations of T-cells expressing the NKR CD56, CD161 and CD94 in the peripheral blood of MS patients, in healthy control subjects (HS) and in patients with other neurological diseases (OND). CD161(+) T-cells and CD94(+) T-cells were significantly decreased in MS patients with primary progressive disease and secondary progressive disease respectively whereas CD56(+) T-cell numbers were unchanged. In contrast NKT-cells that express the invariant Valpha24-Jalpha18(+) T-cell receptor identified here by specific receptor antibody and CD1d-tetrameric PBS57-loaded complexes, were increased in MS patients compared with HS. Reductions in CD161(+) T-cells and CD94(+) T-cells relative to HS were also observed in the OND group and this was particularly prominent in Parkinsonian patients. A striking functional finding was that while NKT-cells in unfractionated peripheral blood from healthy subjects expanded in number and produced IFN-gamma upon stimulation with alpha-galactosylceramide, NKT-cells from MS patients did not. Thus we have identified alterations in a number of potentially important lymphocyte sub-populations warranting further investigation in the immune response in MS.


Asunto(s)
Galactosilceramidas/farmacología , Esclerosis Múltiple/patología , Células T Asesinas Naturales/efectos de los fármacos , Receptores de Células Asesinas Naturales/metabolismo , Adulto , Antígenos CD/metabolismo , Progresión de la Enfermedad , Femenino , Citometría de Flujo/métodos , Humanos , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/complicaciones , Enfermedades del Sistema Nervioso/complicaciones , Enfermedades del Sistema Nervioso/patología , Receptores de Antígenos de Linfocitos T/metabolismo , Estadísticas no Paramétricas , Adulto Joven
14.
Helicobacter ; 13(1): 1-19, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18205661

RESUMEN

Infection by the gastroduodenal pathogen Helicobacter pylori elicits a complex immunologic response in the mucosa involving neutrophils, plasma cells, eosinophils, and lymphocytes, of which T cells are the principal orchestrators of immunity. While so-called classical T cells (e.g. T-helper cells) that are activated by peptide fragments presented on antigen-presenting cells have received much attention in H. pylori infection, there exists a diverse array of other T cell populations that are potentially important for the outcome of the ensuing immune response, some of which have not been extensively studied in H. pylori infection. Pathogen-specific regulatory T cells that control and prevent the development of immunopathology associated with H. pylori infection have been investigated, but these cells can also benefit the bacterium in helping to prolong the chronicity of the infection by suppressing protective immune responses. An overlooked T cell population, the more recently described Th17 cells, may play a role in H. pylori-induced inflammation, due to triggering responses that ultimately lead to the recruitment of polymorphs, including neutrophils. The so-called innate or unconventional T cells, that include two conserved T cell subsets expressing invariant antigen-specific receptors, the CD1d-restricted natural killer T cells which are activated by glycolipids, and the mucosal-associated invariant T cells which play a role in defense against orally acquired pathogens in the intestinal mucosa, have only begun to receive attention. A greater knowledge of the range of T cell responses induced by H. pylori is required for a deeper understanding of the pathogenesis of this bacterium and its ability to perpetuate chronic infection, and could reveal new strategies for therapeutic exploitation.


Asunto(s)
Mucosa Gástrica/inmunología , Infecciones por Helicobacter/inmunología , Helicobacter pylori/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología , Humanos , Mucosa Intestinal/inmunología , Células Asesinas Naturales/inmunología
15.
Helicobacter ; 13(6): 500-5, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19166415

RESUMEN

BACKGROUND: Helicobacter pylori infection is associated with development of chronic inflammation and infiltration of immune cells into the gastric mucosa. As unconventional T-lymphocytes expressing natural killer cell receptors are considered to play central roles in the immune response against infection, a study investigating their frequencies in normal and H. pylori-infected gastric mucosa was undertaken. MATERIALS AND METHODS: Flow cytometry was used to quantify T-cells expressing the natural killer cell markers CD161, CD56, and CD94 in freshly isolated lymphocytes from the epithelial and lamina propria layers of gastric mucosa. Thirteen H. pylori-positive and 24 H. pylori-negative individuals were studied. RESULTS: CD94(+) T-cells were the most abundant (up to 40%) natural killer receptor-positive T-cell population in epithelial and lamina propria layers of H. pylori-negative gastric mucosa. CD161(+) T-cells accounted for about one-third of all T-cells in both compartments, but the lowest proportion were of CD56(+) T-cells. Compared with H. pylori-negative mucosa, in H. pylori-infected mucosa the numbers of CD161(+) T-cells were significantly greater (p = .04) in the epithelium, whereas the numbers of CD56(+) T-cells were lower (p = .01) in the lamina propria. A minor population (< 2%) of T-cells in both mucosal layers of H. pylori-negative subjects were natural killer T-cells, and whose proportions were not significantly different (p > .05) to those in H. pylori-infected individuals. CONCLUSIONS: The predominance, heterogeneity, and distribution of natural killer cell receptor-positive T-cells at different locations within the gastric mucosa reflects a potential functional role during H. pylori infection and warrants further investigation.


Asunto(s)
Mucosa Gástrica/inmunología , Infecciones por Helicobacter/inmunología , Helicobacter pylori/inmunología , Receptores de Células Asesinas Naturales/biosíntesis , Subgrupos de Linfocitos T/química , Subgrupos de Linfocitos T/inmunología , Adolescente , Adulto , Anciano , Antígeno CD56/análisis , Femenino , Citometría de Flujo , Humanos , Masculino , Persona de Mediana Edad , Subfamilia B de Receptores Similares a Lectina de Células NK/análisis , Subfamília D de Receptores Similares a Lectina de las Células NK/análisis
16.
Carbohydr Res ; 343(12): 1952-65, 2008 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-18279843

RESUMEN

Helicobacter pylori is a prevalent bacterial, gastroduodenal pathogen of humans that can express Lewis (Le) and related antigens in the O-chains of its surface lipopolysaccharide. The O-chains of H. pylori are commonly composed of internal Le(x) units with terminal Le(x) or Le(y) units or, in some strains, with additional units of Le(a), Le(b), Le(c), sialyl-Le(x) and H-1 antigens, as well as blood groups A and B, thereby producing a mosaicism of antigenic units expressed. The genetic determination of the Le antigen biosynthetic pathways in H. pylori has been studied, and despite striking functional similarity, low sequence homology occurs between the bacterial and mammalian alpha(1,3/4)- and alpha(1,2)-fucosyltransferases. Factors affecting Le antigen expression in H. pylori, that can influence the biological impact of this molecular mimicry, include regulation of fucosyltransferase genes through slipped-strand mispairing, the activity and expression levels of the functional enzymes, the preferences of the expressed enzyme for distinctive acceptor molecules and the availability of activated sugar intermediates. Le mimicry was initially implicated in immune evasion and gastric adaptation by the bacterium, but more recent studies show a role in gastric colonization and bacterial adhesion with galectin-3 identified as the gastric receptor for polymeric Le(x) on the bacterium. From the host defence aspect, innate immune recognition of H. pylori by surfactant protein D is influenced by the extent of LPS fucosylation. Furthermore, Le antigen expression affects both the inflammatory response and T-cell polarization that develops after infection. Although controversial, evidence suggests that long-term H. pylori infection can induce autoreactive anti-Le antibodies cross-reacting with the gastric mucosa, in part leading to the development of gastric atrophy. Thus, Le antigen expression and fucosylation in H. pylori have multiple biological effects on pathogenesis and disease outcome.


Asunto(s)
Fucosiltransferasas/metabolismo , Helicobacter pylori/metabolismo , Antígenos del Grupo Sanguíneo de Lewis/biosíntesis , Antígeno Lewis X/biosíntesis , Animales , Secuencia de Carbohidratos , Infecciones por Helicobacter/etiología , Helicobacter pylori/química , Helicobacter pylori/patogenicidad , Humanos , Datos de Secuencia Molecular , Estómago/microbiología
17.
Pol J Microbiol ; 57(3): 185-92, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19004238

RESUMEN

The Helicobacter pylori infections are followed by an infiltration of the gastric mucosa by neutrophils and macrophages. Accumulation of phagocytes enables them to interact with H. pylori, but a great number of infected subjects cannot eradicate these bacteria. The H. pylori inhibits its own uptake by blocking the function of phagocytes. The anti-phagocytic mechanism depends on bacterial surface structures and the presence of the cag pathogenicity island (PAI). The role of H. pylori lipopolysaccharide (LPS), during phagocytosis of these bacteria is not clear. LPS may mediate direct bacteria/phagocyte interactions and it may also regulate antibacterial activity of the phagocytes. In this study we investigated the influence of H. pylori LPS on phagocytosis of these bacteria. The H. pylori LPS inhibited an ingestion of these microbes by human peripheral blood granulocytes. This was correlated with a diminished ability of phagocytes to reduce MTT-tetrazolium salt. The anti-phagocytic effect of H. pylori LPS was reduced by recombinant lipopolysaccharide binding protein (rLBP). It is possible that in vivo H. pylori LPS may diminish elimination of these bacteria from the gastric mucosa promoting an infection persistence. However, LBP may modulate the uptake of H. pylori due to neutralization of anti-phagocytic effect of its LPS.


Asunto(s)
Helicobacter pylori/metabolismo , Inmunidad Innata/efectos de los fármacos , Lipopolisacáridos/farmacología , Neutrófilos/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Supervivencia Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Helicobacter pylori/genética , Helicobacter pylori/inmunología , Humanos , Lipopolisacáridos/metabolismo
18.
FEMS Immunol Med Microbiol ; 49(1): 124-32, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17266718

RESUMEN

Campylobacter jejuni is the leading bacterial cause of gastroenteritis worldwide. The present study was undertaken to determine the forms of polysaccharide-related compounds (PRCs) produced by C. jejuni and the culture conditions influencing their production. Expression of polysaccharides by C. jejuni was influenced by culture medium composition and growth phase. In addition to the production of lipooligosaccharide and capsular polysaccharide, a previously undescribed polysaccharide, not related to capsular polysaccharide, was shown to occur in C. jejuni in batch liquid and chemostat cultures. Thus, a variety of PRCs are produced by C. jejuni, and this should be considered when growing the bacterium in vitro for pathogenesis studies.


Asunto(s)
Campylobacter jejuni/crecimiento & desarrollo , Campylobacter jejuni/metabolismo , Polisacáridos Bacterianos/biosíntesis , Técnicas Bacteriológicas , Infecciones por Campylobacter/microbiología , Cromatografía en Capa Delgada , Medios de Cultivo , Electroforesis en Gel de Poliacrilamida , Humanos , Lectinas/metabolismo
20.
J Endotoxin Res ; 12(1): 47-56, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16420743

RESUMEN

Non-steroidal anti-inflammatory drugs (NSAIDs) cause peptic ulcer disease, but whether they interact with Helicobacter pylori to promote damage is controversial. Moreover, the reported induction of apoptosis in gastric cells by H. pylori lipopolysaccharide (LPS) (10(-9) g/ml) contrasts with studies showing low immunological potency of this LPS. Therefore, the effects of LPS from H. pylori NCTC 11637 and Escherichia coli O111:B4 on apoptosis in a primary culture of guinea-pig gastric mucous cells were investigated in the presence and absence of the NSAID, ibuprofen. Cell loss was estimated by a crystal violet assay, and apoptosis determined from caspase activity and from condensation and fragmentation of nuclei. Exposure to E. coli LPS for 24 h caused cell loss and enhanced apoptotic activity at concentrations >or= 10(-9) g/ml, but similar effects were only obtained with H. pylori LPS at concentrations >or= 10(-6) g/ml. Although ibuprofen (250 microM) caused cell loss and apoptosis, addition of either E. coli or H. pylori LPSs further enhanced these effects. In conclusion, LPS and ibuprofen interact to enhance gastric cell loss and apoptosis. In such interactions, E. coli LPS is more potent than that of H. pylori. The low potency of H. pylori LPS may contribute to a chronic low-grade gastritis that can be enhanced by the use of NSAIDs.


Asunto(s)
Antiinflamatorios no Esteroideos/toxicidad , Apoptosis/efectos de los fármacos , Escherichia coli/química , Mucosa Gástrica/citología , Helicobacter pylori/química , Ibuprofeno/toxicidad , Lipopolisacáridos/toxicidad , Animales , Bisbenzimidazol , Caspasas/metabolismo , Recuento de Células , Proliferación Celular/efectos de los fármacos , Colorantes , ADN/biosíntesis , Sinergismo Farmacológico , Células Epiteliales/efectos de los fármacos , Mucosa Gástrica/efectos de los fármacos , Violeta de Genciana , Cobayas , Técnicas In Vitro , Masculino , Timidina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA