Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 785
Filtrar
1.
Helicobacter ; 29(2): e13072, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38686467

RESUMEN

BACKGROUND: Helicobacter pylori infection is one of the main causes of gastric cancer. thioredoxin-1 (Trx1) and arginase (RocF) expressed by H. pylori were found to be closely related to its pathogenicity. However, whether Trx1 and RocF can be used in clinical screening of highly pathogenic H. pylori and the pathogenesis of trx1 high expressing H. pylori remain still unknown. MATERIALS AND METHODS: We investigated the expression level of H. pylori trx1 and H. pylori rocF in human gastric antrum tissues using reverse transcription and quantitative real-time PCR (RT-qPCR) and clarified the clinical application value of trx1 and rocF for screening highly pathogenic H. pylori. The pathogenic mechanism of Trx1 were further explored by RNA-seq of GES-1 cells co-cultured with trx1 high or low expressing H. pylori. Differentially expressed genes and signaling pathways were validated by RT-qPCR, Enzyme-linked immunosorbent assay (ELISA), western blot, immunohistochemistry and immunofluorescence. We also assessed the adherence of trx1 high and low expressing H. pylori to GES-1 cells. RESULTS: We found that H. pylori trx1 and H. pylori rocF were more significantly expressed in the gastric cancer and peptic ulcer group than that in the gastritis group and the parallel diagnosis of H. pylori trx1 and H. pylori rocF had high sensitivity. The trx1 high expressing H. pylori had stronger adhesion ability to GES-1 cells and upregulated the interleukin (IL) 23A/nuclear factor κappaB (NF-κB)/IL17A, IL6, IL8 pathway. CONCLUSIONS: H. pylori trx1 and H. pylori rocF can be used in clinical screening of highly pathogenic H. pylori and predicting the outcome of H. pylori infection. The trx1 high expressing H. pylori has stronger adhesion capacity and promotes the development of gastric diseases by upregulating the activation of NF-κB signaling pathway.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Interleucina-8 , FN-kappa B , Tiorredoxinas , Humanos , Helicobacter pylori/genética , Helicobacter pylori/fisiología , Helicobacter pylori/patogenicidad , Tiorredoxinas/metabolismo , Tiorredoxinas/genética , FN-kappa B/metabolismo , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/metabolismo , Interleucina-8/metabolismo , Interleucina-8/genética , Regulación hacia Arriba , Transducción de Señal , Arginasa/metabolismo , Arginasa/genética , Línea Celular , Gastropatías/microbiología , Gastropatías/metabolismo , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología
3.
Vet Pathol ; 61(4): 604-608, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38440930

RESUMEN

Mannheimia haemolytica-associated abomasitis has been clinically described as a cause of sudden death in lambs, but it is poorly characterized. We describe the pathological features of a severe fibrinonecrotizing abomasitis in 3 lambs that died suddenly. All 3 abomasums had a thickened submucosa due to edema and necrotic areas delimited by bands of degenerate neutrophils with slender nuclei (oat cells) and angiocentric distributions. The overlying mucosa was congested. Myriads of gram-negative coccobacilli were observed within the oat cell bands. M. haemolytica was isolated from the abomasum in all 3 animals and was serotyped as A2 in one of them. Pericarditis and pleuritis were observed in 2 of the lambs. Clostridium spp. were isolated in 1 lamb and detected by immunohistochemistry in the 3 animals, suggesting clostridial co-infection. M. haemolytica should be considered among the differential diagnoses of necrotizing abomasitis in lambs.


Asunto(s)
Abomaso , Mannheimia haemolytica , Necrosis , Infecciones por Pasteurellaceae , Enfermedades de las Ovejas , Animales , Mannheimia haemolytica/aislamiento & purificación , Enfermedades de las Ovejas/patología , Enfermedades de las Ovejas/microbiología , Ovinos , Abomaso/patología , Abomaso/microbiología , Infecciones por Pasteurellaceae/veterinaria , Infecciones por Pasteurellaceae/patología , Infecciones por Pasteurellaceae/microbiología , Necrosis/veterinaria , Necrosis/patología , Necrosis/microbiología , Gastropatías/veterinaria , Gastropatías/patología , Gastropatías/microbiología , Masculino , Femenino , Inmunohistoquímica/veterinaria
5.
Lancet ; 402 Suppl 1: S95, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37997142

RESUMEN

BACKGROUND: The exposure patterns across ethnic groups are unclear for stomach bugs that cause self-limiting symptoms, significantly burdening UK health-care services and the economy. This study seeks to fill this gap by exploring how inequalities arise in managing stomach bugs in UK ethnic groups. METHODS: A qualitative study using semi-structured interviews was undertaken. Ethics approval was given by the University of Liverpool, and data were collected by IZ over 11 months from July 26, 2022, and May 26, 2023. Purposive sampling was used to recruit a general UK population sample (excluding health-care professionals) who were adults, partners, and parents, from an ethnic minority group, with recent diarrhoea, vomiting, or a stomach bug over the past 6 months. Recruitment was conducted through community gatekeepers using flyers. Participants were interviewed in person or virtually and gave written informed consent. An incentive of an Amazon voucher of £10 was imbursed to participants for their time. Interviews were audio-recorded using a password-protected digital recorder, transcribed verbatim, and analysed using reflexive thematic analysis. FINDINGS: 36 interviews (median age 31·5 years) were conducted with 11 women of Pakistani (n=6), Bangladeshi (n=2), Indian (n=2) and Arab ethnicity (n=1), and 25 men of Black (n=22), Pakistani (n=2), and Indian (n=1) ethnicity. This sample enabled an exploration of within-ethnic group experiences of stomach bugs in participants who self-defined their age, sex, and ethnicity. Themes such as managing food preparation (n=16), travel abroad (n=17), and personal cleanliness (n=3) were consistently reported across transcripts. The findings corroborate existing literature that there are more similarities than divergences in the management of stomach bugs across ethnic groups, such as the burden of care disproportionately affecting women and using over-the-counter medication to manage symptoms. INTERPRETATION: We do not know if the impact of these experiences across ethnic groups is entirely representative of the broader ethnic categories (ie, Asian vs Indian, Pakistani, and Bangladeshi) they belong to or if there are inequalities in their impact on ethnic groups living in different circumstances (ie, UK born vs migrant). FUNDING: National Institute for Health and Care Research (NIHR).


Asunto(s)
Diarrea , Grupos Minoritarios , Gastropatías , Estómago , Vómitos , Adulto , Femenino , Humanos , Masculino , Pueblo Asiatico , Etnicidad , Estómago/microbiología , Reino Unido/epidemiología , Investigación Cualitativa , Población Negra , Diarrea/etnología , Diarrea/microbiología , Vómitos/etnología , Vómitos/microbiología , Gastropatías/etnología , Gastropatías/microbiología
6.
Int J Mol Sci ; 24(9)2023 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-37175810

RESUMEN

Genetic predisposition, environmental factors, and infectious agents interact in the development of gastric diseases. Helicobacter pylori (Hp) and Epstein-Barr virus (EBV) infection has recently been shown to be correlated with these diseases. A cross-sectional study was performed on 100 hospitalized Italian patients with and without gastric diseases. The patients were stratified into four groups. Significant methylation status differences among CDH1, DAPK, COX2, hMLH1 and CDKN2A were observed for coinfected (Hp-EBV group) patients; particularly, a significant presence of COX2 (p = 0.0179) was observed. For microsatellite instability, minor stability was described in the Hp-HBV group (69.23%, p = 0.0456). Finally, for p53 mutation in the EBV group, exon 6 was, significantly, most frequent in comparison to others (p = 0.0124), and in the Hp-EBV group exon 8 was, significantly, most frequent in comparison to others (p < 0.0001). A significant positive relationship was found between patients with infection (Hp, EBV or both) and p53 mutation (rho = 0.383, p = 0.0001), methylation status (rho = 0.432, p < 0.0001) and microsatellite instability (rho = 0.285, p = 0.004). Finally, we observed among infection and methylation status, microsatellite instability, and p53 mutation a significant positive relationship only between infection and methylation status (OR = 3.78, p = 0.0075) and infection and p53 mutation (OR = 6.21, p = 0.0082). According to our analysis, gastric disease in the Sicilian population has different pathways depending on the presence of various factors, including infectious agents such as Hp and EBV and genetic factors of the subject.


Asunto(s)
Metilación de ADN , Infecciones por Virus de Epstein-Barr , Genes p53 , Infecciones por Helicobacter , Inestabilidad de Microsatélites , Gastropatías , Helicobacter pylori , Infecciones por Helicobacter/complicaciones , Infecciones por Virus de Epstein-Barr/complicaciones , Herpesvirus Humano 4 , Coinfección , Gastropatías/genética , Gastropatías/microbiología , Gastropatías/virología , Genes p53/genética , Mutación , Sicilia , Humanos , Masculino , Femenino , Adulto Joven , Adulto , Persona de Mediana Edad , Anciano , Anciano de 80 o más Años , Modelos Logísticos
8.
Cells ; 10(9)2021 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-34572132

RESUMEN

Mitochondria are essential organelles that are not only responsible for energy production but are also involved in cell metabolism, calcium homeostasis, and apoptosis. Targeting mitochondria is a key strategy for bacteria to subvert host cells' physiology and promote infection. Helicobacter (H.) pylori targets mitochondria directly. However, mitochondrial genome (mtDNA) polymorphism (haplogroup) is not yet considered an important factor for H. pylori infection. Here, we clarified the association of mitochondrial haplogroups with H. pylori prevalence and the ability to perform damage. Seven mtDNA haplogroups were identified among 28 H. pylori-positive subjects. Haplogroup B was present at a higher frequency and haplotype D at a lower one in the H. pylori population than in that of the H. pylori-negative one. The fibroblasts carrying high-frequency haplogroup displayed a higher apoptotic rate and diminished mitochondrial respiration following H. pylori infection. mtDNA mutations were accumulated more in the H. pylori-positive population than in that of the H. pylori-negative one in old age. Among the mutations, 57% were located in RNA genes or nonsynonymous protein-coding regions in the H. pylori-positive population, while 35% were in the H. pylori-negative one. We concluded that gastric disease caused by Helicobacter virulence could be associated with haplogroups and mtDNA mutations.


Asunto(s)
ADN Mitocondrial/genética , Haplotipos , Infecciones por Helicobacter/epidemiología , Helicobacter pylori/patogenicidad , Mutación , Gastropatías/epidemiología , Anciano , Femenino , Fibroblastos/metabolismo , Fibroblastos/microbiología , Fibroblastos/patología , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Genoma Mitocondrial , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/microbiología , Helicobacter pylori/aislamiento & purificación , Humanos , Masculino , Persona de Mediana Edad , Prevalencia , República de Corea/epidemiología , Gastropatías/complicaciones , Gastropatías/genética , Gastropatías/microbiología
9.
Am J Physiol Gastrointest Liver Physiol ; 321(5): G461-G476, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34431405

RESUMEN

Multiple theories have been proposed describing the pathogenic mechanisms of Helicobacter pylori (H. pylori)-associated gastric motility disorders. We assessed ex vivo pyloric activity in H. pylori-infected rats, and tried to explore the associated ghrelin hormone alteration and pyloric fibrogenesis. In addition, miR-1 was assessed in pyloric tissue samples, being recently accused of having a role in smooth muscle dysfunction. Ninety adult male Wistar albino rats were assigned into nine groups: 1) control group, 2) sterile broth (vehicle group), 3) amoxicillin control, 4) omeperazole control, 5) clarithromycin control, 6) triple therapy control, 7) H. pylori- group, 8) H. pylori-clarithromycin group, and 9) H. pylori-triple therapy group. Urease enzyme activity was applied as an indicator of H. pylori infection. Ex vivo pyloric contractility was evaluated. Serum ghrelin was assessed, and histological tissue evaluation was performed. Besides, pyloric muscle miR-1 expression was measured. The immunological epithelial to mesenchymal transition (EMT) markers; transforming growth factor ß (TGFß), α-smooth muscle actin (α-SMA), and E-cadherin-3 were also evaluated. By H. pylori infection, a significant (P < 0.001) reduced pyloric contractility index was recorded. The miR-1 expression was decreased (P < 0.001) in the H. pylori-infected group, associated with reduced serum ghrelin, elevated TGFß, and α-SMA levels and reduced E-cadherin levels. Decreased miR-1 and disturbed molecular pattern were improved by treatment. In conclusion, H. pylori infection was associated with reduced miR-1, epithelial to mesenchymal transition, and pyloric hypomotility. The miR-1 may be a target for further studies to assess its possible involvement in H. pylori-associated pyloric dysfunction, which might help in the management of human H. pylori manifestations and complications.NEW & NOTEWORTHY This work is investigating functional, histopathological, and molecular changes underlying Helicobacter pylori hypomotility and is correlating these with miR-1, whose disturbance is supposed to be involved in smooth muscle dysfunction and cell proliferation according to literature. Epithelial to mesenchymal transition and reduced ghrelin hormone may contribute to H. pylori infection-associated hypomotility. H. pylori infection was associated with reduced pyloric miR-1 expression. Targeting miR-1 could be valuable in the clinical management of pyloric hypofunction.


Asunto(s)
Transición Epitelial-Mesenquimal , Motilidad Gastrointestinal , Infecciones por Helicobacter/microbiología , Helicobacter pylori/patogenicidad , Músculo Liso/microbiología , Píloro/microbiología , Gastropatías/microbiología , Actinas/metabolismo , Animales , Antibacterianos/farmacología , Cadherinas/metabolismo , Modelos Animales de Enfermedad , Quimioterapia Combinada , Transición Epitelial-Mesenquimal/efectos de los fármacos , Motilidad Gastrointestinal/efectos de los fármacos , Ghrelina/sangre , Infecciones por Helicobacter/tratamiento farmacológico , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/fisiopatología , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Músculo Liso/efectos de los fármacos , Músculo Liso/metabolismo , Músculo Liso/fisiopatología , Inhibidores de la Bomba de Protones/farmacología , Píloro/efectos de los fármacos , Píloro/metabolismo , Píloro/fisiopatología , Ratas Wistar , Gastropatías/tratamiento farmacológico , Gastropatías/metabolismo , Gastropatías/fisiopatología , Factor de Crecimiento Transformador beta/metabolismo
10.
J Med Life ; 14(2): 176-180, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34104240

RESUMEN

The study of the pathogenetic treatment and prevention of Helicobacter pylori (Hp)-associated gastroduodenopathies (GDP) induced by nonsteroidal anti-inflammatory drugs (NSAIDs) in patients with osteoarthritis (OA) is one of the most serious problems in modern clinical medicine. Sixty patients with OA and concomitant Hp-associated GDP induced by NSAIDs were examined. The levels of epidermal growth factor (EDF), sAPO-1/Fas and tumor necrosis factor-α (TNF-α) were determined. Group I included 30 patients who received triple anti-Helicobacter (AHT) therapy, and group II included 30 patients who received rebamipide. Long-term effects were assessed 6 months and 1 year after treatment. All subjects showed a significant increase in TNF-α (4.7 times), EDF (2.2 times) and a decrease in sAPO-1/Fas (3.6 times) levels compared to healthy individuals. After 1 month of treatment, a significantly more significant decrease in TNF-α and an increase in sAPO-1/Fas and EDF was found in group II. In the long-term treatment, a further decrease in TNF-α and an increase in the content of sAPO-1/Fas levels were observed in all groups. However, these changes were significantly more significant in group I compared to group I. The long-term follow-up showed a declining trend of EDF in all groups. The data obtained indicate the effectiveness of rebamipide in the complex pathogenetic treatment and prevention of Hp-associated GDP induced by NSAIDs in patients with OA.


Asunto(s)
Antiinflamatorios no Esteroideos/efectos adversos , Antiinflamatorios no Esteroideos/uso terapéutico , Duodeno/patología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/fisiología , Osteoartritis/tratamiento farmacológico , Gastropatías/inducido químicamente , Gastropatías/microbiología , Factor de Crecimiento Epidérmico/sangre , Infecciones por Helicobacter/sangre , Humanos , Osteoartritis/sangre , Osteoartritis/complicaciones , Gastropatías/sangre , Factor de Necrosis Tumoral alfa/sangre , Receptor fas/sangre
11.
J Gastroenterol Hepatol ; 36(11): 3092-3101, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34089623

RESUMEN

BACKGROUND AND AIM: The biological characterization of microbial environment in early gastric cancer (EGC), other than Helicobacter pylori, is limited. This study aimed to explore the microbial microenvironment in chronic gastritis (CG), fundic gland polyps (FGPs), low-grade intraepithelial neoplasia (LGIN), and EGC. METHODS: 16S-rRNA gene sequencing and bioinformatic analysis were performed on 63 individuals with 252 mucosal biopsies or endoscopic submucosal dissection margin samples from endoscopy. RESULTS: The microbiota in gastric LGIN functions analogously to EGC in terms of functional prediction. Neoplastic lesions showed a significant difference to CG or FGPs in beta diversity of the microbiota. Bacteria genera including Paracoccus, Blautia, Barnesiella, Lactobacillus, Thauera, Collinsella were significantly enriched in gastric neoplastic mucosa (LGIN and EGC) compared with non-neoplastic tissues (CG and FGPs). While Pseudomonas and Kingella were depleted in neoplastic tissues. FGPs showed a distinctive microbial network system that negatively interacted with Helicobacter. CONCLUSIONS: In terms of the mucosal microbial microenvironment, gastric LGIN and EGC showed no significant difference as early neoplastic lesions. We observed a coordinated microbial microenvironment that correlated negatively with Helicobacter.


Asunto(s)
Carcinoma in Situ , Mucosa Gástrica , Gastritis/microbiología , Microbioma Gastrointestinal , Pólipos/microbiología , Neoplasias Gástricas , Infecciones Bacterianas/genética , Infecciones Bacterianas/microbiología , Biopsia , Carcinoma in Situ/microbiología , Carcinoma in Situ/patología , Enfermedad Crónica , Endoscopía Gastrointestinal , Fundus Gástrico/microbiología , Fundus Gástrico/patología , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Gastritis/patología , Microbioma Gastrointestinal/genética , Infecciones por Helicobacter/genética , Helicobacter pylori/genética , Humanos , Pólipos/patología , ARN Ribosómico 16S/genética , Análisis de Secuencia de ARN , Gastropatías/microbiología , Gastropatías/patología , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/patología , Microambiente Tumoral
12.
Int J Mol Sci ; 22(9)2021 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-34062919

RESUMEN

Extracellular vesicles (EVs) are cell-derived vesicles important in intercellular communication that play an essential role in host-pathogen interactions, spreading pathogen-derived as well as host-derived molecules during infection. Pathogens can induce changes in the composition of EVs derived from the infected cells and use them to manipulate their microenvironment and, for instance, modulate innate and adaptive inflammatory immune responses, both in a stimulatory or suppressive manner. Gastric cancer is one of the leading causes of cancer-related deaths worldwide and infection with Helicobacter pylori (H. pylori) is considered the main risk factor for developing this disease, which is characterized by a strong inflammatory component. EVs released by host cells infected with H. pylori contribute significantly to inflammation, and in doing so promote the development of disease. Additionally, H. pylori liberates vesicles, called outer membrane vesicles (H. pylori-OMVs), which contribute to atrophia and cell transformation in the gastric epithelium. In this review, the participation of both EVs from cells infected with H. pylori and H. pylori-OMVs associated with the development of gastric cancer will be discussed. By deciphering which functions of these external vesicles during H. pylori infection benefit the host or the pathogen, novel treatment strategies may become available to prevent disease.


Asunto(s)
Vesículas Extracelulares/metabolismo , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , Gastropatías/metabolismo , Membrana Externa Bacteriana/metabolismo , Progresión de la Enfermedad , Vesículas Extracelulares/genética , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/patología , Helicobacter pylori/patogenicidad , Humanos , Gastropatías/microbiología , Gastropatías/patología
14.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33753513

RESUMEN

Helicobacter suis, a bacterial species naturally hosted by pigs, can colonize the human stomach in the context of gastric diseases such as gastric mucosa-associated lymphoid tissue (MALT) lymphoma. Because H. suis has been successfully isolated from pigs, but not from humans, evidence linking human H. suis infection to gastric diseases has remained incomplete. In this study, we successfully in vitro cultured H. suis directly from human stomachs. Unlike Helicobacter pylori, the viability of H. suis decreases significantly on neutral pH; therefore, we achieved this using a low-pH medium for transport of gastric biopsies. Ultimately, we isolated H. suis from three patients with gastric diseases, including gastric MALT lymphoma. Successful eradication of H. suis yielded significant improvements in endoscopic and histopathological findings. Oral infection of mice with H. suis clinical isolates elicited gastric and systemic inflammatory responses; in addition, progression of gastric mucosal metaplasia was observed 4 mo postinfection. Because H. suis could be isolated from the stomachs of infected mice, our findings satisfied Koch's postulates. Although further prospective clinical studies are needed, H. suis, like H. pylori, is likely a gastric pathogen in humans. Furthermore, comparative genomic analysis of H. suis using complete genomes of clinical isolates revealed that the genome of each H. suis isolate contained highly plastic genomic regions encoding putative strain-specific virulence factors, including type IV secretion system-associated genes, and that H. suis isolates from humans and pigs were genetically very similar, suggesting possible pig-to-human transmission.


Asunto(s)
Infecciones por Helicobacter/genética , Helicobacter heilmannii/genética , Helicobacter heilmannii/patogenicidad , Gastropatías/microbiología , Estómago/microbiología , Factores de Virulencia/genética , Adulto , Animales , Modelos Animales de Enfermedad , Femenino , Genoma Bacteriano , Helicobacter heilmannii/aislamiento & purificación , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Porcinos , Sistemas de Secreción Tipo IV/genética , Virulencia/genética
15.
J Microbiol Biotechnol ; 31(3): 368-379, 2021 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-33622995

RESUMEN

Two virulence factors of Helicobacter pylori, cagA and vacA, have been known to play a role in the development of severe gastric symptoms. However, they are not always associated with peptic ulcer or gastric cancer. To predict the disease outcome more accurately, it is necessary to understand the risk of severe symptoms linked to other virulence factors. Several other virulence factors of H. pylori have also been reported to be associated with disease outcomes, although there are many controversial descriptions. H. pylori isolates from Koreans may be useful in evaluating the relevance of other virulence factors to clinical symptoms of gastric diseases because the majority of Koreans are infected by toxigenic strains of H. pylori bearing cagA and vacA. In this study, a total of 116 H. pylori strains from Korean patients with chronic gastritis, peptic ulcers, and gastric cancers were genotyped. The presence of virulence factors vacAs1c, alpA, babA2, hopZ, and the extremely strong vacuolating toxin was found to contribute significantly to the development of severe gastric symptoms. The genotype combination vacAs1c/alpA/babA2 was the most predictable determinant for the development of severe symptoms, and the presence of babA2 was found to be the most critical factor. This study provides important information on the virulence factors that contribute to the development of severe gastric symptoms and will assist in predicting clinical disease outcomes due to H. pylori infection.


Asunto(s)
Adhesinas Bacterianas/genética , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas Bacterianas/genética , Infecciones por Helicobacter/patología , Factores de Virulencia/genética , Adulto , Animales , Línea Celular , ADN Bacteriano/genética , Endonucleasas/genética , Femenino , Gastritis/microbiología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/genética , Helicobacter pylori/patogenicidad , Humanos , Masculino , Persona de Mediana Edad , Úlcera Péptica/microbiología , Conejos , República de Corea , Gastropatías/microbiología , Neoplasias Gástricas/microbiología
16.
Int J Infect Dis ; 103: 423-430, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33278617

RESUMEN

BACKGROUND: Helicobacter pylori is acquired largely in early childhood, but its association with symptoms and indirect biomarkers of gastric damage in apparently healthy children remains controversial. We aimed to relate persistent H. pylori infection in apparently healthy school-aged children with clinical, laboratory, and noninvasive biomarkers suggestive of gastric damage using a case-control design. MATERIALS AND METHODS: We followed up 83 children aged 4-5 years with persistent H. pylori infection determined by stool antigen detection and/or a urea breath test and 80 noninfected matched controls from a low-income to middle-income, periurban city in Chile for at least 3 years. Monitoring included clinical visits every 4 months and annual assessment by a pediatric gastroenterologist. A blood sample was obtained to determine laboratory parameters potentially associated with gastric damage (hemogram and serum iron and ferritin levels), biomarkers of inflammation (cytokines, pepsinogens I and II, and tissue inhibitor metalloproteinase 1), and expression of cancer-related genes KLK1, BTG3, and SLC5A8. RESULTS: Persistently infected children had higher frequency of epigastric pain on physical examination (40% versus 16%; P = 0.001), especially from 8 to 10 years of age. No differences in anthropometric measurements or iron-deficiency parameters were found. Persistent infection was associated with higher levels of pepsinogen II (median 12.7 ng/mL versus 9.0 ng/mL; P < 0.001); no difference was observed in other biomarkers or gene expression profiles. CONCLUSIONS: H. pylori infection in apparently asymptomatic school-aged children is associated with an increase in clinical symptoms and in the level of one significant biomarker, pepsinogen II, suggesting early gastric involvement.


Asunto(s)
Infecciones por Helicobacter/complicaciones , Helicobacter pylori , Pepsinógeno C/sangre , Gastropatías/microbiología , Anciano , Biomarcadores/sangre , Estudios de Casos y Controles , Niño , Preescolar , Chile/epidemiología , Heces/microbiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pepsinógeno A/sangre , Estómago , Gastropatías/epidemiología
17.
PLoS Pathog ; 16(12): e1008686, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33370399

RESUMEN

Helicobacter pylori chronically infects the stomach of approximately half of the world's population. Manifestation of clinical diseases associated with H. pylori infection, including cancer, is driven by strain properties and host responses; and as chronic infection persists, both are subject to change. Previous studies have documented frequent and extensive within-host bacterial genetic variation. To define how within-host diversity contributes to phenotypes related to H. pylori pathogenesis, this project leverages a collection of 39 clinical isolates acquired prospectively from a single subject at two time points and from multiple gastric sites. During the six years separating collection of these isolates, this individual, initially harboring a duodenal ulcer, progressed to gastric atrophy and concomitant loss of acid secretion. Whole genome sequence analysis identified 1,767 unique single nucleotide polymorphisms (SNPs) across isolates and a nucleotide substitution rate of 1.3x10-4 substitutions/site/year. Gene ontology analysis identified cell envelope genes among the genes with excess accumulation of nonsynonymous SNPs (nSNPs). A maximum likelihood tree based on genetic similarity clusters isolates from each time point separately. Within time points, there is segregation of subgroups with phenotypic differences in bacterial morphology, ability to induce inflammatory cytokines, and mouse colonization. Higher inflammatory cytokine induction in recent isolates maps to shared polymorphisms in the Cag PAI protein, CagY, while rod morphology in a subgroup of recent isolates mapped to eight mutations in three distinct helical cell shape determining (csd) genes. The presence of subgroups with unique genetic and phenotypic properties suggest complex selective forces and multiple niches within the stomach during chronic infection.


Asunto(s)
Úlcera Duodenal/microbiología , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/genética , Helicobacter pylori/genética , Gastropatías/microbiología , Animales , Atrofia/microbiología , Enfermedad Crónica , Ácido Gástrico , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Fenotipo , Polimorfismo de Nucleótido Simple , Gastropatías/patología
18.
Dis Aquat Organ ; 141: 157-169, 2020 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-33030444

RESUMEN

Gastritis and gastric ulcers are well-recognized symptoms in cetaceans, and the genus Helicobacter is considered as the main cause. In this study, we examined the gastric fluid of captive common bottlenose dolphins Tursiops truncatus with gastric diseases in order to isolate the organisms responsible for diagnosis and treatment. Four Gram-negative, rod-shaped isolates (TSBT, TSH1, TSZ, and TSH3) with tightly coiled spirals with 2-4 turns and 2-6 bipolar, sheathed flagella, were obtained from gastric fluids of common bottlenose dolphins with gastric diseases. Phylogenetic analysis, based on 16S rRNA, atpA, and 60 kDa heat-shock protein (hsp60) genes, demonstrated that these isolates form a novel lineage within the genus Helicobacter. Analyses of 16S rRNA, atpA, and hsp60 gene sequences showed that isolate TSBT was most closely related to H. cetorum MIT99-5656T (98.5% similarity), H. pylori ATCC 43504T (76.7% similarity), and H. pylori ATCC 43504T (78.0% similarity), respectively. Type strains of Helicobacter showing resistance to 2% NaCl have not been reported previously; however, these novel isolates were resistant to 2% NaCl. Culture supernatant of some isolates induced intracellular vacuolization in mammalian cultured cells. These data, together with the different morphological and biochemical characteristics of the isolates, reveal that these isolates represent a novel species for which we propose the name Helicobacter delphinicola sp. nov. with type strain TSBT (= JCM 32789T = TSD-183T). Future studies will confirm whether H. delphinicola plays a role in lesion etiopathogenesis in cetaceans.


Asunto(s)
Delfín Mular , Helicobacter , Gastropatías , Animales , Delfín Mular/microbiología , Helicobacter/genética , Helicobacter/aislamiento & purificación , Filogenia , ARN Ribosómico 16S/genética , Gastropatías/microbiología , Gastropatías/veterinaria
19.
Stroke ; 51(12): 3577-3583, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33040706

RESUMEN

BACKGROUND AND PURPOSE: Recent studies suggest that alteration of the normal gut microbiome contributes to atherosclerotic burden and cardiovascular disease. While many gastrointestinal diseases are known to cause disruption of the normal gut microbiome in humans, the clinical impact of gastrointestinal diseases on subsequent cerebrovascular disease remains unknown. We conducted an exploratory analysis evaluating the relationship between gastrointestinal diseases and ischemic stroke. METHODS: We performed a retrospective cohort study using claims between 2008 and 2015 from a nationally representative 5% sample of Medicare beneficiaries. We included only beneficiaries ≥66 years of age. We used previously validated diagnosis codes to ascertain our primary outcome of ischemic stroke. In an exploratory manner, we categorized gastrointestinal disorders by anatomic location, disease chronicity, and disease mechanism. We used Cox proportional hazards models to examine associations of gastrointestinal disorder categories and ischemic stroke with adjustment for demographics and established vascular risk factors. RESULTS: Among a mean of 1 725 246 beneficiaries in each analysis, several categories of gastrointestinal disorders were associated with an increased risk of ischemic stroke after adjustment for established stroke risk factors. The most notable positive associations included disorders of the stomach (hazard ratio, 1.17 [95% CI, 1.15-1.19]) and functional (1.16 [95% CI, 1.15-1.17]), inflammatory (1.13 [95% CI, 1.12-1.15]), and infectious gastrointestinal disorders (1.13 [95% CI, 1.12-1.15]). In contrast, we found no associations with stroke for diseases of the anus and rectum (0.97 [95% CI, 0.94-1.00]) or neoplastic gastrointestinal disorders (0.97 [95% CI, 0.94-1.00]). CONCLUSIONS: In exploratory analyses, several categories of gastrointestinal disorders were associated with an increased risk of future ischemic stroke after adjustment for demographics and established stroke risk factors.


Asunto(s)
Enfermedades Gastrointestinales/epidemiología , Accidente Cerebrovascular Isquémico/epidemiología , Anciano , Anciano de 80 o más Años , Femenino , Gastroenteritis/epidemiología , Gastroenteritis/microbiología , Enfermedades Gastrointestinales/microbiología , Microbioma Gastrointestinal , Humanos , Masculino , Medicare , Modelos de Riesgos Proporcionales , Estudios Retrospectivos , Factores de Riesgo , Gastropatías/epidemiología , Gastropatías/microbiología , Estados Unidos/epidemiología
20.
Cell Rep ; 32(11): 108159, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32937132

RESUMEN

Helicobacter pylori (Hp) is an important human pathogen associated with gastric inflammation and neoplasia. It is commonly believed that this bacterium avoids major immune recognition by Toll-like receptors (TLRs) because of low intrinsic activity of its flagellin and lipopolysaccharides (LPS). In particular, TLR5 specifically detects flagellins in various bacterial pathogens, while Hp evolved mutations in flagellin to evade detection through TLR5. Cancerogenic Hp strains encode a type IV secretion system (T4SS). The T4SS core component and pilus-associated protein CagY, a large VirB10 ortholog, drives effector molecule translocation. Here, we identify CagY as a flagellin-independent TLR5 agonist. We detect five TLR5 interaction sites, promoting binding of CagY-positive Hp to TLR5-expressing cells, TLR5 stimulation, and intracellular signal transduction. Consequently, CagY constitutes a remarkable VirB10 member detected by TLR5, driving crucial innate immune responses by this human pathogen.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Helicobacter pylori/metabolismo , Secuencias Repetitivas de Aminoácido , Receptor Toll-Like 5/metabolismo , Animales , Sitios de Unión , Secuencia Conservada , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Células HEK293 , Humanos , Modelos Biológicos , Mutagénesis/genética , Péptidos/metabolismo , Dominios Proteicos , Gastropatías/microbiología , Gastropatías/patología , Relación Estructura-Actividad , Receptor Toll-Like 5/agonistas , Receptor Toll-Like 5/genética , Regulación hacia Arriba/genética , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...