Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.593
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-38728178

RESUMEN

A Gram-negative, facultative anaerobic, non-motile and rod-shaped bacterium, designated 10c7w1T, was isolated from a human gastrointestinal tract. Colonies on agar plates were small, circular, smooth and beige. The optimal growth conditions were determined to be 37 °C, pH 7.0-7.5 and 0 % (w/v) NaCl. Comparative analysis of complete 16S rRNA gene sequences revealed that strain 10c7w1T showed the highest sequence similarity of 95.8 % to Ottowia beijingensis MCCC 1A01410T, followed by Ottowia thiooxydans (95.2 %) JCM 11629T. The average amino acid identity values between 10c7w1T and O. beijingensis MCCC 1A01410T and O. thiooxydans JCM 11629T were above 60 % (71.4 and 69.5 %). The average nucleotide identity values between strain 10c7w1T and O. beijingensis MCCC 1A01410T and O. thiooxydans JCM 11629T were 76.9 and 72.5 %, respectively. The dominant fatty acids (≥10 %) were straight chain ones, with summed feature 3 (C16 : 1 ω7c/C16 : 1 ω6c), summed feature 8 (C18 : 1 ω7c/C18 : 1 ω6c) and C16 : 00 being the most abundant. Q-8 was the only respiratory quinone. The major polar lipids of strain 10c7w1T were phosphatidylethanolamine, diphosphatidylglycerol and unknown lipids. The DNA G+C content of strain 10c7w1T was 63.6 mol%. On the basis of phylogenetic, phenotypic and chemotaxonomic data, strain 10c7w1T is considered to represent a novel species within the genus Ottowia, for which the name Ottowia cancrivicina sp. nov. is proposed. The type strain is 10c7w1T (=MCCC 1H01399T=KCTC 92200T).


Asunto(s)
Técnicas de Tipificación Bacteriana , Composición de Base , ADN Bacteriano , Ácidos Grasos , Filogenia , ARN Ribosómico 16S , Análisis de Secuencia de ADN , Estómago , ARN Ribosómico 16S/genética , Ácidos Grasos/química , Humanos , ADN Bacteriano/genética , Estómago/microbiología , Hibridación de Ácido Nucleico , Ubiquinona , Fosfolípidos/química
2.
PLoS One ; 19(5): e0302522, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38758940

RESUMEN

Paddlefish has high economic and ecological value. In this study, microbial diversity and community structure in intestine, stomach, and mouth of paddlefish were detected using high-throughput sequencing. The results showed that the diversity and richness indices decreased along the digestive tract, and significantly lower proportion of those were observed in intestine. Firmicutes, Bacteroidetes and Proteobacteria were the dominant phyla. In top 10 phyla, there was no significant difference in mouth and stomach. But compared with intestine, there were significant differences in 8 of the 10 phyla, and Firmicutes and Bacteroidetes increased significantly, while Proteobacteria decreased significantly. There was no dominant genus in mouth and stomach, but Clostridium_sensu_stricto_1 and uncultured_bacterium_o_Bacteroidales was predominant in intestine. In conclusion, the species and abundance of microbiota in the mouth and stomach of paddlefish were mostly the same, but significantly different from those in intestine. Moreover, there was enrichment of the dominant bacteria in intestine.


Asunto(s)
Peces , Microbioma Gastrointestinal , Animales , Peces/microbiología , Tracto Gastrointestinal/microbiología , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Boca/microbiología , Estómago/microbiología , Proteobacteria/aislamiento & purificación , Proteobacteria/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Intestinos/microbiología , Bacteroidetes/aislamiento & purificación , Bacteroidetes/genética , Firmicutes/aislamiento & purificación , Firmicutes/genética , Firmicutes/clasificación , ARN Ribosómico 16S/genética , Biodiversidad
3.
World J Gastroenterol ; 30(16): 2281-2284, 2024 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-38690016

RESUMEN

The following are our views regarding the "letter to the editor" (Helicobacter is preserved in yeast vacuoles! Does Koch's postulates confirm it?) by Alipour and Gaeini, and the response "letter to the editor" (Candida accommodates non-culturable Helicobacter pylori in its vacuole-Koch's postulates aren't applicable) by Siavoshi and Saniee. Alipour and Gaeini rejected the methods, results, discussion, and conclusions summarized in a review article by Siavoshi and Saniee. The present article reviews and discusses evidence on the evolutionary adaptation of Helicobacter pylori (H. pylori) to thrive in Candida cell vacuoles and concludes that Candida could act as a Trojan horse, transporting potentially infectious H. pylori into the stomach of humans.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Helicobacter pylori/patogenicidad , Humanos , Infecciones por Helicobacter/microbiología , Candida/fisiología , Candida/crecimiento & desarrollo , Candida/patogenicidad , Vacuolas/microbiología , Vacuolas/metabolismo , Estómago/microbiología , Mucosa Gástrica/microbiología
4.
J Microbiol Methods ; 221: 106939, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38653334

RESUMEN

BACKGROUND: Fecal samples are commonly used for longitudinal studies of the gut lumen microbiome to track the course of response to infection or drug treatment, but no comparable method has been evaluated for longitudinal analysis of the gastric lumen microbiome in mice. Herein, a buffer flush of the stomach with a flexible gavage needle was used to collect gastric contents at one or several time points without harming the mouse. These samples were compared to samples collected by sacrifice and dissection of the mouse stomach. Microbiota from these samples were sequenced and evaluated in two ways: the composition of samples as measured by beta diversity and the richness of samples as measured by alpha diversity. Additionally, the effect of multiple sampling every two days on these metrics were studied. DNA was extracted from each of these samples and Illumina 16S rRNA gene sequencing was performed. RESULTS: First, taxonomic richness of gavage and dissection samples was compared. A greater number of taxa was detected in gavage samples than in dissection samples. Second, taxonomic richness was analyzed over time. No significant difference in taxonomic richness was observed with repeated gavage flushes. Third, a comparison was made of the taxonomic composition of samples collected by gavage versus dissection followed by a comparison of samples collected over multiple samplings. Nonmetric multidimensional scaling analysis revealed no clear differences between collection by gavage flushing or dissection. Using weighted Unifrac and Aitchison taxonomic distances between gavage and dissection samples were not significantly different from distances between gavage samples themselves, and no significant difference was found in the taxonomic composition of mice which were sampled repeatedly. Finally, relative abundances of specific identified taxa were compared, and eleven taxa were found to differ in frequency between collection methods. Using the more stringent Analysis of Composition of Microbiomes (ANCOM), seven was found to differ. Similarly, no significant differences were uncovered using these analyses over multiple samples by gastric flush. CONCLUSION: In summary, the consistency of the microbiota collected by gastric flushing recommends its use for microbiome analysis of gastric fluid similar to the use of fecal sampling to study the gut lumen microbiome.


Asunto(s)
Microbioma Gastrointestinal , ARN Ribosómico 16S , Manejo de Especímenes , Estómago , Animales , Ratones , ARN Ribosómico 16S/genética , Microbioma Gastrointestinal/genética , Manejo de Especímenes/métodos , Estómago/microbiología , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Estudios Longitudinales , Heces/microbiología , ADN Bacteriano/genética , Análisis de Secuencia de ADN/métodos , Biodiversidad , Ratones Endogámicos C57BL
5.
Benef Microbes ; 15(2): 165-177, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38653476

RESUMEN

The changing of microbiome could precede the development of coeliac disease (CeD). We compared the bacterial profile of microbiota of tissues collected simultaneously from the stomach and duodenum in newly diagnosed patients with CeD. Biopsies were collected from 60 children and adolescents aged 2-18 years: (1) 40 patients with CeD; (2) 20 children as control group. The evaluation of the bacterial microbiota was carried out by sequencing the V3-V4 regions of the 16S rRNA subunit, using next-generation sequencing (NGS). The composition of bacterial microbiota was correlated with clinical and blood parameters. The beta diversity analysis revealed a significant dissimilarity in the gastric samples between the CeD and control group (Bray-Curtis index, P = 0.008, and weighted UniFrac distance, P = 0.024). At L2 (phylum level), Campylobacterota was only present in the stomach of the CeD group. A comparison of the abundance of bacteria between the stomach and duodenum showed significant differences in 10 OTUs (operational taxonomic units) in the control and 9 OTUs in the CeD group at L6 (genus) and in 8 OTUs and in 6 OTUs, respectively, at L7 (species). A significant correlation was observed between the genus Novosphingobium in stomach of CeD group and possession of the DQ2.5 and DQ 8 allele, and in the duodenum - between the DQ 8 allele and the species Blautia wexlerae. Significant differences in selected, little-known genera of bacteria suggest their potential role as new biomarkers in the development of CeD. To fully understand the mechanism of CeD development in genetically predisposed individuals, it is necessary to take into account not only the abundance of a given genus or species of bacteria, but also the anatomical location of its occurrence.


Asunto(s)
Bacterias , Biomarcadores , Enfermedad Celíaca , Duodeno , Microbioma Gastrointestinal , ARN Ribosómico 16S , Estómago , Humanos , Enfermedad Celíaca/microbiología , Enfermedad Celíaca/diagnóstico , Niño , Proyectos Piloto , Preescolar , Duodeno/microbiología , Duodeno/patología , Adolescente , Masculino , ARN Ribosómico 16S/genética , Femenino , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Estómago/microbiología , Estómago/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Biopsia , ADN Bacteriano/genética
7.
Front Cell Infect Microbiol ; 14: 1339750, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38343887

RESUMEN

Infecting about half of the world´s population, Helicobacter pylori is one of the most prevalent bacterial infections worldwide and the strongest known risk factor for gastric cancer. Although H. pylori colonizes exclusively the gastric epithelium, the infection has also been associated with various extragastric diseases, including colorectal cancer (CRC). Epidemiological studies reported an almost two-fold increased risk for infected individuals to develop CRC, but only recently, direct causal and functional links between the chronic infection and CRC have been revealed. Besides modulating the host intestinal immune response, H. pylori is thought to increase CRC risk by inducing gut microbiota alterations. It is known that H. pylori infection not only impacts the gastric microbiota at the site of infection but also leads to changes in bacterial colonization in the distal large intestine. Considering that the gut microbiome plays a driving role in CRC, H. pylori infection emerges as a key factor responsible for promoting changes in microbiome signatures that could contribute to tumor development. Within this review, we want to focus on the interplay between H. pylori infection, changes in the intestinal microbiota, and intestinal immunity. In addition, the effects of H. pylori antibiotic eradication therapy will be discussed.


Asunto(s)
Neoplasias Colorrectales , Microbioma Gastrointestinal , Infecciones por Helicobacter , Helicobacter pylori , Humanos , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/microbiología , Helicobacter pylori/fisiología , Estómago/microbiología , Antibacterianos/uso terapéutico , Antibacterianos/farmacología , Neoplasias Colorrectales/epidemiología , Neoplasias Colorrectales/etiología
8.
Mol Microbiol ; 121(2): 260-274, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38173305

RESUMEN

There is growing evidence that bacterial morphology is closely related to their lifestyle. The helical Helicobacter pylori relies on its unique shape for survival and efficient colonization of the human stomach. Yet, they have been observed to transform into another distinctive morphology, the spherical coccoid. Despite being hypothesized to be involved in the persistence and transmission of this species, years of effort in deciphering the roles of the coccoid form remain fruitless since contrasting observations regarding its lifestyle were reported. Here, we discuss the two forms of H. pylori with a focus on the coccoid form, the molecular mechanism behind its morphological transformation, and experimental approaches to further develop our understanding of this phenomenon. We also propose a putative mechanism of the coccoid formation in H. pylori through induction of a type-I toxin-antitoxin (TA) system recently shown to influence the morphology of this species.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Humanos , Helicobacter pylori/genética , Estómago/microbiología , Infecciones por Helicobacter/microbiología
9.
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
10.
Infect Immun ; 91(12): e0036923, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-37937999

RESUMEN

Bacterial lipoproteins are post-translationally modified with acyl chains, anchoring these proteins to bacterial membranes. In Gram-negative bacteria, three enzymes complete the modifications. Lgt (which adds two acyl chains) and LspA (which removes the signal peptide) are essential. Lnt (which adds a third acyl chain) is not essential in certain bacteria including Francisella tularensis, Neisseria gonorrhoeae, and Acinetobacter baumannii. Deleting lnt results in mild to severe physiologic changes. We previously showed lnt is not essential for Helicobacter pylori growth in vitro. Here, the physiologic consequences of deleting lnt in H. pylori and the role of Lnt in the host response to H. pylori were examined using in vitro and in vivo models. Comparing wild-type, Δlnt, and complemented mutant H. pylori, no changes in growth rates or sensitivity to acid or antibiotics were observed. Since deleting lnt changes the number of acyl chains on lipoproteins and the number of acyl chains on lipoproteins impacts the innate immune response through Toll-like receptor 2 (TLR2) signaling, primary human gastric epithelial cells were treated with a purified lipoprotein from wild-type or lnt mutant H. pylori. Differential gene expression analysis indicated that lipoprotein from the lnt mutant induced a more robust TLR2 response. In a complementary approach, we infected wild-type and Tlr2-/- mice and found that both the wild-type and complemented mutant strains successfully colonized the animals. However, the lnt mutant strain was unable to colonize either mouse strain. These results show that lnt is essential for H. pylori colonization and identifies lipoprotein synthesis as a target for therapeutic intervention.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Animales , Ratones , Humanos , Helicobacter pylori/fisiología , Receptor Toll-Like 2/metabolismo , Estómago/microbiología , Lipoproteínas/genética , Lipoproteínas/metabolismo , Infecciones por Helicobacter/microbiología , Proteínas Bacterianas/metabolismo
11.
Helicobacter ; 28(6): e13020, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37691007

RESUMEN

Helicobacter pylori (H. pylori) is a gram-negative, microaerobic bacterium that colonizes the gastric mucosa in about half of the world's population. H. pylori infection can lead to various diseases. Chronic infection by H. pylori exposes the gastric mucosa to bacterial components such as lipopolysaccharide (LPS), outer membrane vesicles (OMVs), and several toxic proteins. Infected with H. pylori activates the release of pro-inflammatory factors and triggers inflammatory responses that damage the gastric mucosa. As the only microorganism that permanently colonizes the human stomach, H. pylori can suppress host immunity to achieve long-term colonization. Toll-like receptors (TLRs) play a crucial role in T-cell activation, promoting innate immune responses and immune tolerance during H. pylori infection. Among the 10 TLRs found in humans, TLR2, TLR4, TLR5, and TLR9 have been thoroughly investigated in relation to H. pylori-linked immune regulation. In the present review, we provide a comprehensive analysis of the various mechanisms employed by different TLRs in the induction of immune tolerance upon H. pylori infection, which will contribute to the research of pathogenic mechanism of H. pylori.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Humanos , Helicobacter pylori/fisiología , Infecciones por Helicobacter/microbiología , Receptores Toll-Like/metabolismo , Estómago/microbiología , Mucosa Gástrica/patología , Tolerancia Inmunológica
12.
Nat Commun ; 14(1): 1695, 2023 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-36973281

RESUMEN

The complement system has long been appreciated for its role in bloodborne infections, but its activities in other places, including the gastrointestinal tract, remain elusive. Here, we report that complement restricts gastric infection by the pathogen Helicobacter pylori. This bacterium colonized complement-deficient mice to higher levels than wild-type counterparts, particularly in the gastric corpus region. H. pylori uses uptake of the host molecule L-lactate to create a complement-resistant state that relies on blocking the deposition of the active complement C4b component on H. pylori's surface. H. pylori mutants unable to achieve this complement-resistant state have a significant mouse colonization defect that is largely corrected by mutational removal of complement. This work highlights a previously unknown role for complement in the stomach, and has revealed an unrecognized mechanism for microbial-derived complement resistance.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Ratones , Animales , Helicobacter pylori/genética , Mucosa Gástrica/microbiología , Ácido Láctico , Estómago/microbiología , Infecciones por Helicobacter/microbiología
13.
Int J Mol Sci ; 24(3)2023 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-36768541

RESUMEN

The oral cavity is the second most colonized site of Helicobacter pylori after the stomach. This study aimed to compare the genetic relatedness between gastric and oral H. pylori in Japanese patients with early gastric cancer through multilocus sequence typing (MLST) analysis using eight housekeeping genes. Gastric biopsy specimens and oral samples were collected from 21 patients with a fecal antigen test positive for H. pylori. The number of H. pylori allelic profiles ranged from zero to eight since the yield of DNA was small even when the nested PCR was performed. MLST analysis revealed that only one patient had a matching oral and gastric H. pylori genotype, suggesting that different genotypes of H. pylori inhabit the oral cavity and gastric mucosa. The phylogenetic analysis showed that oral H. pylori in six patients was similar to gastric H. pylori, implying that the two strains are related but not of the same origin, and those strains may be infected on separate occasions. It is necessary to establish a culture method for oral H. pylori to elucidate whether the oral cavity acts as the source of gastric infection, as our analysis was based on a limited number of allele sequences.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Boca , Neoplasias Gástricas , Estómago , Humanos , Genotipo , Infecciones por Helicobacter/complicaciones , Helicobacter pylori/genética , Tipificación de Secuencias Multilocus , Filogenia , Neoplasias Gástricas/genética , Boca/microbiología , Estómago/microbiología
14.
Chemistry ; 29(16): e202203672, 2023 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-36562295

RESUMEN

Infecting the stomach of almost 50 % of people, Helicobacter pylori is a causative agent of gastritis, peptic ulcers and stomach cancers. Interactions between bacterial membrane-bound lectin, Blood group Antigen Binding Adhesin (BabA), and human blood group antigens are key in the initiation of infection. Herein, the synthesis of a B-antigen hexasaccharide (B6) and a B-Lewis b heptasaccharide (BLeb7) and Bovine Serum Albumin glycoconjugates thereof is reported to assess the binding properties and preferences of BabA from different strains. From a previously reported trisaccharide acceptor a versatile key Lacto-N-tetraose tetrasaccharide intermediate was synthesized, which allowed us to explore various routes to the final targets, either via initial introduction of fucosyl residues followed by introduction of the B-determinant or vice versa. The first approach proved unsuccessful, whereas the second afforded the target structures in good yields. Protein conjugation using isothiocyanate methodology allowed us to reach high glycan loadings (up to 23 per protein) to mimic multivalent displays encountered in Nature. Protein glycoconjugate inhibition binding studies were performed with H. pylori strains displaying high or low affinity for Lewis b hexasaccharide structures showing that the binding to the high affinity strain was reduced due to the presence of the B-determinant in the Bleb7-conjugates and further reduced by the absence of the Lewis fucose residue in the B6-conjugate.


Asunto(s)
Antígenos de Grupos Sanguíneos , Infecciones por Helicobacter , Helicobacter pylori , Humanos , Adhesinas Bacterianas/química , Estómago/microbiología , Antígenos de Grupos Sanguíneos/metabolismo , Glicoconjugados/química , Infecciones por Helicobacter/microbiología
15.
Cell Mol Gastroenterol Hepatol ; 15(1): 213-236, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36167263

RESUMEN

BACKGROUND & AIMS: Histamine in the stomach traditionally is considered to regulate acid secretion but also has been reported to participate in macrophage differentiation, which plays an important role in tissue homeostasis. Therefore, this study aimed to uncover the precise role of histamine in mediating macrophage differentiation and in maintaining stomach homeostasis. METHODS: Here, we expand on this role using histidine decarboxylase knockout (Hdc-/-) mice with hypertrophic gastropathy. In-depth in vivo studies were performed in Hdc-/- mice, germ-free Hdc-/- mice, and bone-marrow-transplanted Hdc-/- mice. The stomach macrophage populations and function were characterized by flow cytometry. To identify stomach macrophages and find the new macrophage population, we performed single-cell RNA sequencing analysis on Hdc+/+ and Hdc-/- stomach tissues. RESULTS: Single-cell RNA sequencing and flow cytometry of the stomach cells of Hdc-/- mice showed alterations in the ratios of 3 distinct tissue macrophage populations (F4/80+Il1bhigh, F4/80+CD93+, and F4/80-MHC class IIhighCD74high). Tissue macrophages of the stomachs of Hdc-/- mice showed impaired phagocytic activity, increasing the bacterial burden of the stomach and attenuating hypertrophic gastropathy in germ-free Hdc-/- mice. The transplantation of bone marrow cells of Hdc+/+ mice to Hdc-/- mice recovered the normal differentiation of stomach macrophages and relieved the hypertrophic gastropathy of Hdc-/- mice. CONCLUSIONS: This study showed the importance of histamine signaling in tissue macrophage differentiation and maintenance of gastric homeostasis through the suppression of bacterial overgrowth in the stomach.


Asunto(s)
Diferenciación Celular , Histamina , Macrófagos , Estómago , Animales , Ratones , Histamina/fisiología , Histidina Descarboxilasa/genética , Estómago/microbiología , Síndrome del Asa Ciega , Ratones Noqueados
16.
Int J Mol Sci ; 23(24)2022 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-36555092

RESUMEN

Helicobacter pylori infection triggers inflammation that may lead to gastritis, stomach ulcers and cancer. Probiotic bacteria, such as Lactobacillus, have been of interest as treatment options, however, little is known about the molecular mechanisms of Lactobacillus-mediated inhibition of H. pylori pathogenesis. In this work, we investigated the effect of Lactobacillus culture supernatants, so-called conditioned medium (CM), from two gastric isolates, L. gasseri and L. oris, on the expression of transcriptional regulators in H. pylori. Among the four known two-component systems (TCSs), i.e., ArsRS, FlgRS, CheAY and CrdRS, the flagellar regulator gene flgR and the acid resistance associated arsS gene were down-regulated by L. gasseri CM, whereas expression of the other TCS-genes remained unaffected. L. gasseri CM also reduced the motility of H. pylori, which is in line with reduced flgR expression. Furthermore, among six transcription factors of H. pylori only the ferric uptake regulator gene fur was regulated by L. gasseri CM. Deletion of fur further led to dramatically increased sensitivity to the antimicrobial peptide LL-37. Taken together, the results highlight that released/secreted factors of some lactobacilli, but not all, downregulate transcriptional regulators involved in motility, acid tolerance and LL-37 sensitivity of H. pylori.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Humanos , Lactobacillus/fisiología , Helicobacter pylori/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Infecciones por Helicobacter/microbiología , Estómago/microbiología , Medios de Cultivo Condicionados/metabolismo
17.
Gut Microbes ; 14(1): 2152306, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36469575

RESUMEN

Individuals infected with Helicobacter pylori harbor unique and diverse populations of quasispecies, but diversity between and within different regions of the human stomach and the process of bacterial adaptation to each location are not yet well understood. We applied whole-genome deep sequencing to characterize the within- and between-stomach region genetic diversity of H. pylori populations from paired antrum and corpus biopsies of 15 patients, along with single biopsies from one region of an additional 3 patients, by scanning allelic diversity. We combined population deep sequencing with more conventional sequencing of multiple H. pylori single colony isolates from individual biopsies to generate a unique dataset. Single colony isolates were used to validate the scanning allelic diversity pipelines. We detected extensive population allelic diversity within the different regions of each patient's stomach. Diversity was most commonly found within non-coding, hypothetical, outer membrane, restriction modification system, virulence, lipopolysaccharide biosynthesis, efflux systems, and chemotaxis-associated genes. Antrum and corpus populations from the same patient grouped together phylogenetically, indicating that most patients were initially infected with a single strain, which then diversified. Single colonies from the antrum and corpus of the same patients grouped into distinct clades, suggesting mechanisms for within-location adaptation across multiple H. pylori isolates from different patients. The comparisons made available by combined sequencing and analysis of isolates and populations enabled comprehensive analysis of the genetic changes associated with H. pylori diversification and stomach region adaptation.


Asunto(s)
Microbioma Gastrointestinal , Infecciones por Helicobacter , Helicobacter pylori , Humanos , Helicobacter pylori/genética , Infecciones por Helicobacter/microbiología , Estómago/microbiología , Genómica
18.
BMC Vet Res ; 18(1): 407, 2022 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-36397114

RESUMEN

BACKGROUND: Compared with the stomach of ruminant cattle, the stomach of horse is small and mainly for chemical digestion, but the microorganisms in the stomach play an important role in maintaining the homeostasis of the internal environment. Due to the complexity of the microbes in the stomach, little is known about the diversity and structure of bacteria in the equine stomach. Grains are the main energy source for plant-eating livestock and energy is derived through enzymatic hydrolysis of grains into glucose or their microbial fermentation into Volatile fatty acids (VFA). However, the mechanism through which these ingested grains are chemically digested as well as the effect of these grains on the stomach remains elusive. This study explored the effects of feeding different grains (corn, oats, and barley) on bacterial diversity, structure, and composition in the foal's stomach content. Furthermore, the effects of different grains on the vitality of starch digestion-related stomach enzymes were investigated. RESULTS: No significant differences were observed (P > 0.05) in the bacterial rarefaction curves of Operational Taxonomic Units (OTUs) and diversity of the stomach microbiota in all foals. This study also revealed the statistical differences for Firmicutes, Cyanobacteria, Actinobacteria, Fibrobacteres, Lactobacillaceae, Streptococcaceae, Unidentified_Clostridiales, Prevotellaceae, Lactobacillus, Streptococcus, Unidentified_Cyanobacteria, Unidentified_Clostridiales, Lactococcus, Sphingomonas, Lactobacillus_hayakitensis, Lactobacillus_equigenerosi, and Clostridium_perfringens. The linear discriminant analysis effect size analysis revealed 9 bacteria at each classification level. The functional analysis of species information by using FAPROTAX software was able to predict 35 functions, and the top 5 functions were chemoheterotrophy, fermentation, animal_parasites_or_symbionts, nitrate_reduction, and aerobic_chemoheterotrophy. The study also revealed statistical differences for pH, glucose concentration, ß-amylase, maltase, and amylase. CONCLUSIONS: The different grains had no significant effect on the microbial diversity of the stomach content of the foal. However, the relative bacterial abundances differed significantly in response to different diets. Particularly, oats fed to the foals significantly increased the relative abundance of Firmicutes, Lactobacillaceae, Lactobacillus, and Lactobacillus_hayakitensis. The grain had no significant effect on the pH of the stomach content, glucose concentration, and enzyme viability in the foal.


Asunto(s)
Alimentación Animal , Bacterias , Digestión , Almidón , Estómago , Animales , Bacterias/clasificación , Grano Comestible , Glucosa , Caballos , Almidón/metabolismo , Estómago/metabolismo , Estómago/microbiología
19.
Int J Mol Sci ; 23(21)2022 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-36362445

RESUMEN

Helicobacter pylori (H. pylori) is transmitted primarily through the oral-oral route and fecal-oral route. The oral cavity had therefore been hypothesized as an extragastric reservoir of H. pylori, owing to the presence of H. pylori DNA and particular antigens in distinct niches of the oral cavity. This bacterium in the oral cavity may contribute to the progression of periodontitis and is associated with a variety of oral diseases, gastric eradication failure, and reinfection. However, the conditions in the oral cavity do not appear to be ideal for H. pylori survival, and little is known about its biological function in the oral cavity. It is critical to clarify the survival strategies of H. pylori to better comprehend the role and function of this bacterium in the oral cavity. In this review, we attempt to analyze the evidence indicating the existence of living oral H. pylori, as well as potential survival strategies, including the formation of a favorable microenvironment, the interaction between H. pylori and oral microorganisms, and the transition to a non-growing state. Further research on oral H. pylori is necessary to develop improved therapies for the prevention and treatment of H. pylori infection.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Periodontitis , Humanos , Helicobacter pylori/genética , Infecciones por Helicobacter/microbiología , Boca/microbiología , Estómago/microbiología , Periodontitis/microbiología
20.
Front Cell Infect Microbiol ; 12: 976710, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36237432

RESUMEN

Background: Curing refractory Helicobacter pylori infection is difficult. In addition, there is currently no research on the gastric microbiota of refractory H. pylori infection. Methods: We designed a clinical retrospective study involving 32 subjects divided into three groups: 1. nAGHp.a, treatment-naïve patients with H. pylori infection; 2. nAGHp.b, H. pylori-negative patients; and 3. EFHp.a, patients with refractory H. pylori infection. Gastric mucosal samples from the biobank of our research center were collected for 16S rRNA sequencing analysis and bacterial functions were predicted via PICRUSt. Results: There were significant differences between the H. pylori- positive group and the H. pylori-negative group in species diversity, gastric microbiota structure, and bacterial function. The beneficial Lactobacillus in the H. pylori-positive group were significantly enriched compared with those in the refractory H. pylori infection group. The bacterial interaction network diagram suggested that the microbiota interactions in the refractory H. pylori infection group decreased. The gastric microbiota of the refractory H. pylori infection group was enriched in the pathways of metabolism and infectious diseases (energy metabolism, bacterial secretion system, glutathione metabolism, protein folding and associated processing, sulphur metabolism, membrane and intracellular structural molecules, lipopolysaccharide biosynthesis, ubiquinone and other terpenoid-quinone biosynthesis, inorganic ion transport and metabolism, and metabolism of cofactors and vitamins) when compared with the H. pylori-positive group without treatment based on PICRUSt analysis. Conclusion: Significant alterations occurred in the gastric microbiota when eradication of H. pylori failed multiple times. A history of eradication of multiple H. pylori infections leads to an imbalance in the gastric mucosal microbiota to a certain extent, which was mainly reflected in the inhibition of the growth of beneficial Lactobacillus in the stomach. Patients with refractory H. pylori infection may be at a higher risk of developing gastric cancer than other H. pylori-positive patients.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Microbiota , Sistemas de Secreción Bacterianos , Mucosa Gástrica/microbiología , Glutatión , Infecciones por Helicobacter/microbiología , Helicobacter pylori/genética , Humanos , Lactobacillus/genética , Lipopolisacáridos , ARN Ribosómico 16S/genética , Estudios Retrospectivos , Estómago/microbiología , Azufre/uso terapéutico , Terpenos/uso terapéutico , Ubiquinona/uso terapéutico , Vitaminas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA