Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Immunity ; 57(3): 406-408, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38479356

RESUMO

Combined anti-PD-L1+anti-CTLA-4 therapy has shown benefits over anti-PD-L1 monotherapy as a neoadjuvant treatment in head and neck cancer. In this issue of Immunity, Franken et al. report that CD4+ T cell trafficking from lymph nodes to tumors and expansion toward T helper 1 cells are features specific to combination therapy.


Assuntos
Neoplasias de Cabeça e Pescoço , Humanos , Antígeno CTLA-4 , Terapia Combinada , Antígeno B7-H1
3.
Gut Microbes ; 15(2): 2257291, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37747149

RESUMO

Colorectal cancer (CRC) is one of the most commonly diagnosed cancers worldwide. While a close correlation between chronic Helicobacter pylori infection and CRC has been reported, the role of the virome has been overlooked. Here, we infected Apc-mutant mouse models and C57BL/6 mice with H. pylori and conducted a comprehensive metagenomics analysis of H. pylori-induced changes in lower gastrointestinal tract bacterial and viral communities. We observed an expansion of temperate phages in H. pylori infected Apc+/1638N mice at the early stage of carcinogenesis. Some of the temperate phages were predicted to infect bacteria associated with CRC, including Enterococcus faecalis. We also observed a high prevalence of virulent genes, such as flgJ, cwlJ, and sleB, encoded by temperate phages. In addition, we identified phages associated with pre-onset and onset of H. pylori-promoted carcinogenesis. Through co-occurrence network analysis, we found strong associations between the viral and bacterial communities in infected mice before the onset of carcinogenesis. These findings suggest that the expansion of temperate phages, possibly caused by prophage induction triggered by H. pylori infection, may have contributed to the development of CRC in mice by interacting with the bacterial community.


Assuntos
Bacteriófagos , Neoplasias Colorretais , Microbioma Gastrointestinal , Infecções por Helicobacter , Helicobacter pylori , Animais , Camundongos , Bacteriófagos/genética , Viroma , Infecções por Helicobacter/microbiologia , Camundongos Endogâmicos C57BL , Neoplasias Colorretais/microbiologia , Carcinogênese
4.
Gut ; 72(7): 1258-1270, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37015754

RESUMO

OBJECTIVE: Helicobacter pylori infection is the most prevalent bacterial infection worldwide. Besides being the most important risk factor for gastric cancer development, epidemiological data show that infected individuals harbour a nearly twofold increased risk to develop colorectal cancer (CRC). However, a direct causal and functional connection between H. pylori infection and colon cancer is lacking. DESIGN: We infected two Apc-mutant mouse models and C57BL/6 mice with H. pylori and conducted a comprehensive analysis of H. pylori-induced changes in intestinal immune responses and epithelial signatures via flow cytometry, chip cytometry, immunohistochemistry and single cell RNA sequencing. Microbial signatures were characterised and evaluated in germ-free mice and via stool transfer experiments. RESULTS: H. pylori infection accelerated tumour development in Apc-mutant mice. We identified a unique H. pylori-driven immune alteration signature characterised by a reduction in regulatory T cells and pro-inflammatory T cells. Furthermore, in the intestinal and colonic epithelium, H. pylori induced pro-carcinogenic STAT3 signalling and a loss of goblet cells, changes that have been shown to contribute-in combination with pro-inflammatory and mucus degrading microbial signatures-to tumour development. Similar immune and epithelial alterations were found in human colon biopsies from H. pylori-infected patients. Housing of Apc-mutant mice under germ-free conditions ameliorated, and early antibiotic eradication of H. pylori infection normalised the tumour incidence to the level of uninfected controls. CONCLUSIONS: Our studies provide evidence that H. pylori infection is a strong causal promoter of colorectal carcinogenesis. Therefore, implementation of H. pylori status into preventive measures of CRC should be considered.


Assuntos
Neoplasias do Colo , Infecções por Helicobacter , Helicobacter pylori , Microbiota , Neoplasias Gástricas , Humanos , Camundongos , Animais , Helicobacter pylori/genética , Infecções por Helicobacter/complicações , Infecções por Helicobacter/microbiologia , Camundongos Endogâmicos C57BL , Carcinogênese/patologia , Neoplasias Gástricas/patologia , Neoplasias do Colo/patologia , Muco , Mucosa Gástrica/patologia
5.
Gastroenterology ; 164(4): 550-566, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36587707

RESUMO

BACKGROUND & AIMS: Infection with Helicobacter pylori strongly affects global health by causing chronic gastritis, ulcer disease, and gastric cancer. Although extensive research into the strong immune response against this persistently colonizing bacterium exists, the specific role of CD8+ T cells remains elusive. METHODS: We comprehensively characterize gastric H pylori-specific CD8+ T-cell responses in mice and humans by flow cytometry, RNA-sequencing, immunohistochemistry, and ChipCytometry, applying functional analyses including T-cell depletion, H pylori eradication, and ex vivo restimulation. RESULTS: We define CD8+ T-cell populations bearing a tissue-resident memory (TRM) phenotype, which infiltrate the gastric mucosa shortly after infection and mediate pathogen control by executing antigen-specific effector properties. These induced CD8+ tissue-resident memory T cells (TRM cells) show a skewed T-cell receptor beta chain usage and are mostly specific for cytotoxin-associated gene A, the distinctive oncoprotein injected by H pylori into host cells. As the infection progresses, we observe a loss of the TRM phenotype and replacement of CD8+ by CD4+ T cells, indicating a shift in the immune response during the chronic infection phase. CONCLUSIONS: Our results point toward a hitherto unknown role of CD8+ T-cell response in this bacterial infection, which may have important clinical implications for treatment and vaccination strategies against H pylori.


Assuntos
Infecções por Helicobacter , Helicobacter pylori , Humanos , Animais , Camundongos , Linfócitos T CD8-Positivos , Linfócitos T CD4-Positivos , Estômago , Mucosa Gástrica/microbiologia , Infecções por Helicobacter/microbiologia , Antígenos de Bactérias , Proteínas de Bactérias
6.
Helicobacter ; 27(3): e12888, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35363924

RESUMO

BACKGROUND: Murine Helicobacter species have gained increasing awareness in mouse facilities over the last years. Infections with Helicobacter species may have an impact effect on the health of mice and might pose a zoonotic risk to researchers. To minimize the interference with experiments and hence contribute to the 3Rs, a reliable method of monitoring Helicobacter infections in animal facilities needs to be available. The aim of this study was to improve and validate the detection of the most common murine Helicobacter species. MATERIAL AND METHODS: A multiplex PCR assay was developed for identification of Helicobacter hepaticus, H. bilis, H. muridarum, H. rodentium, and H. typhlonius that could simultaneously detect these five strains in fecal samples. To ensure the quality of the results, the method was validated based on recommendations for in-house developed tests. RESULTS: The method established was highly sensitive and specific. All five strains were detectable with a detection limit of 102 bacteria. Eight different mouse facilities were tested with the validated assay, and the following prevalence were found: H. rodentium 57%, H. hepaticus 46%, H. typhlonius 17%, H. bilis 12%, and H. muridarum 0%. CONCLUSION: The multiplex PCR is a reliable, economic, and time-saving diagnostic tool for routine health monitoring. Further prevalence studies are needed to confirm the high prevalence and hence importance of H. rodentium, as until now this agent is not yet listed in FELASA recommendations.


Assuntos
Infecções por Helicobacter , Helicobacter pylori , Helicobacter , Animais , Fezes/microbiologia , Helicobacter/genética , Infecções por Helicobacter/diagnóstico , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/veterinária , Humanos , Camundongos , Reação em Cadeia da Polimerase Multiplex
7.
Microorganisms ; 9(8)2021 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-34442827

RESUMO

The gastric pathogen Helicobacter pylori infects half of the world's population and is a major risk factor for gastric cancer development. In order to attach to human gastric epithelial cells and inject the oncoprotein CagA into host cells, H. pylori utilizes the outer membrane protein HopQ that binds to the cell surface protein CEACAM, which can be expressed on the gastric mucosa. Once bound, H. pylori activates a number of signaling pathways, including canonical and non-canonical NF-κB. We investigated whether HopQ-CEACAM interaction is involved in activating the non-canonical NF-κB signaling pathway. Different gastric cancer cells were infected with the H. pylori wild type, or HopQ mutant strains, and the activation of non-canonical NF-κB was related to CEACAM expression levels. The correlation between CEACAM levels and the activation of non-canonical NF-κB was confirmed in human gastric tissue samples. Taken together, our findings show that the HopQ-CEACAM interaction is important for activation of the non-canonical NF-κB pathway in gastric epithelial cells.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...