Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
1.
Gut ; 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39251326

RESUMEN

BACKGROUND: Tumourigenesis in right-sided and left-sided colons demonstrated distinct features. OBJECTIVE: We aimed to characterise the differences between the left-sided and right-sided adenomas (ADs) representing the early stage of colonic tumourigenesis. DESIGN: Single-cell and spatial transcriptomic datasets were analysed to reveal alterations between right-sided and left-sided colon ADs. Cells, animal experiments and clinical specimens were used to verify the results. RESULTS: Single-cell analysis revealed that in right-sided ADs, there was a significant reduction of goblet cells, and these goblet cells were dysfunctional with attenuated mucin biosynthesis and defective antigen presentation. An impairment of the mucus barrier led to biofilm formation in crypts and subsequent bacteria invasion into right-sided ADs. The regions spatially surrounding the crypts with biofilm occupation underwent an inflammatory response by lipopolysaccharide (LPS) and an apoptosis process, as revealed by spatial transcriptomics. A distinct S100A11+ epithelial cell population in the right-sided ADs was identified, and its expression level was induced by bacterial LPS and peptidoglycan. S100A11 expression facilitated tumour growth in syngeneic immunocompetent mice with increased myeloid-derived suppressor cells (MDSC) but reduced cytotoxic CD8+ T cells. Targeting S100A11 with well-tolerated antagonists of its receptor for advanced glycation end product (RAGE) (Azeliragon) significantly impaired tumour growth and MDSC infiltration, thereby boosting the efficacy of anti-programmed cell death protein 1 therapy in colon cancer. CONCLUSION: Our findings unravelled that dysfunctional goblet cells and consequential bacterial translocation activated the S100A11-RAGE axis in right-sided colon ADs, which recruits MDSCs to promote immune evasion. Targeting this axis by Azeliragon improves the efficacy of immunotherapy in colon cancer.

3.
Gut ; 73(8): 1292-1301, 2024 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-38839272

RESUMEN

OBJECTIVE: There is a strong clinical association between IBD and primary sclerosing cholangitis (PSC), a chronic disease of the liver characterised by biliary inflammation that leads to strictures and fibrosis. Approximately 60%-80% of people with PSC will also develop IBD (PSC-IBD). One hypothesis explaining this association would be that PSC drives IBD. Therefore, our aim was to test this hypothesis and to decipher the underlying mechanism. DESIGN: Colitis severity was analysed in experimental mouse models of colitis and sclerosing cholangitis, and people with IBD and PSC-IBD. Foxp3+ Treg-cell infiltration was assessed by qPCR and flow cytometry. Microbiota profiling was carried out from faecal samples of people with IBD, PSC-IBD and mouse models recapitulating these diseases. Faecal microbiota samples collected from people with IBD and PSC-IBD were transplanted into germ-free mice followed by colitis induction. RESULTS: We show that sclerosing cholangitis attenuated IBD in mouse models. Mechanistically, sclerosing cholangitis causes an altered intestinal microbiota composition, which promotes Foxp3+ Treg-cell expansion, and thereby protects against IBD. Accordingly, sclerosing cholangitis promotes IBD in the absence of Foxp3+ Treg cells. Furthermore, people with PSC-IBD have an increased Foxp3+ expression in the colon and an overall milder IBD severity. Finally, by transplanting faecal microbiota into gnotobiotic mice, we showed that the intestinal microbiota of people with PSC protects against colitis. CONCLUSION: This study shows that PSC attenuates IBD and provides a comprehensive insight into the mechanisms involved in this effect.


Asunto(s)
Colangitis Esclerosante , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Linfocitos T Reguladores , Colangitis Esclerosante/inmunología , Colangitis Esclerosante/complicaciones , Colangitis Esclerosante/microbiología , Animales , Ratones , Linfocitos T Reguladores/inmunología , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/inmunología , Humanos , Factores de Transcripción Forkhead/metabolismo , Colitis/microbiología , Colitis/complicaciones , Masculino , Trasplante de Microbiota Fecal , Femenino , Heces/microbiología , Ratones Endogámicos C57BL
4.
J Hosp Infect ; 148: 189-219, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38609760

RESUMEN

The first British Society of Gastroenterology (BSG) and Healthcare Infection Society (HIS)-endorsed faecal microbiota transplant (FMT) guidelines were published in 2018. Over the past 5 years, there has been considerable growth in the evidence base (including publication of outcomes from large national FMT registries), necessitating an updated critical review of the literature and a second edition of the BSG/HIS FMT guidelines. These have been produced in accordance with National Institute for Health and Care Excellence-accredited methodology, thus have particular relevance for UK-based clinicians, but are intended to be of pertinence internationally. This second edition of the guidelines have been divided into recommendations, good practice points and recommendations against certain practices. With respect to FMT for Clostridioides difficile infection (CDI), key focus areas centred around timing of administration, increasing clinical experience of encapsulated FMT preparations and optimising donor screening. The latter topic is of particular relevance given the COVID-19 pandemic, and cases of patient morbidity and mortality resulting from FMT-related pathogen transmission. The guidelines also considered emergent literature on the use of FMT in non-CDI settings (including both gastrointestinal and non-gastrointestinal indications), reviewing relevant randomised controlled trials. Recommendations are provided regarding special areas (including compassionate FMT use), and considerations regarding the evolving landscape of FMT and microbiome therapeutics.


Asunto(s)
Infecciones por Clostridium , Trasplante de Microbiota Fecal , Trasplante de Microbiota Fecal/métodos , Humanos , Infecciones por Clostridium/terapia , Reino Unido , Clostridioides difficile , COVID-19/terapia , Recurrencia , Gastroenterología/normas , Gastroenterología/métodos , SARS-CoV-2 , Sociedades Médicas
5.
Gut ; 73(7): 1052-1075, 2024 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-38609165

RESUMEN

The first British Society of Gastroenterology (BSG) and Healthcare Infection Society (HIS)-endorsed faecal microbiota transplant (FMT) guidelines were published in 2018. Over the past 5 years, there has been considerable growth in the evidence base (including publication of outcomes from large national FMT registries), necessitating an updated critical review of the literature and a second edition of the BSG/HIS FMT guidelines. These have been produced in accordance with National Institute for Health and Care Excellence-accredited methodology, thus have particular relevance for UK-based clinicians, but are intended to be of pertinence internationally. This second edition of the guidelines have been divided into recommendations, good practice points and recommendations against certain practices. With respect to FMT for Clostridioides difficile infection (CDI), key focus areas centred around timing of administration, increasing clinical experience of encapsulated FMT preparations and optimising donor screening. The latter topic is of particular relevance given the COVID-19 pandemic, and cases of patient morbidity and mortality resulting from FMT-related pathogen transmission. The guidelines also considered emergent literature on the use of FMT in non-CDI settings (including both gastrointestinal and non-gastrointestinal indications), reviewing relevant randomised controlled trials. Recommendations are provided regarding special areas (including compassionate FMT use), and considerations regarding the evolving landscape of FMT and microbiome therapeutics.


Asunto(s)
Infecciones por Clostridium , Trasplante de Microbiota Fecal , Gastroenterología , Trasplante de Microbiota Fecal/métodos , Humanos , Infecciones por Clostridium/terapia , Gastroenterología/normas , COVID-19/terapia , SARS-CoV-2 , Recurrencia , Clostridioides difficile , Reino Unido , Sociedades Médicas
6.
Gut ; 73(9): 1478-1488, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-38599786

RESUMEN

OBJECTIVE: Probiotic Lactococcus lactis is known to confer health benefits to humans. Here, we aimed to investigate the role of L. lactis in colorectal cancer (CRC). DESIGN: L. lactis abundance was evaluated in patients with CRC (n=489) and healthy individuals (n=536). L. lactis was isolated from healthy human stools with verification by whole genome sequencing. The effect of L. lactis on CRC tumourigenesis was assessed in transgenic Apc Min/+ mice and carcinogen-induced CRC mice. Faecal microbiota was profiled by metagenomic sequencing. Candidate proteins were characterised by nano liquid chromatography-mass spectrometry. Biological function of L. lactis conditioned medium (HkyuLL 10-CM) and functional protein was studied in human CRC cells, patient-derived organoids and xenograft mice. RESULTS: Faecal L. lactis was depleted in patients with CRC. A new L. lactis strain was isolated from human stools and nomenclated as HkyuLL 10. HkyuLL 10 supplementation suppressed CRC tumourigenesis in Apc Min/+ mice, and this tumour-suppressing effect was confirmed in mice with carcinogen-induced CRC. Microbiota profiling revealed probiotic enrichment including Lactobacillus johnsonii in HkyuLL 10-treated mice. HkyuLL 10-CM significantly abrogated the growth of human CRC cells and patient-derived organoids. Such protective effect was attributed to HkyuLL 10-secreted proteins, and we identified that α-mannosidase was the functional protein. The antitumourigenic effect of α-mannosidase was demonstrated in human CRC cells and organoids, and its supplementation significantly reduced tumour growth in xenograft mice. CONCLUSION: HkyuLL 10 suppresses CRC tumourigenesis in mice through restoring gut microbiota and secreting functional protein α-mannosidase. HkyuLL 10 administration may serve as a prophylactic measure against CRC.


Asunto(s)
Carcinogénesis , Neoplasias Colorrectales , Heces , Microbioma Gastrointestinal , Lactococcus lactis , Probióticos , alfa-Manosidasa , Animales , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/prevención & control , Microbioma Gastrointestinal/fisiología , Humanos , Ratones , Probióticos/uso terapéutico , Heces/microbiología , alfa-Manosidasa/metabolismo , Ratones Transgénicos , Femenino , Masculino
7.
Gut ; 73(5): 751-769, 2024 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-38331563

RESUMEN

OBJECTIVE: Chronic obstructive pulmonary disease (COPD) is a major cause of global illness and death, most commonly caused by cigarette smoke. The mechanisms of pathogenesis remain poorly understood, limiting the development of effective therapies. The gastrointestinal microbiome has been implicated in chronic lung diseases via the gut-lung axis, but its role is unclear. DESIGN: Using an in vivo mouse model of cigarette smoke (CS)-induced COPD and faecal microbial transfer (FMT), we characterised the faecal microbiota using metagenomics, proteomics and metabolomics. Findings were correlated with airway and systemic inflammation, lung and gut histopathology and lung function. Complex carbohydrates were assessed in mice using a high resistant starch diet, and in 16 patients with COPD using a randomised, double-blind, placebo-controlled pilot study of inulin supplementation. RESULTS: FMT alleviated hallmark features of COPD (inflammation, alveolar destruction, impaired lung function), gastrointestinal pathology and systemic immune changes. Protective effects were additive to smoking cessation, and transfer of CS-associated microbiota after antibiotic-induced microbiome depletion was sufficient to increase lung inflammation while suppressing colonic immunity in the absence of CS exposure. Disease features correlated with the relative abundance of Muribaculaceae, Desulfovibrionaceae and Lachnospiraceae family members. Proteomics and metabolomics identified downregulation of glucose and starch metabolism in CS-associated microbiota, and supplementation of mice or human patients with complex carbohydrates improved disease outcomes. CONCLUSION: The gut microbiome contributes to COPD pathogenesis and can be targeted therapeutically.


Asunto(s)
Neumonía , Enfermedad Pulmonar Obstructiva Crónica , Humanos , Ratones , Animales , Enfermedad Pulmonar Obstructiva Crónica/etiología , Pulmón/metabolismo , Pulmón/patología , Neumonía/etiología , Inflamación/metabolismo , Carbohidratos/farmacología
10.
Gut ; 73(1): 186-202, 2023 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-37734912

RESUMEN

Smart capsules are developing at a tremendous pace with a promise to become effective clinical tools for the diagnosis and monitoring of gut health. This field emerged in the early 2000s with a successful translation of an endoscopic capsule from laboratory prototype to a commercially viable clinical device. Recently, this field has accelerated and expanded into various domains beyond imaging, including the measurement of gut physiological parameters such as temperature, pH, pressure and gas sensing, and the development of sampling devices for better insight into gut health. In this review, the status of smart capsules for sensing gut parameters is presented to provide a broad picture of these state-of-the-art devices while focusing on the technical and clinical challenges the devices need to overcome to realise their value in clinical settings. Smart capsules are developed to perform sensing operations throughout the length of the gut to better understand the body's response under various conditions. Furthermore, the prospects of such sensing devices are discussed that might help readers, especially health practitioners, to adapt to this inevitable transformation in healthcare. As a compliment to gut sensing smart capsules, significant amount of effort has been put into the development of robotic capsules to collect tissue biopsy and gut microbiota samples to perform in-depth analysis after capsule retrieval which will be a game changer for gut health diagnosis, and this advancement is also covered in this review. The expansion of smart capsules to robotic capsules for gut microbiota collection has opened new avenues for research with a great promise to revolutionise human health diagnosis, monitoring and intervention.


Asunto(s)
Biopsia , Tracto Gastrointestinal , Robótica , Humanos , Endoscopía Capsular , Microbioma Gastrointestinal
11.
Gut ; 72(11): 2112-2122, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37491158

RESUMEN

OBJECTIVE: Roseburia intestinalis is a probiotic species that can suppress intestinal inflammation by producing metabolites. We aimed to study the role of R. intestinalis in colorectal tumourigenesis and immunotherapy. DESIGN: R. intestinalis abundance was evaluated in stools of patients with colorectal cancer (CRC) (n=444) and healthy controls (n=575). The effects of R. intestinalis were studied in ApcMin/+ or azoxymethane (AOM)-induced CRC mouse models, and in syngeneic mouse xenograft models of CT26 (microsatellite instability (MSI)-low) or MC38 (MSI-high). The change of immune landscape was evaluated by multicolour flow cytometry and immunohistochemistry staining. Metabolites were profiled by metabolomic profiling. RESULTS: R. intestinalis was significantly depleted in stools of patients with CRC compared with healthy controls. R. intestinalis administration significantly inhibited tumour formation in ApcMin/+ mice, which was confirmed in mice with AOM-induced CRC. R. intestinalis restored gut barrier function as indicated by improved intestinal permeability and enhanced expression of tight junction proteins. Butyrate was identified as the functional metabolite generated by R. intestinalis. R. intestinalis or butyrate suppressed tumour growth by inducing cytotoxic granzyme B+, interferon (IFN)-γ+ and tumour necrosis factor (TNF)-α+ CD8+ T cells in orthotopic mouse models of MC38 or CT26. R. intestinalis or butyrate also significantly improved antiprogrammed cell death protein 1 (anti-PD-1) efficacy in mice bearing MSI-low CT26 tumours. Mechanistically, butyrate directly bound to toll-like receptor 5 (TLR5) receptor on CD8+ T cells to induce its activity through activating nuclear factor kappa B (NF-κB) signalling. CONCLUSION: R. intestinalis protects against colorectal tumourigenesis by producing butyrate, which could also improve anti-PD-1 efficacy by inducing functional CD8+ T cells. R. intestinalis is a potential adjuvant to augment anti-PD-1 efficacy against CRC.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias Colorrectales , Humanos , Ratones , Animales , Butiratos/farmacología , Carcinogénesis , Transformación Celular Neoplásica , Neoplasias Colorrectales/metabolismo
12.
Animals (Basel) ; 13(13)2023 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-37443970

RESUMEN

The aim of the present study was to investigate the effects of Bacillus licheniformis (BL) on the growth performance, antioxidant capacity, ileal morphology, intestinal fecal short-chain fatty acids, and microflora of weaned piglets challenged with lipopolysaccharide (LPS). Piglets were assigned into three groups: basal diet (Con), a basal diet with added 109 CFU B. licheniformis/kg (BLl), and a basal diet with added 1010 CFU B. licheniformis/kg (BLh). On day 28, BLh piglets were intraperitoneally injected with LPS (CBL) and sterilized saline water (BL), Con piglets were injected with LPS (LPS) and sterilized saline water (Con), with the injections being administered for three consecutive days. The average daily gain significantly increased from day 1 to day 28 and the feed: gain ratio decreased with BL supplementation compared with the Con group. Supplementation with BLl and BLh reduced the diarrhea rate in piglets. Serum catalase activity increased and malondialdehyde concentration decreased in the CBL treatment group compared with the LPS treatment group. Both BL and CBL treatments increased the ileal villus length/crypt depth ratio compared with Con and LPS treatments. BL administration significantly increased colonic propionic and isobutyric acid concentrations compared with Con treatment. Both BL and CBL piglets had significantly increased fecal acetic, propionic, and butyric acid levels compared with LPS piglets. Analysis of the colonic microbial metagenome showed that Prevotella species were the predominant bacteria in piglets treated with BL and CBL. The CBL-treated piglets had higher scores for lysine biosynthesis, arginine biosynthesis, sulfur relay system, and histidine metabolism. BL-treated piglets had higher scores for glycosaminoglycan biosynthesis-keratan sulfate, oxidative phosphorylation, and pyruvate and carbon metabolism.

17.
Gut ; 72(7): 1258-1270, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37015754

RESUMEN

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.


Asunto(s)
Neoplasias del Colon , Infecciones por Helicobacter , Helicobacter pylori , Microbiota , Neoplasias Gástricas , Humanos , Ratones , Animales , Helicobacter pylori/genética , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/microbiología , Ratones Endogámicos C57BL , Carcinogénesis/patología , Neoplasias Gástricas/patología , Neoplasias del Colon/patología , Moco , Mucosa Gástrica/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA