Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Theranostics ; 14(7): 2719-2735, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38773969

RESUMEN

Aim: To elucidate dynamics and functions in colonic macrophage subsets, and their regulation by Bifidobacterium breve (B. breve) and its associated metabolites in the initiation of colitis-associated colorectal cancer (CAC). Methods: Azoxymethane (AOM) and dextran sodium sulfate (DSS) were used to create a CAC model. The tumor-suppressive effect of B. breve and variations of macrophage subsets were evaluated. Intestinal macrophages were ablated to determine their role in the protective effects of B. breve. Efficacious molecules produced by B. breve were identified by non-targeted and targeted liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis. The molecular mechanism was further verified in murine bone marrow-derived macrophages (BMDMs), macrophages derived from human peripheral blood mononuclear cells (hPBMCs), and demonstrated in CAC mice. Results: B. breve alleviated colitis symptoms, delayed colonic tumorigenesis, and promoted phenotypic differentiation of immature inflammatory macrophages into mature homeostatic macrophages. On the contrary, the ablation of intestinal macrophages largely annulled the protective effects of B. breve. Microbial analysis of colonic contents revealed the enrichment of probiotics and the depletion of potential pathogens following B. breve supplementation. Moreover, indole-3-lactic acid (ILA) was positively correlated with B. breve in CAC mice and highly enriched in the culture supernatant of B. breve. Also, the addition of ILA directly promoted AKT phosphorylation and restricted the pro-inflammatory response of murine BMDMs and macrophages derived from hPBMCs in vitro. The effects of ILA in murine BMDMs and macrophages derived from hPBMCs were abolished by the aryl hydrocarbon receptor (AhR) antagonist CH-223191 or the AKT inhibitor MK-2206. Furthermore, ILA could protect against tumorigenesis by regulating macrophage differentiation in CAC mice; the AhR antagonist largely abrogated the effects of B. breve and ILA in relieving colitis and tumorigenesis. Conclusion: B. breve-mediated tryptophan metabolism ameliorates the precancerous inflammatory intestinal milieu to inhibit tumorigenesis by directing the differentiation of immature colonic macrophages.


Asunto(s)
Bifidobacterium breve , Diferenciación Celular , Colitis , Indoles , Macrófagos , Probióticos , Animales , Ratones , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Bifidobacterium breve/metabolismo , Indoles/farmacología , Indoles/metabolismo , Humanos , Colitis/inducido químicamente , Colitis/microbiología , Colitis/complicaciones , Diferenciación Celular/efectos de los fármacos , Probióticos/farmacología , Probióticos/administración & dosificación , Modelos Animales de Enfermedad , Carcinogénesis/efectos de los fármacos , Neoplasias Asociadas a Colitis/patología , Neoplasias Asociadas a Colitis/microbiología , Neoplasias Asociadas a Colitis/metabolismo , Ratones Endogámicos C57BL , Colon/microbiología , Colon/patología , Colon/metabolismo , Sulfato de Dextran , Masculino , Microbioma Gastrointestinal , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/microbiología , Azoximetano
2.
Cell Mol Gastroenterol Hepatol ; 18(1): 105-131, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38614455

RESUMEN

BACKGROUND & AIMS: Inflammatory bowel disease is associated with carcinogenesis, which limits the prognosis of the patients. The local expression of proteinases and proteinase-activated receptor 1 (PAR1) increases in inflammatory bowel disease. The present study investigated the therapeutic effects of PAR1 antagonism on colitis-associated carcinogenesis. METHODS: A colitis-associated carcinogenesis model was prepared in mice by treatment with azoxymethane (AOM) and dextran sulfate sodium (DSS). PAR1 antagonist E5555 was administered in long- and short-term protocol, starting on the day of AOM injection and 1 week after completing AOM/DSS treatment, respectively. The fecal samples were collected for metagenome analysis of gut microbiota. The intestinal myofibroblasts of the Crohn's disease patients were used to elucidate underlying cellular mechanisms. Caco-2 cells were used to investigate a possible source of PAR1 agonist proteinases. RESULTS: AOM/DSS model showed weight loss, diarrhea, tumor development, inflammation, fibrosis, and increased production of inflammatory cytokines. The ß-diversity, but not α-diversity, of microbiota significantly differed between AOM/DSS and control mice. E5555 alleviated these pathological changes and altered the microbiota ß-diversity in AOM/DSS mice. The thrombin expression was up-regulated in tumor and non-tumor areas, whereas PAR1 mRNA expression was higher in tumor areas compared with non-tumor areas. E5555 inhibited thrombin-triggered elevation of cytosolic Ca2+ concentration and ERK1/2 phosphorylation, as well as IL6-induced signal transducer and activator of transcription 3 (STAT3) phosphorylation in intestinal myofibroblasts. Caco-2 cell-conditioned medium contained immunoreactive thrombin, which cleaved the recombinant protein containing the extracellular domain of PAR1 at the thrombin cleavage site. CONCLUSIONS: PAR1 antagonism is proposed to be a novel therapeutic strategy for treatment of inflammatory bowel disease and its associated carcinogenesis.


Asunto(s)
Azoximetano , Sulfato de Dextran , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Receptor PAR-1 , Animales , Receptor PAR-1/metabolismo , Receptor PAR-1/antagonistas & inhibidores , Humanos , Ratones , Células CACO-2 , Sulfato de Dextran/toxicidad , Azoximetano/toxicidad , Microbioma Gastrointestinal/efectos de los fármacos , Masculino , Colitis/complicaciones , Colitis/inducido químicamente , Colitis/patología , Colitis/tratamiento farmacológico , Carcinogénesis/efectos de los fármacos , Carcinogénesis/patología , Factor de Transcripción STAT3/metabolismo , Miofibroblastos/metabolismo , Miofibroblastos/patología , Miofibroblastos/efectos de los fármacos , Neoplasias Asociadas a Colitis/patología , Neoplasias Asociadas a Colitis/microbiología , Neoplasias Asociadas a Colitis/tratamiento farmacológico , Neoplasias Asociadas a Colitis/inmunología , Trombina/metabolismo , Ratones Endogámicos C57BL , Enfermedad de Crohn/patología , Enfermedad de Crohn/tratamiento farmacológico , Enfermedad de Crohn/microbiología , Enfermedad de Crohn/inducido químicamente
3.
Sci Rep ; 12(1): 381, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013389

RESUMEN

GPR120 (encoded by FFAR4 gene) is a receptor for long chain fatty acids, activated by ω-3 Polyunsaturated Fatty Acids (PUFAs), and expressed in many cell types. Its role in the context of colorectal cancer (CRC) is still puzzling with many controversial evidences. Here, we explored the involvement of epithelial GPR120 in the CRC development. Both in vitro and in vivo experiments were conducted to mimic the conditional deletion of the receptor from gut epithelium. Intestinal permeability and integrity of mucus layer were assessed by using Evans blue dye and immunofluorescence for MUC-2 protein, respectively. Microbiota composition, presence of lipid mediators and short chain fatty acids were analyzed in the stools of conditional GPR120 and wild type (WT) mice. Incidence and grade of tumors were evaluated in all groups of mice before and after colitis-associated cancer. Finally, GPR120 expression was analyzed in 9 human normal tissues, 9 adenomas, and 17 primary adenocarcinomas. Our work for the first time highlights the role of the receptor in the progression of colorectal cancer. We observed that the loss of epithelial GPR120 in the gut results into increased intestinal permeability, microbiota translocation and dysbiosis, which turns into hyperproliferation of epithelial cells, likely through the activation of ß -catenin signaling. Therefore, the loss of GPR120 represents an early event of CRC, but avoid its progression as invasive cancer. these results demonstrate that the epithelial GPR120 receptor is essential to maintain the mucosal barrier integrity and to prevent CRC developing. Therefore, our data pave the way to GPR120 as an useful marker for the phenotypic characterization of CRC lesions and as new potential target for CRC prevention.


Asunto(s)
Adenocarcinoma/metabolismo , Neoplasias Asociadas a Colitis/metabolismo , Colon/metabolismo , Mucosa Intestinal/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/microbiología , Adenocarcinoma/patología , Animales , Traslocación Bacteriana , Proliferación Celular , Neoplasias Asociadas a Colitis/genética , Neoplasias Asociadas a Colitis/microbiología , Neoplasias Asociadas a Colitis/patología , Colon/microbiología , Colon/patología , Progresión de la Enfermedad , Disbiosis , Microbioma Gastrointestinal , Humanos , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Ratones Noqueados , Permeabilidad , Receptores Acoplados a Proteínas G/genética , Carga Tumoral
4.
Int J Mol Sci ; 22(21)2021 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-34768795

RESUMEN

Ulcerative colitis (UC) is a chronic autoimmune disorder affecting the colonic mucosa. UC is a subtype of inflammatory bowel disease along with Crohn's disease and presents with varying extraintestinal manifestations. No single etiology for UC has been found, but a combination of genetic and environmental factors is suspected. Research has focused on the role of intestinal dysbiosis in the pathogenesis of UC, including the effects of dysbiosis on the integrity of the colonic mucosal barrier, priming and regulation of the host immune system, chronic inflammation, and progression to tumorigenesis. Characterization of key microbial taxa and their implications in the pathogenesis of UC and colitis-associated cancer (CAC) may present opportunities for modulating intestinal inflammation through microbial-targeted therapies. In this review, we discuss the microbiota-immune crosstalk in UC and CAC, as well as the evolution of microbiota-based therapies.


Asunto(s)
Colitis Ulcerosa/microbiología , Neoplasias Asociadas a Colitis/microbiología , Microbiota , Animales , Colitis Ulcerosa/terapia , Neoplasias Asociadas a Colitis/terapia , Interacciones Huésped-Patógeno , Humanos , Probióticos
5.
Front Immunol ; 12: 669747, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34025672

RESUMEN

Patients suffering from ulcerative colitis are at increased risk of developing colorectal cancer. Although the exact underlying mechanisms of inflammation-associated carcinogenesis remain unknown, the intestinal microbiota as well as pathogenic bacteria are discussed as contributors to inflammation and colitis-associated colon cancer (CAC). In the present study, we analyzed the impact of TLR4, the receptor for Gram-negative bacteria derived lipopolysaccharides, on intestinal inflammation and tumorigenesis in a murine model of CAC. During the inflammatory phases of CAC development, we observed a strong upregulation of Tlr4 expression in colonic tissues. Blocking of TLR4 signaling by a small-molecule-specific inhibitor during the inflammatory phases of CAC strongly diminished the development and progression of colonic tumors, which was accompanied by decreased numbers of infiltrating macrophages and reduced colonic pro-inflammatory cytokine levels compared to CAC control mice. Interestingly, inhibiting bacterial signaling by antibiotic treatment during the inflammatory phases of CAC also protected mice from severe intestinal inflammation and almost completely prevented tumor growth. Nevertheless, application of antibiotics involved rapid and severe body weight loss and might have unwanted side effects. Our results indicate that bacterial activation of TLR4 on innate immune cells in the colon triggers inflammation and promotes tumor growth. Thus, the inhibition of the TLR4 signaling during intestinal inflammation might be a novel approach to impede CAC development.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Neoplasias Asociadas a Colitis/prevención & control , Colitis/tratamiento farmacológico , Colon/efectos de los fármacos , Sulfonamidas/farmacología , Receptor Toll-Like 4/antagonistas & inhibidores , Animales , Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Bacterias/metabolismo , Línea Celular Tumoral , Colitis/metabolismo , Colitis/microbiología , Colitis/patología , Neoplasias Asociadas a Colitis/metabolismo , Neoplasias Asociadas a Colitis/microbiología , Neoplasias Asociadas a Colitis/patología , Colon/metabolismo , Colon/microbiología , Colon/patología , Citocinas/metabolismo , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/metabolismo , Ratones Endogámicos BALB C , Transducción de Señal , Receptor Toll-Like 4/metabolismo , Carga Tumoral/efectos de los fármacos , Macrófagos Asociados a Tumores/efectos de los fármacos , Macrófagos Asociados a Tumores/metabolismo
6.
Gut Microbes ; 13(1): 1-20, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33947304

RESUMEN

Chronic inflammation and gut microbiota dysbiosis are risk factors for colorectal cancer. In clinical practice, patients with inflammatory bowel disease (IBD) have a greatly increased risk of developing colitis-associated colorectal cancer (CAC). However, the underlying mechanism of the initiation of CAC remains unknown. Systematic analyses using an existing genome-wide association study (GWAS) and conditional deletion of Zfp90 (encoding zinc finger protein 90 homolog) in a CAC mouse model indicated that Zfp90 is a putative oncogene in CAC development.Strikingly, depletion of the gut microbiota eliminated the tumorigenic effect of Zfp90 in the CAC mouse model. Moreover, fecal microbiota transplantation demonstrated that Zfp90 promoted CAC dependent on the gut microbiota. Analysis of 16s rDNA sequences in fecal specimens from the CAC mouse model allowed us to speculate that a Prevotella copri-defined microbiota might mediate the oncogenic role of Zfp90 in the development of CAC. Mechanistic studies revealed Zfp90 accelerated CAC development through the TLR4-PI3K-AKT-NF-κB pathway. Our findings revealed the crucial role of the Zfp90-microbiota-NF-κB axis in creating a tumor-promoting environment and suggested therapeutic targets for CAC prevention and treatment.


Asunto(s)
Neoplasias Asociadas a Colitis/metabolismo , Microbioma Gastrointestinal , Proteínas Represoras/metabolismo , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Neoplasias Asociadas a Colitis/genética , Neoplasias Asociadas a Colitis/microbiología , Progresión de la Enfermedad , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Represoras/genética
7.
Biomed Pharmacother ; 135: 111206, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33418307

RESUMEN

The gut microbiota has been implicated in the development of colitis-associated cancer (CAC). We investigated how the gut microbiota affects the development of CAC when the composition of the microbial community is altered by the administration of various antibiotics in a murine model. C57BL/6 mice were given intraperitoneal injection of 12.5 mg/kg azoxymethane (AOM), followed by two rounds of 2.0 % dextran sodium sulfate (DSS) exposure. Antibiotics, including ampicillin, neomycin, metronidazole, and/or vancomycin, were administered 14 days prior to AOM injection until the end of the experiment. High-throughput sequencing of mice feces was conducted to evaluate alterations of the gut microbiota. Tumorigenesis and inflammation were most markedly suppressed in the mice treated with an antibiotic cocktail therapy consisting of ampicillin, neomycin, metronidazole, and vancomycin. Individual antibiotic treatments had different effects on tumorigenesis and inflammation. Metronidazole attenuated both tumorigenesis and inflammation. Neomycin suppressed tumorigenesis but did not alleviate inflammation. Ampicillin and vancomycin did not significantly attenuate either tumorigenesis or inflammation. Antimicrobial therapy differentially altered the diversity and composition of the gut microbiota depending on antibiotic type. The phyla Proteobacteria and Tenericutes were positively correlated with tumor burden. Colon tumorigenesis was attenuated through various antibiotics in the AOM/DSS-induced CAC model. Individual antibiotics differentially altered the gut microbial composition and showed different effects on tumor suppression; however, the degree of tumor suppression was less pronounced than that relative to the antibiotic cocktail therapy, suggesting that the global gut microbial community plays an important role in the development of CAC.


Asunto(s)
Neoplasias Asociadas a Colitis/microbiología , Colitis/microbiología , Colon/microbiología , Neoplasias Colorrectales/microbiología , Microbioma Gastrointestinal , Animales , Antibacterianos/farmacología , Antiinflamatorios/farmacología , Anticarcinógenos/farmacología , Colitis/metabolismo , Colitis/patología , Colitis/prevención & control , Neoplasias Asociadas a Colitis/metabolismo , Neoplasias Asociadas a Colitis/patología , Neoplasias Asociadas a Colitis/prevención & control , Colon/efectos de los fármacos , Colon/metabolismo , Colon/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/prevención & control , Modelos Animales de Enfermedad , Heces/microbiología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Ratones Endogámicos C57BL
8.
Carcinogenesis ; 42(2): 210-219, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-32940665

RESUMEN

Fucoxanthin (Fx), a marine carotenoid found in edible brown algae, is well known for having anticancer properties. The gut microbiota has been demonstrated as a hallmark for colorectal cancer progression in both humans and rodents. However, it remains unclear whether the gut microbiota is associated with the anticancer effect of Fx. We investigated the chemopreventive potency of Fx and its effect on gut microbiota in a mouse model of inflammation-associated colorectal cancer (by azoxymethane/dextran sulfate sodium treatment). Fx administration (30 mg/kg bw) during a 14 week period significantly inhibited the multiplicity of colorectal adenocarcinoma in mice. The number of apoptosis-like cleaved caspase-3high cells increased significantly in both colonic adenocarcinoma and mucosal crypts. Fx administration significantly suppressed Bacteroidlales (f_uc; g_uc) (0.3-fold) and Rikenellaceae (g_uc) (0.6-fold) and increased Lachnospiraceae (g_uc) (2.2-fold), compared with those of control mice. Oral administration of a fecal suspension obtained from Fx-treated mice, aimed to enhance Lachnospiraceae, suppress the number of colorectal adenocarcinomas in azoxymethane/dextran sulfate sodium-treated mice with a successful increase in Lachnospiraceae in the gut. Our findings suggested that an alteration in gut microbiota by dietary Fx might be an essential factor in the cancer chemopreventive effect of Fx in azoxymethane/dextran sulfate sodium-treated mice.


Asunto(s)
Adenocarcinoma/prevención & control , Colitis Ulcerosa/tratamiento farmacológico , Neoplasias Asociadas a Colitis/prevención & control , Microbioma Gastrointestinal/efectos de los fármacos , Xantófilas/administración & dosificación , Adenocarcinoma/inmunología , Adenocarcinoma/microbiología , Adenocarcinoma/patología , Animales , Azoximetano/administración & dosificación , Azoximetano/toxicidad , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/microbiología , Neoplasias Asociadas a Colitis/inmunología , Neoplasias Asociadas a Colitis/microbiología , Neoplasias Asociadas a Colitis/patología , Sulfato de Dextran/administración & dosificación , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Ensayos de Selección de Medicamentos Antitumorales , Heces/microbiología , Microbioma Gastrointestinal/inmunología , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Masculino , Ratones
9.
Hum Exp Toxicol ; 40(4): 622-633, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32924602

RESUMEN

Bacteroides fragilis (BF) plays a critical role in developing and maintaining the mammalian immune system. We previously found that BF colonization could prevent inflammation and tumor formation in a germ-free (GF) colitis-associated colorectal cancer (CAC) mouse model. The role of Toll-like receptor 4 (TLR4) in CAC development has not been clearly elucidated in BF mono-colonized gnotobiotic mice. The wild-type (WT) and TLR4 knockout (T4K) germ-free mice were raised with or without BF colonization for 28 days (GF/WT, GF/T4K, BF/WT, and BF/T4K) and then CAC was induced under azoxymethane (AOM)/dextran sulfate sodium (DSS) administration. The results showed that tumor formation and tumor incidence were significantly inhibited in the BF/WT group compared to those observed in the GF/WT group. However, the tumor prevention effect was not observed in the BF/T4K group unlike in the BF/WT group. Moreover, the CAC histological severity of the BF/WT group was ameliorated, but more severe lesions were found in the GF/WT, GF/T4K, and BF/T4K groups. Immunohistochemistry showed decreased cell proliferation (PCNA, ß-catenin) and inflammatory markers (iNOS) in the BF/WT group compared to those in the BF/T4K group. Taken together, BF mono-colonization of GF mice might prevent CAC via the TLR4 signal pathway.


Asunto(s)
Bacteroides fragilis , Neoplasias Asociadas a Colitis , Colitis , Neoplasias Colorrectales , Receptor Toll-Like 4/genética , Animales , Azoximetano , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/microbiología , Colitis/patología , Neoplasias Asociadas a Colitis/metabolismo , Neoplasias Asociadas a Colitis/microbiología , Neoplasias Asociadas a Colitis/patología , Colon/metabolismo , Colon/microbiología , Colon/patología , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/patología , Ciclooxigenasa 2/metabolismo , Sulfato de Dextran , Modelos Animales de Enfermedad , Vida Libre de Gérmenes , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , beta Catenina/metabolismo
10.
J Cell Physiol ; 236(4): 2239-2254, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32853458

RESUMEN

The gut of mammals is considered as a harmonious ecosystem mediated by intestinal microbiota and the host. Both bacteria and mammalian immune cells show region-related distribution characteristics, and the interaction between the two could be demonstrated by synergetic roles in maintaining intestinal homeostasis and dysregulation in intestinal inflammation. The harmonious interplay between bacteria and host requires fine-tuned regulations by environmental and genetic factors. Thus, the disturbed immune response to microbial components or metabolites and dysbiosis related to immunodeficiency are absolute risk factors to intestinal inflammation and cancer. In this review, we discuss the crosstalk between bacteria and host immunity in the gut and highlight the critical roles of bidirectional regulation between bacteria and the mammalian immune system involved in intestinal inflammation.


Asunto(s)
Bacterias/inmunología , Microbioma Gastrointestinal/inmunología , Inmunidad Mucosa , Enfermedades Inflamatorias del Intestino/inmunología , Mucosa Intestinal/inmunología , Animales , Bacterias/metabolismo , Bacterias/patogenicidad , Neoplasias Asociadas a Colitis/inmunología , Neoplasias Asociadas a Colitis/metabolismo , Neoplasias Asociadas a Colitis/microbiología , Disbiosis , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/microbiología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Ratones , Permeabilidad , Transducción de Señal
11.
Gastroenterology ; 160(4): 1256-1268.e9, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33189701

RESUMEN

BACKGROUND & AIMS: Inflammation in the gastrointestinal tract may lead to the development of cancer. Dicarbonyl electrophiles, such as isolevuglandins (isoLGs), are generated from lipid peroxidation during the inflammatory response and form covalent adducts with amine-containing macromolecules. Thus, we sought to determine the role of dicarbonyl electrophiles in inflammation-associated carcinogenesis. METHODS: The formation of isoLG adducts was analyzed in the gastric tissues of patients infected with Helicobacter pylori from gastritis to precancerous intestinal metaplasia, in human gastric organoids, and in patients with colitis and colitis-associated carcinoma (CAC). The effect on cancer development of a potent scavenger of dicarbonyl electrophiles, 5-ethyl-2-hydroxybenzylamine (EtHOBA), was determined in transgenic FVB/N insulin-gastrin (INS-GAS) mice and Mongolian gerbils as models of H pylori-induced carcinogenesis and in C57BL/6 mice treated with azoxymethane-dextran sulfate sodium as a model of CAC. The effect of EtHOBA on mutations in gastric epithelial cells of H pylori-infected INS-GAS mice was assessed by whole-exome sequencing. RESULTS: We show increased isoLG adducts in gastric epithelial cell nuclei in patients with gastritis and intestinal metaplasia and in human gastric organoids infected with H pylori. EtHOBA inhibited gastric carcinoma in infected INS-GAS mice and gerbils and attenuated isoLG adducts, DNA damage, and somatic mutation frequency. Additionally, isoLG adducts were elevated in tissues from patients with colitis, colitis-associated dysplasia, and CAC as well as in dysplastic tumors of C57BL/6 mice treated with azoxymethane-dextran sulfate sodium. In this model, EtHOBA significantly reduced adduct formation, tumorigenesis, and dysplasia severity. CONCLUSIONS: Dicarbonyl electrophiles represent a link between inflammation and somatic genomic alterations and are thus key targets for cancer chemoprevention.


Asunto(s)
Transformación Celular Neoplásica/inmunología , Neoplasias Asociadas a Colitis/inmunología , Lípidos/inmunología , Lesiones Precancerosas/inmunología , Neoplasias Gástricas/inmunología , Animales , Bencilaminas/farmacología , Bencilaminas/uso terapéutico , Núcleo Celular/metabolismo , Transformación Celular Neoplásica/efectos de los fármacos , Neoplasias Asociadas a Colitis/microbiología , Neoplasias Asociadas a Colitis/patología , Neoplasias Asociadas a Colitis/prevención & control , Modelos Animales de Enfermedad , Células Epiteliales , Mucosa Gástrica/citología , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/inmunología , Mucosa Gástrica/patología , Gastritis/inmunología , Gastritis/microbiología , Gastritis/patología , Gerbillinae , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/patología , Helicobacter pylori/inmunología , Helicobacter pylori/aislamiento & purificación , Humanos , Lípidos/antagonistas & inhibidores , Metaplasia/inmunología , Metaplasia/microbiología , Metaplasia/patología , Ratones , Ratones Transgénicos , Organoides , Lesiones Precancerosas/tratamiento farmacológico , Lesiones Precancerosas/microbiología , Lesiones Precancerosas/patología , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/patología , Neoplasias Gástricas/prevención & control
12.
Gastroenterology ; 160(3): 797-808.e6, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33127391

RESUMEN

BACKGROUND & AIMS: Chronic colonic inflammation leads to dysplasia and cancer in patients with inflammatory bowel disease. We have described the critical role of innate immune signaling via Toll-like receptor 4 (TLR4) in the pathogenesis of dysplasia and cancer. In the current study, we interrogate the intersection of TLR4 signaling, epithelial redox activity, and the microbiota in colitis-associated neoplasia. METHODS: Inflammatory bowel disease and colorectal cancer data sets were analyzed for expression of TLR4, dual oxidase 2 (DUOX2), and NADPH oxidase 1 (NOX1). Epithelial production of hydrogen peroxide (H2O2) was analyzed in murine colonic epithelial cells and colonoid cultures. Colorectal cancer models were carried out in villin-TLR4 mice, carrying a constitutively active form of TLR4, their littermates, and villin-TLR4 mice backcrossed to DUOXA-knockout mice. The role of the TLR4-shaped microbiota in tumor development was tested in wild-type germ-free mice. RESULTS: Activation of epithelial TLR4 was associated with up-regulation of DUOX2 and NOX1 in inflammatory bowel disease and colorectal cancer. DUOX2 was exquisitely dependent on TLR4 signaling and mediated the production of epithelial H2O2. Epithelial H2O2 was significantly increased in villin-TLR4 mice; TLR4-dependent tumorigenesis required the presence of DUOX2 and a microbiota. Mucosa-associated microbiota transferred from villin-TLR4 mice to wild-type germ-free mice caused increased H2O2 production and tumorigenesis. CONCLUSIONS: Increased TLR4 signaling in colitis drives expression of DUOX2 and epithelial production of H2O2. The local milieu imprints the mucosal microbiota and imbues it with pathogenic properties demonstrated by enhanced epithelial reactive oxygen species and increased development of colitis-associated tumors. The inter-relationship between epithelial reactive oxygen species and tumor-promoting microbiota requires a 2-pronged strategy to reduce the risk of dysplasia in colitis patients.


Asunto(s)
Colitis Ulcerosa/complicaciones , Neoplasias Asociadas a Colitis/patología , Oxidasas Duales/metabolismo , Microbioma Gastrointestinal/inmunología , Receptor Toll-Like 4/metabolismo , Animales , Azoximetano/administración & dosificación , Azoximetano/toxicidad , Carcinogénesis/inducido químicamente , Carcinogénesis/inmunología , Carcinogénesis/patología , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/microbiología , Neoplasias Asociadas a Colitis/inmunología , Neoplasias Asociadas a Colitis/microbiología , Colon/efectos de los fármacos , Colon/inmunología , Colon/microbiología , Colon/patología , Conjuntos de Datos como Asunto , Sulfato de Dextran/administración & dosificación , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Vida Libre de Gérmenes , Humanos , Peróxido de Hidrógeno/metabolismo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , NADPH Oxidasa 1/metabolismo , Receptor Toll-Like 4/genética
13.
Int Immunopharmacol ; 88: 106862, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32771947

RESUMEN

We investigate the effects of Clostridium butyricum(CB) on gut microbiota and colitis associated colon cancer(CAC) in mice.6-8 weeks old C57BL/6 mice were randomly divided into control, azoxymethane (AOM) + dextran sodium sulphate (DSS) and AOM + DSS + CB groups. Mice in the latter two groups received an intraperitoneal injection of AOM (12.5 mg/kg), followed by three cycles of DSS diluted in water (2.5% w/v). Mice in treatment group received CB (2 × 108 CFU in 200 ul normal saline) by gavage administration three times one week. Microbiota composition was assessed by 16S rRNA high-throughput sequencing. Colon samples were collected to examine severity of colitis and tumorigenesis. Cytokines including TNF-a, IL-6 and Cyclo-oxygenase-2 (COX-2) were detected by RT-qPCR. Expression of Bcl-2, Bax and the state of components of NF-κB signaling pathway were detected by western blot. The results revealed that CB regulated structure of intestinal flora and changed the microbial composition; decreased Firmicutes/ Bacteroidetes ratio in phylum level and increased the relative abundance of probiotics; decreased colitis, decreased incidence and size of colorectal cancer(CRC) and increased apoptosis of tumor cells; decreased cytokines including TNF-a and IL-6; decreased level of COX-2; decreased phosphorylation of NF-κB; decreased level of Bcl-2 and increased expression of Bax. In conclusion, CB could regulate structure and composition of gut microbiota and reduces colitis associated colon cancer in mice, the mechanism may be inhibiting NF-κB pathway and promoting apoptosis.


Asunto(s)
Clostridium butyricum/fisiología , Neoplasias Asociadas a Colitis/microbiología , Neoplasias Asociadas a Colitis/terapia , Colitis/microbiología , Colitis/terapia , Microbioma Gastrointestinal , Animales , Apoptosis , Azoximetano/toxicidad , Bacteroidetes/metabolismo , Peso Corporal , Proliferación Celular , Colitis/inducido químicamente , Colitis/patología , Neoplasias Asociadas a Colitis/inducido químicamente , Neoplasias Asociadas a Colitis/patología , Citocinas/metabolismo , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Heces/microbiología , Firmicutes/metabolismo , Inflamación/microbiología , Masculino , Ratones Endogámicos C57BL , Subunidad p50 de NF-kappa B/antagonistas & inhibidores , ARN Ribosómico 16S
14.
Gut Microbes ; 12(1): 1785803, 2020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-32663105

RESUMEN

Previously, a synbiotic combination of probiotic Lactobacillus gasseri 505 (LG) and a new prebiotic, Cudrania tricuspidata leaf extract (CT) in fermented milk, designated FCT, showed an in vitro immunomodulatory effect and antioxidant activity. Although synbiotic combination might have cancer-protective effects, these activities have not been fully validated in vivo. Ten-week treatment of LG, CT, or FCT to azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced colitis-associated colorectal cancer (CAC) mouse model reduced both the incidence of colonic tumors and damage to the colonic mucosa effectively, suggesting a cancer-protective effect. To understand these, biomarkers associated with inflammation, colon barrier, apoptosis, and cancer cell proliferation were monitored in AOM/DSS group versus LG/CT/FCT groups. A synbiotic combination (FCT) down-regulated pro-inflammatory cytokines (TNF-α, IFN-γ, IL-1ß, and IL-6) and inflammation-associated enzymes (iNOS and COX-2), and up-regulated anti-inflammatory cytokines (IL-4 and IL-10). In addition, colon barrier experiment revealed that biomarkers of mucus layer (MUC-2 and TFF3) and tight junction (occludin and ZO-1) were up-regulated. Subsequent apoptosis experiment showed that pro-apoptotic factors (p53, p21, and Bax) were up-regulated and anti-apoptotic factors (Bcl-2 and Bcl-xL) were down-regulated. Furthermore, comparative metagenome analysis of gut microbiota revealed that Staphylococcus decreased but Lactobacillus, Bifidobacterium, and Akkermansia increased, supporting their protective effects, accompanied by increased short-chain fatty acids (SCFAs). Taken together, the FCT administration showed cancer-protective effects by reducing the risk of colitis-associated colon cancer via regulation of inflammation, carcinogenesis, and compositional change of gut microbiota. Consequently, the synbiotic combination (FCT) could be a novel potential health-protective natural agent against CAC.


Asunto(s)
Neoplasias Asociadas a Colitis/tratamiento farmacológico , Lactobacillus gasseri/fisiología , Maclura/química , Extractos Vegetales/uso terapéutico , Simbióticos , Animales , Antioxidantes/metabolismo , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Asociadas a Colitis/inmunología , Neoplasias Asociadas a Colitis/microbiología , Neoplasias Asociadas a Colitis/patología , Colon/efectos de los fármacos , Colon/metabolismo , Colon/microbiología , Colon/patología , Productos Lácteos Cultivados/análisis , Productos Lácteos Cultivados/microbiología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ácidos Grasos Volátiles/análisis , Heces/química , Heces/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Inmunomodulación , Inflamación , Ratones , Hojas de la Planta/química , Simbióticos/administración & dosificación , Simbióticos/análisis
15.
United European Gastroenterol J ; 8(6): 675-684, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32268844

RESUMEN

Patients with inflammatory bowel disease are at increased risk of colorectal cancer, which has worse prognosis than sporadic colorectal cancer. Until recently, understanding of pathogenesis in inflammatory bowel disease-associated colorectal cancer was restricted to the demonstration of chromosomic/microsatellite instabilities and aneuploidy. The advance of high-throughput sequencing technologies has highlighted the complexity of the pathobiology and revealed recurrently mutated genes involved in the RTK/RAS, PI3K, WNT, and TGFß pathways, leading to potentially new targetable mutations. Moreover, alterations of mitochondrial DNA and the dysregulation of non-coding sequences have also been described, as well as several epigenetic modifications. Although recent studies have brought new insights into pathobiology and raised the prospect of innovative therapeutic approaches, the understanding of colorectal carcinogenesis in inflammatory bowel disease and how it differs from sporadic colorectal cancer remains not fully elucidated. Further studies are required to better understand the pathogenesis and molecular alterations leading to human inflammatory bowel disease-associated colorectal cancer.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinogénesis/genética , Colitis Ulcerosa/complicaciones , Neoplasias Asociadas a Colitis/genética , Enfermedad de Crohn/complicaciones , Carcinogénesis/inmunología , Carcinogénesis/patología , Colitis Ulcerosa/genética , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/microbiología , Neoplasias Asociadas a Colitis/inmunología , Neoplasias Asociadas a Colitis/microbiología , Neoplasias Asociadas a Colitis/patología , Enfermedad de Crohn/genética , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/microbiología , Análisis Mutacional de ADN , ADN Mitocondrial/genética , Epigénesis Genética , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/inmunología , Secuenciación de Nucleótidos de Alto Rendimiento , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/inmunología , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Mutación , ARN no Traducido/genética , Transducción de Señal/genética , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA