Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Proc Natl Acad Sci U S A ; 111(51): 18321-6, 2014 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-25489084

RESUMEN

Environmental factors clearly affect colorectal cancer (CRC) incidence, but the mechanisms through which these factors function are unknown. One prime candidate is an altered colonic microbiota. Here we show that the mucosal microbiota organization is a critical factor associated with a subset of CRC. We identified invasive polymicrobial bacterial biofilms (bacterial aggregates), structures previously associated with nonmalignant intestinal pathology, nearly universally (89%) on right-sided tumors (13 of 15 CRCs, 4 of 4 adenomas) but on only 12% of left-sided tumors (2 of 15 CRCs, 0 of 2 adenomas). Surprisingly, patients with biofilm-positive tumors, whether cancers or adenomas, all had biofilms on their tumor-free mucosa far distant from their tumors. Bacterial biofilms were associated with diminished colonic epithelial cell E-cadherin and enhanced epithelial cell IL-6 and Stat3 activation, as well as increased crypt epithelial cell proliferation in normal colon mucosa. High-throughput sequencing revealed no consistent bacterial genus associated with tumors, regardless of biofilm status. However, principal coordinates analysis revealed that biofilm communities on paired normal mucosa, distant from the tumor itself, cluster with tumor microbiomes as opposed to biofilm-negative normal mucosa bacterial communities also from the tumor host. Colon mucosal biofilm detection may predict increased risk for development of sporadic CRC.


Asunto(s)
Neoplasias Colorrectales/microbiología , Microbiota , Bacterias/clasificación , Bacterias/aislamiento & purificación , Biopelículas , Colonoscopía , Humanos
2.
Cancer Discov ; 12(8): 1873-1885, 2022 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-35678528

RESUMEN

Defining the complex role of the microbiome in colorectal cancer and the discovery of novel, protumorigenic microbes are areas of active investigation. In the present study, culturing and reassociation experiments revealed that toxigenic strains of Clostridioides difficile drove the tumorigenic phenotype of a subset of colorectal cancer patient-derived mucosal slurries in germ-free ApcMin/+ mice. Tumorigenesis was dependent on the C. difficile toxin TcdB and was associated with induction of Wnt signaling, reactive oxygen species, and protumorigenic mucosal immune responses marked by the infiltration of activated myeloid cells and IL17-producing lymphoid and innate lymphoid cell subsets. These findings suggest that chronic colonization with toxigenic C. difficile is a potential driver of colorectal cancer in patients. SIGNIFICANCE: Colorectal cancer is a leading cause of cancer and cancer-related deaths worldwide, with a multifactorial etiology that likely includes procarcinogenic bacteria. Using human colon cancer specimens, culturing, and murine models, we demonstrate that chronic infection with the enteric pathogen C. difficile is a previously unrecognized contributor to colonic tumorigenesis. See related commentary by Jain and Dudeja, p. 1838. This article is highlighted in the In This Issue feature, p. 1825.


Asunto(s)
Toxinas Bacterianas , Clostridioides difficile , Neoplasias del Colon , Neoplasias Colorrectales , Animales , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Carcinogénesis , Clostridioides , Humanos , Inmunidad Innata , Linfocitos/metabolismo , Ratones
3.
Cancer Discov ; 11(7): 1792-1807, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33632774

RESUMEN

Colorectal cancer is multifaceted, with subtypes defined by genetic, histologic, and immunologic features that are potentially influenced by inflammation, mutagens, and/or microbiota. Colorectal cancers with activating mutations in BRAF are associated with distinct clinical characteristics, although the pathogenesis is not well understood. The Wnt-driven multiple intestinal neoplasia (MinApcΔ716/+) enterotoxigenic Bacteroides fragilis (ETBF) murine model is characterized by IL17-dependent, distal colon adenomas. Herein, we report that the addition of the BRAF V600E mutation to this model results in the emergence of a distinct locus of midcolon tumors. In ETBF-colonized BRAF V600E Lgr5 CreMin (BLM) mice, tumors have similarities to human BRAF V600E tumors, including histology, CpG island DNA hypermethylation, and immune signatures. In comparison to Min ETBF tumors, BLM ETBF tumors are infiltrated by CD8+ T cells, express IFNγ signatures, and are sensitive to anti-PD-L1 treatment. These results provide direct evidence for critical roles of host genetic and microbiota interactions in colorectal cancer pathogenesis and sensitivity to immunotherapy. SIGNIFICANCE: Colorectal cancers with BRAF mutations have distinct characteristics. We present evidence of specific colorectal cancer gene-microbial interactions in which colonization with toxigenic bacteria drives tumorigenesis in BRAF V600E Lgr5 CreMin mice, wherein tumors phenocopy aspects of human BRAF-mutated tumors and have a distinct IFNγ-dominant immune microenvironment uniquely responsive to immune checkpoint blockade.This article is highlighted in the In This Issue feature, p. 1601.


Asunto(s)
Bacteroides fragilis/fisiología , Neoplasias Colorrectales/microbiología , Proteínas Proto-Oncogénicas B-raf/genética , Animales , Carcinogénesis , Transformación Celular Neoplásica , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/terapia , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Mutación
4.
mSystems ; 5(1)2020 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-31937674

RESUMEN

Disrupted interactions between host and intestinal bacteria are implicated in colorectal cancer (CRC) development. However, activities derived from these bacteria and their interplay with the host are unclear. Here, we examine this interplay by performing mouse and microbiota RNA sequencing on colon tissues and 16S and small RNA sequencing on stools from germfree (GF) and gnotobiotic ApcMin Δ 850/+ ;Il10-/- mice associated with microbes from biofilm-positive human CRC tumor (BF+T) and biofilm-negative healthy (BF-bx) tissues. The bacteria in BF+T mice differentially expressed (DE) >2,900 genes, including genes related to bacterial secretion, virulence, and biofilms but affected only 62 host genes. Small RNA sequencing of stools from these cohorts revealed eight significant DE host microRNAs (miRNAs) based on biofilm status and several miRNAs that correlated with bacterial taxon abundances. Additionally, computational predictions suggest that some miRNAs preferentially target bacterial genes while others primarily target mouse genes. 16S rRNA sequencing of mice that were reassociated with mucosa-associated communities from the initial association revealed a set of 13 bacterial genera associated with cancer that were maintained regardless of whether the reassociation inoculums were initially obtained from murine proximal or distal colon tissues. Our findings suggest that complex interactions within bacterial communities affect host-derived miRNA, bacterial composition, and CRC development.IMPORTANCE Bacteria and bacterial biofilms have been implicated in colorectal cancer (CRC), but it is still unclear what genes these microbial communities express and how they influence the host. MicroRNAs regulate host gene expression and have been explored as potential biomarkers for CRC. An emerging area of research is the ability of microRNAs to impact growth and gene expression of members of the intestinal microbiota. This study examined the bacteria and bacterial transcriptome associated with microbes derived from biofilm-positive human cancers that promoted tumorigenesis in a murine model of CRC. The murine response to different microbial communities (derived from CRC patients or healthy people) was evaluated through RNA and microRNA sequencing. We identified a complex interplay between biofilm-associated bacteria and the host during CRC in mice. These findings may lead to the development of new biomarkers and therapeutics for identifying and treating biofilm-associated CRCs.

6.
J Clin Invest ; 129(4): 1699-1712, 2019 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-30855275

RESUMEN

Mucus-invasive bacterial biofilms are identified on the colon mucosa of approximately 50% of colorectal cancer (CRC) patients and approximately 13% of healthy subjects. Here, we test the hypothesis that human colon biofilms comprise microbial communities that are carcinogenic in CRC mouse models. Homogenates of human biofilm-positive colon mucosa were prepared from tumor patients (tumor and paired normal tissues from surgical resections) or biofilm-positive biopsies from healthy individuals undergoing screening colonoscopy; homogenates of biofilm-negative colon biopsies from healthy individuals undergoing screening colonoscopy served as controls. After 12 weeks, biofilm-positive, but not biofilm-negative, human colon mucosal homogenates induced colon tumor formation in 3 mouse colon tumor models (germ-free ApcMinΔ850/+;Il10-/- or ApcMinΔ850/+ and specific pathogen-free ApcMinΔ716/+ mice). Remarkably, biofilm-positive communities from healthy colonoscopy biopsies induced colon inflammation and tumors similarly to biofilm-positive tumor tissues. By 1 week, biofilm-positive human tumor homogenates, but not healthy biopsies, displayed consistent bacterial mucus invasion and biofilm formation in mouse colons. 16S rRNA gene sequencing and RNA-Seq analyses identified compositional and functional microbiota differences between mice colonized with biofilm-positive and biofilm-negative communities. These results suggest human colon mucosal biofilms, whether from tumor hosts or healthy individuals undergoing screening colonoscopy, are carcinogenic in murine models of CRC.


Asunto(s)
Biopelículas , Carcinogénesis , Colon/microbiología , Neoplasias del Colon/microbiología , Microbioma Gastrointestinal , Neoplasias Experimentales/microbiología , Animales , Colon/metabolismo , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Humanos , Ratones , Ratones Noqueados , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología
7.
Science ; 359(6375): 592-597, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29420293

RESUMEN

Individuals with sporadic colorectal cancer (CRC) frequently harbor abnormalities in the composition of the gut microbiome; however, the microbiota associated with precancerous lesions in hereditary CRC remains largely unknown. We studied colonic mucosa of patients with familial adenomatous polyposis (FAP), who develop benign precursor lesions (polyps) early in life. We identified patchy bacterial biofilms composed predominately of Escherichia coli and Bacteroides fragilis Genes for colibactin (clbB) and Bacteroides fragilis toxin (bft), encoding secreted oncotoxins, were highly enriched in FAP patients' colonic mucosa compared to healthy individuals. Tumor-prone mice cocolonized with E. coli (expressing colibactin), and enterotoxigenic B. fragilis showed increased interleukin-17 in the colon and DNA damage in colonic epithelium with faster tumor onset and greater mortality, compared to mice with either bacterial strain alone. These data suggest an unexpected link between early neoplasia of the colon and tumorigenic bacteria.


Asunto(s)
Poliposis Adenomatosa del Colon/microbiología , Poliposis Adenomatosa del Colon/patología , Bacteroides fragilis/patogenicidad , Biopelículas , Carcinogénesis , Colon/microbiología , Neoplasias del Colon/microbiología , Escherichia coli/patogenicidad , Interleucina-17/análisis , Animales , Toxinas Bacterianas/genética , Bacteroides fragilis/genética , Bacteroides fragilis/aislamiento & purificación , Colon/patología , Neoplasias del Colon/patología , Daño del ADN , Escherichia coli/genética , Escherichia coli/aislamiento & purificación , Microbioma Gastrointestinal , Humanos , Mucosa Intestinal/química , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Ratones , Péptidos/genética , Péptidos/metabolismo , Policétidos , Lesiones Precancerosas/microbiología
8.
Cell Host Microbe ; 23(2): 203-214.e5, 2018 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-29398651

RESUMEN

Pro-carcinogenic bacteria have the potential to initiate and/or promote colon cancer, in part via immune mechanisms that are incompletely understood. Using ApcMin mice colonized with the human pathobiont enterotoxigenic Bacteroides fragilis (ETBF) as a model of microbe-induced colon tumorigenesis, we show that the Bacteroides fragilis toxin (BFT) triggers a pro-carcinogenic, multi-step inflammatory cascade requiring IL-17R, NF-κB, and Stat3 signaling in colonic epithelial cells (CECs). Although necessary, Stat3 activation in CECs is not sufficient to trigger ETBF colon tumorigenesis. Notably, IL-17-dependent NF-κB activation in CECs induces a proximal to distal mucosal gradient of C-X-C chemokines, including CXCL1, that mediates the recruitment of CXCR2-expressing polymorphonuclear immature myeloid cells with parallel onset of ETBF-mediated distal colon tumorigenesis. Thus, BFT induces a pro-carcinogenic signaling relay from the CEC to a mucosal Th17 response that results in selective NF-κB activation in distal colon CECs, which collectively triggers myeloid-cell-dependent distal colon tumorigenesis.


Asunto(s)
Toxinas Bacterianas/inmunología , Bacteroides fragilis/inmunología , Carcinogénesis/patología , Colon/inmunología , Neoplasias Colorrectales/etiología , Células Epiteliales/inmunología , Interleucina-17/inmunología , Metaloendopeptidasas/inmunología , Factor de Transcripción ReIA/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Toxinas Bacterianas/metabolismo , Bacteroides fragilis/patogenicidad , Línea Celular Tumoral , Colon/citología , Colon/microbiología , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/patología , Activación Enzimática/inmunología , Femenino , Eliminación de Gen , Células HT29 , Humanos , Inflamación/inmunología , Inflamación/microbiología , Interleucina-17/genética , Masculino , Metaloendopeptidasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/inmunología , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/inmunología , Receptores de Interleucina-8B/genética , Factor de Transcripción STAT3/metabolismo
9.
Artículo en Inglés | MEDLINE | ID: mdl-29214046

RESUMEN

Colorectal cancer (CRC) remains the third most common cancer worldwide, with a growing incidence among young adults. Multiple studies have presented associations between the gut microbiome and CRC, suggesting a link with cancer risk. Although CRC microbiome studies continue to profile larger patient cohorts with increasingly economical and rapid DNA sequencing platforms, few common associations with CRC have been identified, in part due to limitations in taxonomic resolution and differences in analysis methodologies. Complementing these taxonomic studies is the newly recognized phenomenon that bacterial organization into biofilm structures in the mucus layer of the gut is a consistent feature of right-sided (proximal), but not left-sided (distal) colorectal cancer. In the present study, we performed 16S rRNA gene amplicon sequencing and biofilm quantification in a new cohort of patients from Malaysia, followed by a meta-analysis of eleven additional publicly available data sets on stool and tissue-based CRC microbiota using Resphera Insight, a high-resolution analytical tool for species-level characterization. Results from the Malaysian cohort and the expanded meta-analysis confirm that CRC tissues are enriched for invasive biofilms (particularly on right-sided tumors), a symbiont with capacity for tumorigenesis (Bacteroides fragilis), and oral pathogens including Fusobacterium nucleatum, Parvimonas micra, and Peptostreptococcus stomatis. Considered in aggregate, species from the Human Oral Microbiome Database are highly enriched in CRC. Although no detected microbial feature was universally present, their substantial overlap and combined prevalence supports a role for the gut microbiota in a significant percentage (>80%) of CRC cases.

10.
Gut Microbes ; 7(1): 54-7, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26939852

RESUMEN

It is now widely recognized that a range of human diseases, including obesity, cancer and inflammatory bowel disease, is strongly linked to the microbiota. For decades, the microbiota has been proposed to contribute to the pathogenesis of colon cancer. Our recent work reveals that the organization of the mucosal microbiota into biofilms marks a subset of human colon cancer. Further, biofilm-positive colon mucosa in the colon cancer host yields an infrequently detected polyamine metabolite, N(1), N(12)-diacetylspermine, that deserves further study to determine its utility as a marker for colon neoplasia.


Asunto(s)
Biopelículas , Neoplasias del Colon/microbiología , Microbioma Gastrointestinal , Mucosa Intestinal/patología , Espermina/análogos & derivados , Toxinas Bacterianas/metabolismo , Bacteroides fragilis/patogenicidad , Neoplasias del Colon/patología , Escherichia coli/patogenicidad , Fusobacterium nucleatum/patogenicidad , Humanos , Mucosa Intestinal/microbiología , Metaloendopeptidasas/metabolismo , Péptidos/metabolismo , Policétidos/metabolismo , Espermina/metabolismo
11.
Cell Metab ; 21(6): 891-7, 2015 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-25959674

RESUMEN

Bacterial biofilms in the colon alter the host tissue microenvironment. A role for biofilms in colon cancer metabolism has been suggested but to date has not been evaluated. Using metabolomics, we investigated the metabolic influence that microbial biofilms have on colon tissues and the related occurrence of cancer. Patient-matched colon cancers and histologically normal tissues, with or without biofilms, were examined. We show the upregulation of polyamine metabolites in tissues from cancer hosts with significant enhancement of N(1), N(12)-diacetylspermine in both biofilm-positive cancer and normal tissues. Antibiotic treatment, which cleared biofilms, decreased N(1), N(12)-diacetylspermine levels to those seen in biofilm-negative tissues, indicating that host cancer and bacterial biofilm structures contribute to the polyamine metabolite pool. These results show that colonic mucosal biofilms alter the cancer metabolome to produce a regulator of cellular proliferation and colon cancer growth potentially affecting cancer development and progression.


Asunto(s)
Bacterias/metabolismo , Fenómenos Fisiológicos Bacterianos , Biopelículas , Neoplasias del Colon , Espermina/análogos & derivados , Neoplasias del Colon/metabolismo , Neoplasias del Colon/microbiología , Neoplasias del Colon/patología , Femenino , Humanos , Masculino , Espermina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA