Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Gut Microbes ; 11(1): 32-50, 2020.
Article in English | MEDLINE | ID: mdl-31179826

ABSTRACT

Iron deficiency, a common comorbidity of gastrointestinal inflammatory disorders such as inflammatory bowel diseases (IBD), is often treated with oral iron supplementation. However, the safety of oral iron supplementation remains controversial because of its association with exacerbated disease activity in a subset of IBD patients. Because iron modulates bacterial growth and function, one possible mechanism by which iron may exacerbate inflammation in susceptible hosts is by modulating the intestinal microbiota. We, therefore, investigated the impact of dietary iron on the intestinal microbiota, utilizing the conventionalization of germ-free mice as a model of a microbial community in compositional flux to recapitulate the instability of the IBD-associated intestinal microbiota. Our findings demonstrate that altering intestinal iron availability during community assembly modulated the microbiota in non-inflamed wild type (WT) and colitis-susceptible interleukin-10-deficient (Il10-/-) mice. Depletion of luminal iron availability promoted luminal compositional changes associated with dysbiotic states irrespective of host genotype, including an expansion of Enterobacteriaceae such as Escherichia coli. Mechanistic in vitro growth competitions confirmed that high-affinity iron acquisition systems in E. coli enhance its abundance over other bacteria in iron-restricted conditions, thereby enabling pathobiont iron scavenging during dietary iron restriction. In contrast, distinct luminal community assembly was observed with dietary iron supplementation in WT versus Il10-/- mice, suggesting that the effects of increased iron on the microbiota differ with host inflammation status. Taken together, shifts in dietary iron intake during community assembly modulate the ecological structure of the intestinal microbiota and is dependent on host genotype and inflammation status.


Subject(s)
Colitis/microbiology , Gastrointestinal Microbiome/drug effects , Inflammatory Bowel Diseases/microbiology , Intestines/microbiology , Iron, Dietary/pharmacology , Animals , Colitis/drug therapy , Colitis/genetics , Colon/microbiology , Disease Models, Animal , Disease Susceptibility , Dysbiosis , Enterobacteriaceae/drug effects , Escherichia coli/drug effects , Genetic Predisposition to Disease , Inflammation/genetics , Inflammation/microbiology , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/genetics , Interleukin-10/genetics , Intestines/pathology , Mice , Mice, Transgenic
2.
J Clin Invest ; 129(4): 1699-1712, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30855275

ABSTRACT

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.


Subject(s)
Biofilms , Carcinogenesis , Colon/microbiology , Colonic Neoplasms/microbiology , Gastrointestinal Microbiome , Neoplasms, Experimental/microbiology , Animals , Colon/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Humans , Mice , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology
3.
Annu Rev Immunol ; 35: 199-228, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28142322

ABSTRACT

Commensal microorganisms (the microbiota) live on all the surface barriers of our body and are particularly abundant and diverse in the distal gut. The microbiota and its larger host represent a metaorganism in which the cross talk between microbes and host cells is necessary for health, survival, and regulation of physiological functions locally, at the barrier level, and systemically. The ancestral molecular and cellular mechanisms stemming from the earliest interactions between prokaryotes and eukaryotes have evolved to mediate microbe-dependent host physiology and tissue homeostasis, including innate and adaptive resistance to infections and tissue repair. Mostly because of its effects on metabolism, cellular proliferation, inflammation, and immunity, the microbiota regulates cancer at the level of predisposing conditions, initiation, genetic instability, susceptibility to host immune response, progression, comorbidity, and response to therapy. Here, we review the mechanisms underlying the interaction of the microbiota with cancer and the evidence suggesting that the microbiota could be targeted to improve therapy while attenuating adverse reactions.


Subject(s)
Immunity, Innate , Immunotherapy/methods , Intestinal Mucosa/immunology , Microbiota/immunology , Neoplasms/immunology , Adaptive Immunity , Animals , Antineoplastic Agents/therapeutic use , Carcinogenesis , Humans , Inflammation , Neoplasms/microbiology , Neoplasms/therapy , Wound Healing
4.
J Vis Exp ; (111)2016 05 11.
Article in English | MEDLINE | ID: mdl-27213580

ABSTRACT

Intestinal ischemia is a life-threatening condition associated with a broad range of clinical conditions including atherosclerosis, thrombosis, hypotension, necrotizing enterocolitis, bowel transplantation, trauma and chronic inflammation. Intestinal ischemia-reperfusion (IR) injury is a consequence of acute mesenteric ischemia, caused by inadequate blood flow through the mesenteric vessels, resulting in intestinal damage. Reperfusion following ischemia can further exacerbate damage of the intestine. The mechanisms of IR injury are complex and poorly understood. Therefore, experimental small animal models are critical for understanding the pathophysiology of IR injury and the development of novel therapies. Here we describe a mouse model of acute intestinal IR injury that provides reproducible injury of the small intestine without mortality. This is achieved by inducing ischemia in the region of the distal ileum by temporally occluding the peripheral and terminal collateral branches of the superior mesenteric artery for 60 min using microvascular clips. Reperfusion for 1 hr, or 2 hr after injury results in reproducible injury of the intestine examined by histological analysis. Proper position of the microvascular clips is critical for the procedure. Therefore the video clip provides a detailed visual step-by-step description of this technique. This model of intestinal IR injury can be utilized to study the cellular and molecular mechanisms of injury and regeneration.


Subject(s)
Disease Models, Animal , Reperfusion Injury , Animals , Ileum , Intestine, Small , Mesenteric Artery, Superior , Mice
5.
Psychosom Med ; 78(5): 610-9, 2016 06.
Article in English | MEDLINE | ID: mdl-27035357

ABSTRACT

OBJECTIVES: The microbiota-gut-brain axis is increasingly implicated in obesity, anxiety, stress, and other health-related processes. Researchers have proposed that gut microbiota may influence dietary habits, and pathways through the microbiota-gut-brain axis make such a relationship feasible; however, few data bear on the hypothesis. As a first step in the development of a model system, the gut microbiome was examined in rat lines selectively outbred on a taste phenotype with biobehavioral profiles that have diverged with respect to energy regulation, anxiety, and stress. METHODS: Occidental low and high-saccharin-consuming rats were assessed for body mass and chow, water, and saccharin intake; littermate controls had shared cages with rats in the experimental group but were not assessed. Cecum and colon microbial communities were profiled using Illumina 16S rRNA sequencing and multivariate analysis of microbial diversity and composition. RESULTS: The saccharin phenotype was confirmed (low-saccharin-consuming rats, 0.7Δ% [0.9Δ%]; high-saccharin-consuming rats, 28.1Δ% [3.6Δ%]). Regardless of saccharin exposure, gut microbiota differed between lines in terms of overall community similarity and taxa at lower phylogenetic levels. Specifically, 16 genera in three phyla distinguished the lines at a 10% false discovery rate. DISCUSSION: The study demonstrates for the first time that rodent lines created through selective pressure on taste and differing on functionally related correlates host different microbial communities. Whether the microbiota are causally related to the taste phenotype or its correlates remains to be determined. These findings encourage further inquiry on the relationship of the microbiome to taste, dietary habits, emotion, and health.


Subject(s)
Behavior, Animal/physiology , Gastrointestinal Microbiome/physiology , Models, Animal , Phenotype , Taste/physiology , Animals , Male , RNA, Ribosomal, 16S , Rats , Sequence Analysis, RNA
6.
Curr Opin Immunol ; 39: 75-81, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26820225

ABSTRACT

The relationship between the host and the commensal microbiota regulates physiological functions including inflammation and immunity and it has been scrutinized in the context of cancer. While viruses and bacterial species have been implicated in oncogenesis, commensal microbes also have a beneficial role in the fight against cancer. Therapy efficacy, including adoptive T cell transfer, alkylating agents and immune checkpoint blockers, relies on immunity that receives its education from the gut microbiota. In cancer therapy with immunostimulating oligonucleotides and platinum salts, the microbiota also modulates the response by priming for the release of pro-inflammatory factors and reactive oxygen species, respectively. This new information offers promising clinical possibilities of modulating cancer therapy and its toxic side effects by targeting the microbiota.


Subject(s)
Gastrointestinal Microbiome/immunology , Neoplasms/therapy , Animals , Humans , Immunity, Innate/immunology , Intestinal Mucosa/microbiology , Symbiosis
7.
Nat Commun ; 5: 4724, 2014 Sep 03.
Article in English | MEDLINE | ID: mdl-25182170

ABSTRACT

Enterobacteria, especially Escherichia coli, are abundant in patients with inflammatory bowel disease or colorectal cancer (CRC). However, it is unclear whether cancer is promoted by inflammation-induced expansion of E. coli and/or changes in expression of specific microbial genes. Here we use longitudinal (2, 12 and 20 weeks) 16S rRNA sequencing of luminal microbiota from ex-germ-free mice to show that inflamed Il10(-/-) mice maintain a higher abundance of Enterobacteriaceae than healthy wild-type mice. Experiments with mono-colonized Il10(-/-) mice reveal that host inflammation is necessary for E. coli cancer-promoting activity. RNA-sequence analysis indicates significant changes in E. coli gene catalogue in Il10(-/-) mice, with changes mostly driven by adaptation to the intestinal environment. Expression of specific genes present in the tumour-promoting E. coli pks island are modulated by inflammation/CRC development. Thus, progression of inflammation in Il10(-/-) mice supports Enterobacteriaceae and alters a small subset of microbial genes important for tumour development.


Subject(s)
Colorectal Neoplasms/genetics , Escherichia coli Infections/genetics , Escherichia coli/genetics , Genes, Bacterial , Genome, Bacterial , Animals , Colorectal Neoplasms/complications , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Escherichia coli/immunology , Escherichia coli/pathogenicity , Escherichia coli Infections/complications , Escherichia coli Infections/immunology , Escherichia coli Infections/pathology , Female , Gene Expression , Genomic Islands , Host-Pathogen Interactions , Inflammation/complications , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukin-10/immunology , Male , Mice , Mice, Knockout , Microbiota/genetics , Microbiota/immunology , RNA, Ribosomal, 16S/genetics
8.
Am J Pathol ; 184(11): 2965-75, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25204845

ABSTRACT

Nucleotide-binding oligomerization domain-containing protein 2 (NOD2), an intracellular pattern recognition receptor, induces autophagy on detection of muramyl dipeptide (MDP), a component of microbial cell walls. The role of bacteria and NOD2 signaling toward ischemia/reperfusion (I/R)-induced intestinal injury response is unknown. Herein, we report that I/R-induced intestinal injury in germ-free (GF) C57BL/6 wild-type (WT) mice is worse than in conventionally derived mice. More important, microbiota-mediated protection against I/R-induced intestinal injury is abrogated in conventionally derived Nod2(-/-) mice and GF Nod2(-/-) mice. Also, WT mice raised in specific pathogen-free (SPF) conditions fared better against I/R-induced injury than SPF Nod2(-/-) mice. Moreover, SPF WT mice i.p. administered 10 mg/kg MDP were protected against injury compared with mice administered the inactive enantiomer, l-MDP, an effect lost in Nod2(-/-) mice. However, MDP administration failed to protect GF mice from I/R-induced intestinal injury compared with control, a phenomenon correlating with undetectable Nod2 mRNA level in the epithelium of GF mice. More important, the autophagy-inducer rapamycin protected Nod2(-/-) mice against I/R-induced injury and increased the levels of LC3(+) puncta in injured tissue of Nod2(-/-) mice. These findings demonstrate that NOD2 protects against I/R and promotes wound healing, likely through the induction of the autophagy response.


Subject(s)
Intestines/microbiology , Microbiota/physiology , Nod2 Signaling Adaptor Protein/metabolism , Reperfusion Injury/prevention & control , Signal Transduction/physiology , Animals , Autophagy/genetics , Intestinal Mucosa/metabolism , Intestines/blood supply , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nod2 Signaling Adaptor Protein/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/microbiology
9.
Bioessays ; 36(7): 658-64, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24752750

ABSTRACT

In humans, the intestine is the major reservoir of microbes. Although the intestinal microbial community exists in a state of homeostasis called eubiosis, environmental and genetics factors can lead to microbial perturbation or dysbiosis, a state associated with various pathologies including inflammatory bowel diseases (IBD) and colorectal cancer (CRC). Dysbiotic microbiota is thought to contribute to the initiation and progression of CRC. At the opposite end of the spectrum, two recently published studies in Science reveal that the microbiota is essential for chemotherapeutic drug efficacy, suggesting a beneficial microbial function in cancer management. The dichotomy between the beneficial and detrimental roles of the microbiota during cancer initiation, progression, and treatment emphasize the interwoven relationship between bacteria and cancer. Moreover, these findings suggest that the microbiota could be considered as a therapeutic target, not only at the level of cancer prevention, but also during management, i.e. by enhancing the efficacy of chemotherapeutics.


Subject(s)
Intestines/microbiology , Microbiota/physiology , Neoplasms/immunology , Neoplasms/therapy , Tumor Microenvironment/immunology , Animals
10.
Am J Pathol ; 184(3): 592-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24418259

ABSTRACT

Regional expression of Wingless/Int (Wnt) genes plays a central role in regulating intestinal development and homeostasis. However, our knowledge of such regional Wnt proteins in the colon remains limited. To understand further the effect of Wnt signaling components in controlling intestinal epithelial homeostasis, we investigated whether the physiological heterogeneity of the proximal and distal colon can be explained by differential Wnt signaling. With the use of a Wnt signaling-specific PCR array, expression of 84 Wnt-mediated signal transduction genes was analyzed, and a differential signature of Wnt-related genes in the proximal versus distal murine colon was identified. Several Wnt agonists (Wnt5a, Wnt8b, and Wnt11), the Wnt receptor frizzled family receptor 3, and the Wnt inhibitory factor 1 were differentially expressed along the colon length. These Wnt signatures were associated with differential epithelial cell proliferation and migration in the proximal versus distal colon. Furthermore, reduced Wnt/ß-catenin activity and decreased Wnt5a and Wnt11 expression were observed in mice lacking commensal bacteria, an effect that was reversed by conventionalization of germ-free mice. Interestingly, myeloid differentiation primary response gene 88 knockout mice showed decreased Wnt5a levels, indicating a role for Toll-like receptor signaling in regulating Wnt5a expression. Our results suggest that the morphological and physiological heterogeneity within the colon is in part facilitated by the differential expression of Wnt signaling components and influenced by colonization with bacteria.


Subject(s)
Bacteria/metabolism , Colon/microbiology , Signal Transduction , Wnt Proteins/metabolism , Animals , Cell Proliferation , Colon/anatomy & histology , Colon/metabolism , Gene Expression Regulation, Bacterial , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota , Specific Pathogen-Free Organisms , Wnt Proteins/genetics , Wnt-5a Protein
11.
Inflamm Bowel Dis ; 19(13): 2857-66, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24141713

ABSTRACT

The Toll-like receptor/MyD88 signaling pathway has been shown to mediate protective functions during intestinal exposure to various noxious events. The goal of this study was to define the role of bacteria and MyD88 signaling in intestinal response to damage using an ischemia-reperfusion (I/R)-induced injury model. We showed that conventionalized mice displayed a better outcome to I/R-induced injury than germ-free mice (3.8 ± 1.98 vs. 11.8 ± 1.83, P < 0.05). However, mice with intestinal epithelial cell (IEC)-specific deletion of Myd88 (Myd88) were protected from I/R-induced injury compared with Myd88 control mice. Myd88 mice also displayed a significantly reduced bacterial translocation (∼85%) into lymph nodes compared with Myd88 mice. Expression of ccl2 and cxcl1 mRNA was significantly reduced (85% and 62%, respectively) in intestinal tissue of Myd88 mice compared with Myd88 mice, which associated with a reduced number of myeloperoxidase-positive cells in intestinal tissues of I/R-exposed Myd88 mice. Immunohistochemistry analysis showed a reduced IgA deposition and complement staining in ischemic tissue of Myd88 mice compared with Myd88 mice. These findings suggest that I/R-induced intestinal injury involves IEC-derived MyD88 signaling leading to increased IgA deposition/degradation, and complement activation in conjunction with an influx of neutrophils mediated by chemokine production.


Subject(s)
Bacteria/pathogenicity , Epithelial Cells/pathology , Intestines/injuries , Myeloid Differentiation Factor 88/physiology , Reperfusion Injury/physiopathology , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Female , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Intestines/microbiology , Intestines/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neutrophils/metabolism , Neutrophils/microbiology , Neutrophils/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reperfusion Injury/metabolism , Reperfusion Injury/microbiology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
12.
Sci Rep ; 3: 2868, 2013 Oct 08.
Article in English | MEDLINE | ID: mdl-24100376

ABSTRACT

Although probiotics have shown success in preventing the development of experimental colitis-associated colorectal cancer (CRC), beneficial effects of interventional treatment are relatively unknown. Here we show that interventional treatment with VSL#3 probiotic alters the luminal and mucosally-adherent microbiota, but does not protect against inflammation or tumorigenesis in the azoxymethane (AOM)/Il10⁻/⁻ mouse model of colitis-associated CRC. VSL#3 (109 CFU/animal/day) significantly enhanced tumor penetrance, multiplicity, histologic dysplasia scores, and adenocarcinoma invasion relative to VSL#3-untreated mice. Illumina 16S sequencing demonstrated that VSL#3 significantly decreased (16-fold) the abundance of a bacterial taxon assigned to genus Clostridium in the mucosally-adherent microbiota. Mediation analysis by linear models suggested that this taxon was a contributing factor to increased tumorigenesis in VSL#3-fed mice. We conclude that VSL#3 interventional therapy can alter microbial community composition and enhance tumorigenesis in the AOM/Il10⁻/⁻ model.


Subject(s)
Colitis/complications , Colitis/microbiology , Colorectal Neoplasms/etiology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Probiotics/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Colitis/genetics , Colitis/pathology , Colorectal Neoplasms/pathology , Disease Models, Animal , Mice , Mice, Knockout , Microbiota , Probiotics/administration & dosage
13.
ISME J ; 7(11): 2116-25, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23823492

ABSTRACT

Maternal transmission and cage effects are powerful confounding factors in microbiome studies. To assess the consequences of cage microenvironment on the mouse gut microbiome, two groups of germ-free (GF) wild-type (WT) mice, one gavaged with a microbiota harvested from adult WT mice and another allowed to acquire the microbiome from the cage microenvironment, were monitored using Illumina 16S rRNA sequencing over a period of 8 weeks. Our results revealed that cage effects in WT mice moved from GF to specific pathogen free (SPF) conditions take several weeks to develop and are not eliminated by the initial gavage treatment. Initial gavage influenced, but did not eliminate a successional pattern in which Proteobacteria became less abundant over time. An analysis in which 16S rRNA sequences are mapped to the closest sequenced whole genome suggests that the functional potential of microbial genomes changes significantly over time shifting from an emphasis on pathogenesis and motility early in community assembly to metabolic processes at later time points. Functionally, mice allowed to naturally acquire a microbial community from their cage, but not mice gavaged with a common biome, exhibit a cage effect in Dextran Sulfate Sodium-induced inflammation. Our results argue that while there are long-term effects of the founding community, these effects are mitigated by cage microenvironment and successional community assembly over time, which must both be explicitly considered in the interpretation of microbiome mouse experiments.


Subject(s)
Biodiversity , Gastrointestinal Tract/microbiology , Animals , Environmental Microbiology , Founder Effect , Housing, Animal , Mice , RNA, Ribosomal, 16S/genetics , Time Factors
14.
Am J Pathol ; 182(3): 776-85, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23291213

ABSTRACT

Intestinal ischemia has a wide variety of causes, including, but not limited to, atherosclerosis, thrombosis, hypotension, and chronic inflammation. In severe cases, ischemic injury can result in death. µ-Opioid receptor (MOR) signaling has previously been shown to protect against chemically induced colitis, but the cellular origin of this effect remains unknown. Herein, we evaluated the role of intestinal epithelial cell (IEC)-derived MOR signaling in host responses to ischemia/reperfusion-induced injury. Ileal ischemia was accomplished through obstruction of the distal branches of the superior mesenteric artery (60 minutes) and reperfusion for 90 minutes (ischemia-reperfusion). Floxed-MOR mice were crossed to Villin-cre transgenic mice to selectively delete the MOR gene in IECs (MOR(IEC-/-)). Radio-ligand binding assays demonstrated selective loss of MOR signaling in IECs of MOR(IEC-/-) mice. The s.c. administration of the MOR agonist, [D-Arg2, Lys4] dermorphin (1-4) amide (DALDA), 10 minutes before surgery protected against both ischemic and reperfusion phases of intestinal injury, an effect abolished in MOR(IEC-/-) mice. This cytoprotective effect was associated with enterocyte-mediated phosphoinositide 3-kinase (PI3K)/glycogen synthase kinase 3ß signaling and decreased apoptosis, as determined by IHC and caspase-3 processing. PI3K blockade with Ly294002 resulted in loss of MOR-mediated cytoprotective function. Together, these data show that IEC-derived µ-opioid signaling uses the PI3K pathway to protect cells against the damaging effect of ischemia-reperfusion. Targeting MOR signaling may represent a novel mean to alleviate intestinal injury and promote the wound-healing response.


Subject(s)
Epithelial Cells/pathology , Intestines/pathology , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Opioid, mu/metabolism , Reperfusion Injury/enzymology , Reperfusion Injury/prevention & control , Signal Transduction , Animals , Apoptosis/drug effects , Cytoprotection/drug effects , Enterocytes/metabolism , Enterocytes/pathology , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Gene Deletion , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Intestines/blood supply , Ligands , Mice , Mice, Inbred C57BL , Oligopeptides/administration & dosage , Oligopeptides/pharmacology , Organ Specificity/drug effects , Phosphorylation/drug effects , Protective Agents/pharmacology , Reperfusion Injury/pathology , Signal Transduction/drug effects
15.
Science ; 338(6103): 120-3, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-22903521

ABSTRACT

Inflammation alters host physiology to promote cancer, as seen in colitis-associated colorectal cancer (CRC). Here, we identify the intestinal microbiota as a target of inflammation that affects the progression of CRC. High-throughput sequencing revealed that inflammation modifies gut microbial composition in colitis-susceptible interleukin-10-deficient (Il10(-/-)) mice. Monocolonization with the commensal Escherichia coli NC101 promoted invasive carcinoma in azoxymethane (AOM)-treated Il10(-/-) mice. Deletion of the polyketide synthase (pks) genotoxic island from E. coli NC101 decreased tumor multiplicity and invasion in AOM/Il10(-/-) mice, without altering intestinal inflammation. Mucosa-associated pks(+) E. coli were found in a significantly high percentage of inflammatory bowel disease and CRC patients. This suggests that in mice, colitis can promote tumorigenesis by altering microbial composition and inducing the expansion of microorganisms with genotoxic capabilities.


Subject(s)
Carcinoma/microbiology , Colitis/complications , Colorectal Neoplasms/microbiology , DNA Damage , Intestines/microbiology , Metagenome/physiology , Animals , Azoxymethane/toxicity , Carcinogens/toxicity , Carcinoma/chemically induced , Carcinoma/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Colitis/genetics , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/pathology , Escherichia coli/genetics , Escherichia coli/pathogenicity , Interleukin-10/genetics , Intestines/pathology , Metagenome/genetics , Mice , Mice, Mutant Strains , Polyketide Synthases/genetics , Sequence Deletion
SELECTION OF CITATIONS
SEARCH DETAIL
...