Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mol Nutr Food Res ; 64(17): e2000162, 2020 09.
Article in English | MEDLINE | ID: mdl-32656952

ABSTRACT

SCOPE: Previous studies have suggested that diets rich in omega-3 and low in omega-6 long-chain polyunsaturated fatty acids (PUFAs) can limit the development of metabolic syndrome (MetS). Transgenic soybeans yielding oils enriched for omega-3 PUFAs represent a new and readily-available option for incorporating omega-3 PUFAs into diets to provide health benefits. METHODS AND RESULTS: Transgenic soybean oils, enriched for either stearidonic acid (SDA) or eicosapentaenoic acid (EPA), are incorporated into diets to test their effects on limiting the development of MetS in a mouse model of diet-induced obesity. Supplementation with SDA- but not EPA-enriched oils improved features of MetS compared to feeding a control wild-type oil. Because previous studies have linked the gut microorganism Akkermansia muciniphila to the metabolic effects of feeding omega-3 PUFAs, the causal contribution of A. muciniphila to mediating the metabolic benefits provided by SDA-enriched diets is investigated. Although A. muciniphila is not required for SDA-induced metabolic improvements, this microorganism does modulate levels of saturated and mono-unsaturated fatty acids in host adipose tissues. CONCLUSION: Together, these findings support the utilization of SDA-enriched diets to modulate weight gain, glucose metabolism, and fatty acid profiles of liver and adipose tissue.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Glucose/metabolism , Obesity/diet therapy , Soybean Oil/pharmacology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Akkermansia/drug effects , Akkermansia/physiology , Animals , Diet, High-Fat/adverse effects , Dietary Supplements , Eicosapentaenoic Acid/pharmacology , Fatty Acids, Unsaturated/pharmacokinetics , Food, Fortified , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Male , Mice, Inbred C57BL , Obesity/metabolism , Obesity/microbiology , Plants, Genetically Modified , Soybean Oil/chemistry , Soybean Oil/genetics , Weight Gain/drug effects
2.
Mucosal Immunol ; 12(1): 164-177, 2019 01.
Article in English | MEDLINE | ID: mdl-30279518

ABSTRACT

Polysaccharide A (PSA), an immunogenic capsular component of non-toxigenic Bacteroides fragilis (NTBF) strain NCTC 9343, is reported to promote mucosal immune development and suppress colitis. Contrastingly, enterotoxigenic Bacteroides fragilis (ETBF) is highly associated with inflammatory bowel disease (IBD) and colorectal cancer (CRC), rapidly inducing IL-17-dependent murine colitis and tumorigenesis. In specific-pathogen-free (SPF) C57BL/6 wild-type (WT) and multiple intestinal neoplasia (MinApc716+/-) mice, we show that sequential treatment of the NTBF strain, 9343, followed by the ETBF strain, 86-5443-2-2 (86), diminished colitis and tumorigenesis. Mice treated simultaneously with 9343 and 86 exhibited both severe colitis and tumorigenesis. Abrogated disease severity in sequentially treated mice was attributed to 9343 strain dominance and decreased IL-17A, but 86 colonization prior to or simultaneous with 9343 mitigated the anti-inflammatory effect of 9343. Remarkably, 9343-mediated protection was independent of PSA, as sequentially treated mice receiving ΔPSA 9343 exhibited similar protection. Further, SPF WT and Min mice colonized with PSA-competent or PSA-deficient 9343 exhibited similar IL-10, IL-17, and IFN-γ responses. Treatment of 86-colonized mice with 9343 failed to disrupt 86 pathogenesis. Our findings demonstrate that 9343 colonization, independent of PSA, offers prophylaxis against colitis-inducing 86 but may not be a valid therapy once colitis is established.


Subject(s)
Bacteroides fragilis/immunology , Colitis/immunology , Colorectal Neoplasms/immunology , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology , Th17 Cells/immunology , Adenomatous Polyposis Coli Protein/genetics , Animals , Bacteroides fragilis/pathogenicity , Carcinogenesis , Cells, Cultured , Colitis/chemically induced , Disease Models, Animal , Humans , Interleukin-17/metabolism , Lipopolysaccharides/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Trinitrobenzenesulfonic Acid
3.
Front Immunol ; 9: 2318, 2018.
Article in English | MEDLINE | ID: mdl-30356663

ABSTRACT

Escherichia coli is a facultative anaerobic symbiont found widely among mammalian gastrointestinal tracts. Several human studies have reported increased commensal E. coli abundance in the intestine during inflammation; however, host immunological responses toward commensal E. coli during inflammation are not well-defined. Here, we show that colonization of gnotobiotic mice with different genotypes of commensal E. coli isolated from healthy conventional microbiota mice and representing distinct populations of E. coli elicited strain-specific disease phenotypes and immunopathological changes following treatment with the inflammatory stimulus, dextran sulfate sodium (DSS). Production of the inflammatory cytokines GM-CSF, IL-6, and IFN-γ was a hallmark of the severe inflammation induced by E. coli strains of Sequence Type 129 (ST129) and ST375 following DSS administration. In contrast, colonization with E. coli strains ST150 and ST468 caused mild intestinal inflammation and triggered only low levels of pro-inflammatory cytokines, a response indistinguishable from that of E. coli-free control mice treated with DSS. The disease development observed with ST129 and ST375 colonization was not directly associated with their abundance in the GI tract as their levels did not change throughout DSS treatment, and no major differences in bacterial burden in the gut were observed among the strains tested. Data mining and in vivo neutralization identified IL-6 as a key cytokine responsible for the observed differential disease severity. Collectively, our results show that the capacity to exacerbate acute intestinal inflammation is a strain-specific trait that can potentially be overcome by blocking the pro-inflammatory immune responses that mediate intestinal tissue damage.


Subject(s)
Disease Susceptibility , Enterocolitis/etiology , Enterocolitis/metabolism , Escherichia coli , Gastrointestinal Microbiome , Interleukin-6/biosynthesis , Animals , Biopsy , Cytokines/biosynthesis , Disease Models, Animal , Disease Progression , Enterocolitis/pathology , Escherichia coli/classification , Escherichia coli/genetics , Female , Gastrointestinal Microbiome/immunology , Immunomodulation , Interleukin-6/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mice , Multilocus Sequence Typing , Phylogeny
5.
Cell Host Microbe ; 23(2): 203-214.e5, 2018 Feb 14.
Article in English | MEDLINE | ID: mdl-29398651

ABSTRACT

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.


Subject(s)
Bacterial Toxins/immunology , Bacteroides fragilis/immunology , Carcinogenesis/pathology , Colon/immunology , Colorectal Neoplasms/etiology , Epithelial Cells/immunology , Interleukin-17/immunology , Metalloendopeptidases/immunology , Transcription Factor RelA/metabolism , Adenomatous Polyposis Coli Protein/genetics , Animals , Bacterial Toxins/metabolism , Bacteroides fragilis/pathogenicity , Cell Line, Tumor , Colon/cytology , Colon/microbiology , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Enzyme Activation/immunology , Female , Gene Deletion , HT29 Cells , Humans , Inflammation/immunology , Inflammation/microbiology , Interleukin-17/genetics , Male , Metalloendopeptidases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/immunology , Receptors, Interleukin-8B/genetics , STAT3 Transcription Factor/metabolism
6.
Science ; 359(6375): 592-597, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29420293

ABSTRACT

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.


Subject(s)
Adenomatous Polyposis Coli/microbiology , Adenomatous Polyposis Coli/pathology , Bacteroides fragilis/pathogenicity , Biofilms , Carcinogenesis , Colon/microbiology , Colonic Neoplasms/microbiology , Escherichia coli/pathogenicity , Interleukin-17/analysis , Animals , Bacterial Toxins/genetics , Bacteroides fragilis/genetics , Bacteroides fragilis/isolation & purification , Colon/pathology , Colonic Neoplasms/pathology , DNA Damage , Escherichia coli/genetics , Escherichia coli/isolation & purification , Gastrointestinal Microbiome , Humans , Intestinal Mucosa/chemistry , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Mice , Peptides/genetics , Peptides/metabolism , Polyketides , Precancerous Conditions/microbiology
7.
Oncoimmunology ; 5(4): e1118601, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27141400

ABSTRACT

Regulatory T cell (Treg) promote IL-17-mediated colon tumorigenesis in multiple intestinal neoplasia (Min) mice colonized with enterotoxigenic Bacteroides fragilis (ETBF). Although they retain their immunosuppressive function, mucosal Treg are instrumental to initiate ETBF-triggered IL-17 colitis by limiting the availability of IL-2 in the local microenvironment. However, the mechanisms that trigger the recruitment of the pro-carcinogenic Treg in ETBF-colonized colon remained to be elucidated.

8.
Cancer Discov ; 5(10): 1098-109, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26201900

ABSTRACT

UNLABELLED: Many epithelial cancers are associated with chronic inflammation. However, the features of inflammation that are procarcinogenic are not fully understood. Regulatory T cells (Treg) typically restrain overt inflammatory responses and maintain intestinal immune homeostasis. Their immune-suppressive activity can inhibit inflammation-associated cancers. Paradoxically, we show that colonic Tregs initiate IL17-mediated carcinogenesis in multiple intestinal neoplasia mice colonized with the human symbiote enterotoxigenic Bacteroides fragilis (ETBF). Depletion of Tregs in ETBF-colonized C57BL/6 FOXP3(DTR) mice enhanced colitis but diminished tumorigenesis associated with shifting of mucosal cytokine profile from IL17 to IFNγ; inhibition of ETBF-induced colon tumorigenesis was dependent on reduced IL17 inflammation and was independent of IFNγ. Treg enhancement of IL17 production is cell-extrinsic. IL2 blockade restored Th17 responses and tumor formation in Treg-depleted animals. Our findings demonstrate that Tregs limit the availability of IL2 in the local microenvironment, allowing the Th17 development necessary to promote ETBF-triggered neoplasia, and thus unveil a new mechanism whereby Treg responses to intestinal bacterial infection can promote tumorigenesis. SIGNIFICANCE: Tregs promote an oncogenic immune response to a common human symbiote associated with inflammatory bowel disease and colorectal cancer. Our data define mechanisms by which mucosal Tregs, despite suppressing excessive inflammation, promote the earliest stages of immune procarcinogenesis via enhancement of IL17 production at the expense of IFNγ production.


Subject(s)
Bacteroides Infections/complications , Bacteroides fragilis/physiology , Cell Transformation, Neoplastic , Colonic Neoplasms/etiology , Colonic Neoplasms/metabolism , Interleukin-17/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Bacteroides Infections/microbiology , Colonic Neoplasms/pathology , Disease Models, Animal , Interleukin-2/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Lymphocyte Depletion , Mice , Mice, Transgenic , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism
9.
J Clin Invest ; 124(10): 4166-72, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25105360

ABSTRACT

The human body comprises fewer host cells than bacterial cells, most of which are obligate anaerobes residing in the gut. The symbiont Bacteroides fragilis constitutes a relatively small proportion (up to 1%-2%) of cultured fecal bacteria, but colonizes most humans. There are 2 classes of B. fragilis distinguished by their ability to secrete a zinc-dependent metalloprotease toxin, B. fragilis toxin (BFT). Strains that do not secrete BFT are nontoxigenic B. fragilis (NTBF), and those that do are called enterotoxigenic B. fragilis (ETBF). ETBF can induce clinical pathology, including inflammatory diarrhea, although asymptomatic colonization may be common. Intestinal inflammation is mediated by BFT, as yet the only known virulence factor of ETBF. Recent experimental evidence demonstrating that ETBF-driven colitis promotes colon tumorigenesis has generated interest in the potential contribution of ETBF to human colon carcinogenesis. Critical questions about the epidemiology of chronic, subclinical human colonization with ETBF and its impact on the biology of the colon need to be addressed.


Subject(s)
Bacteroides fragilis/pathogenicity , Colon/microbiology , Colonic Neoplasms/microbiology , Intestinal Mucosa/microbiology , Animals , Bacteroides Infections/microbiology , Carcinogenesis , Colon/physiopathology , DNA Damage , Humans , Inflammation , Intestinal Mucosa/physiopathology , Metalloendopeptidases/metabolism , Mice , Mucous Membrane/pathology , Signal Transduction , Symbiosis
10.
Immunotargets Ther ; 3: 39-54, 2014.
Article in English | MEDLINE | ID: mdl-27471699

ABSTRACT

Since their recent discovery, T helper 17 (Th17) cells have been frequently detected in the tumor microenvironment of many malignancies, but their clinical implications remain largely unknown. Interleukin-17 (IL-17) detection is commonly related with poor outcomes in colorectal cancers, yet its presence is associated with antitumor responses in ovarian carcinomas. Numerous experimental models illustrate the divergent roles of Th17 cells in tumor immunity, which appears to be mainly dependent on the tumor context (type, location, and stage of cancer). It is recognized that IL-17 is produced by a variety of cell types and that Th17 cells are endowed with a unique functional plasticity. Therefore, when trying to elucidate potential immune biomarkers and immunotargets, it is extremely important to make a clear dissociation between strategies targeting Th17 versus its hallmark cytokine, IL-17. In this review, we will summarize the data regarding the detection of IL-17 and Th17 in human cancers, consider the experimental evidence on their respective roles in antitumor activity, and discuss the potential of IL-17 as an immune target for therapeutic interventions.

SELECTION OF CITATIONS
SEARCH DETAIL
...