Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Transl Med ; 22(1): 631, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38970018

ABSTRACT

BACKGROUND: Wnt/ß-catenin signalling impairment accounts for 85% of colorectal cancers (CRCs), including sporadic and familial adenomatous polyposis (FAP) settings. An altered PI3K/mTOR pathway and gut microbiota also contribute to CRC carcinogenesis. We studied the interplay between the two pathways and the microbiota composition within each step of CRC carcinogenesis. METHODS: Proteins and target genes of both pathways were analysed by RT-qPCR and IHC in tissues from healthy faecal immunochemical test positive (FIT+, n = 17), FAP (n = 17) and CRC (n = 15) subjects. CRC-related mutations were analysed through NGS and Sanger. Oral, faecal and mucosal microbiota was profiled by 16 S rRNA-sequencing. RESULTS: We found simultaneous hyperactivation of Wnt/ß-catenin and PI3K/mTOR pathways in FAP-lesions compared to CRCs. Wnt/ß-catenin molecular markers positively correlated with Clostridium_sensu_stricto_1 and negatively with Bacteroides in FAP faecal microbiota. Alistipes, Lachnospiraceae, and Ruminococcaceae were enriched in FAP stools and adenomas, the latter also showing an overabundance of Lachnoclostridium, which positively correlated with cMYC. In impaired-mTOR-mutated CRC tissues, p-S6R correlated with Fusobacterium and Dialister, the latter also confirmed in the faecal-ecosystem. CONCLUSIONS: Our study reveals an interplay between Wnt/ß-catenin and PI3K/mTOR, whose derangement correlates with specific microbiota signatures in FAP and CRC patients, and identifies new potential biomarkers and targets to improve CRC prevention, early adenoma detection and treatment.


Subject(s)
Carcinogenesis , Colorectal Neoplasms , Phosphatidylinositol 3-Kinases , TOR Serine-Threonine Kinases , Wnt Signaling Pathway , Humans , Colorectal Neoplasms/microbiology , TOR Serine-Threonine Kinases/metabolism , Pilot Projects , Phosphatidylinositol 3-Kinases/metabolism , Male , Female , Adenomatous Polyposis Coli/microbiology , Adenomatous Polyposis Coli/genetics , Middle Aged , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Feces/microbiology , Gastrointestinal Microbiome , Aged , Adult , Mutation/genetics , Microbiota
2.
Medicine (Baltimore) ; 101(49): e32194, 2022 Dec 09.
Article in English | MEDLINE | ID: mdl-36626451

ABSTRACT

The gut microbiome has been increasingly suggested as an underlying cause of various human diseases. In this study, we hypothesized that the gut microbiomes of patients with familial adenomatous polyposis (FAP) are different from those of healthy people and attempted to identify the associations between gut microbiome characteristics and FAP. We collected fecal samples from patients with FAP and healthy volunteers and evaluated the diversity, composition, and distribution of the gut microbiome between the 2 groups via 16S rRNA-based taxonomic profiling of the fecal samples. Fecal samples were collected from 10 patients with FAP (4 men and 6 women, mean age 39.2 ±â€…13.8 years) and 10 healthy volunteers (4 men and 6 women, mean age 40.9 ±â€…9.8 years). The microbial richness in patients with FAP was significantly lower than that in healthy people. Regarding microbial composition, the Firmicutes/Bacteroidetes ratio in patients with FAP was higher than that in healthy people, especially in those with a lower proportion of Bacteroidetes and a higher proportion of Proteobacteria. We also found 7 specific abundant strains in fecal samples of patients with FAP. Patients with FAP had different Firmicutes/Bacteroidetes ratios and Proteobacteria abundance compared to healthy people and showed the presence of specific bacteria. These findings suggest a promising role of the gut microbiome in patients with FAP, although further studies are needed.


Subject(s)
Adenomatous Polyposis Coli , Gastrointestinal Microbiome , Adult , Female , Humans , Male , Middle Aged , Adenomatous Polyposis Coli/microbiology , Bacteria/genetics , Bacteria/isolation & purification , Bacteroidetes/genetics , Bacteroidetes/isolation & purification , Feces/microbiology , Firmicutes/genetics , Firmicutes/isolation & purification , Proteobacteria/genetics , Proteobacteria/isolation & purification , RNA, Ribosomal, 16S/genetics , Healthy Volunteers
3.
Hum Genet ; 140(5): 725-746, 2021 May.
Article in English | MEDLINE | ID: mdl-33180176

ABSTRACT

Metagenomic studies using next-generation sequencing technologies have revealed rich human intestinal microbiome, which likely influence host immunity and health conditions including cancer. Evidence indicates a biological link between altered microbiome and cancers in the digestive system. Escherichia coli and Bacteroides fragilis have been found to be enriched in colorectal mucosal tissues from patients with familial adenomatous polyposis that is caused by germline APC mutations. In addition, recent studies have found enrichment of certain oral bacteria, viruses, and fungi in tumor tissue and fecal specimens from patients with gastrointestinal cancer. An integrative approach is required to elucidate the role of microorganisms in the pathogenic process of gastrointestinal cancers, which develop through the accumulation of somatic genetic and epigenetic alterations in neoplastic cells, influenced by host genetic variations, immunity, microbiome, and environmental exposures. The transdisciplinary field of molecular pathological epidemiology (MPE) offers research frameworks to link germline genetics and environmental factors (including diet, lifestyle, and pharmacological factors) to pathologic phenotypes. The integration of microbiology into the MPE model (microbiology-MPE) can contribute to better understanding of the interactive role of environment, tumor cells, immune cells, and microbiome in various diseases. We review major clinical and experimental studies on the microbiome, and describe emerging evidence from the microbiology-MPE research in gastrointestinal cancers. Together with basic experimental research, this new research paradigm can help us to develop new prevention and treatment strategies for gastrointestinal cancers through targeting of the microbiome.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Gastrointestinal Microbiome/physiology , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/microbiology , Intestinal Mucosa/microbiology , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/microbiology , Bacteroides fragilis/growth & development , Escherichia coli/growth & development , Gastrointestinal Neoplasms/epidemiology , Humans , Molecular Epidemiology
4.
Cell Host Microbe ; 27(4): 659-670.e5, 2020 04 08.
Article in English | MEDLINE | ID: mdl-32101703

ABSTRACT

Secondary bile acids (SBAs) are derived from primary bile acids (PBAs) in a process reliant on biosynthetic capabilities possessed by few microbes. To evaluate the role of BAs in intestinal inflammation, we performed metabolomic, microbiome, metagenomic, and transcriptomic profiling of stool from ileal pouches (surgically created resevoirs) in colectomy-treated patients with ulcerative colitis (UC) versus controls (familial adenomatous polyposis [FAP]). We show that relative to FAP, UC pouches have reduced levels of lithocholic acid and deoxycholic acid (normally the most abundant gut SBAs), genes required to convert PBAs to SBAs, and Ruminococcaceae (one of few taxa known to include SBA-producing bacteria). In three murine colitis models, SBA supplementation reduces intestinal inflammation. This anti-inflammatory effect is in part dependent on the TGR5 bile acid receptor. These data suggest that dysbiosis induces SBA deficiency in inflammatory-prone UC patients, which promotes a pro-inflammatory state within the intestine that may be treated by SBA restoration.


Subject(s)
Bile Acids and Salts/metabolism , Colonic Pouches/microbiology , Dysbiosis/complications , Feces/microbiology , Receptors, G-Protein-Coupled/metabolism , Adenomatous Polyposis Coli/microbiology , Animals , Bile Acids and Salts/pharmacology , Colitis/etiology , Colitis/microbiology , Disease Models, Animal , Humans , Inflammation/drug therapy , Inflammation/etiology , Intestines/drug effects , Intestines/pathology , Metagenome , Mice , Microbiota , Receptors, G-Protein-Coupled/drug effects , Ruminococcus/isolation & purification , Transcriptome
5.
World J Gastroenterol ; 25(36): 5543-5558, 2019 Sep 28.
Article in English | MEDLINE | ID: mdl-31576099

ABSTRACT

BACKGROUND: An altered (dysbiosis) and unhealthy status of the gut microbiota is usually responsible for a reduction of short chain fatty acids (SCFAs) concentration. SCFAs obtained from the carbohydrate fermentation processes are crucial in maintaining gut homeostasis and their determination in stool samples could provide a faster, reliable and cheaper method to highlight the presence of an intestinal dysbiosis and a biomarker for various gut diseases. We hypothesize that different intestinal diseases, such as celiac disease (CD), adenomatous polyposis (AP) and colorectal cancer (CRC) could display a particular fecal SCFAs' signature. AIM: To compare the fecal SCFAs' profiles of CD, AP, CRC patients and healthy controls, using the same analytical method. METHODS: In this cross-sectional study, we defined and compared the SCFAs' concentration in fecal samples of 9 AP, 16 CD, 19 CRC patients and 16 healthy controls (HC). The SCFAs' analysis were performed using a gas-chromatography coupled with mass spectrometry method. Data analysis was carried out using Wilcoxon rank-sum test to assess pairwise differences of SCFAs' profiles, partial least squares-discriminate analysis (PLS-DA) to determine the status membership based on distinct SCFAs' profiles, and Dirichlet regression to determine factors influencing concentration levels of SCFAs. RESULTS: We have not observed any difference in the SCFAs' amount and composition between CD and healthy control. On the contrary, the total amount of SCFAs was significantly lower in CRC patients compared to HC (P = 0.044) and CD (P = 0.005). Moreover, the SCFAs' percentage composition was different in CRC and AP compared to HC. In detail, HC displayed higher percentage of acetic acid (P value = 1.3 × 10-6) and a lower amount of butyric (P value = 0.02192), isobutyric (P value = 7.4 × 10-5), isovaleric (P value = 0.00012) and valeric (P value = 0.00014) acids compared to CRC patients. AP showed a lower abundance of acetic acid (P value = 0.00062) and higher percentages of propionic (P value = 0.00433) and isovaleric (P value = 0.00433) acids compared to HC. Moreover, AP showed higher levels of propionic acid (P value = 0.03251) and a lower level of isobutyric acid (P value = 0.00427) in comparison to CRC. The PLS-DA model demonstrated a significant separation of CRC and AP groups from HC, although some degree of overlap was observed between CRC and AP. CONCLUSION: Analysis of fecal SCFAs shows the potential to provide a non-invasive means of diagnosis to detect patients with CRC and AP, while CD patients cannot be discriminated from healthy subjects.


Subject(s)
Adenomatous Polyposis Coli/diagnosis , Celiac Disease/diagnosis , Colorectal Neoplasms/diagnosis , Dysbiosis/metabolism , Fatty Acids, Volatile/analysis , Adenomatous Polyposis Coli/metabolism , Adenomatous Polyposis Coli/microbiology , Adolescent , Adult , Aged , Aged, 80 and over , Celiac Disease/metabolism , Celiac Disease/microbiology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/microbiology , Cross-Sectional Studies , Dysbiosis/microbiology , Fatty Acids, Volatile/metabolism , Feces/chemistry , Female , Gas Chromatography-Mass Spectrometry , Gastrointestinal Microbiome/physiology , Healthy Volunteers , Humans , Male , Middle Aged , Young Adult
6.
Cell Microbiol ; 21(12): e13099, 2019 12.
Article in English | MEDLINE | ID: mdl-31414579

ABSTRACT

Several commensal and pathogenic Gram-negative bacteria produce DNA-damaging toxins that are considered bona fide carcinogenic agents. The microbiota of colorectal cancer (CRC) patients is enriched in genotoxin-producing bacteria, but their role in the pathogenesis of CRC is poorly understood. The adenomatous polyposis coli (APC) gene is mutated in familial adenomatous polyposis and in the majority of sporadic CRCs. We investigated whether the loss of APC alters the response of colonic epithelial cells to infection by Salmonella enterica, the only genotoxin-producing bacterium associated with cancer in humans. Using 2D and organotypic 3D cultures, we found that APC deficiency was associated with sustained activation of the DNA damage response, reduced capacity to repair different types of damage, including DNA breaks and oxidative damage, and failure to induce cell cycle arrest. The reduced DNA repair capacity and inability to activate adequate checkpoint responses was associated with increased genomic instability in APC-deficient cells exposed to the genotoxic bacterium. Inhibition of the checkpoint response was dependent on activation of the phosphatidylinositol 3-kinase pathway. These findings highlight the synergistic effect of the loss of APC and infection with genotoxin-producing bacteria in promoting a microenvironment conducive to malignant transformation.


Subject(s)
Adenomatous Polyposis Coli/genetics , Colon/metabolism , Epithelial Cells/metabolism , Genomic Instability/genetics , Phosphatidylinositol 3-Kinases/metabolism , Salmonella Infections/metabolism , Salmonella enterica/metabolism , Adenomatous Polyposis Coli/microbiology , Adenomatous Polyposis Coli/pathology , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Cycle Checkpoints/genetics , Cell Line , Colon/microbiology , Colon/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , DNA Damage/genetics , Epithelial Cells/microbiology , Epithelial Cells/pathology , Genes, Tumor Suppressor/physiology , Humans , Mice , Mutagens/metabolism , Salmonella Infections/genetics , Salmonella Infections/microbiology , Salmonella Infections/pathology , Signal Transduction/genetics , Tumor Microenvironment/genetics
7.
Cancer Med ; 7(5): 2003-2012, 2018 05.
Article in English | MEDLINE | ID: mdl-29624892

ABSTRACT

Recent epidemiological evidence suggests that exposure to antibiotics in early-to-middle adulthood is associated with an increased risk of colorectal adenoma. However, mechanistic studies in established preclinical cancer to examine these claims are extremely limited. Therefore, we investigated the effect of long-term exposure of an antibiotic cocktail composed of Vancomycin, Neomycin, and Streptomycin, on tumor development and progression in the ApcMin/+ mouse, an established genetic model for familial adenomatous polyposis. Clinical pathologies related to tumor development as well as intestinal and colon tissue histopathology were studied at ages 8, 12, and 16 weeks of age, which correspond to the approximate ages of development of neoplasia, gut inflammation with polyposis, and cancer progression, respectively, in this animal model. We show that the antibiotics significantly increase the severity of clinical symptoms, including effects on intestinal histology and goblet cell numbers. In addition, they promote small intestinal polyposis. Finally, metagenomic analysis of fecal samples demonstrated that antibiotic exposure is associated with a significant but nonuniform depletion of the animal's natural gut flora. Overall, these findings support the premise that long-term antibiotic exposure mediates the selected depletion of gut microbial communities and the concomitant thinning of the protective mucus layer, resulting in an increase in tumor development.


Subject(s)
Adenomatous Polyposis Coli/microbiology , Adenomatous Polyposis Coli/pathology , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacology , Gastrointestinal Microbiome/drug effects , Goblet Cells/cytology , Intestinal Mucosa/pathology , Adenomatous Polyposis Coli Protein/genetics , Animals , Colon/pathology , Disease Models, Animal , Disease Progression , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neomycin/adverse effects , Neomycin/pharmacology , Streptomycin/adverse effects , Streptomycin/pharmacology , Vancomycin/adverse effects , Vancomycin/pharmacology
8.
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
9.
Cancer Res ; 77(9): 2424-2438, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28428272

ABSTRACT

HuR is an RNA-binding protein implicated in immune homeostasis and various cancers, including colorectal cancer. HuR binding to AU-rich elements within the 3' untranslated region of mRNAs encoding oncogenes, growth factors, and various cytokines leads message stability and translation. In this study, we evaluated HuR as a small-molecule target for preventing colorectal cancer in high-risk groups such as those with familial adenomatosis polyposis (FAP) or inflammatory bowel disease (IBD). In human specimens, levels of cytoplasmic HuR were increased in colonic epithelial cells from patients with IBD, IBD-cancer, FAP-adenoma, and colorectal cancer, but not in patients with IBD-dysplasia. Intraperitoneal injection of the HuR small-molecule inhibitor MS-444 in AOM/DSS mice, a model of IBD and inflammatory colon cancer, augmented DSS-induced weight loss and increased tumor multiplicity, size, and invasiveness. MS-444 treatment also abrogated tumor cell apoptosis and depleted tumor-associated eosinophils, accompanied by a decrease in IL18 and eotaxin-1. In contrast, HuR inhibition in APCMin mice, a model of FAP and colon cancer, diminished the number of small intestinal tumors generated. In this setting, fecal microbiota, evaluated by 16S rRNA gene amplicon sequencing, shifted to a state of reduced bacterial diversity, with an increased representation of Prevotella, Akkermansia, and Lachnospiraceae Taken together, our results indicate that HuR activation is an early event in FAP-adenoma but is not present in IBD-dysplasia. Furthermore, our results offer a preclinical proof of concept for HuR inhibition as an effective means of FAP chemoprevention, with caution advised in the setting of IBD. Cancer Res; 77(9); 2424-38. ©2017 AACR.


Subject(s)
Adenomatous Polyposis Coli/genetics , Colorectal Neoplasms/genetics , ELAV-Like Protein 1/genetics , Inflammatory Bowel Diseases/genetics , Adenomatous Polyposis Coli/microbiology , Adenomatous Polyposis Coli/pathology , Animals , Apoptosis/drug effects , Carcinogenesis/genetics , Cell Proliferation/drug effects , Chemokine CCL11/genetics , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , ELAV-Like Protein 1/antagonists & inhibitors , Feces/microbiology , Furans/administration & dosage , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , HCT116 Cells , Humans , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Interleukin-18/genetics , Mice , Naphthols/administration & dosage , RAW 264.7 Cells
10.
Genet Epidemiol ; 41(3): 210-220, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28019040

ABSTRACT

High-throughput sequencing technologies have enabled large-scale studies of the role of the human microbiome in health conditions and diseases. Microbial community level association test, as a critical step to establish the connection between overall microbiome composition and an outcome of interest, has now been routinely performed in many studies. However, current microbiome association tests all focus on a single outcome. It has become increasingly common for a microbiome study to collect multiple, possibly related, outcomes to maximize the power of discovery. As these outcomes may share common mechanisms, jointly analyzing these outcomes can amplify the association signal and improve statistical power to detect potential associations. We propose the multivariate microbiome regression-based kernel association test (MMiRKAT) for testing association between multiple continuous outcomes and overall microbiome composition, where the kernel used in MMiRKAT is based on Bray-Curtis or UniFrac distance. MMiRKAT directly regresses all outcomes on the microbiome profiles via a semiparametric kernel machine regression framework, which allows for covariate adjustment and evaluates the association via a variance-component score test. Because most of the current microbiome studies have small sample sizes, a novel small-sample correction procedure is implemented in MMiRKAT to correct for the conservativeness of the association test when the sample size is small or moderate. The proposed method is assessed via simulation studies and an application to a real data set examining the association between host gene expression and mucosal microbiome composition. We demonstrate that MMiRKAT is more powerful than large sample based multivariate kernel association test, while controlling the type I error. A free implementation of MMiRKAT in R language is available at http://research.fhcrc.org/wu/en.html.


Subject(s)
Adenomatous Polyposis Coli/genetics , Genetic Association Studies , Genetic Markers/genetics , Microbiota/genetics , Models, Genetic , Polymorphism, Single Nucleotide/genetics , Adenomatous Polyposis Coli/microbiology , Case-Control Studies , Computer Simulation , High-Throughput Nucleotide Sequencing , Humans , Mucous Membrane/microbiology , Phylogeny , Sample Size
11.
Oncotarget ; 7(21): 31226-42, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27121311

ABSTRACT

Saponins derived from medicinal plants have raised considerable interest for their preventive roles in various diseases. Here, we investigated the impacts of triterpenoid saponins isolated from Gynostemma pentaphyllum (GpS) on gut microbiome, mucosal environment, and the preventive effect on tumor growth. Six-week old ApcMin/+ mice and their wild-type littermates were fed either with vehicle or GpS daily for the duration of 8 weeks. The fecal microbiome was analyzed by enterobacterial repetitive intergenic consensus (ERIC)-PCR and 16S rRNA gene pyrosequencing. Study showed that GpS treatment significantly reduced the number of intestinal polyps in a preventive mode. More importantly, GpS feeding strikingly reduced the sulfate-reducing bacteria lineage, which are known to produce hydrogen sulfide and contribute to damage the intestinal epithelium or even promote cancer progression. Meanwhile, GpS also boosted the beneficial microbes. In the gut barrier of the ApcMin/+ mice, GpS treatment increased Paneth and goblet cells, up-regulated E-cadherin and down-regulated N-cadherin. In addition, GpS decreased the pro-oncogenic ß-catenin, p-Src and the p-STAT3. Furthermore, GpS might also improve the inflamed gut epithelium of the ApcMin/+ mice by upregulating the anti-inflammatory cytokine IL-4, while downregulating pro-inflammatory cytokines TNF-α, IL-1ß and IL-18. Intriguingly, GpS markedly stimulated M2 and suppressed M1 macrophage markers, indicating that GpS altered mucosal cytokine profile in favor of the M1 to M2 macrophages switching, facilitating intestinal tissue repair. In conclusion, GpS might reverse the host's inflammatory phenotype by increasing beneficial bacteria, decreasing sulfate-reducing bacteria, and alleviating intestinal inflammatory gut environment, which might contribute to its cancer preventive effects.


Subject(s)
Adenomatous Polyposis Coli/prevention & control , Bacteria/drug effects , Gastrointestinal Microbiome/drug effects , Saponins/pharmacology , Triterpenes/pharmacology , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/microbiology , Adenomatous Polyposis Coli Protein/deficiency , Adenomatous Polyposis Coli Protein/genetics , Animals , Bacteria/classification , Bacteria/genetics , Cytokines/metabolism , Feces/microbiology , Gastrointestinal Microbiome/genetics , Gynostemma/chemistry , Intestinal Mucosa/metabolism , Intestines/drug effects , Intestines/microbiology , Mice, Knockout , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA/methods , Sulfates/metabolism
12.
PLoS One ; 10(6): e0127985, 2015.
Article in English | MEDLINE | ID: mdl-26121046

ABSTRACT

Mutation of the adenomatous polyposis coli (APC gene), an early event in the adenoma-carcinoma sequence, is present in 70-80% of sporadic human colorectal adenomas and carcinomas. To test the hypothesis that mutation of the APC gene alters microbial interactions with host intestinal mucosa prior to the development of polyposis, culture-independent methods (targeted qPCR assays and Illumina sequencing of the 16S rRNA gene V1V2 hypervariable region) were used to compare the intestinal microbial composition of 30 six-week old C57BL/6 APCMin/+ and 30 congenic wild type (WT) mice. The results demonstrate that similar to 12-14 week old APCMin/+ mice with intestinal neoplasia, 6 week old APCMin/+ mice with no detectable neoplasia, exhibit an increased relative abundance of Bacteroidetes spp in the colon. Parallel mouse RNA sequence analysis, conducted on a subset of proximal colonic RNA samples (6 APCMin/+, 6 WT) revealed 130 differentially expressed genes (DEGs, fold change ≥ 2, FDR <0.05). Hierarchical clustering of the DEGs was carried out by using 1-r dissimilarity measurement, where r stands for the Pearson correlation, and Ward minimum variance linkage, in order to reduce the number of input variables. When the cluster centroids (medians) were included along with APC genotype as input variables in a negative binomial (NB) regression model, four of seven mouse gene clusters, in addition to APC genotype, were significantly associated with the increased relative abundance of Bacteroidetes spp. Three of the four clusters include several downregulated genes encoding immunoglobulin variable regions and non-protein coding RNAs. These results support the concept that mutation of the APC gene alters colonic-microbial interactions prior to polyposis. It remains to be determined whether interventions directed at ameliorating dysbiosis in APCMin/+mice, such as through probiotics, prebiotics or antibiotics, could reduce tumor formation.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Adenomatous Polyposis Coli/microbiology , Adenomatous Polyposis Coli/pathology , Gastrointestinal Microbiome , Intestinal Mucosa/microbiology , Animals , Bacteroidetes/physiology , Base Sequence , Cluster Analysis , Female , Gene Expression Profiling , Genotype , Intestinal Mucosa/pathology , Mice, Inbred C57BL , Molecular Sequence Data , Phylogeny , Polymerase Chain Reaction , Principal Component Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Ribosomal, 16S/genetics
13.
Neoplasma ; 61(3): 283-90, 2014.
Article in English | MEDLINE | ID: mdl-24824929

ABSTRACT

Colorectal cancer mortality is one of the most common cause of cancer-related mortality. A multiple risk factors are associated with colorectal cancer, including hereditary, enviromental and inflammatory syndromes affecting the gastrointestinal tract. Familial adenomatous polyposis (FAP) is characterized by the emergence of hundreds to thousands of colorectal adenomatous polyps and FAP syndrome is caused by mutations within the adenomatous polyposis coli (APC) tumor suppressor gene. We analyzed 21 rectal bacterial subclones isolated from FAP patient 41-1 with confirmed 5bp ACAAA deletion within codons 1060-1063 for the presence of APC-like sequences in longest exon 15. The studied section was defined by primers 15Efor-15Erev, what correlates with mutation cluster region (MCR) in which the 75% of all APC germline mutations were detected. More than 90% homology was showed by sequencing and subsequent software comparison. The expression of APC-like sequences was demostrated by Western blot analysis using monoclonal and polyclonal antibodies against APC protein. To study missing link between the DNA analysis (PCR, DNA sequencing) and protein expresion experiments (Western blotting) we analyzed bacterial transcripts containing the 15Efor-15Erev sequence of APC gene by reverse transcription-PCR, what indicated that an APC gene derived fragment may be produced. We observed 97-100 % homology after computer comparison of cDNA PCR products. Our results suggest that presence of APC-like sequences in intestinal/rectal bacteria is enrichment of bacterial genetic information in which horizontal gene transfer between humans and microflora play an important role.


Subject(s)
Adenomatous Polyposis Coli/microbiology , Bacteria/genetics , Genes, APC/physiology , Rectum/microbiology , Adenomatous Polyposis Coli/genetics , Base Sequence , Gene Transfer, Horizontal , Germ-Line Mutation , Humans , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction
14.
Am J Gastroenterol ; 108(2): 216-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23381070

ABSTRACT

A large cross-sectional survey suggests an association between H. pylori gastritis and colonic neoplasms, but the results should be interpreted with caution.


Subject(s)
Adenocarcinoma/microbiology , Adenomatous Polyposis Coli/microbiology , Colonic Neoplasms/microbiology , Gastritis/complications , Gastritis/microbiology , Helicobacter Infections/complications , Helicobacter pylori , Stomach Neoplasms/complications , Female , Humans , Male
15.
Am J Gastroenterol ; 108(2): 208-15, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23208272

ABSTRACT

OBJECTIVES: It has been suggested that Helicobacter pylori (H. pylori) constitutes a risk for the development of adenomatous polyps and adenocarcinoma of the colon. Our aim was to study the association between H. pylori-positive gastritis and the occurrence of any colonic neoplasm. METHODS: From a computerized database of surgical pathology reports, we selected 156,000 subjects who underwent colonoscopy and esophago-gastro-duodenoscopy with biopsy results from both procedures. RESULTS: Compared with normal gastric mucosa, H. pylori gastritis occurred more frequently among patients with hyperplastic polyps (OR=1.24, 95% CI: 1.18-1.30), adenomatous polyps (1.52, 1.46-1.57), advanced adenomas (1.80, 1.69-1.92), villous adenomas or adenomas with high-grade dysplasia (1.97, 1.82-2.14), and adenocarcinomas (2.35, 1.98-2.80). Similarly, the strength of the association between H. pylori-positive gastritis and colonic neoplasm increased with size and number of the adenomas. The association between H. pylori gastritis and the occurrence of colonic neoplasm was similar for different locations of the large bowel. Other gastric conditions etiologically associated with H. pylori, such as intestinal metaplasia, adenoma, lymphoma, and adenocarcinoma, were also significantly associated with an increased risk of colonic neoplasm. CONCLUSIONS: Various forms of gastritis related to H. pylori infection confer an increased risk for colonic neoplasm. In the past, when H. pylori infection was more prevalent, its attributable risk to the occurrence of colorectal neoplasm may have been quite substantial.


Subject(s)
Adenocarcinoma/microbiology , Adenomatous Polyposis Coli/microbiology , Colonic Neoplasms/microbiology , Gastritis/complications , Gastritis/microbiology , Helicobacter Infections/complications , Helicobacter pylori , Stomach Neoplasms/complications , Adenocarcinoma/complications , Adenocarcinoma/pathology , Adenocarcinoma/prevention & control , Adenoma/complications , Adenoma/microbiology , Adenomatous Polyposis Coli/pathology , Adenomatous Polyposis Coli/prevention & control , Adult , Aged , Colonic Neoplasms/pathology , Colonic Neoplasms/prevention & control , Databases, Factual , Female , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Helicobacter Infections/microbiology , Humans , Lymphoma/complications , Lymphoma/microbiology , Male , Metaplasia/microbiology , Middle Aged , Multivariate Analysis , Neoplasm Grading , Public Health/trends , Retrospective Studies , Risk Factors
16.
Med Sci Monit ; 18(8): CR486-492, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22847197

ABSTRACT

BACKGROUND: Familial adenomatous polyposis (FAP) is a hereditary disease induced by germ-line mutations in the tumor suppressor APC gene. These initiate the early stages of the adenoma-carcinoma sequence in familial, but also in sporadic (in 80% to 90%), colon tumorigenesis. We found the presence of APC-like sequences in bacteria of FAP patients. MATERIAL/METHODS: We analyzed bacteria isolated from FAP patients' rectal swabs. Total bacterial DNA was isolated and analyzed for detection of APC-like sequences using PCR. We also tested DNA homology rate and APC-like protein production. RESULTS: We collected blood samples and rectal swabs from patients with confirmed diagnosis of FAP. They were analyzed for presence of sections from exon 15 of the APC gene. Most positive results were found in sections located exactly in the area called the MCR (mutation cluster region), where the highest frequency of APC gene mutations were identified. By sequencing PCR products from bacteria in section F-G together with a patient's DNA sample and human APC gene, we found a more than 90% DNA homology rate. We also confirmed production of APC-like protein using Western blotting. CONCLUSIONS: Our results suggested two hypotheses. The APC-like protein might have same function as a truncated APC product, which is synthesized in most cases of mutations of APC gene in the MCR region in colorectal cancer cells. Alternatively, we can consider the possible existence of horizontal transfer of genetic information between eukaryotic and prokaryotic cells. Our study can be considered as a pilot project. For confirmation of our hypotheses, further research is needed.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/microbiology , Bacteria/genetics , Intestines/microbiology , Base Sequence , Blotting, Western , DNA, Bacterial/genetics , DNA, Bacterial/isolation & purification , Exons/genetics , Humans , Intestines/pathology , Molecular Sequence Data , Polymerase Chain Reaction
17.
Inflamm Bowel Dis ; 18(5): 925-34, 2012 May.
Article in English | MEDLINE | ID: mdl-22114001

ABSTRACT

BACKGROUND: Chronic pouchitis is an important long-term complication following ileal pouch-anal anastomosis for ulcerative colitis. Antibiotic administration reduces symptoms of pouchitis, indicating that bacteria have a role in pathogenesis. The aim of the research was to investigate the bacterial content of pouches using nucleic acid-based methods. METHODS: Stool microbiota of 17 patients with normal pouches (NP), 17 patients with pouchitis (CP) utilizing samples collected from each patient when antibiotic-treated (CP-on, asymptomatic) and when untreated (CP-off, symptomatic), and 14 familial adenomatous polyposis (FAP) patients were analyzed by high-throughput sequencing, fluorescence in situ hybridization technologies, and quantitative polymerase chain reaction (qPCR). RESULTS: Fluorescence in situ hybridization analysis revealed an expanded phylogenetic gap in NP and CP-off patients relative to FAP. Antibiotic treatment reduced the gap in CP stool. The phylogenetic gap of CP-off patients was due to members of the bacterial families Caulobacteriaceae, Sphingomonadaceae, Comamonadaceae, Peptostreptococcaceae, and Clostridiaceae. There was a greater diversity of phylotypes of Clostridiaceae in CP-off subjects. The phylogenetic gap of NP stool was enriched by Ruminococcaceae and Bifidobacteriaceae. CP stool microbiota had reduced diversity relative to NP and FAP stool due largely to a reduction in Lachnospiraceae/Insertae Sedis XIV/clostridial cluster IV groups. CONCLUSIONS: Bacterial groups within the expanded phylogenetic gap of pouch patients may have roles in the pathogenesis of pouchitis. Further research concerning the physiology of cultured members of these groups will be necessary to explain their specific roles. Members of the Lachnospiraceae, Incertae Sedis XIV, and clostridial cluster IV could be useful biomarkers of pouch health.


Subject(s)
Adenomatous Polyposis Coli/microbiology , Bacteria/pathogenicity , Colitis, Ulcerative/microbiology , Colonic Pouches/microbiology , Feces/microbiology , Pouchitis/microbiology , Adenomatous Polyposis Coli/pathology , Adolescent , Adult , Child , Child, Preschool , Chronic Disease , Colitis, Ulcerative/pathology , Colonic Pouches/pathology , DNA/genetics , Female , High-Throughput Nucleotide Sequencing , Humans , In Situ Hybridization, Fluorescence , Infant , Infant, Newborn , Male , Pouchitis/pathology , Real-Time Polymerase Chain Reaction , Young Adult
18.
Inflamm Bowel Dis ; 17(5): 1092-100, 2011 May.
Article in English | MEDLINE | ID: mdl-20845425

ABSTRACT

BACKGROUND: Pouchitis occurs in up to 50% of patients with ulcerative colitis (UC) undergoing ileal pouch anal anastomosis (IPAA). Pouchitis rarely occurs in patients with familial adenomatous polyposis (FAP) who undergo IPAA. Our aim was to compare mucosal and luminal flora in patients with UC-associated pouchitis (UCP), healthy UC pouches (HUC), and healthy FAP pouches (FAP). METHODS: Nineteen patients were enrolled in this cross-sectional study (nine UCP, three HUC, seven FAP). Patients with active pouchitis were identified using the Pouchitis Disease Activity Index (PDAI). Ileal pouch mucosal biopsies and fecal samples were analyzed with a 16S rDNA-based terminal restriction fragment length polymorphism (TRFLP) approach. Pooled fecal DNA from four UCP and four FAP pouches were sequenced for further speciation. RESULTS: TRFLP data revealed statistically significant differences in the mucosal and fecal microbiota between each group of patients. UCP samples exhibited significantly more TRFLP peaks matching Clostridium and Eubacterium genera compared to HUC and FAP pouches and fewer peaks matching Lactobacillus and Streptococcus genera compared to FAP. DNA Sanger sequencing of a subset of luminal samples revealed UCP having more identifiable sequences of Firmicutes (51.2% versus 21.2%) and Verrucomicrobia (20.2% versus 3.2%), and fewer Bacteroidetes (17.9% versus 60.5%) and Proteobacteria (9.8% versus 14.7%) compared to FAP. CONCLUSIONS: The pouch microbial environment appears to be distinctly different in the settings of UC pouchitis, healthy UC, and FAP. These findings suggest that a dysbiosis may exist in pouchitis which may be central to understanding the disease.


Subject(s)
Adenomatous Polyposis Coli/microbiology , Adenomatous Polyposis Coli/surgery , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/surgery , Pouchitis/microbiology , Actinobacteria/genetics , Actinobacteria/isolation & purification , Adult , Bacteroides/genetics , Bacteroides/isolation & purification , Biopsy , DNA, Bacterial/analysis , Feces/microbiology , Female , Fusobacteria/genetics , Fusobacteria/isolation & purification , Gastrointestinal Tract/microbiology , Humans , Male , Metagenome , Middle Aged , Postoperative Complications/microbiology , Postoperative Complications/pathology , Pouchitis/pathology , Proteobacteria/genetics , Proteobacteria/isolation & purification , Young Adult
19.
Ann Surg ; 252(1): 90-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20562611

ABSTRACT

OBJECTIVE: To identify, compare, and contrast the microbiota in patients with and without pouchitis after restorative proctocolectomy (RPC) for ulcerative colitis (UC) and familial adenomatous polyposis (FAP). SUMMARY BACKGROUND DATA: Pouchitis is the most common complication following RPC. An abnormal host-microbial interaction has been implicated. We investigated the pouch microbiota in patients with and without pouchitis undergoing restorative proctocolectomy for UC and FAP. METHODS: Mucosal pouch biopsies, taken from 16 UC (pouchitis 8) and 8 FAP (pouchitis 3) patients were analyzed to the species (or phylotype) level by cloning and sequencing of 3184 full-length bacterial 16S rRNA genes. RESULTS: There was a significant increase in Proteobacteria (P = 0.019) and a significant decrease in Bacteroidetes (P = 0.001) and Faecalibacterium prausnitzii (P = 0.029) in the total UC compared with the total FAP cohort, but only limited differences were found between the UC nonpouchitis and pouchitis groups and the FAP pouchitis and nonpouchitis groups. Bacterial diversity in the FAP nonpouchitis group was significantly greater than in UC nonpouchitis (P = 0.019) and significantly greater in UC nonpouchitis compared with UC pouchitis (P = 0.009). No individual species or phylotype specifically associated with either UC or FAP pouchitis was found. CONCLUSIONS: UC pouch patients have a different, less diverse, gut microbiota than FAP patients. A further reduction in bacterial diversity but no significant dysbiosis occurs in those with pouchitis. The study suggests that a dysbiosis occurs in the ileal pouch of UC RPC patients which predisposes to, but may not directly cause, pouchitis.


Subject(s)
Pouchitis/microbiology , RNA, Ribosomal, 16S/genetics , Adenomatous Polyposis Coli/microbiology , Adenomatous Polyposis Coli/surgery , Adult , Bacteroidetes/genetics , Bacteroidetes/isolation & purification , Base Sequence , Biopsy , Cloning, Molecular , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/surgery , Colonic Pouches/microbiology , DNA, Bacterial/analysis , Humans , Male , Middle Aged , Molecular Sequence Data , Proctocolectomy, Restorative , Proteobacteria/genetics , Proteobacteria/isolation & purification
20.
Gastroenterology ; 139(2): 574-85, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20416302

ABSTRACT

BACKGROUND & AIMS: Commensal bacteria can activate signaling by the Toll-like and interleukin-1 receptors (TLR and IL-1R) to mediate pathogenesis of inflammatory bowel diseases and colitis-associated cancer. We investigated the role of the single immunoglobulin IL-1 receptor-related (SIGIRR) molecule, a negative regulator of TLR and IL-1R signaling, as a tumor suppressor to determine whether SIGIRR controls cell-cycle progression, genetic instability, and colon tumor initiation by modulating commensal TLR signaling in the gastrointestinal tract. METHODS: We analyzed adenomatous polyposis coli (Apc)min/+/Sigirr-/- mice for polyps, microadenomas, and anaphase bridge index. Commensal bacteria were depleted from mice with antibiotics. Akt, mammalian target of rapamycin (mTOR), and beta-catenin pathways were examined by immunoblotting and immunohistochemistry. Loss of heterozygosity of Apc and expression of cytokines and proinflammatory mediators were measured by nonquantitative or quantitative polymerase chain reaction. RESULTS: Apcmin/+/Sigirr-/- mice had increased loss of heterozygosity of Apc and microadenoma formation, resulting in spontaneous colonic polyposis, compared with Apcmin/+/Sigirr+/+ mice. The increased colonic tumorigenesis that occurred in the Apcmin/+/Sigirr-/- mice depended on the presence of commensal bacteria in the gastrointestinal tract. Cell proliferation and chromosomal instability increased in colon crypt cells of the Apcmin/+/Sigirr-/- mice. Akt, mTOR, and their substrates were hyperactivated in colon epithelium of Apcmin/+/Sigirr-/- mice in response to TLR or IL-1R ligands. Inhibition of the mTOR pathway by rapamycin reduced formation of microadenomas and polyps in the Apcmin/+/Sigirr-/- mice. CONCLUSIONS: SIGIRR acts as a tumor suppressor in the colon by inhibiting TLR-induced, mTOR-mediated cell-cycle progression and genetic instability.


Subject(s)
Adenomatous Polyposis Coli/metabolism , Adenomatous Polyposis Coli/prevention & control , Colon/metabolism , Genes, APC , Genes, Tumor Suppressor , Receptors, Interleukin-1/deficiency , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/microbiology , Adenomatous Polyposis Coli/pathology , Animals , Blotting, Western , Cell Proliferation , Chromosomal Instability , Colon/drug effects , Colon/microbiology , Colon/pathology , Cytokines/metabolism , Immunohistochemistry , Inflammation Mediators/metabolism , Intestinal Mucosa/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Loss of Heterozygosity , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Interleukin-1/genetics , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Toll-Like Receptors/metabolism , Tumor Burden , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL