Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 483
1.
Clin Exp Metastasis ; 38(6): 495-510, 2021 12.
Article En | MEDLINE | ID: mdl-34748126

Colorectal carcinoma is the third most common cancer in developed countries and the second leading cause of cancer-related mortality. Interest in the influence of the intestinal microbiota on CRC emerged rapidly in the past few years, and the close presence of microbiota to the tumour mass creates a unique microenvironment in CRC. The gastrointestinal microbiota secrete factors that can contribute to CRC metastasis by influencing, for example, epithelial-to-mesenchymal transition. Although the role of EMT in metastasis is well-studied, mechanisms by which gastrointestinal microbiota contribute to the progression of CRC remain poorly understood. In this review, we will explore bacterial factors that contribute to the migration and invasion of colorectal carcinoma and the mechanisms involved. Bacteria involved in the induction of metastasis in primary CRC include Fusobacterium nucleatum, Enterococcus faecalis, enterotoxigenic Bacteroides fragilis, Escherichia coli and Salmonella enterica. Examples of prominent bacterial factors secreted by these bacteria include Fusobacterium adhesin A and Bacteroides fragilis Toxin. Most of these factors induce EMT-like properties in carcinoma cells and, as such, contribute to disease progression by affecting cell-cell adhesion, breakdown of the extracellular matrix and reorganisation of the cytoskeleton. It is of utmost importance to elucidate how bacterial factors promote CRC recurrence and metastasis to increase patient survival. So far, mainly animal models have been used to demonstrate this interplay between the host and microbiota. More human-based models are needed to study the mechanisms that promote migration and invasion and mimic the progression and recurrence of CRC.


Bacteria/metabolism , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Carcinoma/microbiology , Cell Movement , Colorectal Neoplasms/microbiology , Gastrointestinal Microbiome , Animals , Bacteria/pathogenicity , Carcinoma/metabolism , Carcinoma/secondary , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Dysbiosis , Host-Pathogen Interactions , Humans , Neoplasm Invasiveness , Signal Transduction
2.
BMC Microbiol ; 21(1): 310, 2021 11 09.
Article En | MEDLINE | ID: mdl-34753420

AIMS: To analyze changes in oropharynx microbiota composition after receiving induced chemotherapy followed by surgery for hypopharyngeal squamous cell carcinoma (HPSCC) patients. METHODS: Clinical data and swab samples of 38 HPSCC patients (HPSCC group) and 30 patients with benign disease (control group, CG) were enrolled in the study. HPSCC group was stratified into two groups: induced chemotherapy group (IC) of 10 patients and non-induced chemotherapy group (nIC) of 28 patients. The microbiota from oropharyngeal membrane was analyzed through 16S rRNA sequencing. RESULTS: Alpha-diversity (Shannon and Ace indexes) and weighted UniFrac based beta-diversity severely decreased in the HPSCC group when compared with CG. In pre-operative comparisons, PCoA and NMDS analyses showed microbial structures in the IC group were more similar to CG than nIC. Both IC group and nIC group yielded significantly diverse post-operative communities in contrast to their pre-operative counterparts, evident by the decrease in genera Veillonella and Fusobacterium and increase in genera Streptococcus and Gemella. Given that post-operative oropharynx microbiota showed no difference between IC and nIC groups, the IC group showed less accumulation in anaerobic communities. The abundance of genera Fusobacterium, Parvimonas, Actinomyces were enhanced in the advanced stages (III/IV). CONCLUSIONS: Oropharynx microbiota in the HPSCC group presents dysbiosis with low diversity and abundance. Induced chemotherapy is beneficial in adjusting the oropharynx microbial environment leading to fewer amounts of anaerobe accumulation after operation. Higher amounts of Fusobacterium in advanced stages (III/IV) may influence the progression of HPSCC.


Bacteria/isolation & purification , Carcinoma/microbiology , Microbiota , Oropharynx/microbiology , Pharyngeal Neoplasms/microbiology , Adult , Aged , Antineoplastic Agents/administration & dosage , Bacteria/classification , Bacteria/genetics , Carcinoma/drug therapy , Carcinoma/surgery , Female , Humans , Male , Middle Aged , Pharyngeal Neoplasms/drug therapy , Pharyngeal Neoplasms/surgery , Phylogeny
3.
Int J Biol Sci ; 17(13): 3646-3658, 2021.
Article En | MEDLINE | ID: mdl-34512172

Recently, the impact of microorganisms on tumor growth and metastasis has attracted great attention. The pathogenesis and progression of lung cancer are related to an increase in respiratory bacterial load as well as changes in the bacterial community because the microbiota affects tumors in many ways, including canceration, metastasis, angiogenesis, and treatment. The microbiota may increase tumor susceptibility by altering metabolism and immune responses, promoting inflammation, and increasing toxic effects. The microbiota can regulate tumor metastasis by altering multiple cell signaling pathways and participate in tumor angiogenesis through vascular endothelial growth factors (VEGF), endothelial cells (ECs), inflammatory factors and inflammatory cells. Tumor angiogenesis not only maintains tumor growth at the primary site but also promotes tumor metastasis and invasion. Therefore, angiogenesis is an important mediator of the interaction between microorganisms and tumors. The microbiota also plays a part in antitumor therapy. Alteration of the microbiota caused by antibiotics can regulate tumor growth and metastasis. Moreover, the microbiota also influences the efficacy and toxicity of tumor immunotherapy and chemotherapy. Finally, the effects of air pollution, a risk factor for lung cancer, on microorganisms and the possible role of respiratory microorganisms in the effects of air pollution on lung cancer are discussed.


Carcinoma/microbiology , Lung Neoplasms/microbiology , Microbiota , Respiratory System/microbiology , Air Pollution/adverse effects , Animals , Humans
4.
Theranostics ; 11(10): 4945-4956, 2021.
Article En | MEDLINE | ID: mdl-33754037

Background: Bacterial infection is associated with gastric carcinogenesis. However, the relationship between nonbacterial components and gastric cancer (GC) has not been fully explored. We aimed to characterize the fungal microbiome in GC. Methods: We performed ITS rDNA gene analysis in cancer lesions and adjacent noncancerous tissues of 45 GC cases from Shenyang, China. Obtaining the OTUs and combining effective grouping, we carried out species identifications, alpha and beta diversity analyses, and FUNGuild functional annotation. Moreover, differences were compared and tested between groups to better investigate the composition and ecology of fungi associated with GC and find fungal indicators. Results: We observed significant gastric fungal imbalance in GC. Principal component analysis revealed separate clusters for the GC and control groups, and Venn diagram analysis indicated that the GC group showed a lower OTU abundance than the control. At the genus level, the abundances of 15 fungal biomarkers distinguished the GC group from the control, of which Candida (p = 0.000246) and Alternaria (p = 0.00341) were enriched in GC, while Saitozyma (p = 0.002324) and Thermomyces (p = 0.009158) were decreased. Combining the results of Welch's t test and Wilcoxon rank sum test, Candida albicans (C. albicans) was significantly elevated in GC. The species richness Krona pie chart further revealed that C. albicans occupied 22% and classified GC from the control with an area under the receiver operating curve (AUC) of 0.743. Random forest analysis also confirmed that C. albicans could serve as a biomarker with a certain degree of accuracy. Moreover, compared with that of the control, the alpha diversity index was significantly reduced in the GC group. The Jaccard distance index and the Bray abundance index of the PCoA clarified separate clusters between the GC and control groups at the species level (p = 0.00051). Adonis (PERMANOVA) analysis and ANOVA showed that there were significant differences in fungal structure among groups (p = 0.001). Finally, FUNGuild functional classification predicted that saprotrophs were the most abundant taxa in the GC group. Conclusions: This study revealed GC-associated mycobiome imbalance characterized by an altered fungal composition and ecology and demonstrated that C. albicans can be a fungal biomarker for GC. With the significant increase of C. albicans in GC, the abundance of Fusicolla acetilerea, Arcopilus aureus, Fusicolla aquaeductuum were increased, while Candida glabrata, Aspergillus montevidensis, Saitozyma podzolica and Penicillium arenicola were obviously decreased. In addition, C. albicans may mediate GC by reducing the diversity and richness of fungi in the stomach, contributing to the pathogenesis of GC.


Candida albicans , Carcinogenesis , Carcinoma/microbiology , Gastrointestinal Microbiome/genetics , Mycobiome/genetics , Stomach Neoplasms/microbiology , Aged , Aspergillus , Basidiomycota , Candida glabrata , Carcinoma/pathology , China , DNA, Ribosomal/genetics , Female , Humans , Hypocreales , Male , Metagenomics , Middle Aged , Penicillium , Sordariales , Stomach/microbiology , Stomach Neoplasms/pathology
5.
Nutrients ; 13(2)2021 Feb 15.
Article En | MEDLINE | ID: mdl-33671968

Loop ileostomy closure after colorectal surgery is often associated with Postoperative ileus, with an incidence between 13-20%. The aim of this study is to evaluate the efficacy and safety of preoperative stimulation of the efferent loop with probiotics prior to ileostomy closure in patients operated on for colorectal carcinoma. For this, a prospective, randomized, double-blind, controlled study is designed. All patients who underwent surgery for colorectal carcinoma with loop ileostomy were included. Randomized and divided into two groups, 34 cases and 35 controls were included in the study. Postoperative ileus, the need for nasogastric tube insertion, the time required to begin tolerating a diet, restoration of bowel function, and duration of hospital stay were evaluated. The incidence of Postoperative ileus was similar in both groups, 9/34 patients stimulated with probiotics and 10/35 in the control group (CG) with a p = 0.192. The comparative analysis showed a direct relationship between Postoperative ileus after oncological surgery and Postoperative ileus after reconstruction surgery, independently of stimulation. Postoperative ileus after closure ileostomy is independent of stimulation of the ileostomy with probiotics through the efferent loop. There seem to be a relationship between Postoperative ileus after reconstruction and the previous existence of Postoperative ileus after colorectal cancer surgery.


Ileostomy/adverse effects , Ileus/prevention & control , Intestinal Diseases/prevention & control , Postoperative Complications/prevention & control , Preoperative Care/methods , Probiotics/administration & dosage , Aged , Aged, 80 and over , Carcinoma/microbiology , Carcinoma/surgery , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/surgery , Double-Blind Method , Female , Humans , Ileostomy/methods , Ileus/epidemiology , Ileus/etiology , Incidence , Intestinal Diseases/epidemiology , Intestinal Diseases/etiology , Length of Stay , Male , Middle Aged , Postoperative Complications/epidemiology , Postoperative Complications/etiology , Prospective Studies , Treatment Outcome
6.
Infect Genet Evol ; 84: 104477, 2020 10.
Article En | MEDLINE | ID: mdl-32736040

It is known that Helicobacter pylori is the main cause of peptic ulceration and gastric cancer. However, there is a lack of information on whether H. pylori strains may differ in gastric cancer histological subtypes. This study aimed to investigate different H. pylori strains considering six cag Pathogenicity Island - cagPAI genes (cagA, cagE, cagG, cagM, cagT, and virb11), and vacuolating cytotoxin - vacA alleles, and their relation to gastric cancer histologic subtypes. For this purpose, tumor samples from 285 patients with gastric carcinoma were used. H. pylori infection and genotypes were determined by polymerase chain reaction (PCR). H. pylori was detected in 93.9% of gastric tumors. For comparative analyzes between histopathological subtypes considering H. pylori cagPAI genes the strains were grouped according to the vacA s1/s2 alleles. In the vacAs1 group, the strains cagA(-)cagE(+), cagA(+)cagE(+)cagG(+), cagA(+)cagM(+), or only cagE(+) strains were more frequent in the intestinal subtype (P = .009; P = .024; P = .046, respectively). In contrast, cagM(+)cagG(+)cagA(-) and cagE(-) were associated with diffuse tumors (P = .036), highlighting the presence of cagE in the development of intestinal tumors, and the presence of cagG and absence of cagE in diffuse tumors. Furthermore, WEKA software and Decision Tree (CART) analyses confirmed these findings, in which cagE presence was associated with intestinal tumors, and cagE absence and cagG(+) with diffuse tumors. In conclusion our results showed that vacAs1 (cagG + cagM) strains, mainly cagG positive with cagE absence, were relevant in the studied population for the diffuse outcome, while the presence of cagE was relevant for the intestinal outcome. These findings suggest the relevance of these H. pylori genes as potential markers for gastric cancer histological outcomes.


Bacterial Proteins/metabolism , Biomarkers , Helicobacter Infections/complications , Helicobacter pylori/metabolism , Intestinal Neoplasms/microbiology , Stomach Neoplasms/microbiology , Adult , Aged , Aged, 80 and over , Bacterial Proteins/genetics , Carcinoma/microbiology , Female , Genes, Bacterial , Helicobacter Infections/microbiology , Humans , Intestinal Neoplasms/complications , Intestinal Neoplasms/pathology , Male , Middle Aged , Stomach Neoplasms/complications , Stomach Neoplasms/pathology , Young Adult
7.
Infect Immun ; 88(5)2020 04 20.
Article En | MEDLINE | ID: mdl-32041789

Programmed death-ligand 1 (PD-L1/B7-H1) serves as a cosignaling molecule in cell-mediated immune responses and contributes to chronicity of inflammation and the escape of tumor cells from immunosurveillance. Here, we investigated the molecular mechanisms leading to PD-L1 upregulation in human oral carcinoma cells and in primary human gingival keratinocytes in response to infection with Porphyromonas gingivalis (P. gingivalis), a keystone pathogen for the development of periodontitis. The bacterial cell wall component peptidoglycan uses bacterial outer membrane vesicles to be taken up by cells. Internalized peptidoglycan triggers cytosolic receptors to induce PD-L1 expression in a myeloid differentiation primary response 88 (Myd88)-independent and receptor-interacting serine/threonine-protein kinase 2 (RIP2)-dependent fashion. Interference with the kinase activity of RIP2 or mitogen-activated protein (MAP) kinases interferes with inducible PD-L1 expression.


B7-H1 Antigen/metabolism , Bacteroidaceae Infections/metabolism , Carcinoma/metabolism , Cell Wall/metabolism , Mouth Neoplasms/metabolism , Porphyromonas gingivalis/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Bacteroidaceae Infections/microbiology , Carcinoma/microbiology , Cell Line, Tumor , Gingiva/metabolism , Gingiva/microbiology , Humans , Keratinocytes/metabolism , Keratinocytes/microbiology , Mitogen-Activated Protein Kinases/metabolism , Mouth Neoplasms/microbiology , Periodontitis/metabolism , Periodontitis/microbiology , Up-Regulation/physiology
8.
Anal Cell Pathol (Amst) ; 2019: 3085181, 2019.
Article En | MEDLINE | ID: mdl-32082967

Chronic inflammation induced by Helicobacter pylori (H. pylori) infection plays a major role in development of gastric cancer. However, recent findings suggested that progression of inflammation and neoplastic transformation in H. pylori infection are more complex than previously believed and could involve different factors that modulate gastric microenvironment and influence host-pathogen interaction. Among these factors, gastric myenteric plexus and its potential adaptive changes in H. pylori infection received little attention. This study is aimed at identifying the impact of H. pylori-associated gastritis on number and morphology of nerve cells in the stomach. The distribution of density, inflammation, and programmed cell death in neurons was immunohistochemically assessed in full-thickness archival tissue samples obtained from 40 patients with H. pylori infection who underwent surgery for gastric cancer and were compared with findings on samples collected from 40 age- and sex-matched subjects without bacteria. Overall, significant differences were noted between H. pylori-positive and H. pylori-negative patients. The analysis of tissue specimens obtained from those with infection revealed higher density and larger surface of the myenteric nervous plexus, as well as a significant increase in the number of gastric neuronal cell bodies and glial cells compared to controls. A predominant CD3-immunoreactive T cell infiltrate confined to the myenteric plexus was observed in infected subjects. The presence of mature B lymphocytes, plasma cells, and eosinophils was also noted, but to a lesser extent, within the ganglia. Myenteric ganglionitis was associated with degeneration and neuronal loss. Our results represent the first histopathological evidence supporting the hypothesis that H. pylori-induced gastric inflammation may induce morphological changes in myenteric gastric ganglia. These findings could help gain understanding of some still unclear aspects of pathogenesis of H. pylori infection, with the possibility of having broader implications for gastric cancer progression.


Carcinoma/pathology , Ganglia/pathology , Helicobacter Infections/pathology , Helicobacter pylori/pathogenicity , Myenteric Plexus/cytology , Neurons/cytology , Stomach Neoplasms/pathology , Aged , Apoptosis , B-Lymphocytes/cytology , CD3 Complex/metabolism , Carcinoma/microbiology , Cohort Studies , Eosinophils/cytology , Female , Ganglia/cytology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Humans , Immunohistochemistry , Inflammation/pathology , Male , Middle Aged , Myenteric Plexus/microbiology , Myenteric Plexus/pathology , Neurons/pathology , Retrospective Studies , Stomach Neoplasms/microbiology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
9.
Int J Mol Sci ; 19(12)2018 Dec 18.
Article En | MEDLINE | ID: mdl-30567376

Gastric cancer has reduced prevalence, but poor prognoses. To improve treatment, better knowledge of carcinogenesis and cells of origin should be sought. Stomach cancers are typically localized to one of the three mucosae; cardial, oxyntic and antral. Moreover, not only the stem cell, but the ECL cell may proliferate and give rise to tumours. According to Laurén, the classification of gastric carcinomas seems to reflect biological important differences and possible different cell of origin since the two subtypes, intestinal and diffuse, do not transform into the other and show different epidemiology. The stem cell probably gives rise to the intestinal type, whereas the ECL cell may be important in the diffuse type. Elevation of gastrin may be the carcinogenic factor for Helicobacter pylori as well as the recently described increased risk of gastric cancer due to proton pump inhibitor treatment. Therefore, it is essential to determine the role of the gastrin target cell, the ECL cell, in gastric carcinogenesis. Clinical trials with gastrin antagonists could improve prognoses in those with gastrin receptor positive tumours. However, further studies on gastric carcinomas applying relative available methods and with the highest sensitivity are warranted to improve our knowledge of gastric carcinogenesis.


Carcinogenesis/genetics , Carcinoma/physiopathology , Stomach Neoplasms/physiopathology , Carcinoma/classification , Carcinoma/etiology , Carcinoma/microbiology , Cell Proliferation/genetics , Gastric Mucosa/microbiology , Gastric Mucosa/physiopathology , Gastrins/genetics , Helicobacter Infections/complications , Helicobacter Infections/genetics , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Humans , Stomach Neoplasms/classification , Stomach Neoplasms/etiology , Stomach Neoplasms/microbiology
10.
Article En | MEDLINE | ID: mdl-29904624

Mounting evidence indicates that microbiome plays an important role in the development and progression of cancer. The dogma that urine in healthy individuals must be sterile has been overturned. Dysbiosis of the urinary microbiome has been revealed responsible for various urological disorders, including prostate cancer. The link between chronic inflammation, microbiome and solid tumors has been established for various neoplastic diseases. However, a detailed and comprehensive analysis of urinary microenvironment of bladder cancer has not been yet reported. We performed this study to characterize the potential urinary microbial community possibly associated with bladder cancer. Mid-stream urine was collected from 31 male patients with bladder cancer and 18 non-neoplastic controls. DNA was extracted from urine pellet samples and processed for high throughput 16S rRNA amplicon sequencing of the V4 region using Illumina MiSeq. Sequencing reads were filtered using QIIME and clustered using UPARSE. We observed increased bacterial richness (Observed Species, Chao 1 and Ace indexes; cancer vs. control; 120.0 vs. 56.0; 134.5 vs. 68.3; and 139.6 vs. 72.9, respectively), enrichment of some bacterial genera (e.g., Acinetobacter, Anaerococcus, and Sphingobacterium) and decrease of some bacterial genera (e.g., Serratia, Proteus, and Roseomonas) in cancer group when compared to non-cancer group. Significant difference in beta diversity was found between cancer and non-cancer group, among different risk level, but not among different tumor grade. Enrichment of Herbaspirillum, Porphyrobacter, and Bacteroides was observed in cancer patients with high risk of recurrence and progression, which means these genera maybe potential biomarkers for risk stratification. The PICRUSt showed that various functional pathways were enriched in cancer group, including Staphylococcus aureus infection, glycerolipid metabolism and retinol metabolism. To our knowledge, we performed the most comprehensive study to date to characterize the urinary microbiome associated with bladder cancer. A better understanding of the role of microbiome in the development and progression of bladder cancer could pave a new way for exploring new therapeutic options and biomarkers.


Carcinoma/microbiology , DNA, Bacterial/genetics , Microbiota/genetics , Urinary Bladder Neoplasms/microbiology , Urinary Bladder/microbiology , Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/urine , Carcinoma/pathology , Carcinoma/urine , China , DNA, Bacterial/urine , Disease Progression , Humans , Male , Middle Aged , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/urine , Statistics, Nonparametric , Urinary Bladder/pathology , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/urine , Urothelium/microbiology , Urothelium/pathology
11.
Int J Cancer ; 143(8): 1923-1934, 2018 10 15.
Article En | MEDLINE | ID: mdl-29717480

Chronic gastritis caused by Helicobacter pylori (H. pylori) infection could lead to the development of gastric cancer. The finding that multiple gastric cancers can develop synchronously and/or metachronously suggests the development of field cancerization in chronically inflamed, H. pylori-infected gastric mucosa. The genetic basis of multiple tumorigenesis in the inflamed stomach, however, is not well understood. In this study, we analyzed the microsatellite instability (MSI) status and copy number aberrations (CNAs) of 41 multiple intramucosal early gastric cancers that synchronously or metachronously developed in 19 patients with H. pylori infection. Among the 41 intramucosal gastric carcinomas, 9 (22%) exhibited MSI, and the remaining 32 (78%) exhibited the microsatellite stable (MSS) phenotype. Metachronous multiple intramucosal gastric carcinoma exhibit inter-tumor heterogeneity by individually acquiring genetic aberrations. All synchronous multiple intramucosal gastric carcinoma pairs shared a common MSI/MSS profile, and CNA analysis revealed that synchronous multiple intramucosal gastric carcinoma pairs with the MSS phenotype shared common aberrations of representative tumor-suppressor genes, including focal deletion of APC, TP53, CDKN2A, and CDKN2B. Multiregional CNA analysis revealed that heterogeneous gene amplifications/deletions, including PDL1 amplification, evolved under the presence of shared "trunk" genetic alterations in a subpopulation of individual intramucosal gastric carcinomas. These data suggest that multiple gastric carcinomas develop in a multicentric/multifocal manner exhibiting features of inter- and intra-tumor heterogeneity in H. pylori-infected gastric mucosa, whereas synchronous multiple intramucosal gastric carcinomas could share partially common genetic alterations, possibly via common oncogenic pathways.


Carcinoma/genetics , Gastric Mucosa/pathology , Stomach Neoplasms/genetics , Aged , Aged, 80 and over , Carcinoma/microbiology , DNA Methylation/genetics , Female , Gastric Mucosa/microbiology , Genes, Tumor Suppressor/physiology , Helicobacter Infections/genetics , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Humans , Male , Microsatellite Instability , Microsatellite Repeats/genetics , Middle Aged , Mutation/genetics , Stomach Neoplasms/microbiology
12.
Gut ; 67(2): 226-236, 2018 02.
Article En | MEDLINE | ID: mdl-29102920

OBJECTIVE: Gastric carcinoma development is triggered by Helicobacter pylori. Chronic H. pylori infection leads to reduced acid secretion, which may allow the growth of a different gastric bacterial community. This change in the microbiome may increase aggression to the gastric mucosa and contribute to malignancy. Our aim was to evaluate the composition of the gastric microbiota in chronic gastritis and in gastric carcinoma. DESIGN: The gastric microbiota was retrospectively investigated in 54 patients with gastric carcinoma and 81 patients with chronic gastritis by 16S rRNA gene profiling, using next-generation sequencing. Differences in microbial composition of the two patient groups were assessed using linear discriminant analysis effect size. Associations between the most relevant taxa and clinical diagnosis were validated by real-time quantitative PCR. Predictive functional profiling of microbial communities was obtained with PICRUSt. RESULTS: The gastric carcinoma microbiota was characterised by reduced microbial diversity, by decreased abundance of Helicobacter and by the enrichment of other bacterial genera, mostly represented by intestinal commensals. The combination of these taxa into a microbial dysbiosis index revealed that dysbiosis has excellent capacity to discriminate between gastritis and gastric carcinoma. Analysis of the functional features of the microbiota was compatible with the presence of a nitrosating microbial community in carcinoma. The major observations were confirmed in validation cohorts from different geographic origins. CONCLUSIONS: Detailed analysis of the gastric microbiota revealed for the first time that patients with gastric carcinoma exhibit a dysbiotic microbial community with genotoxic potential, which is distinct from that of patients with chronic gastritis.


Bacteria , Carcinoma/microbiology , Dysbiosis/microbiology , Gastritis/microbiology , Gastrointestinal Microbiome , Helicobacter Infections/microbiology , Stomach Neoplasms/microbiology , Stomach/microbiology , Adult , Aged , Bacteria/genetics , Bacteria/metabolism , Chronic Disease , Female , Gastric Mucosa/microbiology , Helicobacter pylori , Humans , Male , Middle Aged , Nitrosation , RNA, Ribosomal, 16S/analysis , Retrospective Studies
13.
Microbiome ; 5(1): 150, 2017 Nov 16.
Article En | MEDLINE | ID: mdl-29145893

BACKGROUND: Colorectal cancer is a worldwide health problem. Despite growing evidence that members of the gut microbiota can drive tumorigenesis, little is known about what happens to it after treatment for an adenoma or carcinoma. This study tested the hypothesis that treatment for adenoma or carcinoma alters the abundance of bacterial populations associated with disease to those associated with a normal colon. We tested this hypothesis by sequencing the 16S rRNA genes in the feces of 67 individuals before and after treatment for adenoma (N = 22), advanced adenoma (N = 19), and carcinoma (N = 26). RESULTS: There were small changes to the bacterial community associated with adenoma or advanced adenoma and large changes associated with carcinoma. The communities from patients with carcinomas changed significantly more than those with adenoma following treatment (P value < 0.001). Although treatment was associated with intrapersonal changes, the change in the abundance of individual OTUs in response to treatment was not consistent within diagnosis groups (P value > 0.05). Because the distribution of OTUs across patients and diagnosis groups was irregular, we used the random forest machine learning algorithm to identify groups of OTUs that could be used to classify pre and post-treatment samples for each of the diagnosis groups. Although the adenoma and carcinoma models could reliably differentiate between the pre- and post-treatment samples (P value < 0.001), the advanced-adenoma model could not (P value = 0.61). Furthermore, there was little overlap between the OTUs that were indicative of each treatment. To determine whether individuals who underwent treatment were more likely to have OTUs associated with normal colons we used a larger cohort that contained individuals with normal colons and those with adenomas, advanced adenomas, and carcinomas. We again built random forest models and measured the change in the positive probability of having one of the three diagnoses to assess whether the post-treatment samples received the same classification as the pre-treatment samples. Samples from patients who had carcinomas changed toward a microbial milieu that resembles the normal colon after treatment (P value < 0.001). Finally, we were unable to detect any significant differences in the microbiota of individuals treated with surgery alone and those treated with chemotherapy or chemotherapy and radiation (P value > 0.05). CONCLUSIONS: By better understanding the response of the microbiota to treatment for adenomas and carcinomas, it is likely that biomarkers will eventually be validated that can be used to quantify the risk of recurrence and the likelihood of survival. Although it was difficult to identify significant differences between pre- and post-treatment samples from patients with adenoma and advanced adenoma, this was not the case for carcinomas. Not only were there large changes in pre- versus post-treatment samples for those with carcinoma, but also these changes were toward a more normal microbiota.


Bacteria/isolation & purification , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/microbiology , Gastrointestinal Microbiome/genetics , Adenoma/drug therapy , Adenoma/microbiology , Aged , Bacteria/classification , Bacteria/genetics , Carcinoma/drug therapy , Carcinoma/microbiology , Colonic Polyps/drug therapy , Colonic Polyps/microbiology , Feces/microbiology , Female , Gastrointestinal Microbiome/physiology , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/drug therapy , RNA, Ribosomal, 16S/genetics , Risk Factors , Sequence Analysis, DNA
14.
Digestion ; 96(4): 213-219, 2017 11.
Article En | MEDLINE | ID: mdl-29050004

BACKGROUND: The factors associated with the pathogenesis of Helicobacter pylori-uninfected undifferentiated-type early gastric cancer (HPUGC) remain unclear. This study compared patient characteristics, including medical history and alcohol/tobacco use, of HPUGC patients with characteristics of patients with H. pylori-positive undifferentiated-type early gastric cancer (HPPGC) to clarify and gain understanding on those differences that could play a role in the pathogenesis. METHODS: This retrospective study included 282 patients who were treated endoscopically from March 2005 to March 2014. This cohort consisted of 232 patients with HPPGC (82.3%) and 50 patients with HPUGC (17.7%). Patient characteristics were analyzed by subgroups of HPUGC vs. HPPGC, with comparisons for age, gender, cancer history, comorbidity of lifestyle diseases requiring medication (hypertension, type 2 diabetes, and dyslipidemia), cumulative amount of alcohol consumption, and smoking history (Brinkman index [BI]). RESULTS: HPUGC patients were typically younger, had less frequent hypertension, and had higher BI values (p < 0.05 for all parameters). In a younger non-hypertensive subgroup, the OR for high BI (BI ≥340) in the HPUGC group vs. HPPGC group was 5.049 (95% CI 2.458-10.373, p < 0.0001). CONCLUSIONS: The investigation of clinical factors identified smoking history as being possibly contributing to the pathogenesis of HPUGC. Future research is necessary at the cellular and genetic levels.


Carcinoma/microbiology , Helicobacter Infections/complications , Helicobacter pylori , Stomach Neoplasms/microbiology , Aged , Carcinoma/pathology , Early Detection of Cancer , Female , Helicobacter Infections/microbiology , Humans , Male , Middle Aged , Retrospective Studies , Risk Factors , Smoking/adverse effects , Stomach Neoplasms/pathology
15.
Minerva Gastroenterol Dietol ; 63(4): 385-398, 2017 Dec.
Article En | MEDLINE | ID: mdl-28927250

Nowadays liver diseases represent one of major healthy problems in the world in terms of morbidity and mortality. The high prevalence of liver pathologies represents the key point to understand the necessity to identify the pathogenetic mechanisms that support these disorders in order to nurse them. Alterations of intestinal microbiota seem to play an important role in induction and promotion of liver damage progression, in addition to direct injury resulting from different causal agents. Gut microbiota is considered both as a promoting factor and as a potential therapeutic target of a large number of liver pathologies due to the connection between intestine and liver: the gut-liver axis. This connection influences absorption and deposition of nutrients into the liver, but also induces the activation of toll-like receptors due to the passage of pathogen-associated molecular patterns in the portal blood and therefore may start both the development of liver damage and its consequent progression to more advanced stages, including cirrhosis and hepatocarcinoma. The gut liver axis also includes the intestinal permeability (IP) degree. It is very variable and interconnected to several factors, most of them dependent from gut microbiota, that is the "lead" in the control of IP. This revue is aimed to report the more recent knowledges about gut microbiota and chronic liver disease, from liver steatosis to cirrhosis and its complications, including hepatocellular carcinoma. For these reasons, it could be useful to intervene, through suitable therapeutic approaches, on the interruption of mechanisms that underlie dysbiosis, IP increase, activation of liver inflammasome, hepatic stellate cells and hepatocarcinogenic processes in order to reduce the liver damage.


Carcinoma/microbiology , Gastrointestinal Microbiome , Liver Cirrhosis/microbiology , Liver Neoplasms/microbiology , Liver/microbiology , Non-alcoholic Fatty Liver Disease/microbiology , Probiotics/therapeutic use , Bacterial Translocation , Carcinoma/epidemiology , Carcinoma/therapy , Disease Progression , Dysbiosis/therapy , Humans , Italy/epidemiology , Liver Cirrhosis/epidemiology , Liver Cirrhosis/therapy , Liver Neoplasms/epidemiology , Liver Neoplasms/therapy , Non-alcoholic Fatty Liver Disease/epidemiology , Non-alcoholic Fatty Liver Disease/therapy , Prevalence , Treatment Outcome
16.
World J Gastroenterol ; 23(29): 5345-5355, 2017 Aug 07.
Article En | MEDLINE | ID: mdl-28839434

AIM: To clarify the mechanisms of connexin 32 (Cx32) downregulation by potential transcriptional factors (TFs) in Helicobacter pylori (H. pylori)-associated gastric carcinogenesis. METHODS: Approximately 25 specimens at each developmental stage of gastric carcinogenesis [non-atrophic gastritis, chronic atrophic gastritis, intestinal metaplasia, dysplasia and gastric carcinoma (GC)] with H. pylori infection [H. pylori (+)] and 25 normal gastric mucosa (NGM) without H. pylori infection [H. pylori (-)] were collected. After transcriptional factor array analysis, the Cx32 and PBX1 expression levels of H. pylori-infected tissues from the developmental stages of GC and NGM with no H. pylori infection were measured by real-time polymerase chain reaction (RT-PCR) and Western blot analysis. Regarding H. pylori-infected animal models, the Cx32 and PBX1 mRNA expression levels and correlation between the gastric mucosa from 10 Mongolian gerbils with long-term H. pylori colonization and 10 controls were analyzed. PBX1 and Cx32 mRNA and protein levels were further studied under the H. pylori-infected condition as well as PBX1 overexpression and knockdown conditions in vitro. RESULTS: Incremental PBX1 was first detected by TF microarray in H. pylori-related gastric carcinogenesis. The identical trend of PBX1 and Cx32 expression was confirmed in the developmental stages of H. pylori-related clinical specimens. The negative correlation of PBX1 and Cx32 was confirmed in H. pylori-infected Mongolian gerbils. Furthermore, decreased PBX1 expression was detected in the normal gastric epithelial cell line GES-1 with H. pylori infection. Enforced overexpression or RNAi-mediated knockdown of PBX1 contributed to the diminished or restored Cx32 expression in GES-1 and the gastric carcinoma cell line BGC823, respectively. Finally, dual-luciferase reporter assay in HEK293T cells showed that Cx32 promoter activity decreased by 30% after PBX1 vector co-transfection, indicating PBX1 as a transcriptional downregulator of Cx32 by directly binding to its promoters. CONCLUSION: PBX1 is one of the determinants in the Cx32 promoter targeting site, preventing further damage of gap junction protein in H. pylori-associated gastric carcinogenesis.


Carcinogenesis/pathology , Carcinoma/pathology , Connexins/metabolism , Helicobacter Infections/pathology , Pre-B-Cell Leukemia Transcription Factor 1/metabolism , Stomach Neoplasms/pathology , Adolescent , Adult , Aged , Animals , Carcinoma/microbiology , Cell Line, Tumor , Chronic Disease , Coculture Techniques , Connexins/genetics , Disease Models, Animal , Down-Regulation , Female , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gastritis, Atrophic/microbiology , Gastritis, Atrophic/pathology , Gastroscopy , Gene Knockdown Techniques , Gerbillinae , HEK293 Cells , Helicobacter Infections/microbiology , Helicobacter pylori/isolation & purification , Humans , Male , Metaplasia , Middle Aged , Pre-B-Cell Leukemia Transcription Factor 1/genetics , Promoter Regions, Genetic/genetics , RNA, Small Interfering/metabolism , Stomach/microbiology , Stomach/pathology , Stomach Neoplasms/microbiology , Stomach Neoplasms/surgery , Up-Regulation , Young Adult , Gap Junction beta-1 Protein
17.
Sci Rep ; 7(1): 7601, 2017 08 08.
Article En | MEDLINE | ID: mdl-28790448

An increasing number of evidences indicate microbes are implicated in human physiological mechanisms, including complicated disease pathology. Some microbes have been demonstrated to be associated with diverse important human diseases or disorders. Through investigating these disease-related microbes, we can obtain a better understanding of human disease mechanisms for advancing medical scientific progress in terms of disease diagnosis, treatment, prevention, prognosis and drug discovery. Based on the known microbe-disease association network, we developed a semi-supervised computational model of Laplacian Regularized Least Squares for Human Microbe-Disease Association (LRLSHMDA) by introducing Gaussian interaction profile kernel similarity calculation and Laplacian regularized least squares classifier. LRLSHMDA reached the reliable AUCs of 0.8909 and 0.7657 based on the global and local leave-one-out cross validations, respectively. In the framework of 5-fold cross validation, average AUC value of 0.8794 +/-0.0029 further demonstrated its promising prediction ability. In case studies, 9, 9 and 8 of top-10 predicted microbes have been manually certified to be associated with asthma, colorectal carcinoma and chronic obstructive pulmonary disease by published literature evidence. Our proposed model achieves better prediction performance relative to the previous model. We expect that LRLSHMDA could offer insights into identifying more promising human microbe-disease associations in the future.


Asthma/microbiology , Carcinoma/microbiology , Colorectal Neoplasms/microbiology , Gastrointestinal Microbiome/genetics , Models, Statistical , Pulmonary Disease, Chronic Obstructive/microbiology , Actinobacteria/classification , Actinobacteria/genetics , Actinobacteria/isolation & purification , Algorithms , Asthma/diagnosis , Asthma/pathology , Carcinoma/diagnosis , Carcinoma/pathology , Clostridiaceae/classification , Clostridiaceae/genetics , Clostridiaceae/isolation & purification , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/pathology , Comamonadaceae/classification , Comamonadaceae/genetics , Comamonadaceae/isolation & purification , Databases, Factual , Firmicutes/classification , Firmicutes/genetics , Firmicutes/isolation & purification , Humans , Least-Squares Analysis , Oxalobacteraceae/classification , Oxalobacteraceae/genetics , Oxalobacteraceae/isolation & purification , Prognosis , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/pathology , Sphingomonadaceae/classification , Sphingomonadaceae/genetics , Sphingomonadaceae/isolation & purification
18.
J Microbiol ; 55(8): 648-654, 2017 Aug.
Article En | MEDLINE | ID: mdl-28752291

The pharynx is an important site of microbiota colonization, but the bacterial populations at this site have been relatively unexplored by culture-independent approaches. The aim of this study was to characterize the microbiota structure of the pharynx. Pyrosequencing of 16S rRNA gene libraries was used to characterize the pharyngeal microbiota using swab samples from 68 subjects with laryngeal cancer and 28 subjects with vocal cord polyps. Overall, the major phylum was Firmicutes, with Streptococcus as the predominant genus in the pharyngeal communities. Nine core operational taxonomic units detected from Streptococcus, Fusobacterium, Prevotella, Granulicatella, and Veillonella accounted for 21.3% of the total sequences detected. However, there was no difference in bacterial communities in the pharynx from patients with laryngeal cancer and vocal cord polyps. The relative abundance of Firmicutes was inversely correlated with Fusobacteria, Proteobacteria, Actinobacteria, and Bacteroidetes. The correlation was evident at the genus level, and the relative abundance of Streptococcus was inversely associated with Fusobacterium, Leptotrichia, Neisseria, Actinomyces, and Prevotella. This study presented a profile for the overall structure of the microbiota in pharyngeal swab samples. Inverse correlations were found between Streptococcus and other bacterial communities, suggesting that potential antagonism may exist among pharyngeal microbiota.


Bacteria/classification , Bacteria/genetics , Carcinoma/microbiology , Laryngeal Neoplasms/microbiology , Microbiota , Pharynx/microbiology , Polyps/microbiology , Adult , Aged , Aged, 80 and over , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Ribosomal/chemistry , DNA, Ribosomal/genetics , Female , Humans , Male , Middle Aged , Phylogeny , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Young Adult
19.
Afr J Tradit Complement Altern Med ; 14(1): 263-271, 2017.
Article En | MEDLINE | ID: mdl-28480404

BACKGROUND: Gastric cancer is a serious health issue caused by H. pylori and claims more lives in developing and undeveloped countries. Hence, the need for a natural drug with several pharmacological activities with no adverse effect are highly recommended. The target of this study was to verify the anti-H. pyloric efficacy of mangiferin (MF) on H. pylori-infected AGS cells. MATERIALS AND METHODS: AGS cells were co-cultured with H. pylori and incubated with increased concentration of MF (10, 20, 50 and 100 µg/mL) or amoxicillin (AMX) and DMSO (control) group to assess its anti-H. pyloric effect by checking inhibitory zone, bacterial drug sensitivity test (MIC and MBC), adhesion and invasive property and various inflammatory markers. RESULTS: Co-culturing of H. pylori-infected AGS cells with MF (100 µg) considerably increased (p<0.05) the inhibitory zone as well as substantially lowered (p<0.05) in the levels of MBC and MIC with decreased adhesion and invasive property in a dose-dependent manner and thus endorsing its anti H. pyloric activity and are almost equivalent to antibiotic AMX. Meanwhile, inflammatory markers such as NF-κΒ subunit p65, interleukins-1ß, IL-8, and TNF-α were also markedly suppressed (p<0.01) on treatment with MF. In addition, the protein expression of inflammatory enzymes like COX-2 and iNOS were notably downregulated (p<0.05) in AGS cells incubated with MF. CONCLUSION: We, concluded that MF treatment with H. pylori-infected AGS cells significantly suppressed the adhesion and invasion process as well as deactivated NF-p65 thereby blocking inflammatory response and thus lower the incidence of gastric carcinoma.


Anti-Inflammatory Agents/pharmacology , Carcinoma/immunology , Carcinoma/microbiology , Helicobacter Infections/immunology , Helicobacter pylori/drug effects , Stomach Neoplasms/immunology , Xanthones/pharmacology , Carcinoma/drug therapy , Carcinoma/genetics , Cell Line, Tumor , Helicobacter Infections/drug therapy , Helicobacter Infections/genetics , Helicobacter Infections/microbiology , Helicobacter pylori/physiology , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-8/genetics , Interleukin-8/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/microbiology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
20.
Oncologist ; 22(5): 542-548, 2017 05.
Article En | MEDLINE | ID: mdl-28432224

This article reviews the history of the discovery of microbes that increase the risk of cancer of some tissues with a special emphasis on the bacterium Helicobacter pylori and the role played by two Australian physicians, neither schooled in research, who had open minds about the shibboleth that mycobacteria (acid-fast organisms) can survive the acid environment of the stomach, but that other pathogenic bacteria cannot. They discovered one of the most important human pathogens, Helicobacter pylori, and showed it capable of inducing severe gastric inflammatory disease. Subsequently, others built on their observations and showed it capable of inducing two gastric neoplasms: carcinoma and lymphoma. The Oncologist 2017;22:542-548.


Carcinoma/microbiology , Helicobacter pylori/pathogenicity , Lymphoma/microbiology , Stomach Neoplasms/microbiology , Australia , Carcinoma/history , Carcinoma/pathology , History, 20th Century , Humans , Lymphoma/history , Lymphoma/pathology , Stomach Neoplasms/history , Stomach Neoplasms/pathology
...