Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
EBioMedicine ; 93: 104670, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37343363

ABSTRACT

BACKGROUND: Obesity is a risk factor for colorectal cancer (CRC). The role of gut microbiota in mediating the cancer-promoting effect of obesity is unknown. METHODS: Azoxymethane (AOM)-treated, ApcMin/+ and germ-free mice were gavaged with feces from obese individuals and control subjects respectively. The colonic tumor load and number were recorded at the endpoint in two carcinogenic models. The gut microbiota composition and colonic transcriptome were assessed by metagenomic sequencing and RNA sequencing, respectively. The anticancer effects of bacteria depleted in fecal samples of obese individuals were validated. FINDINGS: Conventional AOM-treated and ApcMin/+ mice receiving feces from obese individuals showed significantly increased colon tumor formation compared with those receiving feces from control subjects. AOM-treated mice receiving feces from obese individuals showed impaired intestinal barrier function and significant upregulation of pro-inflammatory cytokines and activation of oncogenic Wnt signaling pathway. Consistently, transferring feces from obese individuals to germ-free mice led to increased colonic cell proliferation, intestinal barrier function impairment, and induction of oncogenic and proinflammatory gene expression. Moreover, germ-free mice transplanted with feces from obese human donors had increased abundance of potential pathobiont Alistipes finegoldii, and reduced abundance of commensals Bacteroides vulgatus and Akkermansia muciniphila compared with those receiving feces from human donors with normal body mass index (BMI). Validation experiments showed that B. vulgatus and A. muciniphila demonstrated anti-proliferative effects in CRC, while A. finegoldii promoted CRC tumor growth. INTERPRETATION: Our results supported the role of obesity-associated microbiota in colorectal carcinogenesis and identified putative bacterial candidates that may mediate its mechanisms. Microbiota modulation in obese individuals may provide new approaches to prevent or treat obesity-related cancers including CRC. FUNDING: This work was funded by National Key Research and Development Program of China (2020YFA0509200/2020YFA0509203), National Natural Science Foundation of China (81922082), RGC Theme-based Research Scheme Hong Kong (T21-705/20-N), RGC Research Impact Fund Hong Kong (R4632-21F), RGC-CRF Hong Kong (C4039-19GF and C7065-18GF), RGC-GRF Hong Kong (14110819, 14111621), and NTU Start-Up Grant (021337-00001).


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Gastrointestinal Microbiome , Humans , Mice , Animals , Carcinogenesis , Obesity/complications , Azoxymethane/toxicity , Colorectal Neoplasms/genetics , Mice, Inbred C57BL , Disease Models, Animal
2.
Sci Rep ; 12(1): 10659, 2022 06 23.
Article in English | MEDLINE | ID: mdl-35739175

ABSTRACT

Emerging evidence of an altered gut microbiome in autism spectrum disorder (ASD) suggests a pathomechanism through the gut-brain axis despite the inconsistent microbiome profile reported across studies. One of the knowledge gaps in the existing ASD microbiota studies is the lack of systematic exploration of the role of comorbid functional gastrointestinal disorder (FGID) in the association of ASD and altered gut microbiome. Consequently, 92 ASD and 112 age-matched typically developing (TD) boys were profiled on general psychopathology, FGID status by Rome IV classification, and gut microbiota using 16S ribosomal RNA amplicon sequencing at the V4 hypervariable region. Compared to TD, a significant decrease in the within-sample abundance of taxa was observed in ASD, regardless of FGID status. The microbiota of ASD FGID+ and ASD FGID- clustered apart from the TD groups. The microbiota of ASD FGID+ also showed qualitative differences from that of ASD FGID- and had the highest-level Firmicutes: Bacteroidetes ratio, which was paralleled by elevated levels of anxiety and overall psychopathology. The altered gastrointestinal microbiota composition in ASD appeared to be independent of comorbid FGID. Further studies should address how FGID may mediate neuropsychiatric symptoms in ASD through inflammation along the microbiota-gut-brain axis.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Gastrointestinal Diseases , Gastrointestinal Microbiome , Case-Control Studies , China , Gastrointestinal Microbiome/genetics , Humans , Male
4.
Emerg Microbes Infect ; 9(1): 631-638, 2020.
Article in English | MEDLINE | ID: mdl-32183606

ABSTRACT

Clostridioides difficile infection (CDI) is a common cause of nosocomial diarrhea and can sometimes lead to pseudo-membranous colitis and toxic megacolon. We previously reported that the PCR ribotype 002 was a common C. difficile ribotype in Hong Kong that was associated with increased mortality. In this study, we assessed in vitro bacteriological characteristics and in vivo virulence of ribotype 002 compared to other common ribotypes, including ribotypes 012, 014 and 046. We observed significantly higher toxin A (p < 0.05) and toxin B (p < 0.05) production, sporulation (p < 0.001) and germination rates (p < 0.0001) in ribotype 002 than other common ribotypes. In a murine model of C. difficile infection, ribotype 002 caused significantly more weight loss (p < 0.001) and histological damage (p < 0.001) than other common ribotypes. These findings may have contributed to the higher prevalence and mortality observed, and provided mechanistic insights that can help public surveillance and develop novel therapeutics to combat against this infection.


Subject(s)
Clostridiales/pathogenicity , Gram-Positive Bacterial Infections/microbiology , Animals , Hong Kong , Male , Mice, Inbred C57BL , Ribotyping , Virulence
5.
Sci Adv ; 5(1): eaau9650, 2019 01.
Article in English | MEDLINE | ID: mdl-30746470

ABSTRACT

A rapid, direct, and low-cost method for detecting bacterial toxins associated with common gastrointestinal diseases remains a great challenge despite numerous studies and clinical assays. Motion-based detection through tracking the emerging micro- and nanorobots has shown great potential in chemo- and biosensing due to accelerated "chemistry on the move". Here, we described the use of fluorescent magnetic spore-based microrobots (FMSMs) as a highly efficient mobile sensing platform for the detection of toxins secreted by Clostridium difficile (C. diff) that were present in patients' stool. These microrobots were synthesized rapidly and inexpensively by the direct deposition of magnetic nanoparticles and the subsequent encapsulation of sensing probes on the porous natural spores. Because of the cooperation effect of natural spore, magnetic Fe3O4 nanoparticles, and functionalized carbon nanodots, selective fluorescence detection of the prepared FMSMs is demonstrated in C. diff bacterial supernatant and even in actual clinical stool samples from infectious patients within tens of minutes, suggesting rapid response and good selectivity and sensitivity of FMSMs toward C. diff toxins.


Subject(s)
Bacterial Proteins/analysis , Bacterial Toxins/analysis , Clostridioides difficile/physiology , Clostridium Infections/diagnosis , Magnetite Nanoparticles/chemistry , Nanomedicine/methods , Remote Sensing Technology/methods , Spores, Bacterial , Carbon/chemistry , Clostridium Infections/microbiology , Feces/chemistry , Feces/microbiology , Ferrosoferric Oxide/chemistry , Fluorescence , Fluorescent Dyes/chemistry , Humans , Sensitivity and Specificity
6.
Semin Cancer Biol ; 55: 28-36, 2019 04.
Article in English | MEDLINE | ID: mdl-29782923

ABSTRACT

The involvement of microorganisms in cancer has been increasing recognized. Collectively, microorganisms have been estimated to account for ∼20% of all cancers worldwide. Recent advances in metagenomics and bioinformatics have provided new insights on the microbial ecology in different tumors, pinpointing the roles of microorganisms in cancer formation, development and response to treatments. Furthermore, studies have emphasized the importance of host-microbial and inter-microbial interactions in the cancer microbiota. These studies have not only revolutionized our understanding of cancer biology, but also opened up new opportunities for cancer prevention, diagnosis, prognostication and treatment. This review article aims to summarize the microbiota in various cancers and their treatments, and explore clinical applications for such relevance.


Subject(s)
Gastrointestinal Microbiome/genetics , Metagenomics , Neoplasms/genetics , Computational Biology , Humans , Neoplasms/diagnosis , Neoplasms/microbiology , Neoplasms/therapy
7.
Gastroenterology ; 156(1): 292, 2019 01.
Article in English | MEDLINE | ID: mdl-30472232
8.
Gastroenterology ; 155(2): 383-390.e8, 2018 08.
Article in English | MEDLINE | ID: mdl-29729257

ABSTRACT

BACKGROUND & AIMS: Colorectal cancer (CRC) development has been associated with increased proportions of Bacteroides fragilis and certain Streptococcus, Fusobacterium, and Peptostreptococcus species in the intestinal microbiota. We investigated associations between bacteremia from specific intestinal microbes and occurrence of CRC. METHODS: We performed a retrospective study after collecting data on 13,096 adult patients (exposed group) in Hong Kong hospitalized with bacteremia (identified by blood culture test) without a previous diagnosis of cancer from January 1, 2006 through December 31, 2015. We collected data on intestinal microbes previously associated with CRC (genera Bacteroides, Clostridium, Filifactor, Fusobacterium, Gemella, Granulicatella, Parvimonas, Peptostreptococcus, Prevotella, Solobacterium, and Streptococcus). Clinical information, including patient demographics, comorbid medical conditions, date of bacteremia, and bacterial species identified, were collected. The incidence of biopsy-proved CRC was compared between the exposed and unexposed (patients without bacteremia matched for age, sex, and comorbidities) groups. RESULTS: The risk of CRC was increased in patients with bacteremia from B fragilis (hazard ratio [HR] = 3.85, 95% CI = 2.62-5.64, P = 5.5 × 10-12) or Streptococcus gallolyticus (HR = 5.73, 95% CI = 2.18-15.1, P = 4.1 × 10-4) compared with the unexposed group. In addition, the risk of CRC was increased in patients with bacteremia from Fusobacterium nucleatum (HR = 6.89, 95% CI = 1.70-27.9, P = .007), Peptostreptococcus species (HR = 3.06, 95% CI = 1.47-6.35, P = .003), Clostridium septicum (HR = 17.1, 95% CI = 1.82-160, P = .013), Clostridium perfringens (HR = 2.29, 95% CI = 1.16-4.52, P = .017), or Gemella morbillorum (HR = 15.2, 95% CI = 1.54-150, P = .020). We observed no increased risk in patients with bacteremia caused by microbes not previously associated with colorectal neoplasms. CONCLUSIONS: In a retrospective analysis of patients hospitalized for bacteremia, we associated later diagnosis of CRC with B fragilis and S gallolyticus and other intestinal microbes. These bacteria might have entered the bloodstream from intestinal dysbiosis and perturbed barrier function. These findings support a model in which specific members of the intestinal microbiota promote colorectal carcinogenesis. Clinicians should evaluate patients with bacteremia from these species for neoplastic lesions in the colorectum.


Subject(s)
Bacteremia/microbiology , Colon/microbiology , Colorectal Neoplasms/blood , Dysbiosis/blood , Gastrointestinal Microbiome , Adult , Aged , Aged, 80 and over , Bacteroides fragilis/isolation & purification , Bacteroides fragilis/pathogenicity , Biopsy , Carcinogenesis , Colon/pathology , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/microbiology , Dysbiosis/diagnosis , Dysbiosis/epidemiology , Dysbiosis/microbiology , Female , Hong Kong/epidemiology , Humans , Incidence , Male , Middle Aged , Retrospective Studies , Streptococcus gallolyticus/isolation & purification , Streptococcus gallolyticus/pathogenicity
9.
J Cell Mol Med ; 22(4): 2469-2477, 2018 04.
Article in English | MEDLINE | ID: mdl-29383879

ABSTRACT

Toxin B (TcdB) is a major pathogenic factor of Clostridum difficile. However, the mechanism by which TcdB exerts its cytotoxic action in host cells is still not completely known. Herein, we report for the first time that TcdB induced autophagic cell death in cultured human colonocytes. The induction of autophagy was demonstrated by the increased levels of LC3-II, formation of LC3+ autophagosomes, accumulation of acidic vesicular organelles and reduced levels of the autophagic substrate p62/SQSTM1. TcdB-induced autophagy was also accompanied by the repression of phosphoinositide 3-kinase (PI3K)/Akt/mechanistic target of rapamycin (mTOR) complex 1 activity. Functionally, pharmacological inhibition of autophagy by wortmannin or chloroquine or knockdown of autophagy-related genes Beclin 1, Atg5 and Atg7 attenuated TcdB-induced cell death in colonocytes. Genetic ablation of Atg5, a gene required for autophagosome formation, also mitigated the cytotoxic effect of TcdB. In conclusion, our study demonstrated that autophagy serves as a pro-death mechanism mediating the cytotoxic action of TcdB in colonocytes. This discovery suggested that blockade of autophagy might be a novel therapeutic strategy for C. difficile infection.


Subject(s)
Autophagy/genetics , Bacterial Proteins/genetics , Bacterial Toxins/genetics , Clostridioides difficile/genetics , Clostridium Infections/therapy , Apoptosis/drug effects , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagosomes/microbiology , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Beclin-1/genetics , Clostridioides difficile/pathogenicity , Clostridium Infections/genetics , Clostridium Infections/microbiology , Colon/cytology , Colon/microbiology , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation , Sequestosome-1 Protein/genetics , TOR Serine-Threonine Kinases/genetics
10.
Sci Rep ; 7(1): 17218, 2017 12 08.
Article in English | MEDLINE | ID: mdl-29222426

ABSTRACT

Incidence of Clostridium difficile infection (CDI) is rapidly increasing and it poses a major health burden globally. However, data regarding the epidemiology of CDI in Asia are limited. We aimed to characterize the antimicrobial susceptibility patterns of common ribotypes of toxigenic C. difficile in Hong Kong. Fifty-three PCR ribotypes were identified among 284 toxigenic C. difficile clinical isolates. The five most prevalent ribotypes were 002 (13%), 017 (12%), 014 (10%), 012 (9.2%), and 020 (9.5%). All tested C. difficile strains remained susceptible to metronidazole, vancomycin, meropenem and piperacillin/tazobactam, but highly resistant to cephalosporins. Of the fluoroquinolones, highest resistance to ciprofloxacin was observed (99%), followed by levofloxacin (43%) and moxifloxacin (23%). The two newly emerged PCR ribotypes, 017 and 002, demonstrated high levels of co-resistance towards clindamycin, tetracycline, erythromycin and moxifloxacin. PCR ribotypes 017 and 002 with multi-drug resistance are rapidly emerging and continuous surveillance is important to monitor the epidemiology of C. difficile to prevent outbreaks of CDI.


Subject(s)
Anti-Bacterial Agents/pharmacology , Clostridioides difficile/classification , Clostridioides difficile/drug effects , Drug Resistance, Bacterial , Ribotyping , Clostridioides difficile/genetics , Hong Kong , Polymerase Chain Reaction
11.
Emerg Infect Dis ; 23(10): 1671-1679, 2017 10.
Article in English | MEDLINE | ID: mdl-28930010

ABSTRACT

Cross-sectional studies suggest an increasing trend in incidence and relatively low recurrence rates of Clostridium difficile infections in Asia than in Europe and North America. The temporal trend of C. difficile infection in Asia is not completely understood. We conducted a territory-wide population-based observational study to investigate the burden and clinical outcomes in Hong Kong, China, over a 9-year period. A total of 15,753 cases were identified, including 14,402 (91.4%) healthcare-associated cases and 817 (5.1%) community-associated cases. After adjustment for diagnostic test, we found that incidence increased from 15.41 cases/100,000 persons in 2006 to 36.31 cases/100,000 persons in 2014, an annual increase of 26%. This increase was associated with elderly patients, for whom incidence increased 3-fold over the period. Recurrence at 60 days increased from 5.7% in 2006 to 9.1% in 2014 (p<0.001). Our data suggest the need for further surveillance, especially in Asia, which contains ≈60% of the world's population.


Subject(s)
Clostridioides difficile/isolation & purification , Clostridium Infections/epidemiology , Cross Infection/epidemiology , Adolescent , Adult , Aged , Aged, 80 and over , Clostridium Infections/microbiology , Clostridium Infections/mortality , Community-Acquired Infections , Cross Infection/microbiology , Cross Infection/mortality , Cross-Sectional Studies , Epidemiological Monitoring , Female , Hong Kong/epidemiology , Humans , Incidence , Male , Middle Aged , Recurrence , Survival Analysis
12.
Gastroenterology ; 153(6): 1621-1633.e6, 2017 12.
Article in English | MEDLINE | ID: mdl-28823860

ABSTRACT

BACKGROUND & AIMS: Altered gut microbiota is implicated in development of colorectal cancer (CRC). Some intestinal bacteria have been reported to potentiate intestinal carcinogenesis by producing genotoxins, altering the immune response and intestinal microenvironment, and activating oncogenic signaling pathways. We investigated whether stool from patients with CRC could directly induce colorectal carcinogenesis in mice. METHODS: We obtained stored stool samples from participants in a metagenome study performed in Hong Kong. Conventional (male C57BL/6) mice were given azoxymethane to induce colon neoplasia after receiving a course of antibiotics in drinking water. Mice were gavaged twice weekly with stool from 5 patients with CRC or 5 healthy individuals (controls) for 5 weeks. Germ-free C57BL/6 mice were gavaged once with stool from 5 patients with CRC or 5 controls. We collected intestinal tissues from mice and performed histology, immunohistochemistry, expression microarray, quantitative polymerase chain reaction, immunoblot, and flow cytometry analyses. We performed 16S ribosomal RNA gene sequencing analysis of feces from mice. RESULTS: Significantly higher proportions of conventional mice fed with stool from individuals with CRC than control stool developed high-grade dysplasia (P < .05) and macroscopic polyps (P < .01). We observed a higher proportion of proliferating (Ki-67-positive) cells in colons of germ-free mice fed with stool from patients with CRC vs those fed with stool from controls (P < .05). Feces from germ-free and conventional mice fed with stool from patients with CRC vs controls contained different microbial compositions, with lower richness in mice fed with stool from patients with CRC. Intestines collected from conventional and germ-free mice fed with stool from patients with CRC had increased expression of cytokines that modulate inflammation, including C-X-C motif chemokine receptor 1, C-X-C motif chemokine receptor 2, interleukin 17A (IL17A), IL22, and IL23A. Intestines from conventional and germ-free mice fed with stool from patients with CRC contained higher proportions of T-helper 1 (Th1) cells (2.25% vs 0.44%) and Th17 cells (2.08% vs 0.31%) (P < .05 for each) than mice fed with stool from controls. Real-time polymerase chain reaction arrays revealed up-regulation of genes involved in cell proliferation, stemness, apoptosis, angiogenesis, invasiveness, and metastasis in mice fed with stool from patients with CRC. CONCLUSIONS: We fed stool samples from patients with CRC and heathy individuals to germ-free mice and conventional mice with azoxymethane. We found stool from patients with CRC to increase the numbers of polyps, levels of intestinal dysplasia and proliferation, markers of inflammation, and proportions of Th1 and Th17 cells in colon, compared with stool from individuals without CRC. This study provides evidence that the fecal microbiota from patients with CRC can promote tumorigenesis in germ-free mice and mice given a carcinogen.


Subject(s)
Cell Transformation, Neoplastic , Colon/microbiology , Colonic Polyps/microbiology , Colorectal Neoplasms/microbiology , Feces/microbiology , Gastrointestinal Microbiome , Animals , Azoxymethane , Case-Control Studies , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Colon/metabolism , Colon/pathology , Colonic Polyps/chemically induced , Colonic Polyps/metabolism , Colonic Polyps/pathology , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Germ-Free Life , Host-Pathogen Interactions , Humans , Inflammation Mediators/metabolism , Ki-67 Antigen/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/microbiology , Male , Mice, Inbred C57BL , Th1 Cells/metabolism , Th1 Cells/microbiology , Th17 Cells/metabolism , Th17 Cells/microbiology
13.
Gut ; 66(8): 1441-1448, 2017 08.
Article in English | MEDLINE | ID: mdl-27797940

ABSTRACT

OBJECTIVE: There is a need for an improved biomarker for colorectal cancer (CRC) and advanced adenoma. We evaluated faecal microbial markers for clinical use in detecting CRC and advanced adenoma. DESIGN: We measured relative abundance of Fusobacterium nucleatum (Fn), Peptostreptococcus anaerobius (Pa) and Parvimonas micra (Pm) by quantitative PCR in 309 subjects, including 104 patients with CRC, 103 patients with advanced adenoma and 102 controls. We evaluated the diagnostic performance of these biomarkers with respect to faecal immunochemical test (FIT), and validated the results in an independent cohort of 181 subjects. RESULTS: The abundance was higher for all three individual markers in patients with CRC than controls (p<0.001), and for marker Fn in patients with advanced adenoma than controls (p=0.022). The marker Fn, when combined with FIT, showed superior sensitivity (92.3% vs 73.1%, p<0.001) and area under the receiver-operating characteristic curve (AUC) (0.95 vs 0.86, p<0.001) than stand-alone FIT in detecting CRC in the same patient cohort. This combined test also increased the sensitivity (38.6% vs 15.5%, p<0.001) and AUC (0.65 vs 0.57, p=0.007) for detecting advanced adenoma. The performance gain for both CRC and advanced adenoma was confirmed in the validation cohort (p=0.0014 and p=0.031, respectively). CONCLUSIONS: This study identified marker Fn as a valuable marker to improve diagnostic performance of FIT, providing a complementary role to detect lesions missed by FIT alone. This simple approach may improve the clinical utility of the current FIT, and takes one step further towards a non-invasive, potentially more accurate and affordable diagnosis of advanced colorectal neoplasia.


Subject(s)
Adenocarcinoma/diagnosis , Adenoma/diagnosis , Colorectal Neoplasms/diagnosis , DNA, Bacterial/analysis , Feces/microbiology , Fusobacterium nucleatum , Occult Blood , Peptostreptococcus , Area Under Curve , Biomarkers, Tumor/analysis , Female , Humans , Male , Middle Aged , ROC Curve
SELECTION OF CITATIONS
SEARCH DETAIL
...