Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Article in English | MEDLINE | ID: mdl-38752653

ABSTRACT

INTRODUCTION: Long-term studies characterizing the natural history of functional bowel disorder (FBD) from community-based settings and exploring association with psychological factors are sparse. We aimed to evaluate the evolution of symptoms, health outcomes, and association of FBD with psychological disorders in Chinese population. METHODS: Individuals identified from random sampling of residents of Hangzhou, China, participated in a baseline survey in January 2010. Follow-up phone survey was conducted in December 2018. FBD was diagnosed based on Rome III criteria. RESULTS: Among 452 individuals (mean age 44.6 ± 15.3 years, 174 [38%] male) who completed the study, the prevalence of FBD was 36.3% (95% confidence interval [CI] 32.6-40.0%) at enrollment and 36.1% (95% CI 32.3-39.8%) at follow-up survey ( P = 0.94). However, 214 individuals (47%) had interval change in diagnosis. Although no difference in incidence of organic disease or death was observed, a higher proportion of patients with FBD (16/164, 9.8% vs 9/288, 3.1%; P = 0.003) compared with those without FBD received non-cancer-related abdominal and/or pelvic surgery during follow-up. FBD was associated with anxiety and/or depression at initial (adjusted odds ratio [AOR] = 1.7, 95% CI 1.7-2.7, P = 0.02) and follow-up (AOR = 8.0, 95% CI 3.2-20.0, P < 0.001) surveys. Diagnosis of FBD at baseline was associated with new-onset anxiety and/or depression at follow-up (AOR = 3.2, 95% CI 1.2-8.3, P = 0.01). DISCUSSION: Although the prevalence of FBD remained stable, transformation of symptoms was common over time. Patients with FBD may have increased risk of receiving non-cancer-related abdominal and/or pelvic surgery. FBD symptoms at baseline increased the risk of new-onset anxiety and/or depression by 3.2-fold over the next 9 years.

2.
Front Pharmacol ; 15: 1371574, 2024.
Article in English | MEDLINE | ID: mdl-38576492

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, resulting in a huge medical burden worldwide. Accumulating evidence suggests that the gut microbiome and bile acids play pivotal roles during the development of NAFLD. Patients with NAFLD exhibit unique signatures of the intestinal microbiome marked by the priority of Gram-negative bacteria, decreased ratio of Firmicutes/Bacteroidetes (F/B), and increased Prevotella and Lachnospiraceae. The intestinal microbiota is involved in the metabolism of bile acids. Ursodeoxycholic acid (UDCA) is a key determinant in maintaining the dynamic communication between the host and gut microbiota. It generally shows surprising therapeutic potential in NAFLD with several mechanisms, such as improving cellular autophagy, apoptosis, and mitochondrial functions. This action is based on its direct or indirect effect, targeting the farnesoid X receptor (FXR) and various other nuclear receptors. This review aims to discuss the current studies on the involvement of the microbiome-UDCA interface in NAFLD therapy and provide prospective insights into future preventative and therapeutic approaches for NAFLD.

3.
Nutr Rev ; 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38553410

ABSTRACT

Context: Research regarding the treatment of inflammatory bowel disease (IBD) with probiotics has not yielded consistent results. OBJECTIVE: The aim of this meta-analysis was to evaluate the efficacy of probiotics supplementation in patients with IBD. DATA SOURCES: Randomized controlled trials (RCTs) evaluating the efficacy of probiotics in patients with IBD were searched in PubMed, the Google Scholar database, Web of Science, and CrossRef for the period July 2003 to June 2023. DATA EXTRACTION: The RCTs were extracted, independently by 2 authors, according to the PICOS criteria. DATA ANALYSIS: Seven studies, including a total of 795 patients, met the study criteria. Five end points were selected to evaluate the efficacy. Of these, 3 indicators showed a statistically significant difference in efficacy: C-reactive protein (odds ratio [OR]: -2.45, 95% confidence interval [CI]: -3.16, -1.73, P < .01), the number of fecal Bifidobacterium (OR: 3.37, 95% CI: 3.28, 3.47, P < .01), and Lactobacillus(OR: 2.00, 95% CI: 1.91, 2.09, P < .01). The other 2 indicators (disease activity for Crohn's disease and for ulcerative colitis) showed no statistically significant difference, while the OR reflected a positive correlation. CONCLUSION: Probiotics supplementation may have a positive effect on IBD by reducing clinical symptoms, reducing the serological inflammatory markers, and increasing favorable gut flora in patients with IBD. Additional RCTs are needed to evaluate the therapeutic effect of probiotics in IBD.

4.
Adv Sci (Weinh) ; 11(12): e2306571, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38235606

ABSTRACT

Most patients with inflammatory bowel disease (IBD) develop anemia, which is attributed to the dysregulation of iron metabolism. Reciprocally, impaired iron homeostasis also aggravates inflammation. How this iron-mediated, pathogenic anemia-inflammation crosstalk is regulated in the gut remains elusive. Herein, it is for the first time revealed that anemic IBD patients exhibit impaired production of short-chain fatty acids (SCFAs), particularly butyrate. Butyrate supplementation restores iron metabolism in multiple anemia models. Mechanistically, butyrate upregulates ferroportin (FPN) expression in macrophages by reducing the enrichment of histone deacetylase (HDAC) at the Slc40a1 promoter, thereby facilitating iron export. By preventing iron sequestration, butyrate not only mitigates colitis-induced anemia but also reduces TNF-α production in macrophages. Consistently, macrophage-conditional FPN knockout mice exhibit more severe anemia and inflammation. Finally, it is revealed that macrophage iron overload impairs the therapeutic effectiveness of anti-TNF-α antibodies in colitis, which can be reversed by butyrate supplementation. Hence, this study uncovers the pivotal role of butyrate in preventing the pathogenic circuit between anemia and inflammation.


Subject(s)
Anemia , Colitis , Inflammatory Bowel Diseases , Humans , Mice , Animals , Iron/metabolism , Butyrates/metabolism , Butyrates/pharmacology , Tumor Necrosis Factor Inhibitors/metabolism , Inflammation/metabolism , Anemia/metabolism , Macrophages/metabolism , Mice, Knockout
5.
Dig Dis Sci ; 69(1): 66-80, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37968554

ABSTRACT

BACKGROUND AND AIMS: Inflammatory bowel disease (IBD) is currently gaining an increasing global interest. Intestinal epithelial barrier dysfunction is crucial toward developing IBD; however, the underlying mechanisms are not yet elucidated. This study is aimed at elucidating the function of CRL4DCAF2, an E3 ligase, toward mediating intestinal homeostasis. METHODS: Colon samples were collected from patients with IBD and healthy individuals to examine the expression of CRL4DCAF2. CRL4DCAF2 conditional knockdown in mouse intestinal epithelial cells (IECs) (DCAF2EKD) were constructed. DCAF2EKD and their littermate control (DCAF2EWT) were treated with dextran sodium sulfate (DSS) to induce acute colitis. Transcriptome analysis was performed on inflamed colon samples obtained from the mice. Cell cycle regulators were evaluated using real-time polymerase chain reaction (PCR), while tight junction and apoptosis proteins were examined via immunofluorescence and western blot. RESULTS: CRL4DCAF2 expression was significantly decreased in the inflamed IBD epithelium, and low expression of CRL4DCAF2 associated with high recurrence risk. Mice with DCAF2 specific knockout in IECs suffer from embryonic death. Multiple genes involved in cell proliferation, immune response, and gap junction were differentially expressed in inflamed colon from DCAF2EKD compared with DCAF2EWT. Furthermore, conditional downregulation of CRL4DCAF2 in the intestinal epithelium induced primarily epithelial damage, increased intestinal permeability, and diminished tight junction protein expression. In vivo and in vitro cell transfection experiments revealed that CRL4DCAF2 enhanced cell proliferation by promoting p21 ubiquitination and degradation, thereby inhibiting G2/M cell cycle. In addition, CRL4DCAF2 can also inhibit IEC apoptosis and promote cell autophagy. CONCLUSIONS: CRL4DCAF2 downregulation in IECs promotes intestinal barrier dysfunction and inhibits IEC proliferation, thus making it more susceptible to inflammation.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Humans , Animals , Mice , Colitis/chemically induced , Colitis/genetics , Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , Cell Proliferation , Homeostasis , Dextran Sulfate/toxicity , Disease Models, Animal , Mice, Inbred C57BL
6.
Front Med (Lausanne) ; 9: 869004, 2022.
Article in English | MEDLINE | ID: mdl-35425770

ABSTRACT

Background: Over the past 40 years, endoscopic ultrasound (EUS) has become a safe and effective tool for both diagnostic and therapeutic applications. A growing number of articles have been published annually. We aimed to explore global scientific outputs and hotspots of EUS published by different countries, organizations, and authors. Methods: The global literature regarding EUS during the 1900-2020 period was identified from the Web of Science (WOS) Core database. "Bibliometrix" and software VOSviewer were applied to perform bibliometric analysis. Results: The annual growth rate of publications from 1980 to 2020 was around 16% and the number of EUS-related articles had experienced a sudden increase in the last decade. Bhutani MS was the most productive author over the past years, with 94 publications. Hawes RH had the highest number of citations, with 6,034 citations. The United States and institutions from United States dominated the EUS research. Among the journals, GASTROINTESTINAL ENDOSCOPY published the highest number of articles, followed by ENDOSCOPY. The majority of top 10 frequently cited references were cited more than 200 times. Carcinoma, diagnosis, fine-needle-aspiration, cytology, and pancreatitis were the important keywords in co-occurrence analysis of keywords. Recent studies focused more on tissue acquisition, size of the needle, lumen-apposing metal stent, and fine-needle- biopsy. Conclusion: Research on EUS has significantly increased in the last decade globally and it will continue to increase. Active collaboration among different authors and countries was observed in the EUS field. Tissue acquisition, size of the needle, apposing metal stent, and fine-needle-biopsy might be the latest research frontiers and should receive more attention.

7.
J Crohns Colitis ; 15(7): 1197-1210, 2021 Jul 05.
Article in English | MEDLINE | ID: mdl-33417675

ABSTRACT

BACKGROUND AND AIMS: Inflammatory bowel disease (IBD) is associated with gut dysbiosis and dysregulation of bile acid metabolism. A high luminal content of deoxycholic acid (DCA) with consumption of a Westernised diet is implicated in the pathogenesis of IBD. The aim of the study is to explore the role of intestinal microbiota and bile acid metabolism in mice with DCA-induced intestinal inflammation. METHODS: Wild-type C57BL mice, 4 weeks old, were fed with AIN-93G (control diet), AIN-93G+0.2% DCA, AIN-93G+0.2% DCA+6 weeks of fexaramine (FXR agonist), or AIN-93G+0.2% DCA+antibiotic cocktail, for 24 weeks. Histopathology, western blotting, and qPCR were performed on the intestinal tissue. Faecal microbiota was analysed by 16S rDNA sequencing. Faecal bile acid and short chain fatty acid (SCFA) levels were analysed by chromatography. RESULTS: Gut dysbiosis and enlarged bile acid pool were observed in DCA-treated mice, accompanied by a lower farnesoid X receptor (FXR) activity in the intestine. Administration of fexaramine mitigated DCA-induced intestinal injury, restored intestinal FXR activity, activated fibroblast growth factor 15, and normalised bile acid metabolism. Furthermore, fexaramine administration increased the abundance of SCFA-producing bacteria. Depletion of the commensal microbiota with antibiotics decreased the diversity of the intestinal microbiota, attenuated bile acid synthesis, and reduced intestinal inflammation induced by DCA. CONCLUSIONS: DCA induced-intestinal inflammation is associated with alterations of gut microbiota and bile acid profiles. Interventions targeting the gut microbiota-FXR signalling pathway may reduce DCA-induced intestinal disease.


Subject(s)
Gastrointestinal Microbiome/drug effects , Inflammatory Bowel Diseases/drug therapy , Receptors, Cytoplasmic and Nuclear/drug effects , Animals , Anti-Bacterial Agents/administration & dosage , Benzene Derivatives/administration & dosage , Bile Acids and Salts/metabolism , Deoxycholic Acid , Female , Inflammatory Bowel Diseases/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction
8.
Dig Dis Sci ; 66(2): 568-576, 2021 02.
Article in English | MEDLINE | ID: mdl-32198567

ABSTRACT

BACKGROUND: A Western diet is a risk factor for the development of inflammatory bowel disease (IBD). High levels of fecal deoxycholic acid (DCA) in response to a Western diet contribute to bowel inflammatory injury. However, the mechanism of DCA in the natural course of IBD development remains unanswered. AIMS: The aim of this study is to investigate the effect of DCA on the induction of gut dysbiosis and its roles in the development of intestinal inflammation. METHODS: Wild-type C57BL/6J mice were fed an AIN-93G diet, either supplemented with or without 0.2% DCA, and killed at 24 weeks. Distal ileum and colon tissues were assessed by histopathological analysis. Hepatic and ileal gene expression was examined by qPCR, and the gut microbiota was analyzed by high-throughput 16S rRNA gene sequencing. HPLC-MS was used for fecal bile acid quantification. RESULTS: Mice fed the DCA-supplemented diet developed focal areas of ileal and colonic inflammation, accompanied by alteration of the composition of the intestinal microbiota and accumulation of fecal bile acids. DCA-induced dysbiosis decreased the deconjugation of bile acids, and this regulation was associated with the repressed expression of target genes in the enterohepatic farnesoid X receptor-fibroblast growth factor (FXR-FGF15) axis, leading to upregulation of hepatic de novo bile acid synthesis. CONCLUSIONS: These results suggest that DCA-induced gut dysbiosis may act as a key etiologic factor in intestinal inflammation, associated with bile acid metabolic disturbance and downregulation of the FXR-FGF15 axis.


Subject(s)
Bile Acids and Salts/metabolism , Deoxycholic Acid/toxicity , Diet, Western/adverse effects , Dysbiosis/metabolism , Enterohepatic Circulation/physiology , Inflammatory Bowel Diseases/metabolism , Animals , Deoxycholic Acid/administration & dosage , Dysbiosis/chemically induced , Dysbiosis/pathology , Enterohepatic Circulation/drug effects , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/pathology , Mice , Mice, Inbred C57BL
9.
Gene ; 753: 144814, 2020 Aug 30.
Article in English | MEDLINE | ID: mdl-32464244

ABSTRACT

AIMS: The goal of our study is to investigate the contribution of the 13 single-nucleotide polymorphisms to inflammatory bowel disease (IBD), including Crohn's disease (CD) and Ulcerative colitis (UC). METHODS: A total of 44 articles were retrieved from bibliographic databases including PubMed, Embase, SpingerLink, Web of Science, Chinese National Knowledge Infrastructure (CNKI), and Wanfang. Through a comprehensive filtering procedure, 13 single-nucleotide polymorphisms were collected in the meta-analysis, which was done by Review Manager 5.0. RESULTS: After a systematic filtration, there were 13 single-nucleotide polymorphisms (SNPs) from 44 articles involved in our meta-analysis. Our results demonstrated that 3 SNPs were found to be significantly associated with CD/UC/IBD: IRF5 rs4728142 (UC: OR = 1.21, 95% CI = 1.09-1.35, P = 0.0003; OR = 1.30, 95% CI = 1.08-1.57, P = 0.006 in Asian), PTGER4 rs4613763 (CD: the overall OR = 1.28, 95% CI = 1.01-1.64, P = 0.04; IBD: OR = 1.31, 95% CI = 1.04-1.65, P = 0.02), IL12B rs6887695 (CD: the overall OR = 1.17, 95% CI = 1.06-1.30, P = 0.002; UC: the overall OR = 1.13, 95% CI = 1.01-1.26, P = 0.03; IBD: the overall OR = 1.15, 95% CI = 1.06-1.24, P = 0.0009). CONCLUSION: Our meta-analyses have indicated the significant associations between SNPs (IRF5 rs4728142, PTGER4 rs4613763, and IL12B rs6887695) and CD/UC/IBD.


Subject(s)
Inflammatory Bowel Diseases/genetics , Interferon Regulatory Factors/genetics , Interleukin-12 Subunit p40/genetics , Receptors, Prostaglandin E, EP4 Subtype/genetics , Asian People/genetics , Case-Control Studies , Colitis, Ulcerative/genetics , Crohn Disease/genetics , Disease Susceptibility , Genetic Association Studies , Genetic Predisposition to Disease/genetics , Genotype , Humans , Phenotype , Polymorphism, Single Nucleotide/genetics , White People/genetics
11.
J Cell Mol Med ; 24(4): 2648-2662, 2020 02.
Article in English | MEDLINE | ID: mdl-31957197

ABSTRACT

High-fat diet (HFD) is a well-known risk factor for gut microbiota dysbiosis and colorectal cancer (CRC). However, evidence relating HFD, gut microbiota and carcinogenesis is limited. Our study aimed to demonstrate that HFD-induced gut dysbiosis promoted intestinal adenoma-adenocarcinoma sequence. In clinical study, we found that HFD increased the incidence of advanced colorectal neoplasia (AN). The expression of monocyte chemoattractant protein 1 (MCP-1), CC chemokine receptor 2 (CCR2) and CD163 in CRC patients with HFD was significantly higher than that in CRC patients with normal diet. When it comes to the Apcmin/+ mice, HFD consumption could induce gut dysbiosis and promote intestinal carcinogenesis, accompanying with activation of MCP-1/CCR2 axis that recruited and polarized M2 tumour-associated macrophages. Interestingly, transfer of faecal microbiota from HFD-fed mice to another batch of Apcmin/+ mice in the absence of HFD could also enhance carcinogenesis without significant body weight gain and induced MCP-1/CCR2 axis activation. HFD-induced dysbiosis could also be transmitted. Meanwhile, antibiotics cocktail treatment was sufficient to inhibit HFD-induced carcinogenesis, indicating the vital role of dysbiosis in cancer development. Conclusively, these data indicated that HFD-induced dysbiosis accelerated intestinal adenoma-adenocarcinoma sequence through activation of MCP-1/CCR2 axis, which would provide new insight into better understanding of the mechanisms and prevention for HFD-related CRC.


Subject(s)
Adenocarcinoma/metabolism , Adenoma/metabolism , Chemokine CCL2/metabolism , Colorectal Neoplasms/metabolism , Diet, High-Fat/adverse effects , Dysbiosis/metabolism , Macrophages/metabolism , Animals , Carcinogenesis/metabolism , Female , Gastrointestinal Microbiome/physiology , Humans , Male , Mice , Middle Aged , Retrospective Studies
12.
Eur J Gastroenterol Hepatol ; 32(1): 48-53, 2020 01.
Article in English | MEDLINE | ID: mdl-31651654

ABSTRACT

BACKGROUND: Data on the endoscopic resection of duodenal and papillary lesions less than 15 mm in size have been well supported by systematic studies. However, for large sessile lesions of the duodenum or papilla (LSL-D/P), surgery is often performed despite significant morbidity and mortality. This study aimed to compare the outcomes and costs between endoscopic and surgical resection of such lesions. METHODS: Patients who underwent endoscopic or surgical resection of LSL-D/P at Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University from 2013 to 2017 were retrospectively analyzed. Endoscopic and surgical outcomes and costs were compared. RESULTS: A total of 68 lesions were evaluated (47.1% of patients were male; mean lesion size 25 mm); 46 were treated by endoscopic resection, and 22 were managed by surgical resection. At the initial procedure, complete resection was achieved in 93.4%. Major complications (perforation, delayed bleeding, pancreatitis, infections and admission to the ICU) occurred in 15.3% of the endoscopic group and 22.6% of the surgical group. For recurrence at the first surveillance endoscopic examination, there was a 12.1% recurrence rate in the endoscopic group and a 5.3% recurrence rate in the surgical group (P = 0.654). Compared with surgical resection, regardless of lesion location, endoscopic resection had a shorter procedural time and hospital stay, a lower morbidity rate and was less costly. CONCLUSION: In centers specialized in complex endoscopic resection, patients with LSL-D/P would likely benefit from advanced endoscopic management, which offers a lower morbidity profile and reduced costs.


Subject(s)
Adenoma , Duodenal Neoplasms , Endoscopic Mucosal Resection , Adenoma/diagnostic imaging , Adenoma/surgery , Duodenal Neoplasms/surgery , Duodenum/surgery , Endoscopic Mucosal Resection/adverse effects , Humans , Male , Neoplasm Recurrence, Local , Retrospective Studies , Treatment Outcome
13.
Cancer Lett ; 469: 456-467, 2020 01 28.
Article in English | MEDLINE | ID: mdl-31734354

ABSTRACT

Gut microbiota dysbiosis is closely involved in intestinal carcinogenesis. A marked reduction in butyrate-producing bacteria has been observed in patients with colorectal cancer (CRC); nevertheless, the potential benefit of butyrate-producing bacteria against intestinal tumor development has not been fully investigated. We found that Clostridium butyricum (C. butyricum, one of the commonly used butyrate-producing bacteria in clinical settings) significantly inhibited high-fat diet (HFD)-induced intestinal tumor development in Apcmin/+ mice. Moreover, intestinal tumor cells treated with C. butyricum exhibited decreased proliferation and increased apoptosis. Additionally, C. butyricum suppressed the Wnt/ß-catenin signaling pathway and modulated the gut microbiota composition, as demonstrated by decreases in some pathogenic bacteria and bile acid (BA)-biotransforming bacteria and increases in some beneficial bacteria, including short-chain fatty acid (SCFA)-producing bacteria. Accordingly, C. butyricum decreased the fecal secondary BA contents, increased the cecal SCFA quantities, and activated G-protein coupled receptors (GPRs), such as GPR43 and GPR109A. The anti-proliferative effect of C. butyricum was blunted by GPR43 gene silencing using small interfering RNA (siRNA). The analysis of clinical specimens revealed that the expression of GPR43 and GPR109A gradually decreased from human normal colonic tissue to adenoma to carcinoma. Together, our results show that C. butyricum can inhibit intestinal tumor development by modulating Wnt signaling and gut microbiota and thus suggest the potential efficacy of butyrate-producing bacteria against CRC.


Subject(s)
Clostridium butyricum/metabolism , Intestinal Neoplasms/metabolism , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled/genetics , Butyrates/metabolism , Cell Proliferation/drug effects , Diet, High-Fat/adverse effects , Fatty Acids, Volatile/biosynthesis , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome/drug effects , Humans , Intestinal Neoplasms/microbiology , Intestinal Neoplasms/prevention & control , Probiotics/metabolism , Probiotics/pharmacology , Wnt Signaling Pathway/drug effects
14.
EBioMedicine ; 48: 301-315, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31594750

ABSTRACT

BACKGROUND: Accumulating evidence points to a close relationship between gut dysbiosis and colorectal cancer (CRC). As >90% of CRC develop from adenoma, we aimed to investigate the crucial role of imbalanced gut microbiota on the progression of intestinal adenoma. METHODS: The Apcmin/+ mice gavage with phosphate-buffered saline (PBS), feces from healthy controls or CRC patients after antibiotic cocktails. The intestinal tissues were isolated for histopathology, western blotting, and RNA-seq. The microbiota of feces and short-chain fatty acids (SCFAs) were analysed by 16S rDNA Amplicon Sequencing and gas chromatography. FINDINGS: The Apcmin/+mice gavaged by feces from CRC patients had more intestinal tumours compared with those fed with feces from healthy controls or PBS. Administration of feces from CRC patients increased tumour proliferation and decreased apoptosis in tumour cells, accompanied by impairment of gut barrier function and up-regulation the pro-inflammatory cytokines profile. The up-regulated the expression of ß-catenin and cyclinD1 further indicating the activation of Wnt signalling pathway. The abundance of pathogenic bacteria was increased after FMT, while producing SCFAs bacteria and SCFAs production were decreased. INTERPRETATION: Gut microbiota of CRC patients disrupted intestinal barrier, induced low-grade inflammation and dysbiosis. The altered gut microbiota enhanced the progression of intestinal adenomas in Apcmin/+mice, suggesting that a new strategy to target gut microbiota against CRC could be noted. FUND: The study was supported by the National Natural Science Foundation of China, Tianjin Research Programme of Application Foundation and Advanced Technology of China, and China Postdoctoral Science Foundation.

15.
Mol Carcinog ; 58(7): 1155-1167, 2019 07.
Article in English | MEDLINE | ID: mdl-30828892

ABSTRACT

The gut microbiota and the bile acid pool play pivotal roles in maintaining intestinal homeostasis. Bile acids are produced in the liver from cholesterol and metabolized in the intestine by the gut microbiota. Gut dysbiosis has been reported to be associated with colorectal cancer. However, the interplay between bile acid metabolism and the gut microbiota during intestinal carcinogenesis remains unclear. In the present study, we investigated the potential roles of bile acids and the gut microbiota in the cholic acid (CA; a primary bile acid)-induced intestinal adenoma-adenocarcinoma sequence. Apc min/+ mice, which spontaneously develop intestinal adenomas, were fed a diet supplemented with 0.4% CA for 12 weeks. Mice that were fed a normal diet were regarded as untreated controls. In CA-treated Apc min/+ mice, the composition of the gut microbiota was significantly altered, and CA was efficiently transformed into deoxycholic acid (a secondary bile acid) by the bacterial 7α-dehydroxylation reaction. The intestinal adenoma-adenocarcinoma sequence was observed in CA-treated Apc min/+ mice and was accompanied by an impaired intestinal barrier function and IL-6/STAT3-related low-grade inflammation. More importantly, microbiota depletion using an antibiotic cocktail globally compromised CA-induced intestinal carcinogenesis, suggesting a leading role for the microbiota during this process. Overall, our data suggested that the crosstalk between bile acids and the gut microbiota mediated intestinal carcinogenesis, which might provide novel therapeutic strategies against intestinal tumor development.


Subject(s)
Adenocarcinoma/pathology , Adenoma/pathology , Carcinogenesis/pathology , Cholic Acid/metabolism , Colorectal Neoplasms/pathology , Gastrointestinal Microbiome/physiology , Adenocarcinoma/microbiology , Adenoma/microbiology , Animals , Anti-Bacterial Agents/pharmacology , Bile Acids and Salts/metabolism , Colorectal Neoplasms/microbiology , Dietary Supplements , Dysbiosis/pathology , Female , Intestines/microbiology , Intestines/pathology , Mice , Mice, Transgenic
16.
Biomed Res Int ; 2019: 6517549, 2019.
Article in English | MEDLINE | ID: mdl-31950048

ABSTRACT

BACKGROUND: Current biomarkers have been routinely used noninvasive methods for assessing disease activity of inflammatory bowel disease (IBD), but none of them are specific. This study was aimed to determine the performance of the serological biomarkers for detecting disease activity in patients with IBD. METHODS: A prospective study that included 73 ulcerative disease (UC) subjects, 141 Crohn's disease (CD) subjects, and 30 of them complicated with C. difficile infection (CDI) were diagnosed at a single-institution IBD center. Disease activity was assessed using by Truelove and Witts criteria for UC and Harvey Bradshaw Simple Index for CD. Serological inflammatory biomarkers were compared in different severity groups. Receiver operator curve analyses assessed the performance of each biomarker in discriminating disease states. RESULTS: For UC patients, elevated monocyte counts, C-reactive protein (CRP), and decreased lymphocyte counts and lymphocyte/monocyte ratio (LMR) significantly differed between subjects with active and inactive UC. LMR of 3.1 was 76% sensitive and had a specificity of 67% for active UC. For CD patients, higher values of neutrophils, monocytes, neutrophil/lymphocyte ratio, CRP, fibrinogen, and lower values of LMR and hemoglobin were significantly different between subjects with active and inactive CD. None of the biomarkers included had a good correlation with disease activity (area under the ROC Curve < 0.70). CONCLUSIONS: A low LMR represents an inexpensive, readily available test with a promising value to identify disease activity in UC patients, whereas none of the inflammatory biomarkers showed a discriminative value in disease activity of CD.


Subject(s)
Biomarkers/blood , C-Reactive Protein/metabolism , Colitis, Ulcerative/blood , Inflammatory Bowel Diseases/blood , Adolescent , Adult , Aged , Clostridioides difficile/pathogenicity , Clostridium Infections/blood , Clostridium Infections/microbiology , Clostridium Infections/pathology , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , Crohn Disease/blood , Crohn Disease/microbiology , Crohn Disease/pathology , Female , Fibrinogen/metabolism , Humans , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Leukocyte Count , Lymphocytes/microbiology , Male , Middle Aged , Monocytes/metabolism , Monocytes/pathology , Neutrophils/metabolism , Neutrophils/pathology , Prospective Studies , Severity of Illness Index , Young Adult
17.
Biomed Res Int ; 2018: 3619748, 2018.
Article in English | MEDLINE | ID: mdl-29977909

ABSTRACT

BACKGROUND: Functional gastrointestinal disorder (FGID) patients are influenced by anxiety, depression, and low sleep quality, which reduce the quality of their life. However, epidemiological data on the quality of sleep in FGID patients were lacking. This study aims to explore the sleep quality and influencing factors of the sleep quality in FGID patients. METHODS: 1200 subjects, diagnosed as FGID in one of the six class-three hospitals in Tianjin, China, from January to December 2014, were recruited. The information about demographic information, the severity of clinical symptoms, psychological status (Zung self-rating depression scale), and sleep quality (evaluated with Pittsburgh sleep quality index) was gathered. RESULTS: The questionnaires from 1117 participants were collected including 920 of functional dyspepsia (FD) patients, 77 of irritable bowel disease (IBS) patients, 26 of functional constipation (FC) patients, and 94 other FGID patients. The results showed that morbidity rate for FD patients who had sleep disorders was higher than those who suffered from IBS or FC (P < 0.001). The proportion of elderly patients suffering from low sleep quality was higher than that of middle-aged and young patients (P < 0.001). The binary logistic regression analysis showed that age, education, and the severity of FGID symptom were influencing factors for poor sleep quality in FGID patients. CONCLUSION: The issue of poor sleep quality in FGID patients in Tianjin area is prominent, and elderly patients suffer lower sleep quality than other FGID patients. Age, education, and the severity of FGID symptoms are critical influencing factors which result in a drop-in sleep quality.


Subject(s)
Gastrointestinal Diseases/complications , Sleep Wake Disorders/etiology , Adolescent , Adult , Aged , Child, Preschool , China , Cross-Sectional Studies , Dyspepsia , Female , Humans , Irritable Bowel Syndrome/complications , Male , Middle Aged , Surveys and Questionnaires , Young Adult
18.
J Cell Mol Med ; 22(9): 4263-4273, 2018 09.
Article in English | MEDLINE | ID: mdl-29956475

ABSTRACT

High fat diet is implicated in the elevated deoxycholic acid (DCA) in the intestine and correlated with increased colon cancer risk. However, the potential mechanisms of intestinal carcinogenesis by DCA remain unclarified. Here, we investigated the carcinogenic effects and mechanisms of DCA using the intestinal tumour cells and Apcmin/+ mice model. We found that DCA could activate epidermal growth factor receptor (EGFR) and promote the release of EGFR ligand amphiregulin (AREG), but not HB-EGF or TGF-α in intestinal tumour cells. Moreover, ADAM-17 was required in DCA-induced promotion of shedding of AREG and activation of EGFR/Akt signalling pathway. DCA significantly increased the multiplicity of intestinal tumours and accelerated adenoma-carcinoma sequence in Apcmin/+ mice. ADAM-17/EGFR signalling axis was also activated in intestinal tumours of DCA-treated Apcmin/+ mice, whereas no significant change occurred in tumour adjacent tissues after DCA exposure. Conclusively, DCA activated EGFR and promoted intestinal carcinogenesis by ADAM17-dependent ligand release.


Subject(s)
ADAM17 Protein/genetics , Adenoma/genetics , Amphiregulin/genetics , Deoxycholic Acid/administration & dosage , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic , Intestinal Neoplasms/genetics , ADAM17 Protein/metabolism , Adenoma/chemically induced , Adenoma/metabolism , Adenoma/pathology , Adenomatous Polyposis Coli Protein/deficiency , Adenomatous Polyposis Coli Protein/genetics , Amphiregulin/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Epithelial Cells/metabolism , Epithelial Cells/pathology , ErbB Receptors/metabolism , HCT116 Cells , Humans , Intestinal Neoplasms/chemically induced , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
19.
Sci Rep ; 7(1): 10322, 2017 09 04.
Article in English | MEDLINE | ID: mdl-28871143

ABSTRACT

Chronic constipation is a prevalent functional gastrointestinal disorder accompanied with intestinal dysbiosis. However, causal relationship between dysbiosis and constipation remains poorly understood. Serotonin transporter (SERT) is a transmembrane transport protein which re-uptakes excessive 5-hydroxytryptamine (5-HT) from effective location to terminate its physiological effects and involves in regulating gastrointestinal motility. In this study, fecal microbiota from patients with constipation and healthy controls were transplanted into the antibiotic depletion mice model. The mice which received fecal microbiota from patients with constipation presented a reducing in intestinal peristalsis and abnormal defecation parameters including the frequency of pellet expulsion, fecal weight and fecal water content. After fecal microbiota transplantation, the SERT expression in the colonic tissue was significantly upregulated, and the content of 5-HT was decreased which negatively correlated with the gastrointestinal transit time. Moverover, fecal microbiota from the mice which received fecal microbiota from patients with constipation also upregulated SERT in Caco-2 cells. Besides, this process accompanied with the decreased abundance of Clostridium, Lactobacillus, Desulfovibrio, and Methylobacterium and an increased tend of Bacteroides and Akkermansia, which also involved in the impairment of intestinal barrier after FMT. Taken together, intestinal dysbiosis may upregulate the SERT expression and contribute to the development of chronic constipation.


Subject(s)
Constipation/etiology , Constipation/metabolism , Disease Susceptibility , Dysbiosis , Gastrointestinal Microbiome , Serotonin Plasma Membrane Transport Proteins/metabolism , Adolescent , Adult , Animals , Constipation/diagnosis , Constipation/physiopathology , Disease Models, Animal , Fecal Microbiota Transplantation , Feces/microbiology , Female , Gene Expression , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , Middle Aged , RNA, Messenger/genetics , Serotonin Plasma Membrane Transport Proteins/genetics , Young Adult
20.
Medicine (Baltimore) ; 96(9): e6233, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28248886

ABSTRACT

Gastric adenomyoma (GA) is a kind of rare gastric submucosal eminence lesions. As the malignant transformation cannot be ruled out, surgery and laparoscopic resection are usually considered. The aim of this study is to evaluate the therapeutic effect and safety of endoscopic submucosal dissection (ESD) for GA.All of the patients with gastric submucosal eminence lesions who underwent ESD from June 2008 to June 2015 in General Hospital, Tianjin Medical University, China, were identified, and patients with GA, which was confirmed by pathological evaluation, were enrolled for further analysis.Among the 571 patients who received ESD, 15 cases with uncertain diagnosis before the procedure were finally confirmed as GA. The mean age of these 15 patients was 46.93 ±â€Š15.56 years (range: 18-73). Most of the lesions were located in antrum (12/15 patients), with 2 in the body of stomach and 1 in cardia, respectively. The mean size of the lesions was 1.47 ±â€Š0.67 cm (range: 0.4-3.0). According to the endoscopic ultrasonography, the lesions of 14 patients originated from submucosa and 1 originated from superficial muscularis, totally with mixed echoes changes. En bloc complete resection was achieved in all of the lesions. No perforation, intraoperative bleeding, delayed bleeding, and mortalities occurred. No recurrence or metastasis was found during 1 to 67 months.ESD appears to be a feasible, safe, and effective treatment for GA with clinical presentation of gastric submucosal eminence lesions.


Subject(s)
Adenomyoma/surgery , Gastroscopy/methods , Stomach Neoplasms/surgery , Adenomyoma/pathology , Adolescent , Adult , Aged , Female , Humans , Male , Middle Aged , Retrospective Studies , Stomach/pathology , Stomach Neoplasms/pathology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...