Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
Clin Microbiol Rev ; : e0013323, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38995034

ABSTRACT

SUMMARYThe human intestinal tract harbors a profound variety of microorganisms that live in symbiosis with the host and each other. It is a complex and highly dynamic environment whose homeostasis directly relates to human health. Dysbiosis of the gut microbiota and polymicrobial biofilms have been associated with gastrointestinal diseases, including irritable bowel syndrome, inflammatory bowel diseases, and colorectal cancers. This review covers the molecular composition and organization of intestinal biofilms, mechanistic aspects of biofilm signaling networks for bacterial communication and behavior, and synergistic effects in polymicrobial biofilms. It further describes the clinical relevance and diseases associated with gut biofilms, the role of biofilms in antimicrobial resistance, and the intestinal host defense system and therapeutic strategies counteracting biofilms. Taken together, this review summarizes the latest knowledge and research on intestinal biofilms and their role in gut disorders and provides directions toward the development of biofilm-specific treatments.

2.
Gut Microbes ; 16(1): 2359500, 2024.
Article in English | MEDLINE | ID: mdl-38825783

ABSTRACT

The gut microbiota has been implicated as a driver of irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD). Recently we described, mucosal biofilms, signifying alterations in microbiota composition and bile acid (BA) metabolism in IBS and ulcerative colitis (UC). Luminal oxygen concentration is a key factor in the gastrointestinal (GI) ecosystem and might be increased in IBS and UC. Here we analyzed the role of archaea as a marker for hypoxia in mucosal biofilms and GI homeostasis. The effects of archaea on microbiome composition and metabolites were analyzed via amplicon sequencing and untargeted metabolomics in 154 stool samples of IBS-, UC-patients and controls. Mucosal biofilms were collected in a subset of patients and examined for their bacterial, fungal and archaeal composition. Absence of archaea, specifically Methanobrevibacter, correlated with disrupted GI homeostasis including decreased microbial diversity, overgrowth of facultative anaerobes and conjugated secondary BA. IBS-D/-M was associated with absence of archaea. Presence of Methanobrevibacter correlated with Oscillospiraceae and epithelial short chain fatty acid metabolism and decreased levels of Ruminococcus gnavus. Absence of fecal Methanobrevibacter may indicate a less hypoxic GI environment, reduced fatty acid oxidation, overgrowth of facultative anaerobes and disrupted BA deconjugation. Archaea and Ruminococcus gnavus could distinguish distinct subtypes of mucosal biofilms. Further research on the connection between archaea, mucosal biofilms and small intestinal bacterial overgrowth should be performed.


Subject(s)
Archaea , Bacteria , Biofilms , Feces , Gastrointestinal Microbiome , Humans , Biofilms/growth & development , Archaea/classification , Archaea/metabolism , Archaea/genetics , Archaea/isolation & purification , Adult , Middle Aged , Female , Male , Bacteria/classification , Bacteria/genetics , Bacteria/metabolism , Bacteria/isolation & purification , Feces/microbiology , Colon/microbiology , Methanobrevibacter/metabolism , Methanobrevibacter/genetics , Methanobrevibacter/growth & development , Methanobrevibacter/isolation & purification , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/metabolism , Irritable Bowel Syndrome/microbiology , Irritable Bowel Syndrome/metabolism , Aged , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Ileum/microbiology , Fatty Acids, Volatile/metabolism , Young Adult , Bile Acids and Salts/metabolism
3.
Gastroenterology ; 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38876174

ABSTRACT

Gastrointestinal biofilms are highly heterogenic and spatially organized polymicrobial communities that can expand and cover large areas in the gastrointestinal tract. Gut microbiota dysbiosis, mucus disruption, and epithelial invasion are associated with pathogenic biofilms that have been linked to gastrointestinal disorders such as irritable bowel syndrome, inflammatory bowel diseases, gastric cancer, and colon cancer. Intestinal biofilms are highly prevalent in ulcerative colitis and irritable bowel syndrome patients, and most endoscopists will have observed such biofilms during colonoscopy, maybe without appreciating their biological and clinical importance. Gut biofilms have a protective extracellular matrix that renders them challenging to treat, and effective therapies are yet to be developed. This review covers gastrointestinal biofilm formation, growth, appearance and detection, biofilm architecture and signalling, human host defence mechanisms, disease and clinical relevance of biofilms, therapeutic approaches, and future perspectives. Critical knowledge gaps and open research questions regarding the biofilm's exact pathophysiological relevance and key hurdles in translating therapeutic advances into the clinic are discussed. Taken together, this review summarizes the status quo in gut biofilm research and provides perspectives and guidance for future research and therapeutic strategies.

4.
Cancer Prev Res (Phila) ; 17(7): 311-324, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38643981

ABSTRACT

Lynch syndrome (LS) is the most prevalent heritable form of colorectal cancer. Its early onset and high lifetime risk for colorectal cancer emphasize the necessity for effective chemoprevention. NFE2L2 (NRF2) is often considered a potential druggable target, and many chemopreventive compounds induce NRF2. However, although NRF2 counteracts oxidative stress, it is also overexpressed in colorectal cancer and may promote tumorigenesis. In this study, we evaluated the role of NRF2 in the prevention of LS-associated neoplasia. We found increased levels of NRF2 in intestinal epithelia of mice with intestinal epithelium-specific Msh2 deletion (MSH2ΔIEC) compared with C57BL/6 (wild-type) mice, as well as an increase in downstream NRF2 targets NAD(P)H dehydrogenase (quinone 1) and glutamate-cysteine ligase catalytic subunit. Likewise, NRF2 levels were increased in human MSH2-deficient LS tumors compared with healthy human controls. In silico analysis of a publicly accessible RNA sequencing LS dataset also found an increase in downstream NRF2 targets. Upon crossing MSH2ΔIEC with Nrf2null (MSH2ΔIECNrf2null) mice, we unexpectedly found reduced tumorigenesis in MSH2ΔIECNrf2null mice compared with MSH2ΔIEC mice after 40 weeks, which occurred despite an increase in oxidative damage in MSH2ΔIECNrf2null mice. The loss of NRF2 impaired proliferation as seen by Ki67 intestinal staining and in organoid cultures. This was accompanied by diminished WNT/ß-catenin signaling, but apoptosis was unaffected. Microbial α-diversity increased over time with the loss of NRF2 based upon 16S rRNA gene amplicon sequencing of murine fecal samples. Altogether, we show that NRF2 protein levels are increased in MSH2 deficiency and associated neoplasia, but the loss of NRF2 attenuates tumorigenesis. Activation of NRF2 may not be a feasible strategy for chemoprevention in LS. Prevention Relevance: Patients with LS have an early onset and high lifetime risk for colorectal cancer. In this study, we show that NRF2 protein levels are increased in MSH2 deficiency and associated neoplasia, but the loss of NRF2 attenuates tumorigenesis. This suggests that NRF2 may not be a tumor suppressor in this specific context.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis , Disease Models, Animal , Mice, Inbred C57BL , MutS Homolog 2 Protein , NF-E2-Related Factor 2 , Animals , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Mice , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/metabolism , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , Humans , Carcinogenesis/genetics , Carcinogenesis/pathology , Mice, Knockout , Female , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Male
5.
Gut Microbes ; 14(1): 2143218, 2022.
Article in English | MEDLINE | ID: mdl-36415023

ABSTRACT

With increasing urbanization and industrialization, the prevalence of inflammatory bowel diseases (IBDs) has steadily been rising over the past two decades. IBD involves flares of gastrointestinal (GI) inflammation accompanied by microbiota perturbations. However, microbial mechanisms that trigger such flares remain elusive. Here, we analyzed the association of the emerging pathogen atypical enteropathogenic E. coli (aEPEC) with IBD disease activity. The presence of diarrheagenic E. coli was assessed in stool samples from 630 IBD patients and 234 age- and sex-matched controls without GI symptoms. Microbiota was analyzed with 16S ribosomal RNA gene amplicon sequencing, and 57 clinical aEPEC isolates were subjected to whole-genome sequencing and in vitro pathogenicity experiments including biofilm formation, epithelial barrier function and the ability to induce pro-inflammatory signaling. The presence of aEPEC correlated with laboratory, clinical and endoscopic disease activity in ulcerative colitis (UC), as well as microbiota dysbiosis. In vitro, aEPEC strains induce epithelial p21-activated kinases, disrupt the epithelial barrier and display potent biofilm formation. The effector proteins espV and espG2 distinguish aEPEC cultured from UC and Crohn's disease patients, respectively. EspV-positive aEPEC harbor more virulence factors and have a higher pro-inflammatory potential, which is counteracted by 5-ASA. aEPEC may tip a fragile immune-microbiota homeostasis and thereby contribute to flares in UC. aEPEC isolates from UC patients display properties to disrupt the epithelial barrier and to induce pro-inflammatory signaling in vitro.


Subject(s)
Colitis, Ulcerative , Enteropathogenic Escherichia coli , Escherichia coli Infections , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Humans , Enteropathogenic Escherichia coli/genetics
6.
Sci Rep ; 12(1): 17571, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36266428

ABSTRACT

The E3 ubiquitin-ligases are important for cellular protein homeostasis and their deregulation is implicated in cancer. The E3 ubiquitin-ligase Hakai is involved in tumour progression and metastasis, through the regulation of the tumour suppressor E-cadherin. Hakai is overexpressed in colon cancer, however, the implication in colitis-associated cancer is unknown. Here, we investigated the potential role of Hakai in intestinal inflammation and cancer bowel disease. Several mouse models of colitis and associated cancer were used to analyse Hakai expression by immunohistochemistry. We also analysed Hakai expression in patients with inflamed colon biopsies from ulcerative colitis and Crohn's disease. By Hakai interactome analysis, it was identified Fatty Acid Synthase (FASN) as a novel Hakai-interacting protein. Moreover, we show that Hakai induces FASN ubiquitination and degradation via lysosome, thus regulating FASN-mediated lipid accumulation. An inverse expression of FASN and Hakai was detected in inflammatory AOM/DSS mouse model. In conclusion, Hakai regulates FASN ubiquitination and degradation, resulting in the regulation of FASN-mediated lipid accumulation, which is associated to the development of inflammatory bowel disease. The interaction between Hakai and FASN may be an important mechanism for the homeostasis of intestinal barrier function and in the pathogenesis of this disease.


Subject(s)
Colitis , Colonic Neoplasms , Ubiquitin-Protein Ligases , Animals , Mice , Cadherins/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Fatty Acid Synthases , Inflammation , Lipids , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitins , Colitis/complications , Colitis/metabolism
8.
Sci Rep ; 11(1): 13699, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34211054

ABSTRACT

Oral iron promotes intestinal tumourigenesis in animal models. In humans, expression of iron transport proteins are altered in colorectal cancer. This study examined whether the route of iron therapy alters iron transport and tumour growth. Colorectal adenocarcinoma patients with pre-operative iron deficiency anaemia received oral ferrous sulphate (n = 15), or intravenous ferric carboxymaltose (n = 15). Paired (normal and tumour tissues) samples were compared for expression of iron loading, iron transporters, proliferation, apoptosis and Wnt signalling using immunohistochemistry and RT-PCR. Iron loading was increased in tumour and distributed to the stroma in intravenous treatment and to the epithelium in oral treatment. Protein and mRNA expression of proliferation and iron transporters were increased in tumours compared to normal tissues but there were no significant differences between the treatment groups. However, intravenous iron treatment reduced ferritin mRNA levels in tumours and replenished body iron stores. Iron distribution to non-epithelial cells in intravenous iron suggests that iron is less bioavailable to tumour cells. Therefore, intravenous iron may be a better option in the treatment of colorectal cancer patients with iron deficiency anaemia due to its efficiency in replenishing iron levels while its effect on proliferation and iron metabolism is similar to that of oral iron treatment.


Subject(s)
Anemia, Iron-Deficiency/complications , Colorectal Neoplasms/complications , Ferric Compounds/therapeutic use , Ferrous Compounds/therapeutic use , Maltose/analogs & derivatives , Administration, Intravenous , Administration, Oral , Aged , Aged, 80 and over , Anemia, Iron-Deficiency/metabolism , Anemia, Iron-Deficiency/therapy , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/therapy , Female , Ferric Compounds/administration & dosage , Ferrous Compounds/administration & dosage , Humans , Iron/metabolism , Male , Maltose/administration & dosage , Maltose/therapeutic use , Middle Aged
9.
Gastroenterology ; 161(4): 1245-1256.e20, 2021 10.
Article in English | MEDLINE | ID: mdl-34146566

ABSTRACT

BACKGROUND & AIMS: Irritable bowel syndrome (IBS) and inflammatory bowel diseases result in a substantial reduction in quality of life and a considerable socioeconomic impact. In IBS, diagnosis and treatment options are limited, but evidence for involvement of the gut microbiome in disease pathophysiology is emerging. Here we analyzed the prevalence of endoscopically visible mucosal biofilms in gastrointestinal disease and associated changes in microbiome composition and metabolism. METHODS: The presence of mucosal biofilms was assessed in 1426 patients at 2 European university-based endoscopy centers. One-hundred and seventeen patients were selected for in-depth molecular and microscopic analysis using 16S ribosomal RNA gene amplicon-sequencing of colonic biopsies and fecal samples, confocal microscopy with deep learning-based image analysis, scanning electron microscopy, metabolomics, and in vitro biofilm formation assays. RESULTS: Biofilms were present in 57% of patients with IBS and 34% of patients with ulcerative colitis compared with 6% of controls (P < .001). These yellow-green adherent layers of the ileum and right-sided colon were microscopically confirmed to be dense bacterial biofilms. 16S-sequencing links the presence of biofilms to a dysbiotic gut microbiome, including overgrowth of Escherichia coli and Ruminococcus gnavus. R. gnavus isolates cultivated from patient biofilms also formed biofilms in vitro. Metabolomic analysis found an accumulation of bile acids within biofilms that correlated with fecal bile acid excretion, linking this phenotype with a mechanism of diarrhea. CONCLUSIONS: The presence of mucosal biofilms is an endoscopic feature in a subgroup of IBS and ulcerative colitis with disrupted bile acid metabolism and bacterial dysbiosis. They provide novel insight into the pathophysiology of IBS and ulcerative colitis, illustrating that biofilm can be seen as a tipping point in the development of dysbiosis and disease.


Subject(s)
Bacteria/growth & development , Biofilms/growth & development , Colitis, Ulcerative/microbiology , Colon/microbiology , Colonoscopy , Gastrointestinal Microbiome , Intestinal Mucosa/microbiology , Irritable Bowel Syndrome/microbiology , Austria , Bacteria/metabolism , Bacteria/ultrastructure , Case-Control Studies , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Colon/metabolism , Colon/pathology , Deep Learning , Germany , Humans , Image Interpretation, Computer-Assisted , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Irritable Bowel Syndrome/metabolism , Irritable Bowel Syndrome/pathology , Metabolomics , Microscopy, Confocal , Microscopy, Electron, Scanning , Predictive Value of Tests , Ribotyping
10.
Sci Rep ; 11(1): 5188, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33664327

ABSTRACT

Inflammatory bowel disease is a group of conditions with rising incidence caused by genetic and environmental factors including diet. The chelator ethylenediaminetetraacetate (EDTA) is widely used by the food and pharmaceutical industry among numerous other applications, leading to a considerable environmental exposure. Numerous safety studies in healthy animals have revealed no relevant toxicity by EDTA. Here we show that, in the presence of intestinal inflammation, EDTA is surprisingly capable of massively exacerbating inflammation and even inducing colorectal carcinogenesis at doses that are presumed to be safe. This toxicity is evident in two biologically different mouse models of inflammatory bowel disease, the AOM/DSS and the IL10-/- model. The mechanism of this effect may be attributed to disruption of intercellular contacts as demonstrated by in vivo confocal endomicroscopy, electron microscopy and cell culture studies. Our findings add EDTA to the list of food additives that might be detrimental in the presence of intestinal inflammation, but the toxicity of which may have been missed by regulatory safety testing procedures that utilize only healthy models. We conclude that the current use of EDTA especially in food and pharmaceuticals should be reconsidered. Moreover, we suggest that intestinal inflammatory models should be implemented in the testing of food additives to account for the exposure of this primary organ to environmental and dietary stress.


Subject(s)
Carcinogenesis/genetics , Colitis/pathology , Colonic Neoplasms/pathology , Edetic Acid/adverse effects , Animals , Carcinogenesis/drug effects , Colitis/chemically induced , Colitis/genetics , Colonic Neoplasms/chemically induced , Colonic Neoplasms/genetics , Disease Models, Animal , Food Additives/adverse effects , Humans , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Interleukin-10/genetics , Mice , Mice, Knockout
11.
Cell Mol Gastroenterol Hepatol ; 11(3): 892-907.e1, 2021.
Article in English | MEDLINE | ID: mdl-33189893

ABSTRACT

BACKGROUND & AIMS: p21-activated kinase-1 (PAK1) belongs to a family of serine-threonine kinases and contributes to cellular pathways such as nuclear factor-κB (NF-κB), mitogen-activated protein kinase (MAPK), phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT), and Wingless-related integration site(Wnt)/ß-catenin, all of which are involved in intestinal homeostasis. Overexpression of PAK1 is linked to inflammatory bowel disease as well as colitis-associated cancer (CAC), and similarly was observed in interleukin (IL)10 knockout (KO) mice, a model of colitis and CAC. Here, we tested the effects of PAK1 deletion on intestinal inflammation and carcinogenesis in IL10 KO mice. METHODS: IL10/PAK1 double-knockout (DKO) mice were generated and development of colitis and CAC was analyzed. Large intestines were measured and prepared for histology or RNA isolation. Swiss rolls were stained with H&E and periodic acid-Schiff. Co-immunoprecipitation and immunofluorescence were performed using intestinal organoids, SW480, and normal human colon epithelial cells 1CT. RESULTS: When compared with IL10 KO mice, DKOs showed longer colons and prolonged crypts, despite having higher inflammation and numbers of dysplasia. Crypt hyperproliferation was associated with Notch1 activation and diminished crypt differentiation, indicated by a reduction of goblet cells. Gene expression analysis indicated up-regulation of the Notch1 target hairy and enhancer of split-1 and the stem cell receptor leucin-rich repeat-containing G-protein-coupled receptor 5 in DKO mice. Interestingly, the stem cell marker olfactomedin-4 was present in colonic tissue. Increased ß-catenin messenger RNA and cytoplasmic accumulation indicated aberrant Wnt signaling. Co-localization and direct interaction of Notch1 and PAK1 was found in colon epithelial cells. Notch1 activation abrogated this effect whereas silencing of PAK1 led to Notch1 activation. CONCLUSIONS: PAK1 contributes to the regulation of crypt homeostasis under inflammatory conditions by controlling Notch1. This identifies a novel PAK1-Notch1 axis in intestinal pathophysiology of inflammatory bowel disease and CAC.


Subject(s)
Colitis-Associated Neoplasms/immunology , Colitis/immunology , Receptor, Notch1/metabolism , p21-Activated Kinases/metabolism , Animals , Cell Line , Colitis/chemically induced , Colitis/complications , Colitis/pathology , Colitis-Associated Neoplasms/pathology , Colon/drug effects , Colon/immunology , Colon/pathology , Disease Models, Animal , Female , Gene Silencing , Humans , Interleukin-10/genetics , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Male , Mice , Mice, Knockout , Organoids , Piroxicam/administration & dosage , Piroxicam/toxicity , Primary Cell Culture , Up-Regulation , Wnt Signaling Pathway/immunology , p21-Activated Kinases/genetics
12.
Haematologica ; 106(3): 782-794, 2021 03 01.
Article in English | MEDLINE | ID: mdl-32079699

ABSTRACT

Iron deficiency (ID) is globally prevalent, and apart from anemia is associated with thrombocytosis. While considered benign, studies linking thrombotic events with prior ID anemia suggest otherwise. Herein we used animal models to assess the influence of ID on thrombotic tendency. Sprague-Dawley rats were fed control or iron deficient diets and ferric carboxymaltose was used to reverse ID. Thrombosis was induced via stenosis of the inferior vena cava or damage to the right carotid artery using ferric chloride. Thrombi were evaluated histologically and via high frequency ultrasound in the venous model. ID consistently induced thrombocytosis alongside anemia. Venous thrombus growth and final dimensions in both arterial and venous thrombi were largest in ID. In both models, platelet numbers correlated with the final thrombus size, with ID thrombi having the largest platelet areas. Platelet function was also evaluated in surgically naive rats. Coagulability on thromboelastography and hemostasis on tail transection were augmented in ID. Platelet and plasma P-selectin expression were both higher in ID. Platelet adhesion and aggregation in ID was impaired under shear flow but was intact on static assays. Iron replacement therapy reversed all ID-related changes in hematological parameters, thrombus dimensions, and platelet assays. In summary, ID alone increases thrombotic tendency. Iron replacement therapy reverses these changes, making it a viable strategy for prevention of ID-related thrombotic disease. This may be of importance in patients with chronic illnesses which may already be at increased risk for thrombosis such as inflammatory bowel disease, chronic kidney disease, or cancer.


Subject(s)
Anemia, Iron-Deficiency , Thrombocytosis , Thrombosis , Anemia, Iron-Deficiency/etiology , Animals , Blood Platelets , Humans , Rats , Rats, Sprague-Dawley , Thrombocytosis/etiology , Thrombosis/etiology
13.
Z Gastroenterol ; 58(12): 1191-1200, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33291175

ABSTRACT

BACKGROUND: Iron deficiency and anemia are common findings in IBD. Treatment of anemia improves quality of life. Neurological symptoms like depression or anxiety are also common in IBD; however, their relationship with ID has not been studied in detail. METHODS: Prospective, single center, non-interventional trial in an IBD cohort (n = 98), which is generally at risk for ID. Quality of sleep (using the Pittsburgh Sleep Quality Index, Epworth Sleepiness Scale, and Insomnia Severity Index) and the presence of fatigue (Piper fatigue scale), depression (Self-rating Depression Scale [SDS]) or anxiety (Self-rating Anxiety Scale [SAS]) were related to ID (ferritin, transferrin saturation), anemia (hemoglobin), and inflammatory disease activity (CRP). RESULTS: ID was present in 35 %, anemia in 16 %, and inflammation in 30 %. The overall quality of sleep in this cohort was similar to that reported for the general population. ID, anemia, or inflammation had no influence on the PSQI (median 4.0 [CI 3.0-5.0]), the ESS 5.5 (5.0-7.0), and the ISI 4.00 (2.5-5.5). Fatigue (PFS; present in 30 %), anxiety (SAS; present in 24 %), and depression (SDS; present in 33 %) were more common than in the general population. Iron deficient and anemic patients were more likely to be depressed (p = 0.02 and p < 0.01) and showed a trend towards presence of fatigue (p = 0.06 and 0.07). Systemic inflammation as measured by CRP had no effect on any of these conditions. CONCLUSION: In this IBD cohort, ID and anemia affect depression and possibly fatigue independent of the presence of inflammation.


Subject(s)
Anemia, Iron-Deficiency/etiology , Depression/etiology , Fatigue/etiology , Inflammatory Bowel Diseases/complications , Quality of Life , Anemia, Iron-Deficiency/epidemiology , Anemia, Iron-Deficiency/psychology , Depression/epidemiology , Depression/psychology , Fatigue/epidemiology , Fatigue/psychology , Humans , Inflammatory Bowel Diseases/epidemiology , Inflammatory Bowel Diseases/psychology , Prospective Studies
14.
Int J Cancer ; 147(8): 2316-2326, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32350866

ABSTRACT

Colorectal cancer is a multifactorial disease involving inherited DNA mutations, environmental factors, gut inflammation and intestinal microbiota. Certain germline mutations within the DNA mismatch repair system are associated with Lynch syndrome tumors including right-sided colorectal cancer with mucinous phenotype and presence of an inflammatory infiltrate. Such tumors are more often associated with bacterial biofilms, which may contribute to disease onset and progression. Inflammatory bowel diseases are also associated with colorectal cancer and intestinal dysbiosis. Herein we addressed the question, whether inflammation can aggravate colorectal cancer development under mismatch repair deficiency. MSH2loxP/loxP Vill-cre mice were crossed into the IL-10-/- background to study the importance of inflammation and mucosal bacteria as a driver of tumorigenesis in a Lynch syndrome mouse model. An increase in large bowel tumorigenesis was found in double knockout mice both under conventional housing and under specific pathogen-free conditions. This increase was mostly due to the development of proximal tumors, a hotspot for tumorigenesis in Lynch syndrome, and was associated with a higher degree of inflammation. Additionally, bacterial invasion into the mucus of tumor crypts was observed in the proximal tumors. Inflammation shifted fecal and mucosal microbiota composition and was associated with enrichment in Escherichia-Shigella as well as Akkermansia, Bacteroides and Parabacteroides genera in fecal samples. Tumor-bearing double knockout mice showed a similar enrichment for Escherichia-Shigella and Parabacteroides. Lactobacilli, Lachnospiraceae and Muribaculaceae family members were depleted upon inflammation. In summary, chronic inflammation aggravates colonic tumorigenesis under mismatch repair deficiency and is associated with a shift in microbiota composition.


Subject(s)
Carcinogenesis/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/microbiology , Colorectal Neoplasms, Hereditary Nonpolyposis/parasitology , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Animals , Bacteria/pathogenicity , Biofilms/growth & development , Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Mismatch Repair/genetics , Disease Models, Animal , Dysbiosis/genetics , Dysbiosis/microbiology , Dysbiosis/pathology , Gastrointestinal Microbiome/genetics , Germ-Line Mutation/genetics , Inflammation/genetics , Inflammation/microbiology , Inflammation/pathology , Interleukin-10/genetics , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout
15.
Clin Gastroenterol Hepatol ; 18(6): 1381-1392, 2020 05.
Article in English | MEDLINE | ID: mdl-32068150

ABSTRACT

Recent evidence points to a plausible role of diet and the microbiome in the pathogenesis of both Crohn's disease (CD) and Ulcerative Colitis (UC). Dietary therapies based on exclusion of table foods and replacement with nutritional formulas and/or a combination of nutritional formulas and specific table foods may induce remission in CD. In UC, specific dietary components have also been associated with flare of disease. While evidence of varying quality has identified potential harmful or beneficial dietary components, physicians and patients at the present time do not have guidance as to which foods are safe, may be protective or deleterious for these diseases. The current document has been compiled by the nutrition cluster of the International Organization for the Study of Inflammatory Bowel Diseases (IOIBD) based on the best current evidence to provide expert opinion regarding specific dietary components, food groups and food additives that may be prudent to increase or decrease in the diet of patients with inflammatory bowel diseases to control and prevent relapse of inflammatory bowel diseases.


Subject(s)
Colitis, Ulcerative , Crohn Disease , Inflammatory Bowel Diseases , Microbiota , Diet , Humans
16.
Nutrients ; 12(1)2019 Dec 23.
Article in English | MEDLINE | ID: mdl-31877961

ABSTRACT

Inflammatory bowel disease increases the odds of developing colitis-associated cancer. We hypothesized that Western-style diet (WD) aggravates azoxymethane (AOM)/dextran sulfate sodium salt (DSS)-induced colitis-associated tumorigenesis and that switching to the standard AIN93G diet will ameliorate disease symptoms even after cancer initiation. Female BALB/c mice received either WD (WD group) or standard AIN93G diet (AIN group) for the whole experimental period. After five weeks, the mice received 12.5 mg/kg AOM intraperitoneally, followed by three DSS cycles. In one group of mice, the WD was switched to AIN93G the day before starting the first DSS cycle (WD/AIN group). Feeding the WD during the whole experimental period aggravated colitis symptoms, shortened the colon (p < 0.05), changed microbiota composition and increased tumor promotion. On molecular level, the WD reduced proliferation (p < 0.05) and increased expression of the vitamin D catabolizing enzyme Cyp24a1 (p < 0.001). The switch to the AIN93G diet ameliorated this effect, reflected by longer colons, fewer (p < 0.05) and smaller (p < 0.01) aberrant colonic crypt foci, comparable with the AIN group. Our results show that switching to a healthy diet, even after cancer initiation is able to revert the deleterious effect of the WD and could be an effective preventive strategy to reduce colitis symptoms and prevent tumorigenesis.


Subject(s)
Colitis/chemically induced , Colitis/complications , Colorectal Neoplasms/prevention & control , Diet, Healthy , Diet, Western/adverse effects , Aberrant Crypt Foci/pathology , Animals , Azoxymethane/administration & dosage , Colon/pathology , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , Dextran Sulfate/administration & dosage , Disease Models, Animal , Female , Gastrointestinal Microbiome/physiology , Liver/enzymology , Mice , Mice, Inbred BALB C , Vitamin D/metabolism
17.
Acta Haematol ; 142(1): 30-36, 2019.
Article in English | MEDLINE | ID: mdl-30970351

ABSTRACT

Inflammatory bowel disease (IBD) is a group of chronic relapsing inflammatory disorders affecting the large and small intestine, with a rising worldwide incidence and prevalence. Anaemia is the most common extraintestinal manifestation of IBD, correlating with disease activity, and tending to relapse even after successful therapy. Iron deficiency is the most common cause; however, it often manifests in combination with anaemia of inflammation. As such, multiple parameters are used for the diagnosis of iron deficiency anaemia in IBD. Timely recognition and selection of appropriate therapy leads to an improvement in the quality of life and prevention of potential sequelae. Oral iron can be effective under specific circumstances; however, as luminal iron changes microbiota and bacterial metabolism, oral administration should be avoided. Intravenous iron is preferred as it bypasses the sites of inflammation. Nevertheless, the optimization of IBD treatment should occur simultaneously, as this improves both patient condition and response to iron therapy. Herein, we discuss the screening, diagnosis, selection of therapy, and follow-up for iron deficiency anaemia in IBD.


Subject(s)
Anemia, Iron-Deficiency/drug therapy , Inflammatory Bowel Diseases/pathology , Iron/administration & dosage , Anemia, Iron-Deficiency/complications , Anemia, Iron-Deficiency/pathology , Humans , Inflammatory Bowel Diseases/complications , Lymphocytes/cytology , Lymphocytes/metabolism , Quality of Life , Severity of Illness Index , Thrombocytosis/complications , Thrombocytosis/pathology
18.
Sci Rep ; 9(1): 2842, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30809073

ABSTRACT

Disruption of mucosal structure and barrier function contribute to the pathogenesis of inflammatory bowel disease (IBD). Efficacy of therapy in IBD is based on endoscopic mucosal healing, which occurs by a dynamic interplay of epithelial cell regeneration, migration and differentiation. Both mesalamine (5-ASA) and azathioprine (AZTP) promote this process through mechanisms not clearly understood. We examined molecular pathways implicated in epithelial barrier function that were altered by 5-ASA and AZTP. Paracellular permeability induced by inflammatory mediators was mitigated by both compounds through restoration of cellular anchoring complexes. 5-ASA and AZTP induced rearrangement and membranous localization of junctional proteins and modulated genes involved in tight junctions. Intestinal organoids from wildtype-mice treated with TNF-α and IL-10- deficient-mice displayed impaired epithelial barrier with loss of membranous E-cadherin and reduced Desmoglein-2 expression. These effects were counteracted by 5-ASA and AZTP. Unlike AZTP that exhibited antiproliferative effects, 5-ASA promoted wound healing in colon epithelial cells. Both affected cellular senescence, cell cycle distribution and restricted cells in G1 or S phase without inducing apoptosis. This study provides mechanistic evidence that molecular actions of 5-ASA and AZTP on intestinal epithelia are fundamental in the resolution of barrier dysfunction.


Subject(s)
Azathioprine/pharmacology , Epithelial Cells/drug effects , Inflammation , Inflammatory Bowel Diseases/physiopathology , Intestines/drug effects , Mesalamine/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal , Azathioprine/therapeutic use , Colitis , Epithelial Cells/physiology , Inflammatory Bowel Diseases/drug therapy , Intestines/physiopathology , Mesalamine/therapeutic use , Mice , Wound Healing
19.
Semin Hematol ; 55(4): 256-261, 2018 10.
Article in English | MEDLINE | ID: mdl-30502855

ABSTRACT

Iron deficiency (ID) workup is a common challenge for gastrointestinal endoscopy. In premenopausal women current guidelines recommend serologic evaluation of coeliac disease only. Here we systematically tested serologic screening for autoimmune gastritis (AIG) in a large cohort of patients with ID. This is a retrospective analysis of patients who attended an out-patient clinic specialized for ID. Patients with ferritin <50 µg/L or transferrin saturation <15% were included. Laboratory workup included endomysial antibodies and parietal-cell antibodies (PCA). Upper gastrointestinal endoscopy with pH-measurement of gastric juice and histology was performed to confirm positive serologic results. Three hundred seventy-three patients with ID were included, about half of whom were anemic. Patients were predominately female with a median age of 40 (confidence interval 11). Positive endomysial antibodies were found in 4 (1%) patients, elevated levels of PCA (>20 U/mL) were found in 69 (18.5%) patients, PCA >100 U/mL in 23 (6.2%). Twenty-six were followed up by gastroscopy; in 12 of 26 patients the diagnosis of AIG was confirmed by histology with 2 additional patients diagnosed as early and/or questionable AIG. A sensitivity of 93% and a specificity of 98% were estimated for a PCA cut-off of 100 U/mL. In 20 patients gastric pH was measured. Achlorhydria was found in 7 patients all diagnosed with AIG. In this ID cohort AIG is by far more common than coeliac disease. PCA above 100 U/mL are a sensitive and specific cut-off for workup of patients with ID prior to endoscopy. Serologic suspicion of AIG helps preselection of patients for endoscopic workup for ID.


Subject(s)
Anemia, Iron-Deficiency/complications , Autoantibodies/metabolism , Gastritis/etiology , Parietal Cells, Gastric/pathology , Adult , Anemia, Iron-Deficiency/pathology , Female , Gastritis/pathology , Humans , Incidence , Male , Retrospective Studies
20.
Int J Surg Case Rep ; 44: 110-113, 2018.
Article in English | MEDLINE | ID: mdl-29499513

ABSTRACT

INTRODUCTION: Intestinal interposition is a term that describes rare anatomic variations where parts of the colon deviate from their normal intraabdominal position, attaching between two organs. Most patients with colonic interpositions are asymptomatic and diagnosed incidentally by computed tomography or ultrasound. Here we present a case of a symptomatic restrogastric colon, interposing kinked between stomach and pancreas. PRESENTATION OF CASE: A 66-year old female patient presented with an eight-year history of intermittent spastic bowel movements, epigastralgia and nausea. Consecutively, the patient lost 12 kg. Physical examination was unremarkable and routine blood tests were within normal limits. Subsequently performed colonoscopy and cross-sectional imaging diagnosed a retrogastric colon. Finally, the patient underwent surgical treatment. The intraoperative findings were consistent with the computed tomography images and showed a kinked retrogastric protrusion of the transverse colon into the lesser sac, adhering to both, the posterior wall of the stomach, and the anterior surface of the pancreas. After adhesiolysis and mobilization, the transverse colon slipped back to the normal position within the abdominal cavity. The patient recovered well after surgery and was discharged on the sixth postoperative day. Six-month follow-up revealed cured bowel function, weight regain and no signs of recurrence. DISCUSSION & CONCLUSION: These rare cases of intestinal interpositions are very often difficult to diagnose, as symptoms are misleading. In case of diagnosis adequate surgical treatment strategies should be considered.

SELECTION OF CITATIONS
SEARCH DETAIL