Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
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
3.
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
4.
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
5.
Cancer Immunol Res ; 3(11): 1227-35, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26130064

ABSTRACT

The mucosal immune response in the setting of intestinal inflammation contributes to colorectal cancer. IL10 signaling has a central role in gut homeostasis and is impaired in inflammatory bowel disease (IBD). Out of two IL10 receptor subunits, IL10R1 and IL10R2, the latter is shared among the IL10 family of cytokines and activates STAT signaling. STAT3 is oncogenic in colorectal cancer; however, knowledge about IL10 signaling upstream of STAT3 in colorectal cancer is lacking. Here, expression of IL10 signaling genes was examined in matched pairs from normal and tumor tissue from colorectal cancer patients showing overexpression (mRNA, protein) of IL10R2 and STAT3 but not IL10R1. IL10R2 overexpression was related to microsatellite stability. Transient overexpression of IL10R2 in HT29 cells increased proliferation upon ligand activation (IL10 and IL22). IL22, and not IL10, phosphorylated STAT3 along with increased phosphorylation of AKT and ERK. A significantly higher expression of IL22R1 and IL10R2 was also confirmed in a separate cohort of colorectal cancer samples. IL22 expression was elevated in gut mucosa from patients with IBD and colitis-associated cancer, which also exhibited increased expression of IL22R1 but not its coreceptor IL10R2. Overall, these data indicate that overexpression of IL10R2 and STAT3 contributes to colorectal carcinogenesis in microsatellite-stable tumors through IL22/STAT3 signaling.


Subject(s)
Carcinogenesis/immunology , Colorectal Neoplasms/immunology , Interleukin-10 Receptor beta Subunit/immunology , Aged , Aged, 80 and over , Carcinogenesis/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic/immunology , Humans , Immunity, Mucosal , Interleukin-10 Receptor beta Subunit/biosynthesis , Interleukin-10 Receptor beta Subunit/genetics , Intestinal Mucosa/immunology , Male , Microsatellite Repeats , Middle Aged , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Receptors, Interleukin/biosynthesis , STAT3 Transcription Factor/biosynthesis , Signal Transduction/immunology
6.
Inflamm Bowel Dis ; 21(2): 287-96, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25569743

ABSTRACT

BACKGROUND: Chronic gut inflammation predisposes to the development of colorectal cancer and increased mortality. Use of mesalamine (5-ASA) in the treatment of ulcerative colitis modulates the risk of neoplastic progression. p21 activated kinase 1 (PAK1) mediates 5-ASA activity by orchestrating MAPK signaling, Wnt-ß catenin pathway, and cell adhesion; all implicated in the colon carcinogenesis. We evaluated the role of PAK1 in IBD and in colitis-associated cancer (CAC). METHODS AND RESULTS: PAK1 expression was scored by immunohistochemistry in human samples from IBD, CAC, and in normal mucosa. Compared with controls, a higher PAK1 expression was detected in IBD which further increased in CAC. The consequence of PAK1 overexpression was investigated using normal diploid colon epithelial cells (HCEC-1CT), which showed higher proliferation and decreased apoptosis on overexpression of PAK1. Analysis of IBD and CAC samples showed activation of AKT (p-AKT). However, mTOR pathway was activated in IBD but not in CAC. Treatment of cells with specific inhibitors (PD98059/LY294002/rapamycin) of growth signaling pathways (MEK/PI3K/mTOR) demonstrated that in HCEC-1CT, PAK1 expression is regulated by MEK, PI3K, and mTOR. In colorectal cancer cell lines, PAK1, and beta-catenin expression correlated and inhibition of PAK1 and addition of 5-ASA elicited similar molecular affects by reducing ERK and AKT activation. Moreover, 5-ASA disrupted PAK1 interaction and colocalization with ß-catenin. CONCLUSIONS: Our data indicate that (1) PAK1 is upregulated in IBD and CAC (2) PAK1 overexpression is associated with activation of PI3K-AKT/mTOR prosurvival pathways in IBD.


Subject(s)
Apoptosis , Cell Proliferation , Colitis/pathology , Colorectal Neoplasms/pathology , Inflammatory Bowel Diseases/pathology , p21-Activated Kinases/metabolism , Blotting, Western , Case-Control Studies , Cells, Cultured , Colitis/complications , Colitis/metabolism , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/etiology , Colorectal Neoplasms/metabolism , Humans , Immunoenzyme Techniques , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Microscopy, Fluorescence , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , p21-Activated Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...