Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Oncogenesis ; 9(8): 74, 2020 Aug 19.
Article in English | MEDLINE | ID: mdl-32814764

ABSTRACT

Inflammation is an established risk factor for colorectal cancer. We and others have shown that colorectal cancer patients with elevated cysteinyl leukotriene receptor 2 (CysLT2R) and 15-hydroxyprostaglandin dehydrogenase (15-PGDH) levels exhibit good prognoses. However, both CysLT2R and 15-PGDH, which act as tumour suppressors, are often suppressed in colorectal cancer. We previously reported that leukotriene C4 (LTC4)-induced differentiation in colon cancer via CysLT2R signalling. Here, we investigated the involvement of Hedgehog (Hh)-GLI1 signalling, which is often hyperactivated in colorectal cancer. We found that the majority of colorectal cancer patients had high-GLI1 expression, which was negatively correlated with CysLT2R, 15-PGDH, and Mucin-2 and overall survival compared with the low-GLI1 group. LTC4-induced 15-PGDH downregulated both the mRNA and protein expression of GLI1 in a protein kinase A (PKA)-dependent manner. Interestingly, the LTC4-induced increase in differentiation markers and reduction in Wnt targets remained unaltered in GLI1-knockdown cells. The restoration of GLI1 in 15-PGDH-knockdown cells did not ameliorate the LTC4-induced effects, indicating the importance of both 15-PGDH and GLI1. LTC4-mediated reduction in the DCLK1 and LGR5 stemness markers in colonospheres was abolished in cells lacking 15-PGDH or GLI1. Both DCLK1 and LGR5 were highly increased in tumour tissue compared with the matched controls. Reduced Mucin-2 levels were observed both in zebrafish xenografts with GLI1-knockdown cells and in the cysltr2-/- colitis-associated colon cancer (CAC) mouse model. Furthermore, GLI1 expression was positively correlated with stemness and negatively correlated with differentiation in CRC patients when comparing tumour and mucosal tissues. In conclusion, restoring 15-PGDH expression via CysLT2R activation might benefit colorectal cancer patients.

2.
Anticancer Res ; 39(4): 1719-1728, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30952711

ABSTRACT

BACKGROUND: The wingless-type mammary tumour virus integration site 5A (WNT5A) agonist Foxy5 was shown in vitro to affect intracellular signalling implicated in the regulation of colonic cancer stem cells (CSCs). MATERIALS AND METHODS: In order to study whether Foxy5 can modulate CSCs, either HT-29 or Caco-2 human colonic cancer cells, both lacking endogenous WNT5A expression, were inoculated subcutaneously into nude mice. RESULTS: Foxy5 reduced the expression of the stem-cell marker aldehyde dehydrogenase and, interestingly, the specific colon CSC marker double cortin-like kinase 1. Foxy5 also reduced active ß-catenin and the expression of its downstream target Achaete Scute complex homolog 2, a CSC-preserving transcription factor. Foxy5 also reduced cyclo-oxygenase 2 expression, responsible for the formation of the CSC-promoting prostaglandin E2 (PGE2), but increased that of 15-hydroxyprostaglandin dehydrogenase expression, a PGE2-degrading enzyme. Accordingly, Foxy5 impairs both ß-catenin and PGE2 signalling, both of which have been implicated in promoting the niche of colonic CSCs. CONCLUSION: Our data suggest that Foxy5 can complement the traditional adjuvant chemotherapeutic treatment to which CSCs are resistant.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Oligopeptides/pharmacology , Wnt-5a Protein/agonists , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Caco-2 Cells , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Doublecortin-Like Kinases , Female , HT29 Cells , Humans , Hydroxyprostaglandin Dehydrogenases/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Retinal Dehydrogenase , Tumor Burden/drug effects , Tumor Microenvironment , Wnt Signaling Pathway/drug effects , Wnt-5a Protein/metabolism , Xenograft Model Antitumor Assays , beta Catenin/metabolism
3.
Oncotarget ; 8(21): 34773-34786, 2017 May 23.
Article in English | MEDLINE | ID: mdl-28410235

ABSTRACT

Cysteinyl leukotriene receptor 1 (CysLT1R) has been shown to be up-regulated in the adenocarcinomas of colorectal cancer patients, which is associated with a poor prognosis. In a spontaneous model of colon cancer, CysLT1R disruption was associated with a reduced tumor burden in double-mutant female mice (ApcMin/+/Cysltr1-/-) compared to ApcMin/+ littermates. In the current study, we utilized a genetic approach to investigate the effect of CysLT1R in the induced azoxymethane/dextran sulfate sodium (AOM/DSS) model of colitis-associated colon cancer. We found that AOM/DSS female mice with a global disruption of the Cysltr1 gene (Cysltr1-/-) had a higher relative body weight, a more normal weight/length colon ratio and smaller-sized colonic polyps compared to AOM/DSS wild-type counterparts. The Cysltr1-/- colonic polyps exhibited low-grade dysplasia, while wild-type polyps had an adenoma-like phenotype. The Cysltr1-/- colonic polyps exhibited significant decreases in nuclear ß-catenin and COX-2 protein expression, while the normal crypts surrounding the polyps exhibited increased Mucin 2 expression. Furthermore, Cysltr1-/- mice exhibited an overall reduction in inflammation, with a significant decrease in proinflammatory cytokines, polyp 5-LOX expression and infiltration of CD45 leukocytes and F4/80 macrophages. In conclusion, the present genetic approach in an AOM/DSS model further supports an important role for CysLT1R in colon tumorigenesis.


Subject(s)
Colitis/chemically induced , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Receptors, Leukotriene/genetics , Animals , Azoxymethane , Body Weight , Colitis/complications , Colitis/genetics , Colonic Neoplasms/genetics , Cyclooxygenase 2/metabolism , Dextran Sulfate , Disease Models, Animal , Mice , beta Catenin/metabolism
4.
BMC Cancer ; 16: 425, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27388564

ABSTRACT

BACKGROUND: Colorectal cancer is one of the most common types of cancers worldwide. Recent studies have identified cancer-initiating cells (CICs) as a subgroup of replication-competent cells in the development of colorectal cancer. Although it is understood that an inflammation-rich tumor microenvironment presumably supports CIC functions, the contributory factors are not very well defined. The present study advances our understanding of the role of the eicosanoids leukotriene D4 (LTD4) and prostaglandin E2 (PGE2) in the tumorigenic ability of CICs and investigates the consequential changes occurring in the tumor environment that might support tumor growth. METHODS: In this study we used human HCT-116 colon cancer ALDH(+) cells in a nude mouse xenograft model. Protein expression and immune cell was determined in tumor-dispersed cells by flow cytometry and in tumor sections by immunohistochemistry. mRNA expressions were quantified using RT-q-PCR and plasma cytokine levels by Multiplex ELISA. RESULTS: We observed that LTD4 and PGE2 treatment augmented CIC-induced tumor growth. LTD4-and PGE2-treated xenograft tumors revealed a robust increase in ALDH and Dclk1 protein expression, coupled with activated ß-catenin signaling and COX-2 up-regulation. Furthermore, LTD4 or PGE2 accentuated the accumulation of CD45 expressing cells within xenograft tumors. Further analysis revealed that these infiltrating immune cells consisted of neutrophils (LY6G) and M2 type macrophages (CD206(+)). In addition, LTD4 and PGE2 treatment significantly elevated the plasma levels of cysteinyl leukotrienes and PGE2, as well as levels of IL-1ß, IL-2, IL-6, TNF-α and CXCL1/KC/GRO. In addition, increased mRNA expression of IL-1ß, IL-6 and IL-10 were detected in tumors from mice that had been treated with LTD4 or PGE2. CONCLUSION: Our data suggest that both LTD4 and PGE2 promote CICs in initiating tumor growth by allowing modifications in the tumor environment. Our data indicate that new therapeutic strategies targeting eicosanoids, specifically LTD4 and PGE2, could be tested for better therapeutic management of colon cancer.


Subject(s)
Colonic Neoplasms/metabolism , Cytokines/genetics , Dinoprostone/toxicity , Leukotriene D4/toxicity , Neoplastic Stem Cells/metabolism , Animals , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Cytokines/metabolism , Doublecortin-Like Kinases , Female , HCT116 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Nude , Neoplasms, Experimental , Neoplastic Stem Cells/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Tumor Microenvironment
5.
Acta Oncol ; 55(12): 1434-1442, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27355473

ABSTRACT

BACKGROUND: Inflammatory cells and inflammatory mediators play an important role in colorectal cancer (CRC). Previous studies have shown that CRC patients with increased expression of cysteinyl leukotriene receptor 1 (CysLTR1) have a poorer prognosis, and Cysltr1-/- mice display fewer intestinal polyps. However, the role of mast cells (MCs) in colon cancer progression remains unclear. The aim of the present study was to explore the relevance of MCs in CRC. MATERIAL AND METHODS: A tissue microarray from 72 CRC patients was stained with MC anti-tryptase and -chymase antibodies. Mouse colon tissue was stained with MC anti-tryptase antibody. Immunohistochemistry was used to identify MCs in patients and mice. RESULTS: Patient colon cancer tissue had in comparison with normal colon tissue a reduced number of MCs, predominantly of chymase-positive cells. Further analysis revealed that patients with a relative high MCD in their cancer tissues showed significantly longer overall survival compared to those with a low MCD [hazard ratio (HR) 0.539; 95% confidence interval (CI), 0.302-0.961]. Similar results were observed in subgroups of patients with either no distant metastasis (p = 0.004), or <75 years (p = 0.015) at time of diagnosis. Multivariate Cox analysis showed that MCD independently correlated with reduced risk of death in colon cancer patients (HR 0.380; 95% CI 0.202-0.713). Additionally, a negative correlation was found between cytoplasmic CysLTR1 expression and number of MCs. In agreement, in the CAC mouse model, Cysltr1-/- mice showed significantly higher MCs in their polyp/tumor areas compared with wild-type mice. CONCLUSION: A high MCD in cancer tissue correlated with longer patient survival independently from other risk factors for CRC. The concept that MCs have an anti-tumor effect in CRC is further supported by the findings of a negative correlation with CysLTR1 expression in patients and a high MCD in colon polyps/tumors from CysLTR1-/- mice.


Subject(s)
Biomarkers, Tumor/analysis , Colitis/complications , Colonic Neoplasms/mortality , Colorectal Neoplasms/mortality , Mast Cells/pathology , Receptors, Leukotriene/physiology , Adult , Aged , Aged, 80 and over , Animals , Case-Control Studies , Chymases/immunology , Colitis/chemically induced , Colonic Neoplasms/etiology , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Disease Models, Animal , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Lymphatic Metastasis , Male , Mast Cells/enzymology , Mast Cells/immunology , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neoplasm Staging , Prognosis , Retrospective Studies , Survival Rate , Tissue Array Analysis , Tryptases/immunology
6.
Carcinogenesis ; 37(5): 491-9, 2016 05.
Article in English | MEDLINE | ID: mdl-26979937

ABSTRACT

There is emerging literature emphasizing the role of inflammatory eicosanoids, including prostaglandins and leukotrienes, in cancer development. Increased expression of both the cysteinyl leukotriene receptor 1 (CysLTR1) and the enzyme responsible for the production of leukotrienes, 5-lipoxygenase, is associated with poor prognosis in patients with colorectal adenocarcinomas. Apc mutation is an early event in the development of sporadic and hereditary (familial adenomatous polyposis) colorectal cancer. We utilized the Apc(Min/+) mouse model of familial adenomatous polyposis/sporadic colorectal cancer to investigate the role of CysLTR1 in intestinal tumorigenesis by crossing Apc(Min/+) mice with mice lacking the Cysltr1 gene. We could observe a reduced tumor burden in the small intestine of double-mutant female (Cysltr1 (-/-) Apc (Min/+) ) but not double-mutant male mice, compared with gender-matched single-mutant (Cysltr1 (+/+) Apc (Min/+) ) mice. This reduction was in a Cysltr1-dependent manner, female double-mutant mice having significantly reduced tumor formation compared with control littermates. The female double-mutant phenotype was accompanied with decreased systemic inflammation, as evidenced by significantly reduced serum levels of prostaglandin E2 and CysLTs, as well as increased CD3(+)CD8(+) T-cell tumor infiltration. Furthermore, the reduced formation of polyps in double-mutant (Cysltr1 (-/-) Apc (Min/+) ) female mice could in part be explained by the cytotoxic action of CD3(+)CD8(+) T cells in the polyp and reduced nuclear accumulation of ß-catenin in the epithelium of small intestinal polyps. Our results stress the important role that CysLTR1 plays in colorectal cancer and its potential as a therapeutic target in cancer therapy.


Subject(s)
Intestinal Polyps/genetics , Adenomatous Polyposis Coli Protein/genetics , Animals , Colorectal Neoplasms/genetics , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dinoprostone/blood , Dinoprostone/genetics , Female , Gene Expression Regulation, Neoplastic , Intestinal Polyps/epidemiology , Intestinal Polyps/pathology , Intestine, Small/metabolism , Intestine, Small/pathology , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Mucin-2/genetics , Mucin-2/metabolism , Neoplasms, Experimental/genetics , Receptors, Leukotriene/genetics , beta Catenin/metabolism
7.
J Biol Chem ; 291(14): 7242-55, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26846851

ABSTRACT

Retinal angiogenesis is tightly regulated to meet oxygenation and nutritional requirements. In diseases such as proliferative diabetic retinopathy and neovascular age-related macular degeneration, uncontrolled angiogenesis can lead to blindness. Our goal is to better understand the molecular processes controlling retinal angiogenesis and discover novel drugs that inhibit retinal neovascularization. Phenotype-based chemical screens were performed using the ChemBridge Diverset(TM)library and inhibition of hyaloid vessel angiogenesis in Tg(fli1:EGFP) zebrafish. 2-[(E)-2-(Quinolin-2-yl)vinyl]phenol, (quininib) robustly inhibits developmental angiogenesis at 4-10 µmin zebrafish and significantly inhibits angiogenic tubule formation in HMEC-1 cells, angiogenic sprouting in aortic ring explants, and retinal revascularization in oxygen-induced retinopathy mice. Quininib is well tolerated in zebrafish, human cell lines, and murine eyes. Profiling screens of 153 angiogenic and inflammatory targets revealed that quininib does not directly target VEGF receptors but antagonizes cysteinyl leukotriene receptors 1 and 2 (CysLT1-2) at micromolar IC50values. In summary, quininib is a novel anti-angiogenic small-molecule CysLT receptor antagonist. Quininib inhibits angiogenesis in a range of cell and tissue systems, revealing novel physiological roles for CysLT signaling. Quininib has potential as a novel therapeutic agent to treat ocular neovascular pathologies and may complement current anti-VEGF biological agents.


Subject(s)
Angiogenesis Inhibitors , Drug Discovery , Phenols , Quinolines , Retinal Neovascularization/drug therapy , Signal Transduction/drug effects , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/pharmacology , Animals , Animals, Genetically Modified , Cell Line , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/metabolism , Humans , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Mice , Phenols/chemistry , Phenols/pharmacokinetics , Phenols/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...