Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Clin Sci (Lond) ; 127(5): 341-50, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24641356

ABSTRACT

Intestinal fibrosis with stricture formation is a complication of CD (Crohn's disease) that may mandate surgical resection. Accurate biomarkers that reflect the relative contribution of fibrosis to an individual stricture are an unmet need in managing patients with CD. The miRNA-29 (miR-29) family has been implicated in cardiac, hepatic and pulmonary fibrosis. In the present study, we investigated the expression of miR-29a, miR-29b and miR-29c in mucosa overlying a stricture in CD patients (SCD) paired with mucosa from non-strictured areas (NSCD). There was significant down-regulation of the miR-29 family in mucosa overlying SCD compared with mucosa overlying NSCD. miR-29b showed the largest fold-decrease and was selected for functional analysis. Overexpression of miR-29b in CD fibroblasts led to a down-regulation of collagen I and III transcripts and collagen III protein, but did not alter MMP (matrix metalloproteinase)-3, MMP-12 and TIMP (tissue inhibitor of metalloproteinase)-1 production. TGF (transforming growth factor)-ß1 up-regulated collagen I and III transcripts and collagen III protein as a consequence of the down-regulation of miR-29b, and TGF-ß1-induced collagen expression was reversed by exogenous overexpression of miR-29b. Furthermore, serum levels of miR-29 were lower in patients with stricturing disease compared with those without. These findings implicate the miR-29 family in the pathogenesis of intestinal fibrosis in CD and provide impetus for the further evaluation of the miR-29 family as biomarkers.


Subject(s)
Collagen Type III/biosynthesis , Collagen Type I/biosynthesis , Crohn Disease/pathology , MicroRNAs/biosynthesis , Adolescent , Adult , Aged , Collagen Type I/genetics , Collagen Type III/genetics , Constriction, Pathologic/metabolism , Crohn Disease/genetics , Down-Regulation , Fibrosis , Humans , Intestinal Mucosa/metabolism , MicroRNAs/metabolism , Middle Aged , Transforming Growth Factor beta1/pharmacology , Up-Regulation
2.
Gastro Hep Adv ; 3(3): 385-395, 2024.
Article in English | MEDLINE | ID: mdl-39131151

ABSTRACT

Background and Aims: Survival rates for esophageal squamous cell carcinoma (ESCC) are extremely low due to the late diagnosis of most cases. An understanding of the early molecular processes that lead to ESCC may facilitate opportunities for early diagnosis; however, these remain poorly defined. Tylosis with esophageal cancer (TOC) is a rare syndrome associated with a high lifetime risk of ESCC and germline mutations in RHBDF2, encoding iRhom2. Using TOC as a model of ESCC predisposition, this study aimed to identify early-stage transcriptional changes in ESCC development. Methods: Esophageal biopsies were obtained from control and TOC individuals, the latter undergoing surveillance endoscopy, and adjacent diagnostic biopsies were graded as having no dysplasia or malignancy. Bulk RNA-Seq was performed, and findings were compared with sporadic ESCC vs normal RNA-Seq datasets. Results: Multiple transcriptional changes were identified in TOC samples, relative to controls, and many were detected in ESCC. Accordingly, pathway analyses predicted an enrichment of cancer-associated processes linked to cellular proliferation and metastasis, and several transcription factors were predicted to be associated with TOC and ESCC, including negative enrichment of GRHL2. Subsequently, a filtering strategy revealed 22 genes that were significantly dysregulated in both TOC and ESCC. Moreover, Keratin 17, which was upregulated in TOC and ESCC, was also found to be overexpressed at the protein level in 'normal' TOC esophagus tissue. Conclusion: Transcriptional changes occur in TOC esophagus prior to the onset of dysplasia, many of which are associated with ESCC. These findings support the utility of TOC to help reveal the early molecular processes that lead to sporadic ESCC.

3.
BMC Cell Biol ; 14: 52, 2013 Nov 26.
Article in English | MEDLINE | ID: mdl-24279897

ABSTRACT

BACKGROUND: BORIS (CTCFL), a paralogue of the multifunctional and ubiquitously expressed transcription factor CTCF, is best known for its role in transcriptional regulation. In the nucleus, BORIS is particularly enriched in the nucleolus, a crucial compartment for ribosomal RNA and RNA metabolism. However, little is known about cytoplasmic BORIS, which represents the major pool of BORIS protein. RESULTS: We show, firstly, that BORIS has a putative nuclear export signal in the C-terminal domain. Furthermore, BORIS associates with mRNA in both neural stem cells and young neurons. The majority of the BORIS-associated transcripts are different in the two cell types. Finally, by using polysome profiling we show that BORIS is associated with actively translating ribosomes. CONCLUSION: We have demonstrated the RNA binding properties of cellular BORIS and its association with actively translating ribosomes. We suggest that BORIS is involved in gene expression at both the transcriptional and post-transcriptional levels.


Subject(s)
Cell Nucleolus/genetics , Cytoplasm/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation , Polyribosomes/genetics , RNA, Messenger/genetics , RNA, Ribosomal/genetics , Amino Acid Sequence , Cell Line, Tumor , Cell Nucleolus/metabolism , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , Humans , Molecular Sequence Data , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Polyribosomes/metabolism , Protein Binding , Protein Biosynthesis , Protein Sorting Signals , Protein Structure, Tertiary , RNA, Messenger/metabolism , RNA, Ribosomal/metabolism , Signal Transduction , Transcription, Genetic
4.
J Natl Cancer Inst ; 108(6): djv394, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26719345

ABSTRACT

BACKGROUND: The antibody cetuximab, targeting epidermal growth factor receptor (EGFR), is used to treat metastatic colorectal cancer (mCRC). Clinical trials suggest reduced benefit from the combination of cetuximab with oxaliplatin. The aim of this study was to investigate potential negative interactions between cetuximab and oxaliplatin. METHODS: Thiazolyl blue tetrazolium bromide (MTT) assay and Calcusyn software were used to characterize drug interactions. Reactive oxygen species (ROS) were measured by flow cytometry and real-time polymerase chain reaction oxidative stress arrays identified genes regulating ROS production. Chromatin immunoprecipitation (ChIP) measured signal transducer and activator of transcription 1 (STAT-1) binding to dual oxidase 2 (DUOX2) promoter. SW48, DLD-1 KRAS wild-type cell lines and DLD-1 xenograft models exposed to cetuximab, oxaliplatin, or oxaliplatin + cetuximab (control [saline]; n = 3 mice per treatment group) were used. Statistical tests were two-sided. RESULTS: Cetuximab and oxaliplatin exhibited antagonistic effects on cellular proliferation and apoptosis (caspase 3/7 activity reduced by 1.4-fold, 95% confidence interval [CI] = 0.78 to 2.11, P = .003) as opposed to synergistic effects observed with the irinotecan metabolite 7-Ethyl-10-hydroxycamptothecin (SN-38). Although both oxaliplatin and SN-38 produced ROS, only oxaliplatin-mediated apoptosis was ROS dependent. Production of ROS by oxaliplatin was secondary to STAT1-mediated transcriptional upregulation of DUOX2 (3.1-fold, 95% CI = 1.75 to 2.41, P < .001). Inhibition of DUOX2 induction and p38 activation by cetuximab reduced oxaliplatin cytotoxicity. CONCLUSIONS: Inhibition of STAT1 and DUOX2-mediated ROS generation by cetuximab impairs p38-dependent apoptosis by oxaliplatin in preclinical models and may contribute to reduced efficacy in clinical settings. Understanding the rationale for unexpected trial results will inform improved rationales for combining EGFR inhibitors with chemotherapeutic agents in future therapeutic use.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cetuximab/pharmacology , Colorectal Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , Organoplatinum Compounds/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cetuximab/therapeutic use , Chromatin Immunoprecipitation , Dual Oxidases , Heterografts , Humans , MAP Kinase Signaling System/drug effects , NADPH Oxidases/metabolism , Organoplatinum Compounds/therapeutic use , Oxaliplatin , STAT1 Transcription Factor/metabolism
5.
Cancer Epidemiol Biomarkers Prev ; 14(6): 1460-3, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15941956

ABSTRACT

Whereas a recent study reported an increased risk of colorectal cancer associated with any HFE germ line mutation (C282Y or H63D), other investigators have concluded there is no increased risk, or that any increase is dependent on polymorphisms in HFE-interacting genes such as the transferrin receptor (TFR). We have established the frequency of HFE mutations in colorectal cancer patients (n = 327) with a family history of the disease and randomly selected controls (n = 322); this design increases greatly the study's power. Genotyping for the TRF S142G polymorphism was also conducted on a large proportion of the study group. Using PCR, restriction enzyme mapping, sequencing followed by data analysis with Fisher's exact test and logistic regression, we show that the presence of any HFE mutation (Y282 or D63) was not associated with colorectal cancer risk (P = 0.57). In contrast, individuals compound heterozygous for both mutations (15 cases versus 5 controls) had thrice the odds of developing colorectal cancer (odds ratio, 3.03; 95% confidence interval, 1.06-8.61) compared with those with a single mutation. This finding did not quite reach statistical significance after allowing for multiple post hoc testing (P(observed) = 0.038 versus P = 0.025, with Bonferonni correction). Overall, our data indicate that individuals with a single HFE mutation, C282Y or H63D, are unlikely predisposed to develop colorectal cancer. However, risk of colorectal cancer might be increased by compound heterozygosity for the HFE mutations in the small number of subjects studied. TFR gene polymorphism was not an independent risk factor and did not modify the disease risk associated with HFE mutation.


Subject(s)
Colorectal Neoplasms/etiology , Colorectal Neoplasms/genetics , Germ-Line Mutation , Histocompatibility Antigens Class I/genetics , Membrane Proteins/genetics , Polymorphism, Genetic , Adult , Aged , Case-Control Studies , Female , Genotype , Hemochromatosis Protein , Heterozygote , Humans , Male , Middle Aged , Odds Ratio , Polymerase Chain Reaction , Risk Factors
6.
Cancer Res ; 75(18): 3760-70, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26206557

ABSTRACT

Mast cells (MC) are immune cells located next to the intestinal epithelium with regulatory function in maintaining the homeostasis of the mucosal barrier. We have investigated MC activities in colon inflammation and cancer in mice either wild-type (WT) or MC-deficient (Kit(W-sh)) reconstituted or not with bone marrow-derived MCs. Colitis was chemically induced with dextran sodium sulfate (DSS). Tumors were induced by administering azoxymethane (AOM) intraperitoneally before DSS. Following DSS withdrawal, Kit(W-sh) mice showed reduced weight gain and impaired tissue repair compared with their WT littermates or Kit(W-sh) mice reconstituted with bone marrow-derived MCs. MCs were localized in areas of mucosal healing rather than damaged areas where they degraded IL33, an alarmin released by epithelial cells during tissue damage. Kit(W-sh) mice reconstituted with MC deficient for mouse mast cell protease 4 did not restore normal mucosal healing or reduce efficiently inflammation after DSS withdrawal. In contrast with MCs recruited during inflammation-associated wound healing, MCs adjacent to transformed epithelial cells acquired a protumorigenic profile. In AOM- and DSS-treated WT mice, high MC density correlated with high-grade carcinomas. In similarly treated Kit(W-sh) mice, tumors were less extended and displayed lower histologic grade. Our results indicate that the interaction of MCs with epithelial cells is dependent on the inflammatory stage, and on the activation of the tissue repair program. Selective targeting of MCs for prevention or treatment of inflammation-associated colon cancer should be timely pondered to allow tissue repair at premalignant stages or to reduce aggressiveness at the tumor stage.


Subject(s)
Carcinoma/immunology , Colitis/immunology , Colonic Neoplasms/immunology , Intestinal Mucosa/physiology , Mast Cells/physiology , Regeneration/immunology , Animals , Animals, Congenic , Azoxymethane/toxicity , Carcinoma/chemically induced , Carcinoma/pathology , Cell Count , Cell Transformation, Neoplastic/immunology , Cells, Cultured , Colitis/chemically induced , Colitis/pathology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Dextran Sulfate/toxicity , Epithelial Cells/pathology , Humans , Inflammatory Bowel Diseases/pathology , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33/physiology , Mast Cells/transplantation , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Proto-Oncogene Proteins c-kit/deficiency , Proto-Oncogene Proteins c-kit/genetics , Receptors, Interleukin/physiology , Serine Endopeptidases/deficiency , Species Specificity , Specific Pathogen-Free Organisms
7.
Pathol Res Pract ; 206(1): 1-8, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19945228

ABSTRACT

The combination of host protective and tumor-promoting actions of the immune system throughout tumor development is termed cancer immunoediting. This review briefly summarizes the currently vast evidence supporting the immune system's role in not only protecting against developing cancer, but also sculpting tumor immunogenicity and immune escape. We also briefly summarize the history of immunotherapy and discuss the immunoediting process in the context of spontaneous tumor regression and whether this observation can be utilized in future treatment regimens.


Subject(s)
Immunotherapy/methods , Neoplasm Regression, Spontaneous/immunology , Neoplasms/immunology , Animals , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Humans
8.
PLoS One ; 5(5): e10714, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20502714

ABSTRACT

CD133 is a membrane molecule that has been, controversially, reported as a CSC marker in colorectal cancer (CRC). In this study, we sought to clarify the expression and role of CD133 in CRC. Initially the size of the CD133-expressing (CD133+) population in eight well-described CRC cell lines was measured by flow cytometry and was found to range from 0% to >95%. The cell line HT29 has a CD133+ population of >95% and was chosen for functional evaluation of CD133 after gene knockdown by RNA interference. A time course assay showed that CD133 inhibition had no significant effect on cell proliferation or apoptosis. However, CD133 knockdown did result in greater susceptibility to staurosporine-induced apoptosis (p = 0.01) and reduction in cell motility (p<0.04). Since gene knockdown may cause "off-target" effects, the cell line SW480 (which has a CD133+ population of 40%) was sorted into pure CD133+ and CD133- populations to allow functional comparison of isogenic populations separated only by CD133 expression. In concordance with the knockdown experiments, a time course assay showed no significant proliferative differences between the CD133+/CD133- populations. Also greater resistance to staurosporine-induced apoptosis (p = 0.008), greater cell motility (p = 0.03) and greater colony forming efficiency was seen in the CD133+ population than the CD133- population in both 2D and 3D culture (p<0.0001 and p<0.003 respectively). Finally, the plasticity of CD133 expression in tumour cells was tested. Quantitative PCR analysis showed there was transcriptional repression in the CD133- population of SW480. Prolonged culture of a pure CD133- population resulted in re-emergence of CD133+ cells. We conclude that CD133 expression in CRCs is associated with some features attributable to stemness and that there is plasticity of CD133 expression. Further studies are necessary to delineate the mechanistic basis of these features.


Subject(s)
Antigens, CD/metabolism , Biomarkers, Tumor/metabolism , Cell Movement , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Glycoproteins/metabolism , Neoplastic Stem Cells/metabolism , Peptides/metabolism , Tumor Stem Cell Assay , AC133 Antigen , Alternative Splicing/drug effects , Alternative Splicing/genetics , Antigens, CD/genetics , Apoptosis/drug effects , Base Sequence , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/genetics , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Glycoproteins/genetics , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Molecular Sequence Data , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Peptides/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Staurosporine/pharmacology
9.
Pathol Res Pract ; 206(9): 616-24, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20630662

ABSTRACT

Death-associated protein kinase (DAPK) has pro-apoptotic functions and participates in various apoptotic systems. DAPK acts as a tumor suppressor, and its inactivation by promoter hypermethylation has been frequently observed in various human cancers. As alterations of pro-apoptotic genes might cause instability in the balance of cell-turnover during chronic inflammatory processes, epigenetic silencing of DAPK might be involved in the carcinogenesis of ulcerative colitis-associated carcinoma (UCC). To evaluate the role of DAPK in the inflammation-driven carcinogenesis of ulcerative colitis (UC), we analyzed promoter hypermethylation and protein expression of DAPK using methylation-specific PCR and immunohistochemistry in 43 UCCs and paired UC-background mucosa, as well as in UC-background mucosa of 50 patients without UCC. The frequency of methylation of DAPK in UCCs was low (27.6%) compared to overall non-neoplastic UC-background mucosa (48.3%; p=0.02) and sporadic colorectal carcinoma (57.4%, p=0.019). The difference in the methylation frequency in UC-background mucosa in patients without UCC (54.2%), compared to those with UCC (40.0%), was not significant (p=0.141). Promoter methylation correlated significantly with decreased DAPK protein expression (p<0.001) and severity of inflammatory activity (p=0.024). In unmethylated UC-background mucosa, DAPK protein expression increased with activity of UC-associated inflammation, suggesting a protective role of the pro-apoptotic DAPK during the chronic inflammatory process of UC. Thus, inactivation of DAPK by promoter hypermethylation might be crucial for accumulation of DNA damage in inflamed mucosa of UC, and might therefore contribute to the initiation of the neoplastic process and development of UC-associated carcinoma.


Subject(s)
Adenocarcinoma/genetics , Apoptosis Regulatory Proteins/genetics , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cell Transformation, Neoplastic/genetics , Colitis, Ulcerative/genetics , Colonic Neoplasms/genetics , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Colitis, Ulcerative/complications , Colitis, Ulcerative/pathology , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , DNA Methylation , Death-Associated Protein Kinases , Female , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Staging , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics , Tissue Array Analysis , Young Adult
10.
Int J Colorectal Dis ; 22(1): 1-5, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16133005

ABSTRACT

PURPOSE: A comprehensive literature review was performed to examine the prevalence of anal cancer, anal intraepithelial neoplasia (AIN) and anal human papillomavirus (HPV) infection in renal transplant recipients who are at risk of anal cancer due to iatrogenic immunosuppression. METHODS: Pertinent articles were identified from searches performed on the National Center for Biotechnology Information database using the following keywords: anal cancer, AIN, screening, renal transplant (or kidney transplant), organ transplant recipients and post-transplant malignancies. RESULTS: The prevalence of AIN is 20% in renal transplant patients. The prevalence of anal HPV infection in established transplant patients is 47%, and the prevalence of anal HPV infection in new transplant patients is 23%. The relative risk for anal cancer in renal transplant patients is 10. CONCLUSIONS: As compared to HIV-positive male patients who practise anal intercourse, renal transplant patients showed a modest rise in relative risk for anal cancer. Screening programmes to detect AIN in HIV-positive patients who practise anal intercourse have been introduced on a preliminary basis in sexual health clinics in the US and may become standard practise in this population. The case for screening in renal transplant patients is unclear and would merit further investigation, especially with reference to the prevalence of anal HPV infection in this population. It may transpire that renal transplant patients would benefit more from HPV prophylaxis rather than screening for AIN.


Subject(s)
Anus Neoplasms/etiology , Carcinoma in Situ/etiology , Graft Rejection/prevention & control , Immunosuppression Therapy/adverse effects , Immunosuppressive Agents/adverse effects , Kidney Failure, Chronic/surgery , Kidney Transplantation , Anus Diseases/epidemiology , Anus Diseases/virology , Anus Neoplasms/epidemiology , Carcinoma in Situ/epidemiology , Graft Rejection/complications , Humans , Papillomavirus Infections/epidemiology , Papillomavirus Infections/etiology , Prevalence , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL