Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
2.
Cancer Prev Res (Phila) ; 8(3): 222-30, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25604134

ABSTRACT

Colorectal cancer progresses through multiple distinct stages that are potentially amenable to chemopreventative intervention. Epidermal growth factor receptor (EGFR) inhibitors are efficacious in advanced tumors including colorectal cancer. There is significant evidence that EGFR also plays important roles in colorectal cancer initiation, and that EGFR inhibitors block tumorigenesis. We performed a double-blind randomized clinical trial to test whether the EGFR inhibitor erlotinib given for up to 30 days had an acceptable safety and efficacy profile to reduce EGFR signaling biomarkers in colorectal aberrant crypt foci (ACF), a subset of which progress to colorectal cancer, and normal rectal tissue. A total of 45 patients were randomized to one of three erlotinib doses (25, 50, and 100 mg) with randomization stratified by nonsteroidal anti-inflammatory drug (NSAID) use. There were no unanticipated adverse events with erlotinib therapy. Erlotinib was detected in both normal rectal mucosa and ACFs. Colorectal ACF phosphorylated ERK (pERK), phosphorylated EGFR (pEGFR), and total EGFR signaling changes from baseline were modest and there was no dose response. Overall, this trial did not meet is primary efficacy endpoint. Colorectal EGFR signaling inhibition by erlotinib is therefore likely insufficient to merit further studies without additional prescreening stratification or potentially longer duration of use.


Subject(s)
Aberrant Crypt Foci/drug therapy , Colorectal Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Rectum/drug effects , Aberrant Crypt Foci/metabolism , Aberrant Crypt Foci/pathology , Case-Control Studies , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Double-Blind Method , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Staging , Phosphorylation/drug effects , Prognosis , Rectum/metabolism , Rectum/pathology , Signal Transduction/drug effects
3.
Cancer Res ; 73(12): 3771-82, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23610444

ABSTRACT

Colon cancer is one of the deadliest cancers worldwide because of its metastasis to other essential organs. Metastasis of colon cancer involves a complex set of events, including epithelial-to-mesenchymal transition (EMT) that increases invasiveness of the tumor cells. Here, we show that the xeroderma pigmentosum group E (XPE) gene product, damaged DNA-binding protein (DDB)-2, is downregulated in high-grade colon cancers, and it plays a dominant role in the suppression of EMT of the colon cancer cells. Depletion of DDB2 promotes mesenchymal phenotype, whereas expression of DDB2 promotes epithelial phenotype. DDB2 constitutively represses genes that are the key activators of EMT, indicating that DDB2 is a master regulator of EMT of the colon cancer cells. Moreover, we observed evidence that DDB2 functions as a barrier for EMT induced by hypoxia and TGF-ß. Also, we provide evidence that DDB2 inhibits metastasis of colon cancer. The results presented here identify a transcriptional regulatory pathway of DDB2 that is directly linked to the mechanisms that suppress metastasis of colon cancer.


Subject(s)
Colonic Neoplasms/genetics , DNA-Binding Proteins/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Animals , Blotting, Western , Cadherins/metabolism , Cell Hypoxia , Cell Line, Tumor , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , DNA-Binding Proteins/metabolism , HCT116 Cells , Humans , Immunohistochemistry , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Male , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Mice, SCID , Neoplasm Invasiveness , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous
4.
Int J Cancer ; 131(11): 2553-61, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22488198

ABSTRACT

Estrogen receptor-beta (ERß) has been suggested to exert anti-inflammatory and anti-tumorigenic effects in the colon, providing a translational potential to prevent and/or treat inflammatory bowel disease (IBD) and its progression to colitis-associated colorectal cancer (CAC). However, the specific direct role of ERß in CAC has not yet been tested. We assessed the effects of ERß deficiency in the azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced CAC model using ERß knockout (ßERKO) mice and wild-type (WT) littermates. These mice were injected with AOM followed by 1 week of DSS treatment, and sacrificed on weeks 9 or 16. ßERKO mice developed more severe clinical colitis compared to WT mice, as evidenced by significantly higher disease activity index after DSS treatment, weight to length ratio of the colons, inflammation score and grade of dysplasia. ERß-deficient colons presented greater number and size of polyps at weeks 9 and 16, respectively, and were characterized by a significant increase in interleukin (IL)-6, IL-17, tumor necrosis factor alpha and interferon-gamma mRNA levels. Furthermore, higher protein expression levels of nuclear factor-kappa B, inducible nitric oxide synthase, ß-catenin, proliferating cell nuclear antigen, mucin-1 and significantly lower caveolin-1 and mucin-2 protein levels were shown in ßERKO mice compared to WT mice. These data suggest a possible anti-inflammatory and anti-neoplastic mechanism of action of ERß in CAC. These results demonstrate for the first time that ERß provides protection in the AOM/DSS-induced CAC model in mice, suggesting a preventive and/or therapeutic potential for the use of ERß-selective agonists in IBD.


Subject(s)
Colitis/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Estrogen Receptor beta/metabolism , Neoplasms/genetics , Neoplasms/pathology , Animals , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Differentiation/genetics , Colitis/genetics , Colitis/metabolism , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/metabolism , Estradiol/blood , Estrogen Receptor beta/genetics , Female , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-17/genetics , Interleukin-17/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucin-1/genetics , Mucin-1/metabolism , Mucin-2/genetics , Mucin-2/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasms/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , beta Catenin/genetics , beta Catenin/metabolism
5.
J Endocrinol ; 212(2): 207-15, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22068926

ABSTRACT

CYP24 is a well-established vitamin D receptor (VDR) target gene. The active VDR ligand 1,25(OH)2D3 regulates its own catabolism by increasing CYP24 expression. It is well known that in the presence of 1,25(OH)2D3, VDR binds to VDREs in the promoter region of CYP24 and initiates CYP24 transcription. However, little is known about the role of 1,25(OH)2D3 in the posttranscriptional modulation of CYP24. In this study, we investigated the functional significance of 1,25(OH)2D3 in CYP24 RNA splicing in colon cancer cells. Using RT-PCR, we found that 1,25(OH)2D3 actively induces CYP24 splicing in a time-dependent manner and CYP24 splicing pattern could be cell type or tissue specific. The induction of RNA splicing by 1,25(OH)2D3 was mainly CYP24 selective. Treatment of cells with parathyroid hormone inhibited basal CYP24 splicing, but failed to inhibit 1,25(OH)2D3-induced CYP24 splicing. Further experiments demonstrated that new RNA synthesis was required for the induction of CYP24 splicing by vitamin D. In addition, alteration of multiple signaling pathways also affected CYP24 splicing and cellular sensitivity in response to vitamin D appeared to correlate with the induction of CYP24 splicing. These results suggest that 1,25(OH)2D3 not only regulates CYP24 transcription, but also plays an important role in posttranscriptional modulation of CYP24 by inducing its splicing. Our findings reveal an additional regulatory step that makes the vitamin D mediated action more prompt and efficient.


Subject(s)
Calcitriol/metabolism , Colonic Neoplasms/metabolism , Neoplasm Proteins/metabolism , RNA Splicing , Steroid Hydroxylases/metabolism , Aberrant Crypt Foci/metabolism , Aberrant Crypt Foci/pathology , Biopsy , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Colon/metabolism , Colon/pathology , Colonic Neoplasms/pathology , Humans , Molecular Weight , Neoplasm Proteins/genetics , Osmolar Concentration , Parathyroid Hormone/analogs & derivatives , Parathyroid Hormone/metabolism , Protein Kinase Inhibitors/pharmacology , RNA Splicing/drug effects , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Signal Transduction/drug effects , Steroid Hydroxylases/chemistry , Steroid Hydroxylases/genetics , Vitamin D3 24-Hydroxylase
6.
Cancer Prev Res (Phila) ; 4(3): 354-64, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21372035

ABSTRACT

Curcumin is derived from the spice tumeric and has antiinflammatory and antineoplastic effects in vitro and in animal models, including preventing aberrant crypt foci (ACF) and adenomas in murine models of colorectal carcinogenesis. Inhibiting the production of the procarcinogenic eicosanoids prostaglandin E2 (PGE2) and 5-hydroxyeicosatetraenoic acid (5-HETE) can suppress carcinogenesis in rodents. Curcumin reduces mucosal concentrations of PGE2 (via inhibition of cyclooxygenases 1 and 2) and 5-HETE (via inhibition of 5-lipoxygenase) in rats. Although preclinical data support curcumin activity in many sites, the poor bioavailability reported for this agent supports its use in the colorectum. We assessed the effects of oral curcumin (2 g or 4 g per day for 30 days) on PGE2 within ACF (primary endpoint), 5-HETE, ACF number, and proliferation in a nonrandomized, open-label clinical trial in 44 eligible smokers with eight or more ACF on screening colonoscopy. We assessed pre- and posttreatment concentrations of PGE2 and 5-HETE by liquid chromatography tandem mass spectroscopy in ACF and normal-tissue biopsies; ACF number via rectal endoscopy; proliferation by Ki-67 immunohistochemistry; and curcumin concentrations by high-performance liquid chromatography in serum and rectal mucosal samples. Forty-one subjects completed the study. Neither dose of curcumin reduced PGE2 or 5-HETE within ACF or normal mucosa or reduced Ki-67 in normal mucosa. A significant 40% reduction in ACF number occurred with the 4-g dose (P < 0.005), whereas ACF were not reduced in the 2-g group. The ACF reduction in the 4-g group was associated with a significant, five-fold increase in posttreatment plasma curcumin/conjugate levels (versus pretreatment; P = 0.009). Curcumin was well tolerated at both 2 g and 4 g. Our data suggest that curcumin can decrease ACF number, and this is potentially mediated by curcumin conjugates delivered systemically.


Subject(s)
Colorectal Neoplasms/pathology , Colorectal Neoplasms/prevention & control , Curcumin/pharmacology , Gene Expression Regulation, Neoplastic , Adult , Aged , Anticarcinogenic Agents/pharmacology , Biopsy/methods , Dinoprostone/antagonists & inhibitors , Endoscopy/methods , Female , Humans , Ki-67 Antigen/biosynthesis , Male , Middle Aged , Smoking , Treatment Outcome
7.
Am J Pathol ; 178(2): 672-8, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21281799

ABSTRACT

Epithelial cells lining the adult colon do not normally express gastrin-releasing peptide (GRP) or its receptor (GRPR). In contrast, GRP/GRPR can be aberrantly expressed in colon cancer where they are associated with improved patient survival rates. However, the mechanism of action whereby these proteins mediate their beneficial effects is not known. Heterochromatin protein 1 is an epigenetic modifier of gene transcription for which three different isoforms exist in humans: HP1(Hsα), HP1(Hsß), and HP1(Hsγ). In breast cancer and melanoma, respectively, HP1(Hsα) and HP1(Hsß) have been shown to modulate the aggressiveness of tumor cells in vivo. In contrast, the role of HP1 in colon cancer has not been elucidated, and a mechanism of regulating the expression of any HP1 isoform in any context has not yet been identified. In this article we demonstrate that abrogating GRP/GRPR signaling specifically down-regulates HP1(Hsß) expression and that inhibiting GRPR signaling, or ablating HP1(Hsß) expression, increases colon cancer cell invasiveness in vitro. These findings identify for the first time a signaling pathway regulating heterochromatin protein expression and suggest a mechanism whereby aberrantly expressed GRPR might alter the outcome of patients with colorectal cancer.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Gastrin-Releasing Peptide/metabolism , Signal Transduction , Adult , Cell Line, Tumor , Cell Nucleus/metabolism , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Collagen/metabolism , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness , RNA, Small Interfering/metabolism , Receptors, Bombesin/metabolism , Time Factors
8.
Cancer Prev Res (Phila) ; 4(2): 259-69, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21209397

ABSTRACT

Sulindac, atorvastatin, or prebiotic dietary fiber may reduce colorectal cancer (CRC) risk. However, clinical trial data are currently limited. We conducted a randomized, phase II chemoprevention trial involving subjects 40 years or older, with previously resected colon cancer or multiple/advanced colorectal adenomas. Magnification chromoendoscopy (MCE) was performed to identify and characterize rectal aberrant crypt foci (ACF); eligibility criteria required five or more rectal ACFs at baseline. Intervention assignments were as follows: (a) atorvastatin 20 mg qd; (b) sulindac 150 mg bid; (c) oligofructose-enriched inulin (as ORAFTI®Synergy1) 6 gm bid; or (d) control (maltodextrin) 6 gm bid, for 6 months. Percent change in rectal ACF number (%ΔACF) within arm was the primary endpoint. Secondary endpoints included changes in proliferation (Ki67) and apoptosis (caspase-3), as measured from normal mucosa biopsy samples. Among 85 eligible randomized subjects, 76 (86%) completed the trial per protocol. The median (range) of rectal ACF was 9 (5-34) and 8 (0-37) at baseline and postintervention, respectively. The median (SD) for %ΔACF was 5.6 (-69% to 143%), -18.6 (-83% to 160%), -3.6 (-88% to 83%), and -10.0 (-100% to 117%) in the atorvastatin, sulindac, ORAFTI®Synergy1 and control arms, respectively. Neither within-arm (P = 0.12-0.59) nor between-arm (P = 0.30-0.92) comparisons of %ΔACF were statistically significant. The active and control interventions also seemed to have similar effects on mucosal proliferation and apoptosis (P > 0.05 for each comparison). Data from this multicenter, phase II trial do not provide convincing evidence of CRC risk reduction from 6-month interventions with atorvastatin, sulindac, or ORAFTI®Synergy1, although statistical power was limited by the relatively small sample size.


Subject(s)
Aberrant Crypt Foci/prevention & control , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/prevention & control , Dietary Fiber/therapeutic use , Heptanoic Acids/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Pyrroles/therapeutic use , Sulindac/therapeutic use , Aberrant Crypt Foci/pathology , Aged , Atorvastatin , Colorectal Neoplasms/pathology , Female , Humans , Intestinal Mucosa/drug effects , Male , Middle Aged , Survival Rate , Treatment Outcome
9.
Clin Epigenetics ; 2(2): 331-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-22704345

ABSTRACT

Epithelial cells lining the adult colon do not normally express gastrin-releasing peptide (GRP) or its receptor (GRPR). In contrast, GRP/GRPR can be aberrantly expressed in human colorectal cancer (CRC) including Caco-2 cells. We have previously shown that GRPR activation results in the up-regulation of HP1ß, an epigenetic modifier of gene transcription. The aim of this study was to identify the genes whose expression is altered by HP1ß subsequent to GRPR activation. We determined HP1ß binding positions throughout the genome using chromatin immunoprecipitation followed by massively parallel DNA sequencing (ChIP-seq). After exposure to GRP, we identified 9,625 genomic positions occupied by HP1ß. We performed gene microarray analysis on Caco-2 cells in the absence and presence of a GRPR specific antagonist as well as siRNA to HP1ß. The expression of 97 genes was altered subsequent to GRPR antagonism, while the expression of 473 genes was altered by HP1ß siRNA exposure. When these data were evaluated in concert with our ChIP-seq findings, 9 genes showed evidence of possible altered expression as a function of GRPR signaling via HP1ß. Of these, genomic PCR of immunoprecipitated chromatin demonstrated that GRPR signaling affected the expression of IL1RAPL2, FAM13A, GBE1, PLK3, and SLCO1B3. These findings provide the first evidence by which GRPR aberrantly expressed in CRC might affect tumor progression.

10.
Dig Dis Sci ; 56(2): 339-51, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21140215

ABSTRACT

BACKGROUND: Lubiprostone, used clinically (b.i.d.) to treat constipation, has been reported to increase transepithelial Cl(-) transport in T84 cells by activating ClC-2 channels. AIM: To identify the underlying signaling pathway, we explored the effects of short-term and overnight lubiprostone treatment on second messenger signaling and Cl(-) transport. METHODS: Cl(-) transport was assessed either as I(sc) across T84 monolayers grown on Transwells and mounted in Ussing chambers or by the iodide efflux assay. [cAMP](i) was measured by enzyme immunoassay, and [Ca(2+)](i) by Fluo-3 fluorescence. Quantitation of apical cell surface CFTR protein levels was assessed by Western blotting and biotinylation with the EZ-Link Sulfo-NHS-LC-LC-Biotin. ClC-2 mRNA level was studied by RT-PCR. RESULTS: Lubiprostone and the cAMP stimulator, forskolin, caused comparable and maximal increases of I(sc) in T84 cells. The I(sc) effects of lubiprostone and forskolin were each suppressed if the tissue had previously been treated with the other agent. These responses were unaltered even if the monolayers were treated with lubiprostone overnight. Lubiprostone-induced increases in iodide efflux were ~80% of those obtained with forskolin. Lubiprostone increased [cAMP](i). H89, bumetanide, or CFTR(inh)-172 greatly attenuated lubiprostone-stimulated Cl(-) secretion, whereas the ClC-2 inhibitor CdCl(2) did not. Compared to controls, FSK-treatment increased membrane-associated CFTR by 1.9 fold, and lubiprostone caused a 2.6-fold increase in apical membrane CFTR as seen by immunoblotting following cell surface biotinylation. CONCLUSIONS: Lubiprostone activates Cl(-) secretion in T84 cells via cAMP, protein kinase A, and by increasing apical membrane CFTR protein.


Subject(s)
Alprostadil/analogs & derivatives , Carcinoma/metabolism , Chlorides/metabolism , Cyclic AMP/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Alprostadil/pharmacology , Benzoates/pharmacology , Biological Transport/drug effects , Bumetanide/pharmacology , Cathartics/pharmacology , Cell Line, Tumor , Cell Membrane/metabolism , Colforsin/pharmacology , Colonic Neoplasms/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Humans , Lubiprostone , Thiazolidines/pharmacology
11.
Gastroenterology ; 138(7): 2410-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20197068

ABSTRACT

BACKGROUND & AIMS: Mechanisms for age restriction of rotavirus diarrhea are unclear. Because rotavirus primarily infects small intestine, colonic contribution has not been widely studied. Recent data suggest that colonic secretion postbacterial infection is mediated by galanin-1 receptors (Gal1-R). We evaluated age-dependent expression of Gal1-R in Rhesus rotavirus (RRV)-infected mice and its contribution to fluid secretion. METHODS: Twenty-four hours after infection of C57BL/6J mice (wild type or Gal1-R knockout) with RRV or vehicle, closed small intestinal and colon loops were constructed. Net fluid content of the loops was calculated (milligrams/centimeters) at 2 hours post-treatment with galanin, galanin antibody, or lidocaine. Gal1-R expression was quantified by automated chromogen analysis. RESULTS: Viral antigen was detected in small intestinal epithelial cells but not in colon. Developmental Gal1-R was widely expressed in the small intestine but minimally in the colon. Postinfection, markedly increased Gal1-R was seen in the colon but not after day 25. Galanin caused a significantly higher increase in the net fluid content of infected colon than small intestine. Treatment with lidocaine reduced net fluid secretion in the small intestine and the colon. Mean diarrheal scores were significantly reduced in Gal1-R knockout mice compared with wild type (1.19 +/- 0.31, n = 22 vs 3.36 +/- 0.50, n = 35, P = .0001). CONCLUSIONS: These data show that RRV infection of the small intestine increases colonic secretion through Gal1-R and provide a promising start toward understanding the age restriction of rotavirus diarrhea.


Subject(s)
Colon/metabolism , Intestine, Small/virology , Receptor, Galanin, Type 1/physiology , Rotavirus Infections/metabolism , Age Factors , Animals , Colon/virology , Diarrhea/etiology , Galanin/pharmacology , Lidocaine/pharmacology , Mice , Mice, Inbred C57BL , Rotavirus Infections/complications
12.
Clin Exp Metastasis ; 26(7): 663-71, 2009.
Article in English | MEDLINE | ID: mdl-19430935

ABSTRACT

Epithelial cells lining the adult human colon do not normally express gastrin releasing peptide (GRP) or its receptor (GRPR), but both can be up regulated post malignant transformation. However, controversy exists as to the contribution these proteins make to tumor cell behavior once present. Since GRPR activation promotes proliferation, it has been assumed that their aberrant expression promotes colon cancer (CC) growth and progression. Yet we have contended that when expressed, GRP/GRPR benefits the host since in vitro studies demonstrate they enhance tumor cell attachment to the extracellular matrix and promote CC cytolysis by natural killer lymphocytes. Thus the aim of this study was to ascertain the effect of aberrant GRP/GRPR expression on patient survival. To do this we identified all CC diagnosed at a single institution from 1998 to 2002 that were classified as AJCC stage II or III (n = 88); of these 50 (57%) had sufficient tissues remaining for study. GRP/GRPR expression and natural killer cell density were determined immunohistochemically at the leading edge of each CC, and survival assessed by Kaplan Meier analysis. Expression of high levels of GRPR alone, or both GRP and GRPR, was associated with delayed CC recurrence (14.1-17.0 months, respectfully; P = 0.005) and increased survival (10.1-13.1 months, respectfully; P = 0.0124). CC expressing GRP/GRPR were associated with significantly fewer lymph node metastases than tumors not expressing these proteins, and contained significantly more CD16 + natural killer cells, than tumors not expressing these proteins. These findings demonstrate that patients whose CC express GRPR are associated with a survival advantage as compared to those whose CC do not express these proteins.


Subject(s)
Colonic Neoplasms/metabolism , Gastrin-Releasing Peptide/metabolism , Receptors, Bombesin/metabolism , Amino Acid Sequence , Colonic Neoplasms/pathology , Humans , Immunohistochemistry , Lymphatic Metastasis , Molecular Sequence Data , Receptors, Bombesin/chemistry , Survival Analysis
13.
Cancer Epidemiol Biomarkers Prev ; 17(5): 1155-62, 2008 May.
Article in English | MEDLINE | ID: mdl-18483337

ABSTRACT

Aberrant crypt foci (ACF) are the earliest histopathologic lesion associated with colorectal cancer. ACFs are commonly used as a surrogate marker for colorectal cancer chemoprevention studies in rodents and, more recently, in humans. However, ACF prevalence in unselected populations is not known, nor which ACF features are important for predicting polyp histopathology. To address these questions, we did magnification chromo-colonoscopy on all patients undergoing routine colorectal cancer screening over a 31-month period. ACFs were classified by location, size (small, <20 crypts/ACF; medium, 20-100 crypts/ACF; large, >100 crypts/ACF), and whether they were elevated above the tissue plane. Overall, 802 magnification chromo-colonoscopies with ACF enumeration were done. Mean patient age was 58.6 +/- 8.5 years, of whom 56% were female, 58% were African American, 21% were Caucasian, and 16% were Latino. Total ACF number, along with increasing ACF size and elevation, correlated with the presence of distal hyperplastic polyps and were higher in African Americans. In contrast, ever-smaller ACFs correlated with the presence of distal adenomas and were independent of age and race. The odds ratio for patients with >or=6 small ACFs and adenomas was 6.02 (95% confidence interval, 2.64-13.70) compared with patients with or=1 large ACF and hyperplastic polyps was 5.88 (95% confidence interval, 3.00-11.67) compared with patients with none. Small flat ACFs correlate with the presence of distal adenomas, whereas large raised ACFs correlate with the presence of hyperplastic polyps.


Subject(s)
Colonic Polyps/pathology , Colorectal Neoplasms/pathology , Precancerous Conditions/pathology , Adult , Aged , Aged, 80 and over , Analysis of Variance , Colonoscopy , Coloring Agents , Disease Progression , Female , Humans , Intestinal Mucosa/pathology , Logistic Models , Male , Middle Aged , Risk Factors
14.
Clin Exp Metastasis ; 25(4): 451-63, 2008.
Article in English | MEDLINE | ID: mdl-18350254

ABSTRACT

Gastrin-releasing peptide (GRP) and its receptor (GRPR) are not normally expressed by epithelial cells lining the adult human colon. However post malignant transformation both GRP and its receptor are aberrantly expressed in the colon where we have previously shown they act to retard metastasis by enhancing tumor cell attachment to the extracellular matrix. In the present study, we show that GRP signaling via its cognate receptor when both are aberrantly expressed in human colon cancer cells causes heat shock protein 72 (Hsp72) to be expressed. We show that GRP/GRPR induces expression of Hsp72 by signaling via focal adhesion kinase. When expressed, Hsp72 promotes the binding of CD16+ and CD94+ natural killer cells, resulting in tumor cell cytolysis. These findings demonstrate the presence of a novel mechanism whereby aberrantly expressed GRP/GRPR in human colorectal cancer attenuates tumor progression and may promote a favorable outcome.


Subject(s)
Colonic Neoplasms/immunology , Cytotoxicity, Immunologic , Gastrin-Releasing Peptide/physiology , HSP72 Heat-Shock Proteins/physiology , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily D/analysis , Receptors, IgG/analysis , Cell Line, Tumor , Focal Adhesion Protein-Tyrosine Kinases/physiology , Humans , Receptors, Bombesin/physiology , Signal Transduction , Up-Regulation
15.
J Histochem Cytochem ; 55(12): 1257-64, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17875655

ABSTRACT

Vitamin D deficiency is strongly associated with the risk of developing colorectal cancer (CRC). Because of the propensity of bioactive 1,25-dihydroxyvitamin D3 to cause toxic hypercalcemia, considerable effort has been directed to identifying safer drugs while retaining the efficacy of the parent compound. However, vitamin D precursors do not present toxicity concerns and may be sufficient for CRC chemoprevention or chemotherapy, providing the appropriate enzymes are present in colonic epithelia. We previously showed that CYP27B1 is present at equally high levels in the colon and CRC irrespective of differentiation but was not present in metastases. In this study we used quantitative immunohistochemistry to show that CYP27A1, converting D3 to 25-hydroxycholecalciferol, is present in increasing concentrations in the nuclei of normal colonic epithelia, aberrant crypt foci (ACF), and adenomatous polyps. Whereas total cellular CYP27A1 remains high in CRC and lymph node metastases, the amount of enzyme present in the nuclei decreases with tumor cell dedifferentiation while rising in the cytoplasm. Similarly, increasing amounts of the deactivating enzyme CYP24 are present in the nuclei of normal colonic epithelia, ACFs, and adenomatous polyps. Although the amount of total CYP24 decreases slightly in CRC as a function of tumor cell dedifferentiation and metastasis, location of this enzyme shifts almost entirely from the nuclear compartment to the cytoplasmic compartment. These data indicate that non-toxic vitamin D precursors should be sufficient for CRC chemoprevention, but that neither vitamin D nor its precursors may be sufficient for CRC chemotherapy.


Subject(s)
Cell Transformation, Neoplastic , Cholestanetriol 26-Monooxygenase/biosynthesis , Colorectal Neoplasms/enzymology , Steroid Hydroxylases/biosynthesis , Adenomatous Polyps/enzymology , Adenomatous Polyps/ultrastructure , Colon/enzymology , Colon/pathology , Colon/ultrastructure , Colorectal Neoplasms/pathology , Colorectal Neoplasms/ultrastructure , Humans , Immunohistochemistry , Intestinal Mucosa/enzymology , Intestinal Mucosa/ultrastructure , Lymphatic Metastasis , Vitamin D3 24-Hydroxylase
16.
J Steroid Biochem Mol Biol ; 103(3-5): 763-7, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17257827

ABSTRACT

Recently, it has been reported that 25-hydroxyvitamin D3-1alpha-hydroxylase [1alpha(OH)ase, CYP27B1], required to convert non-toxic 25-hyxdroxyvitamin D3 [25(OH)D(3)] to its active metabolite [1alpha,25(OH)(2)D(3)], is present in the epithelial cells of the human colon. In the present study, the potential chemoprotective role of 25(OH)D(3) was evaluated for colon cancer using the HT-29, human colon cancer cell line. Colon cancer cells were treated with 25(OH)D(3) (500nM or 1muM), 1alpha,25(OH)(2)D(3) (500nM), cholecalciferol (D3, 1muM) or vehicle and cell number determined at days 2 and 5 post-treatment. Results showed that both 25(OH)D(3) and 1alpha,25(OH)(2)D(3) induced dose- and time-dependent anti-proliferative effects on the HT-29 cells, with maximum inhibition noted at day 5. Western blot analyses revealed an up-regulation of VDR and 1alpha(OH)ase expression following 24h of treatment with 25(OH)D(3), and 1alpha,25(OH)(2)D(3). These results are consistent with the expression of VDR and 1alpha(OH)ase in samples of normal colonic tissue, aberrant crypt foci (ACFs) and colon adenocarcinomas. The VDR expression was sequentially increased from normal to pre-cancerous lesions to well-differentiated tumors and then decreased in poorly differentiated tumors. Expression of 1alpha(OH)ase was equally expressed in normal, pre-cancerous lesions and malignant human colon tissues. The increased expression of 1alpha(OH)ase in colon cancer cells treated with the pro-hormone and its anti-proliferative effects, suggest that 25(OH)D(3) may offer possible therapeutic and chemopreventive option in colon cancer.


Subject(s)
Calcifediol/pharmacology , Colonic Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Cell Differentiation , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/classification , Colonic Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Humans , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism
17.
Am J Physiol Gastrointest Liver Physiol ; 292(1): G182-90, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16920698

ABSTRACT

Gastrin-releasing peptide (GRP) and its receptor (GRPR) act as morphogens when expressed in colorectal cancer (CRC), promoting the assumption of a better differentiated phenotype by regulating cell motility in the context of remodeling and retarding tumor cell metastasis by enhancing cell-matrix attachment. Although we have shown that these processes are mediated by focal adhesion kinase (FAK), the downstream target(s) of GRP-induced FAK activation are not known. Since osteoblast differentiation is mediated by FAK-initiated upregulation of ICAM-1 (Nakayamada S, Okada Y, Saito K, Tamura M, Tanaka Y. J Biol Chem 278: 45368-45374, 2003), we determined whether GRP-induced activation of FAK alters ICAM-1 expression in CRC and, if so, determined the contribution of ICAM-1 to mediating GRP's morphogenic properties. Caco-2 and HT-29 cells variably express GRP/GRPR. These cells only express ICAM-1 when GRPR are present. In human CRC, GRPR and ICAM-1 are only expressed by better differentiated tumor cells, with ICAM-1 located at the basolateral membrane. ICAM-1 expression was only observed subsequent to GRPR signaling via FAK. To study the effect of ICAM-1 expression on tumor cell motility, CRC cells expressing GRP, GRPR, and ICAM-1 were cultured in the presence and absence of GRPR antagonist or monoclonal antibody to ICAM-1. CRC cells engaged in directed motility in the context of remodeling and were highly adherent to the extracellular matrix, only in the absence of antagonist or ICAM-1 antibody. These data indicate that GRP upregulation of ICAM-1 via FAK promotes tumor cell motility and attachment to the extracellular matrix.


Subject(s)
Colorectal Neoplasms/physiopathology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Intercellular Adhesion Molecule-1/genetics , Morphogenesis/physiology , Amino Acid Sequence , Cell Line , Cell Movement , Colonic Neoplasms , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Extracellular Matrix/metabolism , Gastrin-Releasing Peptide , Humans , Molecular Sequence Data , Peptide Fragments , Receptors, Bombesin/chemistry , Receptors, Bombesin/physiology
18.
J Proteome Res ; 5(6): 1460-8, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16739997

ABSTRACT

Gastrin-releasing peptide (GRP) and its receptor (GRPR) are aberrantly up-regulated in colon cancer. When expressed, they act as morphogens, retaining tumor cells in a better differentiated state and retarding metastasis. To identify targets activated in response to GRPR signaling we studied Caco-2 and HT-29 cells, colon cancer cell lines that expresses GRPR as a function of confluence. Total cell protein was extracted from pre-confluent cells (expressing GRP/GRPR) cultured in serum-free media in the presence or absence of GRPR-specific antagonist; as well as from confluent cells that do not express GRPR. Overall, we identified 5 proteins that are specifically down-regulated after GRP/GRPR expression: Bach2, creatine kinase B, p47, and two that could not be identified; and 6 proteins that are up-regulated: gephyrin, HSP70, HP1, ICAM-1, ACAT, and one that could not be identified. These findings suggest that the mechanism(s) by which GRP/GRPR mediate its morphogenic effects in colon cancer involve the actions of a number of hitherto unappreciated proteins.


Subject(s)
Gastrin-Releasing Peptide/physiology , Proteome/metabolism , Receptors, Bombesin/biosynthesis , Cell Line, Tumor , Colonic Neoplasms , Culture Media, Serum-Free , Electrophoresis, Gel, Two-Dimensional , Humans , Protein Binding , Receptors, Bombesin/agonists , Signal Transduction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
19.
J Nutr ; 135(12 Suppl): 3021S-3024S, 2005 12.
Article in English | MEDLINE | ID: mdl-16317164

ABSTRACT

Selenium has been shown to reduce cancer incidence in animal models and more recent data indicate that it may be protective in humans as well. However, little is known about the mechanism by which selenium prevents cancer. Cytosolic glutathione peroxidase (GPX1), a selenium-containing antioxidant enzyme, has been implicated in the development of cancer of the head and neck, lung, and breast, in part because of allelic loss at the GPX1 locus. The study of allelic loss at the GPX1 locus in colon cancer was investigated by examining loss of heterozygosity (LOH) in DNA extracted from both tumor and adjacent histopathologically normal tissue obtained by laser capture microdissection. Tissue samples were obtained from 53 colon cancer patients. Two highly polymorphic markers, alanine codon repeats and a proline-leucine polymorphism (198P/L) present in the GPX1 gene, were used to examine LOH at this locus. Analysis of both polymorphisms identified LOH at GPX1 in a significant percentage of colorectal cancer (42%). These results indicated that LOH at the GPX1 locus is a common event in cancer development and that GPX1 or other tightly linked genes may be involved in the etiology of this disease.


Subject(s)
Colonic Neoplasms/prevention & control , Glutathione Peroxidase/genetics , Loss of Heterozygosity/genetics , Selenoproteins/therapeutic use , Animals , Colonic Neoplasms/genetics , Genotype , Humans , Polymorphism, Genetic , Glutathione Peroxidase GPX1
20.
Cancer Epidemiol Biomarkers Prev ; 14(10): 2370-6, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16214919

ABSTRACT

Considerable evidence exists to support the use of vitamin D to prevent and/or treat colorectal cancer. However, the routine use of bioactive vitamin D, 1,25-dihydroxyvitamin D3, is limited by the side effect of toxic hypercalcemia. Recent studies, however, suggest that colonic epithelial cells express 25-hydroxyvitamin D3-1alpha-hydroxylase, an enzyme that converts nontoxic pro-vitamin D, 25-hydroxycholecalciferol [25(OH)D3], to its bioactive form. Yet, nothing is known as to the cellular expression of 1alpha-hydroxylase and the vitamin D receptor (VDR) in the earliest histopathologic structures associated with malignant transformation such as aberrant crypt foci (ACF) and polyps [addressing the possibility of using nontoxic 25(OH)D3 for chemoprevention], nor is anything known as to the expression of these proteins in colorectal cancer as a function of tumor cell differentiation or metastasis [relevant to using 25(OH)D3 for chemotherapy]. In this study, we show that 1alpha-hydroxylase is present at equal high levels in normal colonic epithelium as in ACFs, polyps, and colorectal cancer irrespective of tumor cell differentiation. In contrast, VDR levels were low in normal colonic epithelial cells; were increased in ACFs, polyps, and well-differentiated tumor cells; and then declined as a function of tumor cell de-differentiation. Both 1alpha-hydroxylase and VDR levels were negligible in tumor cells metastasizing to regional lymph nodes. Overall, these data support using 25(OH)D3 for colorectal cancer chemoprevention but suggest that pro-vitamin D is less likely to be useful for colorectal cancer chemotherapy.


Subject(s)
Colon/enzymology , Colonic Neoplasms/enzymology , Receptors, Calcitriol/metabolism , Steroid Hydroxylases/metabolism , Blotting, Western , Caco-2 Cells , Cholestanetriol 26-Monooxygenase , Colonic Neoplasms/pathology , Colonic Neoplasms/prevention & control , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...