Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
FEBS Lett ; 588(5): 829-35, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24492008

ABSTRACT

End-binding protein (EB1) is a microtubule protein that binds to the tumor suppressor adenomatous polyposis coli (APC). While EB1 is implicated as a potential oncogene, its role in cancer progression is unknown. Therefore, we analyzed EB1/APC expression at the earliest stages of colorectal carcinogenesis and in the uninvolved mucosa ("field effect") of human and animal tissue. We also performed siRNA-knockdown in colon cancer cell lines. EB1 is up-regulated in early and field carcinogenesis in the colon, and the cellular/nano-architectural effect of EB1 knockdown depended on the genetic context. Thus, dysregulation of EB1 is an important early event in colon carcinogenesis.


Subject(s)
Adenocarcinoma/metabolism , Carcinogenesis/metabolism , Colorectal Neoplasms/metabolism , Microtubule-Associated Proteins/metabolism , Up-Regulation , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Animals , Apoptosis , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Cytoskeleton/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Male , Microtubule-Associated Proteins/genetics , Rats , Tissue Array Analysis
2.
Cancer Prev Res (Phila) ; 6(10): 1111-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23983085

ABSTRACT

Current fecal tests (occult blood, methylation, DNA mutations) target minute amounts of tumor products among a large amount of fecal material and thus have suboptimal performance. Our group has focused on exploiting field carcinogenesis as a modality to amplify the neoplastic signal. Specifically, we have shown that endoscopically normal rectal brushings have striking nano-architectural alterations which are detectable using a novel optical technique, partial wave spectroscopic microscopy (PWS). We therefore wished to translate this approach to a fecal assay. We examined mucus layer fecal colonocytes (MLFC) at preneoplastic and neoplastic time points (confirmed with rat colonoscopy) in the azoxymethane (AOM)-treated rat model and conducted PWS analysis to derive the nano-architectural parameter, disorder strength (Ld). We confirmed these results with studies in a genetic model (the Pirc rat). We showed that MLFC appeared microscopically normal, consistent with field carcinogenesis. Ld was elevated at an early time point (5 weeks post-AOM injection, effect size = 0.40, P = 0.024) and plateaued before adenoma formation (10 weeks post-AOM, effect size = 0.66, P = 0.001), with no dramatic increase once tumors developed. We replicated these data in the preneoplastic Pirc rat with an effect size in the MLFC that replicated the rectal brushings (increase vs. age-matched controls of 62% vs. 74%, respectively). We provide the first demonstration of a biophotonics approach to fecal assay. Furthermore, targeting the nano-architectural changes of field carcinogenesis rather than the detection of tumor products may provide a novel paradigm for colorectal cancer screening.


Subject(s)
Adenoma/ultrastructure , Carcinogenesis , Colon/cytology , Colorectal Neoplasms/ultrastructure , Intestinal Mucosa/ultrastructure , Adenoma/pathology , Animals , Azoxymethane/chemistry , Colonoscopy , Colorectal Neoplasms/pathology , Disease Models, Animal , Early Detection of Cancer , Endoscopy , Feces , Intestinal Mucosa/pathology , Male , Mass Screening , Microscopy , Occult Blood , Optics and Photonics , Rats , Rats, Inbred F344
3.
PLoS One ; 8(5): e64600, 2013.
Article in English | MEDLINE | ID: mdl-23724067

ABSTRACT

Normal cell function is dependent on the proper maintenance of chromatin structure. Regulation of chromatin structure is controlled by histone modifications that directly influence chromatin architecture and genome function. Specifically, the histone deacetylase (HDAC) family of proteins modulate chromatin compaction and are commonly dysregulated in many tumors, including colorectal cancer (CRC). However, the role of HDAC proteins in early colorectal carcinogenesis has not been previously reported. We found HDAC1, HDAC2, HDAC3, HDAC5, and HDAC7 all to be up-regulated in the field of human CRC. Furthermore, we observed that HDAC2 up-regulation is one of the earliest events in CRC carcinogenesis and observed this in human field carcinogenesis, the azoxymethane-treated rat model, and in more aggressive colon cancer cell lines. The universality of HDAC2 up-regulation suggests that HDAC2 up-regulation is a novel and important early event in CRC, which may serve as a biomarker. HDAC inhibitors (HDACIs) interfere with tumorigenic HDAC activity; however, the precise mechanisms involved in this process remain to be elucidated. We confirmed that HDAC inhibition by valproic acid (VPA) targeted the more aggressive cell line. Using nuclease digestion assays and transmission electron microscopy imaging, we observed that VPA treatment induced greater changes in chromatin structure in the more aggressive cell line. Furthermore, we used the novel imaging technique partial wave spectroscopy (PWS) to quantify nanoscale alterations in chromatin. We noted that the PWS results are consistent with the biological assays, indicating a greater effect of VPA treatment in the more aggressive cell type. Together, these results demonstrate the importance of HDAC activity in early carcinogenic events and the unique role of higher-order chromatin structure in determining cell tumorigenicity.


Subject(s)
Carcinogenesis/pathology , Chromatin/metabolism , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Histone Deacetylases/metabolism , Intestinal Mucosa/pathology , Up-Regulation , Animals , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Cell Survival/drug effects , Chromatin/ultrastructure , HT29 Cells , Histone Deacetylase Inhibitors/pharmacology , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/enzymology , Rats , Rats, Inbred F344 , Spectrum Analysis , Up-Regulation/drug effects , Valproic Acid/pharmacology
4.
PLoS One ; 7(9): e45591, 2012.
Article in English | MEDLINE | ID: mdl-23049818

ABSTRACT

Colorectal cancer (CRC) screening tests often have a trade-off between efficacy and patient acceptability/cost. Fecal tests (occult blood, methylation) engender excellent patient compliance but lack requisite performance underscoring the need for better population screening tests. We assessed the utility of microRNAs (miRNAs) as markers of field carcinogenesis and their potential role for CRC screening using the azoxymethane (AOM)-treated rat model. We found that 63 miRNAs were upregulated and miR-122, miR-296-5p and miR-503# were downregulated in the uninvolved colonic mucosa of AOM rats. We monitored the expression of selected miRNAs in colonic biopsies of AOM rats at 16 weeks and correlated it with tumor development. We noted that the tumor bearing rats had significantly greater miRNA modulation compared to those without tumors. The miRNAs showed good diagnostic performance with an area under the receiver operator curve (AUROC) of >0.7. We also noted that the miRNA induction in the colonic mucosa was mirrorred in the mucus layer fecal colonocytes isolated from AOM rat stool and the degree of miRNA induction was greater in the tumor bearing rats compared to those without tumors. Lastly, we also noted significant miRNA modulation in the Pirc rats- the genetic model of colon carcinogenesis, both in the uninvolved colonic mucosa and the fecal colonocytes. We thus demonstrate that miRNAs are excellent markers of field carcinogenesis and could accurately predict future neoplasia. Based on our results, we propose an accurate, inexpensive, non-invasive miRNA test for CRC risk stratification based on rectal brushings or from abraded fecal colonocytes.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma/diagnosis , Carcinoma/genetics , Colonic Neoplasms/diagnosis , Colonic Neoplasms/genetics , Early Detection of Cancer , MicroRNAs/genetics , Animals , Area Under Curve , Azoxymethane , Biomarkers, Tumor/metabolism , Biopsy , Carcinogens , Carcinoma/chemically induced , Carcinoma/pathology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Down-Regulation , Feces/cytology , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , MicroRNAs/metabolism , Predictive Value of Tests , Rats , Up-Regulation
5.
PLoS One ; 7(6): e38047, 2012.
Article in English | MEDLINE | ID: mdl-22675506

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is a major cause of morbidity and mortality underscoring the need for safe and effective chemopreventive strategies. Targeting epidermal growth factor receptor (EGFR) is attractive in that it is an early critical event in HNSCC pathogenesis. However, current agents lack efficacy or have unacceptable toxicity. Several groups have demonstrated that the over-the-counter medication, polyethylene glycol (PEG) has remarkable chemopreventive efficacy against colon carcinogenesis. Importantly, we reported that this effect is mediated through EGFR internalization/degradation. In the current study, we investigated the chemopreventive efficacy of this agent against HNSCC, using both the well validated animal model 4-NQO (4-nitroquinoline 1-oxide) rat model and cell culture with the human HNSCC cell line SCC-25. We demonstrated that daily topical application of 10% PEG-8000 in the oral cavity (tongue and cavity wall) post 4NQO initiation resulted in a significant reduction in tumor burden (both, tumor size and tumors/tumor bearing rat) without any evidence of toxicity. Immunohistochemical studies depicted decreased proliferation (number of Ki67-positive cells) and reduced expression of EGFR and its downstream effectors cyclin D1 in the tongue mucosa of 4NQO-rats treated with PEG. We showed that EGFR was also markedly downregulated in SCC-25 cells by PEG-8000 with a concomitant induction of G1-S phase cell-cycle arrest, which was potentially mediated through upregulated p21(cip1/waf1). In conclusion, we demonstrate, for the first time, that PEG has promising efficacy and safety as a chemopreventive efficacy against oral carcinogenesis.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Epidermal Growth Factor/antagonists & inhibitors , Molecular Targeted Therapy , Mouth Neoplasms/drug therapy , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/therapeutic use , 4-Nitroquinoline-1-oxide , Administration, Oral , Administration, Topical , Animals , Blotting, Western , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/pathology , Chemoprevention , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Disease Progression , Down-Regulation/drug effects , Epidermal Growth Factor/metabolism , Epithelial Cells/drug effects , Epithelial Cells/pathology , ErbB Receptors/metabolism , Humans , Male , Mouth Mucosa/drug effects , Mouth Mucosa/pathology , Polyethylene Glycols/pharmacology , Rats , Rats, Inbred F344
6.
Oncol Rep ; 26(5): 1127-32, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21769438

ABSTRACT

This study was undertaken to examine the gender-sensitivity and chemopreventive responsiveness of celecoxib on intestinal stem-like cells as a biomarker of colon carcino-genesis, using the MIN mouse model. Male and female MIN mice (6-7-weeks old) were randomized to either control diet or to a diet supplemented with celecoxib (1,500 ppm). The animals were euthanized ten weeks later and the intestines were flushed and opened longitudinally to assess tumor count. Small intestinal segments were formalin-fixed and tissue sections were subjected to immunohistochemical evaluation of DCAMKL1, a known marker of stem-like cells. We found that in animals receiving control (AIN 76A diet) alone, female MIN mice had a higher polyp count than males (52.32 ± 13.89 vs. 35.43 ± 16.05; p<0.0005). However, compared to control diet groups, celecoxib supplementation caused a larger reduction in the number of polyps in females than their male cohorts (6.38 ± 1.43 vs. 12.83 ± 6.74; a reduction of 88% in females to 64% in males). Significant differences (p=0.013) were observed in the number of DCAMKL1-stained cells in the crypts of the wild-type (WT) (10.01 ± 1.07 stem cells per high powered field; HPF) compared to the MIN mice (24.15 ± 8.08 stem cells per HPF), illustrating increased stem-like cells in animals that are more prone to neoplasia. DCAMKL1 labeled stem-like cells were equal in number in the male and female groups receiving the control AIN 76A diet alone (females, 25.73 stem-like cells/HPF); males, 24.15 stem-like cells/HPF). However, females showed a greater reduction in the number of DCAMKL1-labeled stem-like cells with celecoxib supplementation than the respective males (16.63 ± 4.23 vs. 21.56 ± 9.06; a reduction of 35.4% in females to 10.7% in males). We conclude that a higher number of stem-like cells in the uninvolved mucosa paralleled tumorigenesis and mirrored greater chemopreventive responsiveness of female MIN mice compared to males.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Intestinal Mucosa/pathology , Intestinal Neoplasms/pathology , Intestinal Neoplasms/prevention & control , Pyrazoles/pharmacology , Stem Cells/pathology , Sulfonamides/pharmacology , Animals , Celecoxib , Chemoprevention , Disease Models, Animal , Doublecortin-Like Kinases , Female , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Neoplasms/genetics , Intestinal Neoplasms/metabolism , Male , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/biosynthesis , Sex Factors , Stem Cells/drug effects
7.
Cancer Lett ; 306(2): 205-13, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21493000

ABSTRACT

Spectroscopic techniques have demonstrated that in the microscopically normal mucosa, there is an increase in mucosal micro-circulation in patients harboring neoplasia elsewhere in the colon (i.e. marker of field carcinogenesis). However, the physiological and molecular basis of this early increase in blood supply (EIBS) has not been elucidated. We, therefore, investigated the microvessel density (MVD) and angiogenic gene expression in the premalignant colonic mucosa from the well-validated azoxymethane (AOM)-treated rat experimental model of colon carcinogenesis. Fisher 344 rats were treated with AOM (15 mg/kg i.p.) or saline and euthanized 14 weeks later (a time-point that precedes carcinoma development). Colon sections were studied for MVD via immunohistochemical assessment for CD31 and location was compared with optical assessment of mucosal hemoglobin with low-coherence enhanced backscattering spectroscopy (LEBS). Finally, we performed a pilot real-time PCR angiogenesis microarray (84 genes) from the microscopically normal colonic mucosa of AOM and age-matched saline treated rats. AOM treatment increased MVD in both the mucosa and submucosa of the rats (125% increase in mucosa; p<0.007, and 96% increase in submucosa; p<0.02) but the increase was most pronounced at the cryptal base consistent with the LEBS data showing maximal hemoglobin augmentation at 200-225 µm depth. Microarray analysis showed striking dysregulation of angiogenic and anti-angiogenic factors. We demonstrate, for the first time, that neo-angiogenesis occurs in the microscopically normal colonic mucosa and was accentuated at the bottom of the crypt. This finding has potential implications as a biomarker for risk-stratification and target for chemoprevention.


Subject(s)
Cell Transformation, Neoplastic/pathology , Colon/pathology , Colonic Neoplasms/blood supply , Colonic Neoplasms/pathology , Intestinal Mucosa/pathology , Neovascularization, Pathologic , Adenoma/blood supply , Adenoma/chemically induced , Adenoma/pathology , Animals , Azoxymethane/toxicity , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinogens/toxicity , Colonic Neoplasms/chemically induced , Gene Expression Profiling , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction
8.
Int J Oncol ; 38(2): 529-36, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21170505

ABSTRACT

Polyethylene glycol (PEG) is a safe and effective chemopreventive agent against colorectal carcinogenesis in cell culture, animal models and human subjects. Although the precise molecular mechanism is unclear, we previously reported that PEG suppresses colonic epithelial proliferation. As cellular proliferation is driven by complex G1-S phase transition, we now characterize the role of PEG on cell cycle regulation. We focused our attention on the effect of PEG on the CDK inhibitor p21cip1/waf1, which is implicated in early colon carcinogenesis and is upregulated by non-steroidal anti-inflammatory drugs. These studies were done in the azoxymethane-treated (AOM) rat model as well as in HT-29 colon cancer cells. Immunohistochemical analysis revealed that while AOM decreased the p21 expression (75%, p<0.01) in the premalignant colonic mucosa, PEG induced p21 levels back to normal. These findings paralleled a decreased BrdUrd incorporation (78%, p<0.001) and hypophosphorylated retinoblastoma protein (Rb; by 47%) signifying PEG's antiproliferative activity. Furthermore, in HT-29 cells, PEG decreased proliferation as measured by PCNA (68% reduction), increased p21 expression (2.3-fold), induced cell cycle arrest during G0/G1 phase (45% reduction in S phase cells) and inhibited the phosphorylation of Rb (by 52% compared to untreated). PEG caused greater than a 2-fold induction of protein and mRNA level of p21cip1/waf1 in HT-29 cells. These results demonstrate for the first time that PEG is involved in p21 regulation concomitant with G1S phase cell cycle arrest and it is through these effects that it can exert its anti-proliferative and hence chemopreventive role.


Subject(s)
Cell Proliferation/drug effects , Colonic Neoplasms/prevention & control , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , G1 Phase/drug effects , Polyethylene Glycols/pharmacology , S Phase/drug effects , Animals , Azoxymethane/toxicity , Blotting, Western , Carcinogens/toxicity , Colon/drug effects , Colon/metabolism , Colonic Neoplasms/chemically induced , Colonic Neoplasms/drug therapy , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , HT29 Cells , Humans , Male , RNA, Messenger/genetics , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Surface-Active Agents/pharmacology
9.
Biomed Opt Express ; 1(4): 1196-1208, 2010 Oct 20.
Article in English | MEDLINE | ID: mdl-21258541

ABSTRACT

Low-coherence enhanced backscattering (LEBS) spectroscopy is a light scattering technique which uses partial spatial coherence broadband illumination to interrogate the optical properties at sub-diffusion length scales. In this work, we present a post-processing technique which isolates the hemoglobin concentration at different depths within a sample using a single spectroscopic LEBS measurement with a fixed spatial coherence of illumination. We verify the method with scattering (spectralon reflectance standard and polystyrene microspheres) and absorbing (hemoglobin) phantoms. We then demonstrate the relevance of this method for quantifying hemoglobin content as a function of depth within biological tissue using the azoxymethane treated animal model of colorectal cancer.

10.
Mol Cancer Ther ; 7(9): 3103-11, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18790788

ABSTRACT

Polyethylene glycol (PEG) is a clinically widely used agent with profound chemopreventive properties in experimental colon carcinogenesis. We reported previously that Snail/beta-catenin signaling may mediate the suppression of epithelial proliferation by PEG, although the upstream events remain unclear. We report herein the role of epidermal growth factor receptor (EGFR), a known mediator of Snail and overexpressed in approximately 80% of human colorectal cancers, on PEG-mediated antiproliferative and hence antineoplastic effects in azoxymethane (AOM) rats and HT-29 colon cancer cells. AOM rats were randomized to either standard diet or one with 10% PEG-3350 and euthanized 8 weeks later. The colonic samples were subjected to immunohistochemical or Western blot analyses. PEG decreased mucosal EGFR by 60% (P < 0.001). Similar PEG effects were obtained in HT-29 cells. PEG suppressed EGFR protein via lysosmal degradation with no change in mRNA levels. To show that EGFR antagonism per se was responsible for the antiproliferative effect, we inhibited EGFR by either pretreating cells with gefitinib or stably transfecting with EGFR-short hairpin RNA and measured the effect of PEG on proliferation. In either case, PEG effect was blunted, suggesting a vital role of EGFR. Flow cytometric analysis revealed that EGFR-short hairpin RNA cells, besides having reduced membrane EGFR, also expressed low Snail levels (40%), corroborating a strong association. Furthermore, in EGFR silenced cells, PEG effect on EGFR or Snail was muted, similar to that on proliferation. In conclusion, we show that EGFR is the proximate membrane signaling molecule through which PEG initiates antiproliferative activity with Snail/beta-catenin pathway playing the central intermediary function.


Subject(s)
Chemoprevention , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/prevention & control , ErbB Receptors/metabolism , Polyethylene Glycols/therapeutic use , Transcription Factors/metabolism , Animals , Azoxymethane , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Down-Regulation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , ErbB Receptors/antagonists & inhibitors , Gefitinib , HT29 Cells , Humans , Intestinal Mucosa/pathology , Lysosomes/drug effects , Lysosomes/metabolism , Male , Models, Biological , Polyethylene Glycols/pharmacology , Quinazolines/pharmacology , Rats , Rats, Inbred F344 , Repressor Proteins/genetics , Repressor Proteins/metabolism , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription, Genetic/drug effects
11.
Mol Cancer Ther ; 7(7): 1797-806, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18644992

ABSTRACT

Although the nonsteroidal anti-inflammatory drugs (NSAID) protection against colorectal cancer is well established, the molecular mechanisms remain unclear. We show herein that induction of the tumor suppressor gene COOH-terminal Src kinase (Csk) by NSAID is important for their antiproliferative and hence chemopreventive effects. In the azoxymethane-treated rat model of experimental colon carcinogenesis, sulindac treatment markedly induced Csk with a corresponding increase in inhibitory phosphorylation of Src (Tyr(527)). Sulindac-mediated Csk induction was replicated in the human colorectal cancer cell line HT-29, with a corresponding suppression of both Src kinase activity (63% of vehicle; P < 0.05) and E-cadherin tyrosine phosphorylation (an in vivo Src target). To determine the importance of Csk in NSAID antiproliferative activity, we stably transfected a Csk-specific short hairpin RNA (shRNA) vector into HT-29 cells, thereby blunting the sulindac-mediated Csk induction. These transfectants were significantly less responsive to the antiproliferative effect of sulindac sulfide (suppression of proliferating cell nuclear antigen was 21 +/- 2.3% in transfectants versus 45 +/- 4.23% in wild-type cells), with a corresponding mitigation of the sulindac-mediated G(1)-S-phase arrest (S-phase cells 48 +/- 3.6% versus 14 +/- 2.8% of vehicle respectively). Importantly, the Csk shRNA cells had a marked decrease in the cyclin-dependent kinase inhibitor p21(cip/waf1), a critical regulator of G(1)-S-phase progression (49% of wild-type cells). Moreover, although sulindac-mediated induction of p21(cip/waf1) was 113% in wild-type HT-29, this induction was alleviated in the Csk shRNA transfectants (65% induction; P < 0.01). Thus, this is the first demonstration that the antiproliferative activity of NSAID is modulated, at least partly, through the Csk/Src axis.


Subject(s)
Colonic Neoplasms/enzymology , Colonic Neoplasms/prevention & control , Protein-Tyrosine Kinases/metabolism , Sulindac/analogs & derivatives , Animals , Antineoplastic Agents/pharmacology , Azoxymethane , CSK Tyrosine-Protein Kinase , Cell Cycle/drug effects , Cell Proliferation/drug effects , Colon/drug effects , Colon/enzymology , Colon/pathology , Colonic Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Down-Regulation/drug effects , Drug Screening Assays, Antitumor , Enzyme Induction/drug effects , HT29 Cells , Humans , Male , Models, Biological , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/biosynthesis , Rats , Rats, Inbred F344 , Sulindac/pharmacology , src-Family Kinases
12.
FEBS Lett ; 581(20): 3857-62, 2007 Aug 07.
Article in English | MEDLINE | ID: mdl-17658518

ABSTRACT

We have recently demonstrated that dramatic alteration in mucosal microvascular blood content termed early increase in blood supply (EIBS) is a hallmark of early colon carcinogenesis. In the current study, we elucidate the mechanism of EIBS by assessing iNOS/nitric oxide axis in the histologically normal colonic mucosa of rats treated with the colon-specific carcinogen, azoxymethane. We demonstrate that there was a strong temporal correlation between EIBS and iNOS expression/activity. Importantly, we also observed that short-term treatment with nitric oxide inhibitor abrogated EIBS. These data indicate that iNOS induction may have a critical role in augmenting the predysplastic mucosal blood supply and thereby fostering colon carcinogenesis.


Subject(s)
Colonic Neoplasms/blood supply , Colonic Neoplasms/etiology , Nitric Oxide Synthase Type II/metabolism , Animals , Azoxymethane/toxicity , Carcinogens/toxicity , Colonic Neoplasms/pathology , Enzyme Inhibitors/pharmacology , Immunohistochemistry , Light , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Rats , Rats, Inbred F344 , Scattering, Radiation , Time Factors
13.
Mol Cancer Ther ; 5(8): 2060-9, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16928827

ABSTRACT

Polyethylene glycol (PEG) is one of the most potent chemopreventive agents against colorectal cancer; however, the mechanisms remain largely unexplored. In this study, we assessed the ability of PEG to target cyclin D1-beta-catenin-mediated hyperproliferation in the azoxymethane-treated rat model and the human colorectal cancer cell line, HT-29. Azoxymethane-treated rats were randomized to AIN-76A diet alone or supplemented with 5% PEG-8000. After 30 weeks, animals were euthanized and biopsies of aberrant crypt foci and uninvolved crypts were subjected to immunohistochemical and immunoblot analyses. PEG markedly suppressed both early and late markers of azoxymethane-induced colon carcinogenesis (fractal dimension by 80%, aberrant crypt foci by 64%, and tumors by 74%). In both azoxymethane-treated rats and HT-29 cells treated with 5% PEG-3350 for 24 hours, PEG decreased proliferation (45% and 52%, respectively) and cyclin D1 (78% and 56%, respectively). Because beta-catenin is the major regulator of cyclin D1 in colorectal cancer, we used the T-cell factor (Tcf)-TOPFLASH reporter assay to show that PEG markedly inhibited beta-catenin transcriptional activity. PEG did not alter total beta-catenin expression but rather its nuclear localization, leading us to assess E-cadherin expression (a major determinant of beta-catenin subcellular localization), which was increased by 73% and 71% in the azoxymethane-rat and HT-29 cells, respectively. We therefore investigated the effect of PEG treatment on levels of the negative regulator of E-cadherin, SNAIL, and observed a 50% and 75% decrease, respectively. In conclusion, we show, for the first time, a molecular mechanism through which PEG imparts its antiproliferative and hence profound chemopreventive effect.


Subject(s)
Colon/pathology , Colonic Neoplasms/prevention & control , Polyethylene Glycols/pharmacology , Transcription Factors/metabolism , beta Catenin/metabolism , Animals , Anticarcinogenic Agents/pharmacology , Azoxymethane/adverse effects , Cadherins/drug effects , Cadherins/metabolism , Cell Proliferation/drug effects , Colon/drug effects , Colon/metabolism , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Cyclin D1/drug effects , Cyclin D1/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Male , Rats , Rats, Inbred F344 , Signal Transduction , Snail Family Transcription Factors , Transcription Factors/drug effects , Tumor Cells, Cultured , beta Catenin/drug effects
14.
Cancer Epidemiol Biomarkers Prev ; 14(7): 1639-45, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16030095

ABSTRACT

BACKGROUND: We have reported recently that microarchitectural analysis of the histologically normal mucosa using a novel optics technology, four-dimensional elastic light scattering fingerprinting (ELF), provided unprecedented sensitivity for early detection of colon carcinogenesis. In the present study, we explored the ability of four-dimensional ELF to identify an inherited predisposition to colorectal cancer, an issue of considerable importance for optimizing population screening strategies. METHODS: We used the MIN mouse, a model whose germ line adenomatous polyposis coli truncation leads to spontaneous intestinal tumorigenesis, thus replicating the human syndrome, familial adenomatous polyposis. Spectral markers were assessed by four-dimensional ELF analysis in MIN mice at preneoplastic time points and compared with age-matched controls (C57BL6 mice with wild-type adenomatous polyposis coli). To assess the responsiveness of spectral markers to chemopreventive agents, a subset of MIN mice was supplemented with celecoxib 1,500 ppm. RESULTS: Spectral slope, fractal dimension, and principal component 3 were dramatically altered in the uninvolved MIN mouse mucosa at the earliest time points. Furthermore, alteration in spectral variables increased over time, consonant with the microarchitectural underpinnings of subsequent tumorigenesis. Additionally, these markers spatially correlated with future adenoma development (small intestine > colon). Short-term treatment with the potent chemopreventive agent, celecoxib, resulted in near normalization of fractal dimension and principal component 3. CONCLUSIONS: We report, for the first time, that spectral markers, assayed by four-dimensional ELF, were able to sensitively identify a genetic predisposition for intestinal tumorigenesis before the occurrence of phenotypic manifestations. Moreover, the reversal of spectral markers by celecoxib treatment supports the neoplastic relevance.


Subject(s)
Colonic Neoplasms/diagnosis , Intestinal Mucosa/metabolism , Animals , Celecoxib , Colonic Neoplasms/prevention & control , Cyclooxygenase Inhibitors/therapeutic use , Intestinal Mucosa/drug effects , Male , Mice , Mice, Inbred C57BL , Predictive Value of Tests , Pyrazoles/therapeutic use , Sulfonamides/therapeutic use
15.
FEBS Lett ; 579(17): 3497-502, 2005 Jul 04.
Article in English | MEDLINE | ID: mdl-15961079

ABSTRACT

Hyperproliferation of the premalignant epithelium is critical for colonic carcinogenesis; however the mechanisms remain largely unexplored. We report herein that prior to occurrence of neoplastic lesions in the azoxymethane-rat model of colon carcinogenesis; the tumor suppressor gene C-terminal Src kinase (Csk) was down-regulated with a concomitant increase in Src activity. Furthermore, pharmacological or genetic (RNA interference) inhibition of Csk resulted in increased proliferation in colon cancer cell lines through the mitogen-activated protein kinase dependent pathway. Thus, we demonstrate, for the first time, that Csk suppression is an important early event in colorectal cancer pathogenesis.


Subject(s)
Cell Transformation, Neoplastic , Colonic Neoplasms/enzymology , Down-Regulation , Genes, Tumor Suppressor , Intestinal Mucosa/pathology , Precancerous Conditions/enzymology , Protein-Tyrosine Kinases/genetics , Animals , CSK Tyrosine-Protein Kinase , Cell Proliferation , Colon/pathology , Colonic Neoplasms/genetics , HT29 Cells , Humans , Intestinal Mucosa/immunology , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Male , Precancerous Conditions/pathology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/analysis , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA Interference , Rats , Rats, Inbred F344 , src-Family Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...