Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
PLoS One ; 13(4): e0193544, 2018.
Article in English | MEDLINE | ID: mdl-29617381

ABSTRACT

Chemoprevention represents an attractive modality against colorectal cancer (CRC) although widespread clinical implementation of promising agents (e.g. aspirin/NSAIDS) have been stymied by both suboptimal efficacy and concerns over toxicity. This highlights the need for better agents. Several groups, including our own, have reported that the over-the-counter laxative polyethylene glycol (PEG) has remarkable efficacy in rodent models of colon carcinogenesis. In this study, we undertook the first randomized human trial to address the role of PEG in prevention of human colonic neoplasia. This was a double-blind, placebo-controlled, three-arm trial where eligible subjects were randomized to 8g PEG-3350 (n = 27) or 17g PEG-3350 (n = 24), or placebo (n = 24; maltodextrin) orally for a duration of six months. Our initial primary endpoint was rectal aberrant crypt foci (ACF) but this was changed during protocol period to rectal mucosal epidermal growth factor receptor (EGFR). Of the 87 patients randomized, 48 completed study primary endpoints and rectal EGFR unchanged PEG treatment. Rectal ACF had a trend suggesting potentially reduction with PEG treatment (pre-post change 1.7 in placebo versus -0.3 in PEG 8+ 17g doses, p = 0.108). Other endpoints (proliferation, apoptosis, expression of SNAIL and E-cadherin), previously noted to be modulated in rodent models, appeared unchanged with PEG treatment in this clinical trial. We conclude that PEG was generally well tolerated with the trial failing to meet primary efficacy endpoints. However, rectal ACFs demonstrated a trend (albeit statistically insignificant) for suppression with PEG. Moreover, all molecular assays including EGFR were unaltered with PEG underscoring issues with lack of translatability of biomarkers from preclinical to clinical trials. This data may provide the impetus for future clinical trials on PEG using more robust biomarkers of chemoprevention. TRIAL REGISTRATION: ClinicalTrials.gov NCT00828984.


Subject(s)
Aberrant Crypt Foci/prevention & control , Anticarcinogenic Agents/therapeutic use , Colorectal Neoplasms/prevention & control , ErbB Receptors/analysis , Laxatives/therapeutic use , Polyethylene Glycols/therapeutic use , Aberrant Crypt Foci/pathology , Adult , Aged , Aged, 80 and over , Anticarcinogenic Agents/administration & dosage , Biomarkers, Tumor/analysis , Chemoprevention , Colonic Neoplasms/pathology , Colonic Neoplasms/prevention & control , Colorectal Neoplasms/pathology , Double-Blind Method , Female , Humans , Laxatives/administration & dosage , Male , Middle Aged , Placebo Effect , Polyethylene Glycols/administration & dosage , Rectum/pathology
2.
Oncotarget ; 8(13): 20543-20557, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28423551

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is the second leading cause of cancer-related mortality in the United States. There is an increasing need for the identification of biomarkers of pre-malignant and early stage CRC to improve risk-stratification and screening recommendations. In this study, we investigated the possibility of metabolic and mitochondrial reprogramming early in the pre-malignant colorectal field. METHODS: Rectal biopsies were taken from 81 patients undergoing screening colonoscopy, and gene expression of metabolic and mitochondrial markers were assessed using real time quantitative PCR. Validation studies were performed in two different animal models of colon carcinogenesis: Pirc rats and AOM-treated rats. RESULTS: We found evidence of a Warburg effect in the normal-appearing rectal mucosa of patients harboring precancerous lesions elsewhere in the colon compared to control patients, with a significant increase in HIF1α, SLC2A1 (referred to as GLUT1), PKM2, and LDHA. We also found evidence of early mitochondrial changes in the colorectal field of patients harboring pre-cancerous lesions, with significantly increased mitochondrial gene expression of DRP1 (fission), OPA1 (fusion), PGC1-α (biogenesis), UCP2 (uncoupling) and mtND1 (copy number). Similar results were observed in the two different animal models. CONCLUSIONS: These results demonstrate for the first time evidence of early Warburg-like metabolic changes as well as changes in mitochondrial function, dynamics and mtDNA copy number in endoscopically normal premalignant colorectal mucosal field. These findings provide an opportunity for the development of metabolic biomarkers that could be used for improving screening recommendations and risk-stratification. This also provides a potential target for novel chemopreventive strategies in the pre-malignant colorectal field.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Colorectal Neoplasms/metabolism , Precancerous Conditions/metabolism , Animals , Cell Transformation, Neoplastic/pathology , Colorectal Neoplasms/pathology , Humans , Immunohistochemistry , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Precancerous Conditions/pathology , Rats , Real-Time Polymerase Chain Reaction , Transcriptome
3.
Cancer Prev Res (Phila) ; 9(11): 844-854, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27549371

ABSTRACT

Alterations in high order chromatin, with concomitant modulation in gene expression, are one of the earliest events in the development of colorectal cancer. Cohesins are a family of proteins that modulate high-order chromatin, although the role in colorectal cancer remains incompletely understood. We, therefore, assessed the role of cohesin SA1 in colorectal cancer biology and as a biomarker focusing in particular on the increased incidence/mortality of colorectal cancer among African-Americans. Immunohistochemistry on tissue arrays revealed dramatically decreased SA1 expression in both adenomas (62%; P = 0.001) and adenocarcinomas (75%; P = 0.0001). RT-PCR performed in endoscopically normal rectal biopsies (n = 78) revealed a profound decrease in SA1 expression in adenoma-harboring patients (field carcinogenesis) compared with those who were neoplasia-free (47%; P = 0.03). From a racial perspective, colorectal cancer tissues from Caucasians had 56% higher SA1 expression than in African-Americans. This was mirrored in field carcinogenesis where healthy Caucasians expressed more SA1 at baseline compared with matched African-American subjects (73%; P = 0.003). However, as a biomarker for colorectal cancer risk, the diagnostic performance as assessed by area under ROC curve was greater in African-Americans (AUROC = 0.724) than in Caucasians (AUROC = 0.585). From a biologic perspective, SA1 modulation of high-order chromatin was demonstrated with both biophotonic (nanocytology) and chromatin accessibility [micrococcal nuclease (MNase)] assays in SA1-knockdown HT29 colorectal cancer cells. The functional consequences were underscored by increased proliferation (WST-1; P = 0.0002, colony formation; P = 0.001) in the SA1-knockdown HT29 cells. These results provide the first evidence indicating a tumor suppressor role of SA1 in early colon carcinogenesis and as a risk stratification biomarker giving potential insights into biologic basis of racial disparities in colorectal cancer. Cancer Prev Res; 9(11); 844-54. ©2016 AACR.


Subject(s)
Adenocarcinoma/ethnology , Adenoma/ethnology , Biomarkers, Tumor/analysis , Colonic Neoplasms/ethnology , Nuclear Proteins/metabolism , Adenocarcinoma/diagnosis , Adenocarcinoma/metabolism , Adenoma/diagnosis , Adenoma/metabolism , Black or African American , Carcinogenesis , Chromatin , Colonic Neoplasms/diagnosis , Colonic Neoplasms/metabolism , Female , Humans , Incidence , Male , Middle Aged , White People
4.
Cancer Epidemiol Biomarkers Prev ; 23(11): 2413-21, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25155760

ABSTRACT

BACKGROUND: We have previously reported that colonic pericryptal microvascular blood flow is augmented in the premalignant colonic epithelium, highlighting the increased metabolic demand of the proliferative epithelium as a marker of field carcinogenesis. However, its molecular basis is unexplored. In this study, we assessed the expression of a regulator of the "lipogenic switch," fatty acid synthase (FASN), in early colon carcinogenesis for its potential biomarker utility for concurrent neoplasia. METHODS: FASN expression (IHC) in the colonic epithelium from azoxymethane and polyposis in rat colon (Pirc) models of colorectal cancer was studied. FASN mRNA expression from endoscopically normal rectal mucosa was evaluated and correlated with colonoscopic findings (pathologic confirmation of neoplasia). RESULTS: FASN expression progressively increased from premalignant to malignant stage in the azoxymethane model (1.9- to 2.5-fold; P < 0.0001) and was also higher in the adenomas compared with adjacent uninvolved mucosa (1.8- to 3.4-fold; P < 0.001) in the Pirc model. Furthermore, FASN was significantly overexpressed in rectal biopsies from patients harboring adenomas compared with those with no adenomas. These effects were accentuated in male (∼2-fold) and obese patients (1.4-fold compared with those with body mass index < 30). Overall, the performance of rectal FASN was excellent (AUROC of 0.81). CONCLUSIONS: FASN is altered in the premalignant colonic mucosa and may serve as a marker for colonic neoplasia present elsewhere. The enhanced effects in men and obesity may have implications for identifying patient subgroups at risk for early-onset neoplasia. IMPACT: These findings support the role of rectal FASN expression as a reliable biomarker of colonic neoplasia.


Subject(s)
Colorectal Neoplasms/metabolism , Fatty Acid Synthases/metabolism , Obesity/complications , Animals , Biomarkers, Tumor , Colorectal Neoplasms/pathology , Disease Models, Animal , Humans , Male , Obesity/pathology , Rats
5.
Int J Oncol ; 45(3): 1209-15, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24919547

ABSTRACT

MicroRNAs (miRNAs) have been shown to be reliable early biomarkers in a variety of cancers including that of lung. We ascertained whether the biomarker potential of miRNAs could be validated in microscopically normal and easily accessible buccal epithelial brushings from cigarette smokers as a consequence of lung cancer linked 'field carcinogenesis'. We found that compared to neoplasia-free subjects, a panel of 68 miRNAs were upregulated and 3 downregulated in the normal appearing buccal mucosal cells collected from patients harboring lung cancer (n=76). The performance characteristics of selected miRNAs (with ≥ 1-fold change) were excellent with an average under the receiver operator characteristic curve (AUROC) of >0.80. Several miRNAs also displayed gender specificity between the groups. These results provide the first proof-of-concept scenario in which minimally intrusive cheek brushings could provide an initial screening tool in a large at-risk population.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Early Detection of Cancer/methods , Lung Neoplasms/genetics , MicroRNAs/genetics , Mouth Mucosa/metabolism , Adult , Aged , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Case-Control Studies , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Sex Characteristics , Smoking/genetics , Smoking/pathology
6.
BMC Cancer ; 14: 189, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24629088

ABSTRACT

BACKGROUND: Nuclear alterations are a well-known manifestation of cancer. However, little is known about the early, microscopically-undetectable stages of malignant transformation. Based on the phenomenon of field cancerization, the tissue in the field of a tumor can be used to identify and study the initiating events of carcinogenesis. Morphological changes in nuclear organization have been implicated in the field of colorectal cancer (CRC), and we hypothesize that characterization of chromatin alterations in the early stages of CRC will provide insight into cancer progression, as well as serve as a biomarker for early detection, risk stratification and prevention. METHODS: For this study we used transmission electron microscopy (TEM) images of nuclei harboring pre-neoplastic CRC alterations in two models: a carcinogen-treated animal model of early CRC, and microscopically normal-appearing tissue in the field of human CRC. We quantify the chromatin arrangement using approaches with two levels of complexity: 1) binary, where chromatin is separated into areas of dense heterochromatin and loose euchromatin, and 2) grey-scale, where the statistics of continuous mass-density distribution within the nucleus is quantified by its spatial correlation function. RESULTS: We established an increase in heterochromatin content and clump size, as well as a loss of its characteristic peripheral positioning in microscopically normal pre-neoplastic cell nuclei. Additionally, the analysis of chromatin density showed that its spatial distribution is altered from a fractal to a stretched exponential. CONCLUSIONS: We characterize quantitatively and qualitatively the nanoscale structural alterations preceding cancer development, which may allow for the establishment of promising new biomarkers for cancer risk stratification and diagnosis. The findings of this study confirm that ultrastructural changes of chromatin in field carcinogenesis represent early neoplastic events leading to the development of well-documented, microscopically detectable hallmarks of cancer.


Subject(s)
Adenoma/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/ultrastructure , Chromatin Assembly and Disassembly , Colorectal Neoplasms/pathology , Animals , Chromatin/pathology , Chromatin/ultrastructure , Humans , Microscopy, Electron, Transmission , Rats
7.
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
8.
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
9.
World J Gastroenterol ; 18(33): 4507-16, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22969223

ABSTRACT

Gastrointestinal (GI) malignancies are notorious for frequently progressing to advanced stages even in the absence of serious symptoms, thus leading to delayed diagnoses and dismal prognoses. Secondary prevention of GI malignancies through early detection and treatment of cancer-precursor/premalignant lesions, therefore, is recognized as an effective cancer prevention strategy. In order to efficiently detect these lesions, systemic application of screening tests (surveillance) is needed. However, most of the currently used non-invasive screening tests for GI malignancies (for example, serum markers such as alpha-fetoprotein for hepatocellular carcinoma, and fecal occult blood test, for colon cancer) are only modestly effective necessitating the use of highly invasive endoscopy-based procedures, such as esophagogastroduodenoscopy and colonoscopy for screening purposes. Even for hepatocellular carcinoma where non-invasive imaging (ultrasonography) has become a standard screening tool, the need for repeated liver biopsies of suspicious liver nodules for histopathological confirmation can't be avoided. The invasive nature and high-cost associated with these screening tools hinders implementation of GI cancer screening programs. Moreover, only a small fraction of general population is truly predisposed to developing GI malignancies, and indeed needs surveillance. To spare the average-risk individuals from superfluous invasive procedures and achieve an economically viable model of cancer prevention, it's important to identify cohorts in general population that are at substantially high risk of developing GI malignancies (risk-stratification), and select suitable screening tests for surveillance in these cohorts. We herein provide a brief overview of such high-risk cohorts for different GI malignancies, and the screening strategies that have commonly been employed for surveillance purpose in them.


Subject(s)
Early Detection of Cancer/methods , Gastrointestinal Neoplasms/diagnosis , Population Surveillance/methods , Biomarkers, Tumor/metabolism , Cohort Studies , Colonoscopy , Endoscopy, Digestive System , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/prevention & control , Humans , Secondary Prevention
10.
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
11.
J Biomed Opt ; 17(4): 047005, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22559696

ABSTRACT

Polarization-gated spectroscopy is an established method to depth-selectively interrogate the structural properties of biological tissue. We employ this method in vivo in the azoxymethane (AOM)-treated rat model to monitor the morphological changes that occur in the field of a tumor during early carcinogenesis. The results demonstrate a statistically significant change in the shape of the refractive-index correlation function for AOM-treated rats versus saline-treated controls. Since refractive index is linearly proportional to mass density, these refractive-index changes can be directly linked to alterations in the spatial distribution patterns of macromolecular density. Furthermore, we found that alterations in the shape of the refractive-index correlation function shape were an indicator of both present and future risk of tumor development. These results suggest that noninvasive measurement of the shape of the refractive-index correlation function could be a promising marker of early cancer development.


Subject(s)
Colonic Neoplasms/chemistry , Refractometry/methods , Spectrum Analysis/methods , Algorithms , Animals , Azoxymethane , Case-Control Studies , Colonic Neoplasms/chemically induced , Colonic Neoplasms/diagnosis , Colonoscopy , Male , Monte Carlo Method , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/chemistry , Neoplasms, Experimental/diagnosis , Rats , Rats, Inbred F344 , Refractometry/instrumentation , Spectrum Analysis/instrumentation
12.
Gut Microbes ; 2(5): 267-73, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-22067938

ABSTRACT

Infections from enteric bacteria such as enteropathogenic Escherichia coli (EPEC) and enterohemorrhagic Escherichia coli (EHEC) are a public health threat worldwide. EPEC and EHEC are extracellular pathogens, and their interaction with host surface receptors is critical to the infection process. We previously demonstrated that polyethylene glycol (PEG) downregulates surface receptors in intestinal cells. Here we show that PEG decreases ß1-integrin, the surface receptor in intestinal cells that is critical for EPEC and EHEC attachment. We hypothesized that PEG would inhibit the attachment of these enteric pathogens to host cells and improve clinical signs of infection. We found that attachment of the mouse enteric pathogen Citrobacter rodentium, which belongs to the same group of pathogens as EPEC and EHEC, was attenuated by the concurrent presence of PEG. Pretreatment with PEG, without concurrent presence during infection, also reduced bacterial attachment. This finding was further supported in vivo such as that PEG administered by gavage daily during infection as well as prior to infection significantly decreased C. rodentium in the colon and improved the appearance of the infected colon in mice. In addition, PEG decreased the ß1-integrin in colonic mucosa and reduced the C. rodentium-induced activation of epidermal growth factor receptors. PEG also significantly reduced infection-induced colonic inflammation. Finally, PEG efficiently reduced C. rodentium shedding from the colon during infection. In conclusion, PEG can be an efficient and safe preventive agent against EPEC and EHEC infections.


Subject(s)
Bacterial Adhesion/drug effects , Citrobacter rodentium/physiology , Colon/microbiology , Enterobacteriaceae Infections/microbiology , Epithelium/microbiology , Polyethylene Glycols/pharmacology , Animals , Citrobacter rodentium/drug effects , Colon/metabolism , Colon/pathology , Down-Regulation/drug effects , Enterobacteriaceae Infections/metabolism , Enterobacteriaceae Infections/pathology , Epithelium/metabolism , Humans , Integrin beta1/metabolism , Mice , Mice, Inbred C57BL
13.
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
14.
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
15.
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
16.
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.

17.
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
18.
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
19.
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
20.
J Biomed Opt ; 11(4): 041125, 2006.
Article in English | MEDLINE | ID: mdl-16965153

ABSTRACT

The phenomenon of enhanced backscattering (EBS) of light, also known as coherent backscattering (CBS) of light, has been the object of intensive investigation in nonbiological media over the last two decades. However, there have been only a few attempts to explore EBS for tissue characterization and diagnosis. We have recently made progress in the EBS measurements in tissue by taking advantage of low spatial coherence illumination, which has led us to the development of low-coherence enhanced backscattering (LEBS) spectroscopy. In this work, we review the current state of research on LEBS. After a brief discussion of the basic principle of EBS and LEBS, we present an overview of the unique features of LEBS for tissue characterization, and show that LEBS enables depth-selective spectroscopic assessment of mucosal tissue. Then, we demonstrate the potential of LEBS spectroscopy for predicting the risk of colon carcinogenesis and colonoscopy-free screening for colorectal cancer (CRC).


Subject(s)
Colonic Neoplasms/diagnosis , Refractometry/methods , Spectrum Analysis/methods , Tomography, Optical Coherence/methods , Animals , Drug Evaluation, Preclinical/methods , Humans , Light , Reproducibility of Results , Scattering, Radiation , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...