Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Badal Roy is active.

Publication


Featured researches published by Badal Roy.


Oncogene | 2014

Utility of a bacterial infection model to study epithelial-mesenchymal transition, mesenchymal-epithelial transition or tumorigenesis.

P Chandrakesan; Badal Roy; Laxmi Uma Maheswar Rao Jakkula; Ishfaq Ahmed; P Ramamoorthy; Ossama Tawfik; R Papineni; C Houchen; Shrikant Anant; Shahid Umar

DCLK1 and Lgr5 have recently been identified as markers of quiescent and cycling stem cells in the small intestinal crypts, respectively. Epithelial–mesenchymal transition (EMT) is a key development program that is often activated during cancer invasion and metastasis, and also imparts a self-renewal capability to disseminating cancer cells. Utilizing the Citrobacter rodentium (CR)-induced transmissible murine colonic hyperplasia (TMCH) model, we observed a relative decrease in DCLK1 expression in the colonic crypts, with significant shift towards stromal staining at peak (12 days post infection) hyperplasia, whereas staining for Lgr5 and Msi-1 increased several fold. When hyperplasia was regressing (days 20–34), an expansion of DCLK1+ve cells in the CR-infected crypts compared with that seen in uninfected control was recorded. Purified colonic crypt cells exhibiting epigenetic modulation of the transforming growth factor-β (TGFβ), Wnt and Notch pathways on 12 or 34 days post infection formed monolayers in vitro, and underwent trans-differentiation into fibroblast-like cells that stained positive for vimentin, fibronectin and DCLK1. These cells when trypsinized and regrown in soft agar, formed colonospheres/organoids that developed into crypt-like structures (colonoids) in Matrigel and stained positive for DCLK1. Mice exhibiting 12 or 34 days of TMCH were given azoxymethane once for 8 h (Gp1) or weekly for 3 weeks (Gp2), and subjected to crypt isolation. Crypt cells from Gp1 animals formed monolayers as well as colonospheres in soft agar and nodules/tumors in nude mice. Crypt cells isolated from Gp2 animals failed to form the monolayers, but developed into colonospheres in soft agar and nodules/tumors in nude mice. Thus, both hyperplasia and increased presence of DCLK1+ve cells promote cellular transformation in response to a second hit. The TMCH model, therefore, provides an excellent template to study how alterations in intestinal stem cells promote trans-differentiation, crypt regeneration or colon carcinogenesis following bacterial infection.


Oncogene | 2015

Role of bacterial infection in the epigenetic regulation of Wnt antagonist WIF1 by PRC2 protein EZH2.

Badal Roy; Dharmalingam Subramaniam; Ishfaq Ahmed; Jala Vr; Hester Cm; Greiner Ka; Haribabu B; Shrikant Anant; Shahid Umar

The enhancer of zeste homolog-2 (EZH2) represses gene transcription through histone H3 lysine-27-trimethylation (H3K27me3). Citrobacter rodentium (CR) promotes crypt hyperplasia and tumorigenesis by aberrantly regulating Wnt/β-catenin signaling. We aimed at investigating EZH2’s role in epigenetically regulating Wnt/β-catenin signaling following bacterial infection. NIH:Swiss outbred and ApcMin/+ mice were infected with CR (108 CFU); BLT1−/−ApcMin/+ mice, azoxymethane (AOM)/dextran sodium sulfate (DSS)-treated mice and de-identified human adenocarcinoma samples were the models of colon cancer. Following infection with wild-type but not mutant CR, elevated EZH2 levels in the crypt at days 6 and 12 (peak hyperplasia) coincided with increases in H3K27me3 and β-catenin levels, respectively. Chromatin immunoprecipitation revealed EZH2 and H3K27me3’s occupancy on WIF1 (Wnt inhibitory factor 1) promoter resulting in reduced WIF1 mRNA and protein expression. Following EZH2 knockdown via small interfering RNA or EZH2-inhibitor deazaneplanocin A (Dznep) either alone or in combination with histone deacetylase inhibitor suberoylanilide hydroxamic acid, WIF1 promoter activity increased significantly while the overexpression of EZH2 attenuated WIF1 reporter activity. Ectopic overexpression of SET domain mutant (F681Y) almost completely rescued WIF1 reporter activity and partially rescued WIF1 protein levels, whereas H3K27me3 levels were significantly attenuated suggesting that an intact methyltransferases activity is required for EZH2-dependent effects. Interestingly, although β-catenin levels were lower in EZH2-knocked down cells, F681Y mutants exhibited only partial reduction in β-catenin levels. Besides EZH2, increases in miR-203 expression in the crypts at days 6 and 12 post infection correlated with reduced levels of its target WIF1; overexpression of miR-203 in primary colonocytes decreased WIF1 mRNA and protein levels. Elevated levels of EZH2 and β-catenin with concomitant decrease in WIF1 expression in the polyps of CR-infected ApcMin/+ mice paralleled changes recorded in BLT1−/−ApcMin/+, AOM/DSS and human adenocarcinomas. Thus, EZH2-induced downregulation of WIF1 expression may partially regulate Wnt/β-catenin-dependent crypt hyperplasia in response to CR infection.


Molecular Carcinogenesis | 2016

RNA binding protein RBM3 increases β‐catenin signaling to increase stem cell characteristics in colorectal cancer cells

Anand Venugopal; Dharmalingam Subramaniam; Julia Balmaceda; Badal Roy; Dan A. Dixon; Shahid Umar; Scott Weir; Shrikant Anant

Colorectal cancer (CRC) is the second leading cause of cancer deaths in the United States. It arises from loss of intestinal epithelial homeostasis and hyperproliferation of the crypt epithelium. In order to further understand the pathogenesis of CRC it is important to further understand the factors regulating intestinal epithelial proliferation and more specifically, regulation of the intestinal epithelial stem cell compartment. Here, we investigated the role of the RNA binding protein RBM3 in stem cell homeostasis in colorectal cancers. Using a doxycycline (Dox) inducible RBM3 overexpressing cell lines HCT 116 and DLD‐1, we measured changes in side population (SP) cells that have high xenobiotic efflux capacity and increased capacity for self‐renewal. In both cell lines, RBM3 induction showed significant increases in the percentage of side population cells. Additionally, we observed increases in spheroid formation and in cells expressing DCLK1, LGR5 and CD44Hi. As the Wnt/β‐catenin signaling pathway is important for both physiologic and cancer stem cells, we next investigated the effects of RBM3 overexpression on β‐catenin activity. RBM3 overexpression increased levels of nuclear β‐catenin as well as TCF/LEF transcriptional activity. In addition, there was inactivation of GSK3β leading to decreased β‐catenin phosphorylation. Pharmacologic inhibition of GSK3β using (2′Z,3′E)‐6‐Bromoindirubin‐3′‐oxime (BIO) also recapitulates the RBM3 induced β‐catenin activity. In conclusion, we see that RNA binding protein RBM3 induces stemness in colorectal cancer cells through a mechanism involving suppression of GSK3β activity thereby enhancing β‐catenin signaling.


PLOS ONE | 2013

Differential Effects of β-catenin and NF-κB Interplay in the Regulation of Cell Proliferation, Inflammation and Tumorigenesis in Response to Bacterial Infection

Parthasarathy Chandrakesan; Laxmi Uma Maheswar Rao Jakkula; Ishfaq Ahmed; Badal Roy; Shrikant Anant; Shahid Umar

Both β-catenin and NF-κB have been implicated in our laboratory as candidate factors in driving proliferation in an in vivo model of Citrobacter rodentium (CR)-induced colonic crypt hyper-proliferation and hyperplasia. Herein, we test the hypothesis that β-catenin and not necessarily NF-κB regulates colonic crypt hyperplasia or tumorigenesis in response to CR infection. When C57Bl/6 wild type (WT) mice were infected with CR, sequential increases in proliferation at days 9 and 12 plateaued off at day 19 and paralleled increases in NF-κB signaling. In Tlr4−/− (KO) mice, a sequential but sustained proliferation which tapered off only marginally at day 19, was associated with TLR4-dependent and independent increases in NF-κB signaling. Similarly, increases in either activated or total β-catenin in the colonic crypts of WT mice as early as day 3 post-infection coincided with cyclinD1 and c-myc expression and associated crypt hyperplasia. In KO mice, a delayed kinetics associated predominantly with increases in non-phosphorylated (active) β-catenin coincided with increases in cyclinD1, c-myc and crypt hyperplasia. Interestingly, PKCζ-catalyzed Ser-9 phosphorylation and inactivation of GSK-3β and not loss of wild type APC protein accounted for β-catenin accumulation and nuclear translocation in either strain. In vitro studies with Wnt2b and Wnt5a further validated the interplay between the Wnt/β-catenin and NF-κB pathways, respectively. When WT or KO mice were treated with nanoparticle-encapsulated siRNA to β-catenin (si- β-Cat), almost complete loss of nuclear β-catenin coincided with concomitant decreases in CD44 and crypt hyperplasia without defects in NF-κB signaling. si-β-Cat treatment to Apc Min/+ mice attenuated CR-induced increases in β-catenin and CD44 that halted the growth of mutated crypts without affecting NF-κB signaling. The predominant β-catenin-induced crypt proliferation was further validated in a Castaneus strain (B6.CAST.11M) that exhibited significant crypt hyperplasia despite an attenuated NF-κB signaling. Thus, β-catenin and not necessarily NF-κB regulates crypt hyperplasia in response to bacterial infection.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2013

Evidence of functional cross talk between the Notch and NF-κB pathways in nonneoplastic hyperproliferating colonic epithelium

Ishfaq Ahmed; Badal Roy; Parthasarathy Chandrakesan; Anand Venugopal; Lijun Xia; Roy A. Jensen; Shrikant Anant; Shahid Umar

The Notch and NF-κB signaling pathways regulate stem cell function and inflammation in the gut, respectively. We investigate whether a functional cross talk exists between the two pathways during transmissible murine colonic hyperplasia (TMCH) caused by Citrobacter rodentium (CR). During TMCH, NF-κB activity and subunit phosphorylation in colonic crypts of NIH Swiss mice at days 6 and 12 were associated with increases in downstream target CXC chemokine ligand (CXCL)-1/keratinocyte-derived chemokine (KC) expression. Blocking Notch signaling acutely for 5 days with the Notch blocker dibenzazepine (DBZ) failed to inhibit crypt NF-κB activity or CXCL-1/KC expression. Chronic DBZ administration for 10 days, however, blocked Notch and NF-κB signaling in the crypts and abrogated hyperplasia. Intriguingly, chronic Notch inhibition was associated with significant increases in IL-1α, granulocyte colony-stimulating factor, monocyte chemoattractant protein 1, macrophage inflammatory protein 2, and KC in the crypt-denuded lamina propria or whole distal colon, with concomitant increases in myeloperoxidase activity. In core-3(-/-) mice, which are defective in intestinal mucin, DBZ administration replicated the results of NIH Swiss mice; in Apc(Min/+) mice, which are associated with CR-induced elevation of NF-κB-p65(276) expression, DBZ reversed the increase in NF-κB-p65(276), which may have blocked rapid proliferation of the mutated crypts. DBZ further blocked reporter activities involving the NF-κB-luciferase reporter plasmid or the Toll-like receptor 4/NF-κB/SEAPorter HEK-293 reporter cell line, while ectopic expression of Notch-N(ICD) reversed the inhibitory effect. Dietary bael (Aegle marmelos) extract (4%) and curcumin (4%) restored Notch and NF-κB cross talk in NIH Swiss mice, inhibited CR/DBZ-induced apoptosis in the crypts, and promoted crypt regeneration. Thus functional cross talk between the Notch and NF-κB pathways during TMCH regulates hyperplasia and/or inflammation in response to CR infection.


Carcinogenesis | 2016

An ornamental plant targets epigenetic signaling to block cancer stem cell-driven colon carcinogenesis

Ishfaq Ahmed; Badal Roy; Dharmalingam Subramaniam; Showkat Ahmad Ganie; Deep Kwatra; Dan A. Dixon; Shrikant Anant; Mohammad Afzal Zargar; Shahid Umar

Phytochemicals modulate key cellular signaling pathways and have proven anticancer effects. Alcea rosea(AR; Hollyhock) is an ornamental plant with known anti-inflammatory properties. This study explored its role as an anticancer agent. The AR seed extract (AR extract) inhibited proliferation and colony formation in a dose- and time-dependent manner and promoted apoptosis as was evidenced by cleavage of PARP and increased expression of Bax accompanying reduced levels of BCL-xl protein in HCT116 and SW480 cells, respectively. In addition, AR extract-arrested cells at Go/G1 phase of cell cycle and exhibited decreases in Cyclin D1. AR extract-treated cells exhibited reduced number and size of colonospheres in a dose-dependent manner concomitant with decreases in cancer stem cell (CSC) markers ALDH1A1 and Dclk1. Relative levels of β-catenin, Notch-ICD, Hes1 and EZH2 were also attenuated by AR extract. TOP-flash reporter activity, a measure of Wnt signaling, decreased significantly in response to treatment while overexpression of wild type but not mutant EZH2, reversed the inhibitory effects. Moreover, WIF1 (a Wnt antagonist) promoter activity increased dramatically following treatment with AR extract which phenocopied increases in WIF1 reporter activity following EZH2 knockdown.In vivo, AR extract attenuated tumor growth due probably to reduced levels of EZH2, β-catenin, CyclinD1 and Ki-67 along with reduced levels of CSC markers. Since partial purification via HPLC yielded a prominent peak, efforts are underway to identify the active ingredient(s). Taken together, the results clearly suggest that AR extract/active component(s) can be an effective preventative/therapeutic agent to target colon cancer.


Nature Communications | 2016

Genome-wide RNAi screening identifies TMIGD3 isoform1 as a suppressor of NF-κB and osteosarcoma progression.

Swathi V. Iyer; Atul Ranjan; Harold K. Elias; Alejandro Parrales; Hiromi Sasaki; Badal Roy; Shahid Umar; Ossama Tawfik; Tomoo Iwakuma

The ability of cancer cells to survive and grow in anchorage- and serum-independent conditions is well correlated with their aggressiveness. Here, using a human whole-genome shRNA library, we identify TMIGD3 isoform1 (i1) as a factor that suppresses this ability in osteosarcoma (OS) cells, mainly by inhibiting NF-κB activity. Knockdown of TMIGD3 increases proliferation, tumour formation and metastasis of OS cells. Overexpression of TMIGD3 isoform1 (i1), but not isoform3 (i3) which shares a common C-terminal region, suppresses these malignant properties. Adenosine A3 receptor (A3AR) having an identical N-terminal region shows similar biological profiles to TMIGD3 i1. Protein expression of TMIGD3 and A3AR is lower in human OS tissues than normal tissues. Mechanistically, TMIGD3 i1 and A3AR commonly inhibit the PKA−Akt−NF-κB axis. However, TMIGD3 i1 only partially rescues phenotypes induced by A3AR knockdown, suggesting the presence of distinct pathways. Our findings reveal an unappreciated role for TMIGD3 i1 as a suppressor of NF-κB activity and OS progression.


Cancer Research | 2016

Abstract 3297: Altered mucus composition and bacterial dysbiosis promote development of colitis following chronic Notch inhibition

Ishfaq Ahmed; Badal Roy; Rita-marie McFadden; Shrikant Anant; Seth Septer; Shahid Umar

Proceedings: AACR 107th Annual Meeting 2016; April 16-20, 2016; New Orleans, LA Background. Intestinal mucus layer disruption and gut microflora modification in conjunction with tight junction (TJ) changes can increase colonic permeability that allows bacterial dissemination and intestinal and systemic disease. Employing Citrobacter rodentium (CR) as a model system to mimic the pathogenic mechanisms of Enteropathogenic and Enterohaemorrhagic E. coli, we showed previously that a functional cross-talk between the Notch and Wnt/β-catenin pathways regulates hyperplasia and/or colitis following CR infection. Aim: To test the hypothesis that CR infection combined with chronic Notch inhibition will alter the composition of the colonic mucus to promote dysbiosis, tight junction disruption, loss of multipotent intestinal stem cells (ISCs) and colitis. Material/methods: Immune-competent NIH:Swiss outbred mice were infected with CR (108 CFUs) and treated with Notch blocker Dibenzazepine [DBZ, i.p. @ 10μmol/Kg body weight]. Mucus composition, fecal 16S rDNA analysis and components of TJ integrity were measured and correlated with changes in components of the Notch pathway. 6% Pectin diet served as a SCFA-delivery system to the colon. Results: In the colons of CR-infected/DBZ-treated mice, mucus analysis revealed dramatic alterations in the composition of trace O-glycans and complex type and hybrid N-glycans, compared to CR-infected alone that preceded alterations in 16S rDNA microbial community structure. Indeed, mucin degrading and colitogenic bacterium, Akkermansia muciniphila exhibited dramatic increases in the feces of CR-infected and DBZ-treated mice. At the same time, colons from CR-infected/DBZ-treated mice had decreased expression of antimicrobial peptides Angiogenin-4, Retnlβ, Intelectin-1 and Intelectin-2, respectively. Expression levels of both TJ and adherens junction proteins (e.g., Claudin-5, ZO-1, ZO-2, E-Cadherin, β-catenin) also decreased significantly in the colonic crypts of CR-infected/DBZ-treated mice that paralleled loss of Notch signaling. Both C3H/HeN mice that exhibit exaggerated response to CR infection and T- and B-cell deficient Rag-1 mice following CR-infection and DBZ-treatment, exhibited dramatic increases in paracellular permeability, goblet cell metaplasia and exacerbation of colitis. Chronic blockade of the Notch pathway depleted colonic epithelium of ISC markers Dclk1 and Lgr5 concomitant with exacerbation of colitis. 6% Pectin diet ameliorated colitis by restoring barrier integrity via recruitment of Bacteroides vulgatus, Streptococcus gordonii and Lactococcus lactis, bacteria with potential anti-inflammatory functions. Conclusions: 1. These studies help us delineate the mechanistic basis of colitis development in the aftermath of chronic Notch inhibition. 2. Our findings also caution against the use of Notch inhibitors for patients suffering from colitis-associated colon cancer. Citation Format: Ishfaq Ahmed, Badal Roy, Rita-marie McFadden, Shrikant Anant, Seth Septer, Shahid Umar. Altered mucus composition and bacterial dysbiosis promote development of colitis following chronic Notch inhibition. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3297.


Gastroenterology | 2015

Sa1829 Phytochemicals Target Epigenetic Signaling to Block Cancer Stem Cell-Driven Colon Carcinogenesis

Ishfaq Ahmed; Badal Roy; Dharmalingam Subramaniam; Deep Kwatra; Scott Weir; Dan A. Dixon; Shrikant Anant; Shahid Umar

Background: Phytochemicals modulate key cellular signaling pathways and have proven anticancer effects. Alcea rosea (AR; Hollyhock) is an ornamental plant in the Malvaceae family with known anti-inflammatory properties. Its role as an anti-cancer agent however, has not yet been confirmed. The present investigation is focused on the mechanistic insight of AR seed extract as an anti-cancer agent for colon cancer. Methodology: Colon cancer cell lines HCT116 and SW480 and normal colon epithelial cells were used in the study. Cell proliferation, cell cycle and stemness were determined by hexoseaminidase, flow cytometry and spheroid assays, respectively. TOP-Flash reporter and WIF1 promoter activities were performed in HEK-293T cells. For in vivo effects, we used the HCT116-tumor xenograft model for colon tumorigenesis. Real Time PCR, Western blot and immunohistochemistry studies were performed for determining gene expression. Results: The AR seed extract inhibited proliferation and colony formation in a dose and time dependent manner in colon cancer cell lines. The anti-proliferative activity appeared to be mediated by apoptotic mechanisms, as cleavage of PARP and increased expression of Bax accompanied decreased levels of BCL-xl protein. In addition, AR seed extract arrested cells at Go/G1 phase of cell cycle and exhibited decreases in Cyclin D1. Treatment of cells with AR seed extract also reduced the number and size of colonospheres in a dose-dependent manner which coincided with decreases in the expression of ALDH1A1 and Dclk1, markers of CSCs (Cancer Stem Cells). During analyses of pathways critical for CSC maintenance and colon cancer progression, AR seed extract attenuated levels of β-catenin, Jagged1, Notch-ICD, Hes1 and EZH2. TOP-flash reporter activity, a measure of Wnt signaling, decreased significantly in response to treatment while overexpression of wild type but not mutant EZH2, reversed the inhibitory effects. Moreover,WIF1 (aWnt antagonist) promoter activity increased dramatically following treatment with AR seed extract which phenocopied increases in WIF1 reporter activity following EZH2 knockdown. To determine the effect of AR seed extract on tumor growth in vivo, athymic nude mice bearing tumor xenografts were administered AR seed extract intraperitoneally at 200 mg/Kg daily for 21 days. The AR seed extract significantly inhibited tumor xenograft growth. In addition, there were reduced levels of EZH2, b-catenin, CyclinD1 and Ki-67 along with attenuated levels of Dclk1 in the xenograft tissues. Finally, preliminary run through HPLC yielded three prominent peaks and further identification via NMR spectroscopy is underway to identify the active ingredient(s). Conclusion: These results clearly suggest that AR seed extracts can be an effective preventative/therapeutic agent for colon cancer.


Cancer Prevention Research | 2013

Abstract A30: Epigenetic regulation of Wnt/β-catenin signaling in vivo by PRC2 protein EZH2

Badal Roy; Dharmalingam Subramaniam; Ishfaq Ahmed; Shrikant Anant; Shahid Umar

Introduction: Colon cancer results from the accumulation of both acquired genetic and epigenetic changes that transform normal glandular epithelium into invasive adenocarcinomas. The Enhancer of Zeste Homolog 2 (EZH2) protein is a core component of the Polycomb Repressive Complex 2 (PRC2) that represses gene transcription through histone H3 lysine 27 trimethylation (H3K27me3). EZH2 is overexpressed in several malignancies including colon cancer; EZH29s role in tumor initiation however, is less clear. Citrobacter rodentium (CR)-induced transmissible murine colonic hyperplasia (TMCH) that utilizes a type-three secretion system (T3SS) to activate Wnt/β-catenin signaling, represents the earliest molecular and functional changes associated with colon carcinogenesis; the increases in β-catenin recorded during TMCH however, are neither due to mutations in Adenomatous polyposis coli (Apc) or β-catenin (CTNNB1) gene. Aim: To investigate if EZH2 is critical in epigenetically regulating Wnt/β-catenin signaling in response to bacterial infection. Experimental Procedures: TMCH or tumorigenesis were induced by CR infection (108cfu) in wild type and ApcMin/+ mice, respectively. AOM (@10mg/Kg body wt.)/DSS (2.5%; 3 cycles) model of colitis-associated cancer and de-identified human adenoma and adenocarcinoma samples were used to further examine evidence of epigenetically-regulated Wnt/β-catenin signaling. Results: Following CR infection, EZH2 protein increased significantly in the crypt at day 6 and particularly at day 12 (peak hyperplasia) and coincided with expression of EZH2 target protein H3K27me3. The changes accrued were specific to CR as T3SS mutant escV failed to elicit alterations in either EZH2, H3K27me3 or crypt hyperplasia. Mechanistically, siRNA-mediated knockdown of EZH2 led to: i) attenuated cell migration, ii) reduced colonosphere formation and, iii) decreased β-catenin/TCF4-dependent TOPflash reporter activity due to loss of β-catenin. Chromatin immunoprecipitation revealed EZH29s occupancy on WIF1 (Wnt Inhibitory Factor 1) promoter resulting in reduced WIF1 mRNA and protein expression. Following EZH2 knockdown either via siRNA or EZH2-inhibitor DZNep (5μM), relative levels of H3K27me3 decreased which led to the transcriptional upregulation of WIF1 as was evidenced by WIF1 promoter activity. Besides EZH2, we also discovered increases in miR-203 expression in the crypts at days-6 and 12 post-infection that correlated with reduced levels of its target WIF1, at these time points; overexpression of miR-203 in Young Adult Mouse Colon primary cells resulted in decreased WIF1 transcription. Tumors in the distal colons of ApcMin/+ mice following CR infection expressed high levels of EZH2 and β-catenin and a concomitant decrease in WIF1. In response to AOM/DSS, colonic tumors at 24 weeks expressed high levels of EZH2 and β-catenin and a concomitant decrease in WIF1. Pharmacological intervention with honokiol (HNK; @5mg/Kg body wt. p.o.) a week prior to and during AOM/DSS-induced colon carcinogenesis reduced both tumor burden and tumor size. Immunohistochemical analysis of HNK-treated tissues revealed significant reduction in EZH2 and β-catenin staining while WIF1 expression was dramatically upregulated. In human adenomas/adenocarcinomas, simultaneous increases in EZH2 and β-catenin staining coincided with a corresponding decrease in WIF1 positivity. Conclusions: 1. CR infection induces EZH2 overexpression that correlates with aberrant Wnt/β-catenin signaling in the colonic crypts.2. EZH2 downregulates WIF1 expression which may correlate with earliest stages of colon tumorigenesis. 3. Honokiol-induced upregulation of WIF1 to block aberrant Wnt/β-catenin signaling may be a novel strategy to target epigenetic abnormalities for colon cancer prevention and therapeutics. Citation Format: Badal C. Roy, Dharmalingam Subramaniam, Ishfaq Ahmed, Shrikant Anant, Shahid Umar. Epigenetic regulation of Wnt/β-catenin signaling in vivo by PRC2 protein EZH2. [abstract]. In: Proceedings of the Twelfth Annual AACR International Conference on Frontiers in Cancer Prevention Research; 2013 Oct 27-30; National Harbor, MD. Philadelphia (PA): AACR; Can Prev Res 2013;6(11 Suppl): Abstract nr A30.

Collaboration


Dive into the Badal Roy's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Seth Septer

Children's Mercy Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Audrey Seamons

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge