43 results on '"Sureban SM"'
Search Results
2. Editor's Note: Diphenyl Difluoroketone: A Curcumin Derivative with Potent In Vivo Anticancer Activity.
- Author
-
Subramaniam D, May R, Sureban SM, Lee KB, George R, Kuppusamy P, Ramanujam RP, Hideg K, Dieckgraefe BK, Houchen CW, and Anant S
- Published
- 2022
- Full Text
- View/download PDF
3. Could Nutraceutical Approaches Possibly Attenuate the Cytokine Storm in COVID-19 Patients?
- Author
-
Vignesh R, Velu V, and Sureban SM
- Subjects
- Cytokine Release Syndrome, Dietary Supplements, Humans, SARS-CoV-2, COVID-19, Gastrointestinal Microbiome
- Abstract
Competing Interests: SS is an inventor on several patents with a commercial interest and also has an ownership interest in COARE Holdings Inc. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2021
- Full Text
- View/download PDF
4. DCLK1 Monoclonal Antibody-Based CAR-T Cells as a Novel Treatment Strategy against Human Colorectal Cancers.
- Author
-
Sureban SM, Berahovich R, Zhou H, Xu S, Wu L, Ding K, May R, Qu D, Bannerman-Menson E, Golubovskaya V, and Houchen CW
- Abstract
CAR-T (chimeric antigen receptor T cells) immunotherapy is effective in many hematological cancers; however, efficacy in solid tumors is disappointing. Doublecortin-like kinase 1 (DCLK1) labels tumor stem cells (TSCs) in genetic mouse models of colorectal cancer (CRC). Here, we describe a novel CAR-T targeting DCLK1 (CBT-511; with our proprietary DCLK1 single-chain antibody variable fragment) as a treatment strategy to eradicate CRC TSCs. The cell surface expression of DCLK1 and cytotoxicity of CBT-511 were assessed in CRC cells (HT29, HCT116, and LoVo). LoVo-derived tumor xenografts in NOD Scid gamma (NSG
TM )mice were treated with CBT-511 or mock CAR-T cells. Adherent CRC cells express surface DCLK1 (two-dimensional, 2D). A 4.5-fold increase in surface DCLK1 was observed when HT29 cells were grown as spheroids (three-dimensional, 3D). CBT-511 induced cytotoxicity (2D; p < 0.0001), and increased Interferon gamma (IFN-γ) release in CRC cells (2D) compared to mock CAR-T ( p < 0.0001). Moreover, an even greater increase in IFN-γ release was observed when cells were grown in 3D. CBT-511 reduced tumor growth by approximately 50 percent compared to mock CAR-T. These data suggest that CRC cells with increased clonogenic capacity express increased surface DCLK1. A DCLK1-targeted CAR-T can induce cytotoxicity in vitro and inhibit xenograft growth in vivo., Competing Interests: S.M.S., D.Q., R.M., E.B.-M., and C.W.H. have ownership interests in COARE Holdings Inc. All the other authors have no conflict of interest.- Published
- 2019
- Full Text
- View/download PDF
5. Dclk1, a tumor stem cell marker, regulates pro-survival signaling and self-renewal of intestinal tumor cells.
- Author
-
Chandrakesan P, Yao J, Qu D, May R, Weygant N, Ge Y, Ali N, Sureban SM, Gude M, Vega K, Bannerman-Menson E, Xia L, Bronze M, An G, and Houchen CW
- Subjects
- Adenocarcinoma genetics, Adenocarcinoma metabolism, Adenocarcinoma pathology, Animals, Cluster Analysis, Colonic Neoplasms pathology, Disease Models, Animal, Doublecortin-Like Kinases, Epithelial-Mesenchymal Transition genetics, Female, Gene Expression Profiling, Gene Knockdown Techniques, Genes, APC, Humans, Male, Mice, Mice, Transgenic, Mutation, Receptor, Notch1 metabolism, Cell Self Renewal genetics, Cell Survival genetics, Colonic Neoplasms genetics, Colonic Neoplasms metabolism, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Neoplastic Stem Cells metabolism, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Signal Transduction
- Abstract
Background: More than 80% of intestinal neoplasia is associated with the adenomatous polyposis coli (APC) mutation. Doublecortin-like kinase 1 (Dclk1), a kinase protein, is overexpressed in colorectal cancer and specifically marks tumor stem cells (TSCs) that self-renew and increased the tumor progeny in Apc
Min/+ mice. However, the role of Dclk1 expression and its contribution to regulating pro-survival signaling for tumor progression in Apc mutant cancer is poorly understood., Methods: We analyzed DCLK1 and pro-survival signaling gene expression datasets of 329 specimens from TCGA Colon Adenocarcinoma Cancer Data. The network of DCLK1 and pro-survival signaling was analyzed utilizing the GeneMANIA database. We examined the expression levels of Dclk1 and other stem cell-associated markers, pro-survival signaling pathways, cell self-renewal in the isolated intestinal epithelial cells of ApcMin/+ mice with high-grade dysplasia and adenocarcinoma. To determine the functional role of Dclk1 for tumor progression, we knocked down Dclk1 and determined the pro-survival signaling pathways and stemness. We used siRNA technology to gene silence pro-survival signaling in colon cancer cells in vitro. We utilized FACS, IHC, western blot, RT-PCR, and clonogenic (self-renewal) assays., Results: We found a correlation between DCLK1 and pro-survival signaling expression. The expression of Dclk1 and stem cell-associated markers Lgr5, Bmi1, and Musashi1 were significantly higher in the intestinal epithelial cells of ApcMin/+ mice than in wild-type controls. Intestinal epithelial cells of ApcMin/+ mice showed increased expression of pro-survival signaling, pluripotency and self-renewal ability. Furthermore, the enteroids formed from the intestinal Dclk1+ cells of ApcMin/+ mice display higher pluripotency and pro-survival signaling. Dclk1 knockdown in ApcMin/+ mice attenuates intestinal adenomas and adenocarcinoma, and decreases pro-survival signaling and self-renewal. Knocking down RELA and NOTCH1 pro-survival signaling and DCLK1 in HT29 and DLD1 colon cancer cells in vitro reduced the tumor cells' ability to self-renew and survive., Conclusion: Our results indicate that Dclk1 is essential in advancing intestinal tumorigenesis. Knocking down Dclk1 decreases tumor stemness and progression and is thus predicted to regulate pro-survival signaling and tumor cell pluripotency. This study provides a strong rationale to target Dclk1 as a treatment strategy for colorectal cancer.- Published
- 2017
- Full Text
- View/download PDF
6. Intestinal tuft cells regulate the ATM mediated DNA Damage response via Dclk1 dependent mechanism for crypt restitution following radiation injury.
- Author
-
Chandrakesan P, May R, Weygant N, Qu D, Berry WL, Sureban SM, Ali N, Rao C, Huycke M, Bronze MS, and Houchen CW
- Subjects
- Animals, Apoptosis radiation effects, Biomarkers metabolism, Cell Membrane Permeability radiation effects, Cell Proliferation radiation effects, Cell Survival radiation effects, Cyclooxygenase 2 metabolism, Dinoprostone metabolism, Doublecortin-Like Kinases, Enterocytes metabolism, Enterocytes radiation effects, Epithelial Cells metabolism, Integrases metabolism, Mice, Knockout, Microfilament Proteins metabolism, Phosphorylation radiation effects, Protein Serine-Threonine Kinases deficiency, Signal Transduction, Stem Cells metabolism, Survival Analysis, Whole-Body Irradiation, Ataxia Telangiectasia Mutated Proteins metabolism, DNA Damage, Intestines pathology, Protein Serine-Threonine Kinases metabolism, Radiation Injuries metabolism, Radiation Injuries pathology
- Abstract
Crypt epithelial survival and regeneration after injury require highly coordinated complex interplay between resident stem cells and diverse cell types. The function of Dclk1 expressing tuft cells regulating intestinal epithelial DNA damage response for cell survival/self-renewal after radiation-induced injury is unclear. Intestinal epithelial cells (IECs) were isolated and purified and utilized for experimental analysis. We found that small intestinal crypts of Villin
Cre ;Dclk1f/f mice were hypoplastic and more apoptotic 24 h post-total body irradiation, a time when stem cell survival is p53-independent. Injury-induced ATM mediated DNA damage response, pro-survival genes, stem cell markers, and self-renewal ability for survival and restitution were reduced in the isolated intestinal epithelial cells. An even greater reduction in these signaling pathways was observed 3.5 days post-TBI, when peak crypt regeneration occurs. We found that interaction with Dclk1 is critical for ATM and COX2 activation in response to injury. We determined that Dclk1 expressing tuft cells regulate the whole intestinal epithelial cells following injury through paracrine mechanism. These findings suggest that intestinal tuft cells play an important role in regulating the ATM mediated DNA damage response, for epithelial cell survival/self-renewal via a Dclk1 dependent mechanism, and these processes are indispensable for restitution and function after severe radiation-induced injury., Competing Interests: C.W. Houchen is a founder of COARE Biotechnology, Inc. The other authors declare no competing financial interests.- Published
- 2016
- Full Text
- View/download PDF
7. Plasma DCLK1 is a marker of hepatocellular carcinoma (HCC): Targeting DCLK1 prevents HCC tumor xenograft growth via a microRNA-dependent mechanism.
- Author
-
Sureban SM, Madhoun MF, May R, Qu D, Ali N, Fazili J, Weygant N, Chandrakesan P, Ding K, Lightfoot SA, and Houchen CW
- Subjects
- Animals, Biomarkers, Tumor genetics, Carcinoma, Hepatocellular blood, Carcinoma, Hepatocellular enzymology, Carcinoma, Hepatocellular genetics, Carcinoma, Hepatocellular pathology, Cell Line, Tumor, Cell Proliferation, Databases, Genetic, Doublecortin-Like Kinases, Gene Expression Regulation, Neoplastic, Humans, Intracellular Signaling Peptides and Proteins genetics, Kruppel-Like Factor 4, Liver Cirrhosis blood, Liver Cirrhosis enzymology, Liver Neoplasms blood, Liver Neoplasms enzymology, Liver Neoplasms genetics, Liver Neoplasms pathology, Mice, Nude, MicroRNAs genetics, Neoplastic Stem Cells enzymology, Neoplastic Stem Cells pathology, Phenotype, Protein Serine-Threonine Kinases genetics, RNA Interference, Retrospective Studies, Signal Transduction, Time Factors, Transcription Factors genetics, Transcription Factors metabolism, Transfection, Tumor Burden, Up-Regulation, Xenograft Model Antitumor Assays, Biomarkers, Tumor blood, Carcinoma, Hepatocellular therapy, Gene Knockdown Techniques, Genetic Therapy methods, Intracellular Signaling Peptides and Proteins blood, Liver Neoplasms therapy, MicroRNAs metabolism, Protein Serine-Threonine Kinases blood, RNAi Therapeutics
- Abstract
Tumor stem cell marker Doublecortin-like kinase1 (DCLK1) is upregulated in several solid tumors. The role of DCLK1 in hepatocellular carcinoma (HCC) is unclear. We immunostained tissues from human livers with HCC, cirrhosis controls (CC), and non-cirrhosis controls (NCC) for DCLK1. Western blot and ELISA analyses for DCLK1 were performed with stored plasma samples. We observed increased immunoreactive DCLK1 in epithelia and stroma in HCC and CCs compared with NCCs, and observed a marked increase in plasma DCLK1 from patients with HCC compared with CC and NCC. Analysis of the Cancer Genome Atlas' HCC dataset revealed that DCLK1 is overexpressed in HCC tumors relative to adjacent normal tissues. High DCLK1-expressing cells had more epithelial-mesenchymal transition (EMT). Various tumor suppressor miRNAs were also downregulated in HCC tumors. We evaluated the effects of DCLK1 knockdown on Huh7.5-derived tumor xenograft growth. This was associated with growth arrest and a marked downregulation of cMYC, and EMT transcription factors ZEB1, ZEB2, SNAIL, and SLUG via let-7a and miR-200 miRNA-dependent mechanisms. Furthermore, upregulation of miR-143/145, a corresponding decrease in pluripotency factors OCT4, NANOG, KLF4, and LIN28, and a reduction of let-7a, miR-143/145, and miR-200-specific luciferase activity was observed. These findings suggest that the detection of elevated plasma DCLK1 may provide a cost-effective, less invasive tool for confirmation of clinical signs of cirrhosis, and a potential companion diagnostic marker for patients with cirrhosis and HCC. Our results support evaluating DCLK1 as a biomarker for detection and as a therapeutic target for eradicating HCC.
- Published
- 2015
- Full Text
- View/download PDF
8. Dclk1+ small intestinal epithelial tuft cells display the hallmarks of quiescence and self-renewal.
- Author
-
Chandrakesan P, May R, Qu D, Weygant N, Taylor VE, Li JD, Ali N, Sureban SM, Qante M, Wang TC, Bronze MS, and Houchen CW
- Subjects
- Animals, Apoptosis, Cells, Cultured, Doublecortin-Like Kinases, Epithelial Cells physiology, Humans, Immunoenzyme Techniques, In Situ Hybridization, Fluorescence, Intestine, Small physiology, Longevity, Mice, Mice, Transgenic, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Cell Differentiation, Cell Proliferation, Epithelial Cells cytology, Intestine, Small cytology, Protein Serine-Threonine Kinases physiology
- Abstract
To date, no discrete genetic signature has been defined for isolated Dclk1+ tuft cells within the small intestine. Furthermore, recent reports on the functional significance of Dclk1+ cells in the small intestine have been inconsistent. These cells have been proposed to be fully differentiated cells, reserve stem cells, and tumor stem cells. In order to elucidate the potential function of Dclk1+ cells, we FACS-sorted Dclk1+ cells from mouse small intestinal epithelium using transgenic mice expressing YFP under the control of the Dclk1 promoter (Dclk1-CreER;Rosa26-YFP). Analysis of sorted YFP+ cells demonstrated marked enrichment (~6000 fold) for Dclk1 mRNA compared with YFP- cells. Dclk1+ population display ~6 fold enrichment for the putative quiescent stem cell marker Bmi1. We observed significantly greater expression of pluripotency genes, pro-survival genes, and quiescence markers in the Dclk1+ population. A significant increase in self-renewal capability (14-fold) was observed in in vitro isolated Dclk1+ cells. The unique genetic report presented in this manuscript suggests that Dclk1+ cells may maintain quiescence, pluripotency, and metabolic activity for survival/longevity. Functionally, these reserve characteristics manifest in vitro, with Dclk1+ cells exhibiting greater ability to self-renew. These findings indicate that quiescent stem-like functionality is a feature of Dclk1-expressing tuft cells.
- Published
- 2015
- Full Text
- View/download PDF
9. Inflammatory and oncogenic roles of a tumor stem cell marker doublecortin-like kinase (DCLK1) in virus-induced chronic liver diseases.
- Author
-
Ali N, Chandrakesan P, Nguyen CB, Husain S, Gillaspy AF, Huycke M, Berry WL, May R, Qu D, Weygant N, Sureban SM, Bronze MS, Dhanasekaran DN, and Houchen CW
- Subjects
- Animals, Cell Line, Tumor, Disease Models, Animal, Doublecortin-Like Kinases, Humans, Liver Neoplasms pathology, Mice, Mice, Nude, Transcriptome, Inflammation metabolism, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Liver Neoplasms genetics, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism
- Abstract
Hepatocellular carcinoma (HCC) is the third most common cause of cancer-related mortality worldwide. We previously showed that a tumor/cancer stem cell (CSC) marker, doublecortin-like kinase (DCLK1) positively regulates hepatitis C virus (HCV) replication, and promotes tumor growth in colon and pancreas. Here, we employed transcriptome analysis, RNA interference, tumor xenografts, patient's liver tissues and hepatospheroids to investigate DCLK1-regulated inflammation and tumorigenesis in the liver. Our studies unveiled novel DCLK1-controlled feed-forward signaling cascades involving calprotectin subunit S100A9 and NFκB activation as a driver of inflammation. Validation of transcriptome data suggests that DCLK1 co-expression with HCV induces BRM/SMARCA2 of SW1/SNF1 chromatin remodeling complexes. Frequently observed lymphoid aggregates including hepatic epithelial and stromal cells of internodular septa extensively express DCLK1 and S100A9. The DCLK1 overexpression also correlates with increased levels of S100A9, c-Myc, and BRM levels in HCV/HBV-positive patients with cirrhosis and HCC. DCLK1 silencing inhibits S100A9 expression and hepatoma cell migration. Normal human hepatocytes (NHH)-derived spheroids exhibit CSC properties. These results provide new insights into the molecular mechanism of the hepatitis B/C-virus induced liver inflammation and tumorigenesis via DCLK1-controlled networks. Thus, DCLK1 appears to be a novel therapeutic target for the treatment of inflammatory diseases and HCC.
- Published
- 2015
- Full Text
- View/download PDF
10. Ablation of Doublecortin-Like Kinase 1 in the Colonic Epithelium Exacerbates Dextran Sulfate Sodium-Induced Colitis.
- Author
-
Qu D, Weygant N, May R, Chandrakesan P, Madhoun M, Ali N, Sureban SM, An G, Schlosser MJ, and Houchen CW
- Subjects
- Animals, Apoptosis drug effects, Apoptosis genetics, Cell Proliferation drug effects, Cell Proliferation genetics, Colitis chemically induced, Colitis enzymology, Colitis pathology, Colon metabolism, Colon pathology, Doublecortin-Like Kinases, Intestinal Mucosa metabolism, Intestinal Mucosa pathology, Male, Mice, Stem Cells drug effects, Colitis genetics, Colon drug effects, Dextran Sulfate adverse effects, Gene Deletion, Intestinal Mucosa drug effects, Protein Serine-Threonine Kinases deficiency, Protein Serine-Threonine Kinases genetics
- Abstract
Doublecortin-like kinase 1 (Dclk1), a microtubule-associated kinase, marks the fifth lineage of intestinal epithelial cells called tuft cells that function as tumor stem cells in Apc mutant models of colon cancer. In order to determine the role of Dclk1 in dextran sulfate sodium (DSS) induced colonic inflammation both intestinal epithelial specific Dclk1 deficient (VillinCre;Dclk1f/f) and control (Dclk1f/f) mice were fed 3% DSS in drinking water for 9 days, allowed to recover for 2 days, and killed. The clinical and histological features of DSS-induced colitis were scored and immunohistochemical, gene expression, pro-inflammatory cytokines/chemokines, and immunoblotting analyses were used to examine epithelial barrier integrity, inflammation, and stem and tuft cell features. In DSS-induced colitis, VillinCre;Dclk1f/f mice demonstrated exacerbated injury including higher clinical colitis scores, increased epithelial barrier permeability, higher levels of pro-inflammatory cytokines and chemokines, decreased levels of Lgr5, and dysregulated Wnt/b-Catenin pathway genes. These results suggest that Dclk1 plays an important role in regulating colonic inflammatory response and colonic epithelial integrity.
- Published
- 2015
- Full Text
- View/download PDF
11. Dietary Pectin Increases Intestinal Crypt Stem Cell Survival following Radiation Injury.
- Author
-
Sureban SM, May R, Qu D, Chandrakesan P, Weygant N, Ali N, Lightfoot SA, Ding K, Umar S, Schlosser MJ, and Houchen CW
- Subjects
- Animals, Cell Survival drug effects, Dietary Supplements analysis, Doublecortin-Like Kinases, Female, Gastric Mucosa cytology, Gastric Mucosa metabolism, Gene Expression Regulation drug effects, Mice, Mice, Inbred C57BL, Mucositis diet therapy, Mucositis etiology, Mucositis pathology, Pectins pharmacology, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Radiation Injuries, Experimental diet therapy, Radiation Injuries, Experimental pathology, Radiation-Protective Agents pharmacology, Stem Cells metabolism, Stem Cells radiation effects, Survival Analysis, Whole-Body Irradiation, Mucositis prevention & control, Pectins administration & dosage, Radiation Injuries, Experimental prevention & control, Radiation-Protective Agents administration & dosage, Stem Cells drug effects
- Abstract
Gastrointestinal (GI) mucosal damage is a devastating adverse effect of radiation therapy. We have recently reported that expression of Dclk1, a Tuft cell and tumor stem cell (TSC) marker, 24h after high dose total-body gamma-IR (TBI) can be used as a surrogate marker for crypt survival. Dietary pectin has been demonstrated to possess chemopreventive properties, whereas its radioprotective property has not been studied. The aim of this study was to determine the effects of dietary pectin on ionizing radiation (IR)-induced intestinal stem cell (ISC) deletion, crypt and overall survival following lethal TBI. C57BL/6 mice received a 6% pectin diet and 0.5% pectin drinking water (pre-IR mice received pectin one week before TBI until death; post-IR mice received pectin after TBI until death). Animals were exposed to TBI (14 Gy) and euthanized at 24 and 84h post-IR to assess ISC deletion and crypt survival respectively. Animals were also subjected to overall survival studies following TBI. In pre-IR treatment group, we observed a three-fold increase in ISC/crypt survival, a two-fold increase in Dclk1+ stem cells, increased overall survival (median 10d vs. 7d), and increased expression of Dclk1, Msi1, Lgr5, Bmi1, and Notch1 (in small intestine) post-TBI in pectin treated mice compared to controls. We also observed increased survival of mice treated with pectin (post-IR) compared to controls. Dietary pectin is a radioprotective agent; prevents IR-induced deletion of potential reserve ISCs; facilitates crypt regeneration; and ultimately promotes overall survival. Given the anti-cancer activity of pectin, our data support a potential role for dietary pectin as an agent that can be administered to patients receiving radiation therapy to protect against radiation-induces mucositis.
- Published
- 2015
- Full Text
- View/download PDF
12. Regulation of miRNAs by agents targeting the tumor stem cell markers DCLK1, MSI1, LGR5, and BMI1.
- Author
-
Sureban SM, Qu D, and Houchen CW
- Abstract
Gastrointestinal cancers such as colorectal, pancreatic, liver, gastric, and esophageal, are the most common forms of malignant cancers. MicroRNAs (miRNA) play important role in regulating gastrointestinal cancer progress either as potent oncogenes or tumor suppressors. In this report, we will discuss the importance of several tumor suppressors involved in colon or pancreatic cancer. Some recent studies on tumor stem cells and regulation of these miRNAs via agents targeting the tumor stem cell markers doublecortin-like kinase 1 (DCLK1), Musashi-1 (MSI1), polycomb protein BMI1, and WNT genes (LGR5 and ASCL2) will also be discussed. Agents such as siRNA/shRNA, small molecule kinase inhibitors, and general herbal drugs (curcumin) targeting these tumor stem cell markers and tumor suppressor miRNAs could be the perfect therapeutic agents for the treatment of these cancers.
- Published
- 2015
- Full Text
- View/download PDF
13. Doublecortin-like kinase 1 is elevated serologically in pancreatic ductal adenocarcinoma and widely expressed on circulating tumor cells.
- Author
-
Qu D, Johnson J, Chandrakesan P, Weygant N, May R, Aiello N, Rhim A, Zhao L, Zheng W, Lightfoot S, Pant S, Irvan J, Postier R, Hocker J, Hanas JS, Ali N, Sureban SM, An G, Schlosser MJ, Stanger B, and Houchen CW
- Subjects
- Adult, Aged, Animals, CA-19-9 Antigen blood, Carcinoma, Pancreatic Ductal pathology, Case-Control Studies, Doublecortin-Like Kinases, Epithelial Cells metabolism, Epithelial Cells pathology, Female, Gene Expression, Humans, Male, Mice, Middle Aged, Neoplasm Staging, Pancreatic Neoplasms pathology, Stromal Cells metabolism, Stromal Cells pathology, Carcinoma, Pancreatic Ductal blood, Carcinoma, Pancreatic Ductal genetics, Intracellular Signaling Peptides and Proteins blood, Intracellular Signaling Peptides and Proteins genetics, Neoplastic Cells, Circulating metabolism, Pancreatic Neoplasms blood, Pancreatic Neoplasms genetics, Protein Serine-Threonine Kinases blood, Protein Serine-Threonine Kinases genetics
- Abstract
Doublecortin-like kinase 1 (DCLK1) is a putative pancreatic stem cell marker and is upregulated in pancreatic cancer, colorectal cancer, and many other solid tumors. It marks tumor stem cells in mouse models of intestinal neoplasia. Here we sought to determine whether DCLK1 protein can be detected in the bloodstream and if its levels in archived serum samples could be quantitatively assessed in pancreatic cancer patients. DCLK1 specific ELISA, western blotting, and immunohistochemical analyses were used to determine expression levels in the serum and staining intensity in archived tumor tissues of pancreatic ductal adenocarcinoma (PDAC) patients and in pancreatic cancer mouse models. DCLK1 levels in the serum were elevated in early stages of PDAC (stages I and II) compared to healthy volunteers (normal controls). No differences were observed between stages III/IV and normal controls. In resected surgical tissues, DCLK1 expression intensity in the stromal cells was significantly higher than that observed in tumor epithelial cells. Circulating tumor cells were isolated from KPCY mice and approximately 52% of these cells were positive for Dclk1 staining. Dclk1 levels in the serum of KPC mice were also elevated. We have previously demonstrated that DCLK1 plays a potential role in regulating epithelial mesenchymal transition (EMT). Given the increasingly recognized role of EMT derived stem cells in cancer progression and metastasis, we hypothesize that DCLK1 may contribute to the metastatic process. Taken together, our results suggest that DCLK1 serum levels and DCLK1 positive circulating tumor cells should be further assessed for their potential diagnostic and prognostic significance.
- Published
- 2015
- Full Text
- View/download PDF
14. DCLK1 is a broadly dysregulated target against epithelial-mesenchymal transition, focal adhesion, and stemness in clear cell renal carcinoma.
- Author
-
Weygant N, Qu D, May R, Tierney RM, Berry WL, Zhao L, Agarwal S, Chandrakesan P, Chinthalapally HR, Murphy NT, Li JD, Sureban SM, Schlosser MJ, Tomasek JJ, and Houchen CW
- Subjects
- Blotting, Western, Carcinoma, Renal Cell metabolism, Carcinoma, Renal Cell pathology, Cell Line, Tumor, Cell Movement genetics, DNA Methylation, Doublecortin-Like Kinases, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Humans, Intracellular Signaling Peptides and Proteins metabolism, Kidney Neoplasms metabolism, Kidney Neoplasms pathology, Male, Middle Aged, Neoplastic Stem Cells metabolism, Promoter Regions, Genetic genetics, Protein Serine-Threonine Kinases metabolism, RNA Interference, Reverse Transcriptase Polymerase Chain Reaction, Carcinoma, Renal Cell genetics, Epithelial-Mesenchymal Transition genetics, Focal Adhesions genetics, Intracellular Signaling Peptides and Proteins genetics, Kidney Neoplasms genetics, Protein Serine-Threonine Kinases genetics
- Abstract
Renal clear cell carcinoma (RCC) is the most common type of kidney cancer and the 8th most common cancer overall in the US. RCC survival rates drop precipitously with regional and distant spread and recent studies have demonstrated that RCC presents an epithelial-mesenchymal transition (EMT) phenotype linked to increased recurrence and decreased survival. EMT is a key characteristic of tumor stem cells (TSCs) along with chemo-resistance and radio-resistance, which are also phenotypic of RCC. Targeting these factors is key to increasing the survival of RCC patients. Doublecortin-like kinase 1 (DCLK1) marks TSCs in pancreatic and colorectal cancer and regulates EMT and stemness. Analysis of the Cancer Genome Atlas' RCC dataset revealed that DCLK1 is overexpressed and dysregulated on the mRNA and epigenetic level in more than 93% of RCC tumors relative to adjacent normal tissue. Immunohistochemistry using α-DCLK1 antibody confirmed overexpression and demonstrated a major increase in immunoreactivity in stage II-III tumors compared to normal kidney and stage I tumors. Small-interfering RNA (siRNA) mediated knockdown of DCLK1 resulted in decreased expression of EMT and pluripotency factors and significantly reduced invasion, migration, focal adhesion, drug-resistance, and clonogenic capacity. These findings suggest that DCLK1 is a novel, overexpressed factor in RCC progression that may be targeted to suppress EMT, metastasis, and stemness in early-stage and advanced RCC to increase patient survival. Moreover, the possibility that DCLK1 may mark a population of tumor stem-like cells in RCC should be further investigated in light of these findings.
- Published
- 2015
- Full Text
- View/download PDF
15. DCLK1 is detectable in plasma of patients with Barrett's esophagus and esophageal adenocarcinoma.
- Author
-
Whorton J, Sureban SM, May R, Qu D, Lightfoot SA, Madhoun M, Johnson M, Tierney WM, Maple JT, Vega KJ, and Houchen CW
- Subjects
- Adenocarcinoma blood, Adenocarcinoma pathology, Barrett Esophagus blood, Barrett Esophagus pathology, Blotting, Western, Case-Control Studies, Doublecortin-Like Kinases, Enzyme-Linked Immunosorbent Assay, Epithelial Cells enzymology, Esophageal Neoplasms blood, Esophageal Neoplasms pathology, Esophagoscopy, Humans, Immunohistochemistry, Predictive Value of Tests, Prognosis, Prospective Studies, Stromal Cells enzymology, Up-Regulation, Adenocarcinoma enzymology, Barrett Esophagus enzymology, Biomarkers, Tumor blood, Esophageal Neoplasms enzymology, Intracellular Signaling Peptides and Proteins blood, Protein Serine-Threonine Kinases blood
- Abstract
Background: Doublecortin-like kinase 1 (DCLK1), a putative tumor stem cell marker has been shown to be highly expressed in the stromal and epithelial compartments in colon and pancreatic cancer as well as Barrett's esophagus (BE) and esophageal adenocarcinoma (EAC)., Aim: To prospectively investigate whether the immunohistochemical expression of DCLK1 was associated with detectable DCLK1 plasma expression in patients with existing BE and EAC., Methods: Immunohistochemistry was performed on paraffin-embedded sections using DCLK1 antibody and scored based on staining intensity and tissue involvement. Purified human plasma samples were subjected to Western blot and ELISA analysis., Results: Forty (40) patients were enrolled: 10 controls (normal endoscopy) and 30 with BE/EAC (13 nondysplastic BE [NDBE], 9 dysplastic BE [DBE] and 8 EAC). Mean epithelial DCLK1 staining was as follows: controls = 0.11, NDBE = 3.83, DBE = 6.0, EAC = 7.17. Mean stromal DCLK1 staining was as follows: NDBE = 5.83, DBE = 5.375, EAC = 10.83. DCLK1 was detected by plasma Western blot in 1 control and in all patients with BE/EAC p < 0.0005. Plasma DCLK1 was elevated by ELISA in EAC compared to other groups, p < 0.05., Conclusions: Increased expression of DCLK1 was observed in the epithelium, stroma and plasma of patients with BE/EAC. Furthermore, the presence of detectable DCLK1 in plasma of BE/EAC patients may provide a less invasive, detection tool in those patients as well as represent a novel molecular marker distinguishing between normal esophageal mucosa and BE or EAC.
- Published
- 2015
- Full Text
- View/download PDF
16. DCLK1 facilitates intestinal tumor growth via enhancing pluripotency and epithelial mesenchymal transition.
- Author
-
Chandrakesan P, Weygant N, May R, Qu D, Chinthalapally HR, Sureban SM, Ali N, Lightfoot SA, Umar S, and Houchen CW
- Subjects
- Adenocarcinoma genetics, Adenoma genetics, Animals, Cell Transformation, Neoplastic genetics, Doublecortin-Like Kinases, Epithelial Cells pathology, Intestinal Mucosa metabolism, Intestinal Neoplasms genetics, Intestines cytology, Mice, Mice, Inbred C57BL, Mice, Transgenic, MicroRNAs genetics, Protein Serine-Threonine Kinases biosynthesis, RNA Interference, RNA, Small Interfering, Spheroids, Cellular, Tumor Cells, Cultured, Adenomatous Polyposis Coli Protein genetics, Epithelial-Mesenchymal Transition genetics, Intestinal Neoplasms pathology, Neoplastic Stem Cells pathology, Protein Serine-Threonine Kinases genetics
- Abstract
Doublecortin-like kinase 1 (Dclk1) is overexpressed in many cancers including colorectal cancer (CRC) andit specifically marks intestinal tumor stem cells. However, the role of Dclk1 in intestinal tumorigenesis in Apc mutant conditions is still poorly understood. We demonstrate that Dclk1 expression and Dclk1+ cells are significantly increased in the intestinal epithelium of elderly ApcMin/+ mice compared to young ApcMin/+ mice and wild type mice. Intestinal epithelial cells of ApcMin/+ mice demonstrate increased pluripotency, self-renewing ability, and EMT. Furthermore, miRNAs are dysregulated, expression of onco-miRNAs are significantly increased with decreased tumor suppressor miRNAs. In support of these findings, knockdown of Dclk1 in elderly ApcMin/+ mice attenuates intestinal adenomas and adenocarcinoma by decreasing pluripotency, EMT and onco-miRNAs indicating that Dclk1 overexpression facilitates intestinal tumorigenesis. Knocking down Dclk1 weakens Dclk1-dependent intestinal processes for tumorigenesis. This study demonstrates that Dclk1 is critically involved in facilitating intestinal tumorigenesis by enhancing pluripotency and EMT factors in Apc mutant intestinal tumors and it also provides a potential therapeutic target for the treatment of colorectal cancer.
- Published
- 2014
- Full Text
- View/download PDF
17. XMD8-92 inhibits pancreatic tumor xenograft growth via a DCLK1-dependent mechanism.
- Author
-
Sureban SM, May R, Weygant N, Qu D, Chandrakesan P, Bannerman-Menson E, Ali N, Pantazis P, Westphalen CB, Wang TC, and Houchen CW
- Subjects
- Animals, Carcinoma, Pancreatic Ductal enzymology, Carcinoma, Pancreatic Ductal pathology, Cell Line, Tumor, Cell Proliferation drug effects, Doublecortin-Like Kinases, Epithelial-Mesenchymal Transition drug effects, Gene Expression drug effects, Intracellular Signaling Peptides and Proteins genetics, Kruppel-Like Factor 4, Mice, Mice, Inbred NOD, Mice, SCID, MicroRNAs genetics, MicroRNAs metabolism, Pancreatic Neoplasms enzymology, Pancreatic Neoplasms pathology, Protein Serine-Threonine Kinases genetics, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins c-myc metabolism, Proto-Oncogene Proteins p21(ras), RNA, Messenger genetics, RNA, Messenger metabolism, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, Receptor, Notch1 genetics, Receptor, Notch1 metabolism, Tumor Burden drug effects, Xenograft Model Antitumor Assays, ras Proteins genetics, ras Proteins metabolism, Antineoplastic Agents pharmacology, Benzodiazepinones pharmacology, Carcinoma, Pancreatic Ductal drug therapy, Intracellular Signaling Peptides and Proteins metabolism, Pancreatic Neoplasms drug therapy, Protein Serine-Threonine Kinases metabolism
- Abstract
XMD8-92 is a kinase inhibitor with anti-cancer activity against lung and cervical cancers, but its effect on pancreatic ductal adenocarcinoma (PDAC) remains unknown. Doublecortin-like kinase1 (DCLK1) is upregulated in various cancers including PDAC. In this study, we showed that XMD8-92 inhibits AsPC-1 cancer cell proliferation and tumor xenograft growth. XMD8-92 treated tumors demonstrated significant downregulation of DCLK1 and several of its downstream targets (including c-MYC, KRAS, NOTCH1, ZEB1, ZEB2, SNAIL, SLUG, OCT4, SOX2, NANOG, KLF4, LIN28, VEGFR1, and VEGFR2) via upregulation of tumor suppressor miRNAs let-7a, miR-144, miR-200a-c, and miR-143/145; it did not however affect BMK1 downstream genes p21 and p53. These data taken together suggest that XMD8-92 treatment results in inhibition of DCLK1 and downstream oncogenic pathways (EMT, pluripotency, angiogenesis and anti-apoptotic), and is a promising chemotherapeutic agent against PDAC., (Copyright © 2014. Published by Elsevier Ireland Ltd.)
- Published
- 2014
- Full Text
- View/download PDF
18. Wnt inhibitory factor 1 suppresses cancer stemness and induces cellular senescence.
- Author
-
Ramachandran I, Ganapathy V, Gillies E, Fonseca I, Sureban SM, Houchen CW, Reis A, and Queimado L
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adenoma, Pleomorphic genetics, Adenoma, Pleomorphic pathology, Biomarkers, Tumor genetics, Carcinoma genetics, Carcinoma pathology, Cell Cycle Checkpoints, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Cell Line, Tumor, Cell Proliferation, DNA Methylation, Disease Progression, Down-Regulation, Gene Expression Regulation, Neoplastic, Humans, Loss of Heterozygosity, MicroRNAs metabolism, Neoplastic Stem Cells pathology, Promoter Regions, Genetic, Repressor Proteins genetics, Salivary Gland Neoplasms genetics, Salivary Gland Neoplasms pathology, Time Factors, Transfection, Adaptor Proteins, Signal Transducing metabolism, Adenoma, Pleomorphic metabolism, Biomarkers, Tumor metabolism, Carcinoma metabolism, Cellular Senescence, Neoplastic Stem Cells metabolism, Repressor Proteins metabolism, Salivary Gland Neoplasms metabolism, Wnt Signaling Pathway
- Abstract
Hyperactivation of the Wingless-type (Wnt)/β-catenin pathway promotes tumor initiation, tumor growth and metastasis in various tissues. Although there is evidence for the involvement of Wnt/β-catenin pathway activation in salivary gland tumors, the precise mechanisms are unknown. Here we report for the first time that downregulation of the Wnt inhibitory factor 1 (WIF1) is a widespread event in salivary gland carcinoma ex-pleomorphic adenoma (CaExPA). We also show that WIF1 downregulation occurs in the CaExPA precursor lesion pleomorphic adenoma (PA) and indicates a higher risk of progression from benign to malignant tumor. Our results demonstrate that diverse mechanisms including WIF1 promoter hypermethylation and loss of heterozygosity contribute to WIF1 downregulation in human salivary gland tumors. In accordance with a crucial role in suppressing salivary gland tumor progression, WIF1 re-expression in salivary gland tumor cells inhibited cell proliferation, induced more differentiated phenotype and promoted cellular senescence, possibly through upregulation of tumor-suppressor genes, such as p53 and p21. Most importantly, WIF1 significantly diminished the number of salivary gland cancer stem cells and the anchorage-independent cell growth. Consistent with this observation, WIF1 caused a reduction in the expression of pluripotency and stemness markers (OCT4 and c-MYC), as well as adult stem cell self-renewal and multi-lineage differentiation markers, such as WNT3A, TCF4, c-KIT and MYB. Furthermore, WIF1 significantly increased the expression of microRNAs pri-let-7a and pri-miR-200c, negative regulators of stemness and cancer progression. In addition, we show that WIF1 functions as a positive regulator of miR-200c, leading to downregulation of BMI1, ZEB1 and ZEB2, with a consequent increase in downstream targets such as E-cadherin. Our study emphasizes the prognostic and therapeutic potential of WIF1 in human salivary gland CaExPA. Moreover, our findings demonstrate a novel mechanism by which WIF1 regulates cancer stemness and senescence, which might have major implications in the field of cancer biology.
- Published
- 2014
- Full Text
- View/download PDF
19. Small molecule kinase inhibitor LRRK2-IN-1 demonstrates potent activity against colorectal and pancreatic cancer through inhibition of doublecortin-like kinase 1.
- Author
-
Weygant N, Qu D, Berry WL, May R, Chandrakesan P, Owen DB, Sureban SM, Ali N, Janknecht R, and Houchen CW
- Subjects
- Antineoplastic Agents chemistry, Benzodiazepinones chemistry, Binding Sites, Cell Line, Tumor, Cell Movement drug effects, Cell Proliferation drug effects, Cell Survival drug effects, Collagen, Diffusion Chambers, Culture, Doublecortin-Like Kinases, Drug Combinations, Gene Expression, Genetic Vectors, Humans, Intracellular Signaling Peptides and Proteins chemistry, Intracellular Signaling Peptides and Proteins genetics, Isoenzymes antagonists & inhibitors, Isoenzymes chemistry, Isoenzymes genetics, Laminin, Lentivirus genetics, Molecular Docking Simulation, Molecular Dynamics Simulation, Protein Binding, Protein Kinase Inhibitors chemistry, Protein Serine-Threonine Kinases chemistry, Protein Serine-Threonine Kinases genetics, Proteoglycans, Pyrimidines chemistry, Antineoplastic Agents pharmacology, Benzodiazepinones pharmacology, Intracellular Signaling Peptides and Proteins antagonists & inhibitors, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors, Pyrimidines pharmacology
- Abstract
Background: Doublecortin-like kinase 1 (DCLK1) is emerging as a tumor specific stem cell marker in colorectal and pancreatic cancer. Previous in vitro and in vivo studies have demonstrated the therapeutic effects of inhibiting DCLK1 with small interfering RNA (siRNA) as well as genetically targeting the DCLK1+ cell for deletion. However, the effects of inhibiting DCLK1 kinase activity have not been studied directly. Therefore, we assessed the effects of inhibiting DCLK1 kinase activity using the novel small molecule kinase inhibitor, LRRK2-IN-1, which demonstrates significant affinity for DCLK1., Results: Here we report that LRRK2-IN-1 demonstrates potent anti-cancer activity including inhibition of cancer cell proliferation, migration, and invasion as well as induction of apoptosis and cell cycle arrest. Additionally we found that it regulates stemness, epithelial-mesenchymal transition, and oncogenic targets on the molecular level. Moreover, we show that LRRK2-IN-1 suppresses DCLK1 kinase activity and downstream DCLK1 effector c-MYC, and demonstrate that DCLK1 kinase activity is a significant factor in resistance to LRRK2-IN-1., Conclusions: Given DCLK1's tumor stem cell marker status, a strong understanding of its biological role and interactions in gastrointestinal tumors may lead to discoveries that improve patient outcomes. The results of this study suggest that small molecule inhibitors of DCLK1 kinase should be further investigated as they may hold promise as anti-tumor stem cell drugs.
- Published
- 2014
- Full Text
- View/download PDF
20. Brief report: Dclk1 deletion in tuft cells results in impaired epithelial repair after radiation injury.
- Author
-
May R, Qu D, Weygant N, Chandrakesan P, Ali N, Lightfoot SA, Li L, Sureban SM, and Houchen CW
- Subjects
- Animals, Doublecortin-Like Kinases, Gene Expression Regulation, Mice, RNA, Messenger genetics, RNA, Messenger metabolism, Radiation Injuries metabolism, Receptors, Notch genetics, Receptors, Notch metabolism, Stem Cells cytology, Stem Cells metabolism, Survival Analysis, Whole-Body Irradiation, Epithelial Cells metabolism, Epithelial Cells pathology, Gene Deletion, Intestinal Mucosa pathology, Protein Serine-Threonine Kinases genetics, Radiation Injuries pathology, Wound Healing
- Abstract
The role of Dclk1(+) tuft cells in the replacement of intestinal epithelia and reestablishing the epithelial barrier after severe genotoxic insult is completely unknown. Successful restoration requires precise coordination between the cells within each crypt subunit. While the mechanisms that control this response remain largely uncertain, the radiation model remains an exceptional surrogate for stem cell-associated crypt loss. Following the creation of Dclk1-intestinal-epithelial-deficient Villin-Cre;Dclk1(flox/flox) mice, widespread gene expression changes were detected in isolated intestinal epithelia during homeostasis. While the number of surviving crypts was unaffected, Villin-Cre;Dclk1(flox/flox) mice failed to maintain tight junctions and died at approximately 5 days, where Dclk1(flox/flox) mice lived until day 10 following radiation injury. These findings suggest that Dclk1 plays a functional role critical in the epithelial restorative response., (© AlphaMed Press.)
- Published
- 2014
- Full Text
- View/download PDF
21. Inhibition of Notch signaling reduces the number of surviving Dclk1+ reserve crypt epithelial stem cells following radiation injury.
- Author
-
Qu D, May R, Sureban SM, Weygant N, Chandrakesan P, Ali N, Li L, Barrett T, and Houchen CW
- Subjects
- Animals, Biomarkers, Doublecortin-Like Kinases, Epithelial Cells, Female, Intestinal Mucosa cytology, Mice, Mice, Inbred C57BL, Protein Serine-Threonine Kinases genetics, RNA, Messenger genetics, RNA, Messenger metabolism, Receptors, Notch genetics, Stem Cells, Gene Expression Regulation radiation effects, Protein Serine-Threonine Kinases metabolism, Receptors, Notch metabolism, Signal Transduction radiation effects, Whole-Body Irradiation adverse effects
- Abstract
We have previously reported that doublecortin-like kinase 1 (Dclk1) is a putative intestinal stem cell (ISC) marker. In this report, we evaluated the use of Dclk1 as a marker of surviving ISCs in response to treatment with high-dose total body irradiation (TBI). Both apoptotic and mitotic Dclk1(+) cells were observed 24 h post-TBI associated with a corresponding loss of intestinal crypts observed at 84 h post-TBI. Although the Notch signaling pathway plays an important role in regulating proliferation and lineage commitment within the intestine, its role in ISC function in response to severe genotoxic injury is not yet fully understood. We employed the microcolony assay to functionally assess the effects of Notch inhibition with difluorophenacetyl-l-alanyl-S-phenylglycine t-butyl ester (DAPT) on intestinal crypt stem cell survival following severe (>8 Gy) radiation injury. Following treatment with DAPT, we observed a nearly 50% reduction in the number of surviving Dclk1(+) crypt epithelial cells at 24 h after TBI and similar reduction in the number of surviving small intestinal crypts at 84 h. These data indicate that inhibition of Notch signaling decreases ISC survival following radiation injury, suggesting that the Notch signaling pathway plays an important role in ISC-mediated crypt regeneration. These results also suggest that crypt epithelial cell Dclk1 expression can be used as one potential marker to evaluate the early survival of ISCs following severe radiation injury.
- Published
- 2014
- Full Text
- View/download PDF
22. Fluvastatin interferes with hepatitis C virus replication via microtubule bundling and a doublecortin-like kinase-mediated mechanism.
- Author
-
Ali N, Allam H, Bader T, May R, Basalingappa KM, Berry WL, Chandrakesan P, Qu D, Weygant N, Bronze MS, Umar S, Janknecht R, Sureban SM, Huycke M, and Houchen CW
- Subjects
- Antineoplastic Agents pharmacology, Antiviral Agents pharmacology, Carcinoma, Hepatocellular drug therapy, Carcinoma, Hepatocellular genetics, Carcinoma, Hepatocellular metabolism, Carcinoma, Hepatocellular virology, Cell Cycle Checkpoints drug effects, Cell Cycle Checkpoints genetics, Cell Line, Tumor, Cell Survival drug effects, Cell Survival genetics, Doublecortin-Like Kinases, Down-Regulation drug effects, Down-Regulation genetics, Fluvastatin, Hepacivirus genetics, Hepacivirus metabolism, Hepatocytes drug effects, Hepatocytes metabolism, Humans, Intracellular Signaling Peptides and Proteins genetics, Microtubules genetics, Protein Serine-Threonine Kinases genetics, Virus Replication genetics, Fatty Acids, Monounsaturated pharmacology, Hepacivirus drug effects, Indoles pharmacology, Intracellular Signaling Peptides and Proteins metabolism, Microtubules metabolism, Protein Serine-Threonine Kinases metabolism, Virus Replication drug effects
- Abstract
Hepatitis C virus (HCV)-induced alterations in lipid metabolism and cellular protein expression contribute to viral pathogenesis. The mechanism of pleiotropic actions of cholesterol-lowering drugs, statins, against HCV and multiple cancers are not well understood. We investigated effects of fluvastatin (FLV) on microtubule-associated and cancer stem cell marker (CSC), doublecortin-like kinase 1 (DCLK1) during HCV-induced hepatocarcinogenesis. HCV replication models, cancer cell lines and normal human hepatocytes were used to investigate the antiviral and antitumor effects of statins. FLV treatment resulted in induction of microtubule bundling, cell-cycle arrest and alterations in cellular DCLK1 distribution in HCV-expressing hepatoma cells. These events adversely affected the survival of liver-derived tumor cells without affecting normal human hepatocytes. FLV downregulated HCV replication in cell culture where the ATP pool and cell viability were not compromised. Pravastatin did not exhibit these effects on HCV replication, microtubules and cancer cells. The levels of miR-122 that regulates liver homeostasis and provides HCV genomic stability remained at steady state whereas DCLK1 mRNA levels were considerably reduced during FLV treatment. We further demonstrated that HCV replication was increased with DCLK1 overexpression. In conclusion, unique effects of FLV on microtubules and their binding partner DCLK1 are likely to contribute to its anti-HCV and antitumor activities in addition to its known inhibitory effects on 3-hydroxy-3-methylglutary-CoA reductase (HMGCR).
- Published
- 2013
- Full Text
- View/download PDF
23. DCLK1 regulates pluripotency and angiogenic factors via microRNA-dependent mechanisms in pancreatic cancer.
- Author
-
Sureban SM, May R, Qu D, Weygant N, Chandrakesan P, Ali N, Lightfoot SA, Pantazis P, Rao CV, Postier RG, and Houchen CW
- Subjects
- Animals, Cell Line, Tumor, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Disease Models, Animal, Doublecortin-Like Kinases, Epithelial-Mesenchymal Transition genetics, Gene Expression Regulation, Neoplastic, Gene Silencing, Humans, Intracellular Signaling Peptides and Proteins metabolism, Kruppel-Like Factor 4, Mice, Neoplastic Stem Cells metabolism, Pancreatic Neoplasms pathology, Protein Serine-Threonine Kinases metabolism, RNA Processing, Post-Transcriptional, RNA-Binding Proteins, Tumor Burden genetics, Xenograft Model Antitumor Assays, Intracellular Signaling Peptides and Proteins genetics, MicroRNAs genetics, MicroRNAs metabolism, Neovascularization, Pathologic genetics, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Protein Serine-Threonine Kinases genetics
- Abstract
Stem cell pluripotency, angiogenesis and epithelial-mesenchymal transition (EMT) have been shown to be significantly upregulated in pancreatic ductal adenocarcinoma (PDAC) and many other aggressive cancers. The dysregulation of these processes is believed to play key roles in tumor initiation, progression, and metastasis, and is contributory to PDAC being the fourth leading cause of cancer-related deaths in the US. The tumor suppressor miRNA miR-145 downregulates critical pluripotency factors and oncogenes and results in repressed metastatic potential in PDAC. Additionally, the miR-200 family regulates several angiogenic factors which have been linked to metastasis in many solid tumors. We have previously demonstrated that downregulation of DCLK1 can upregulate critical miRNAs in both in vitro and in vivo cancer models and results in downregulation of c-MYC, KRAS, NOTCH1 and EMT-related transcription factors. A recent report has also shown that Dclk1 can distinguish between normal and tumor stem cells in Apc (min/+) mice and that ablation of Dclk1(+) cells resulted in regression of intestinal polyps without affecting homeostasis. Here we demonstrate that the knockdown of DCLK1 using poly(lactide-co-glycolide)-encapsulated-DCLK1-siRNA results in AsPC1 tumor growth arrest. Examination of xenograft tumors revealed, (a) increased miR-145 which results in decreased pluripotency maintenance factors OCT4, SOX2, NANOG, KLF4 as well as KRAS and RREB1; (b) increased let-7a which results in decreased pluripotency factor LIN28B; and (c) increased miR-200 which results in decreased VEGFR1, VEGFR2 and EMT-related transcription factors ZEB1, ZEB2, SNAIL and SLUG. Specificity of DCLK1 post-transcriptional regulation of the downstream targets of miR-145, miR-200 and let-7a was accomplished utilizing a luciferase-based reporter assay. We conclude that DCLK1 plays a significant master regulatory role in pancreatic tumorigenesis through the regulation of multiple tumor suppressor miRNAs and their downstream pro-tumorigenic pathways. This novel concept of targeting DCLK1 alone has several advantages over targeting single pathway or miRNA-based therapies for PDAC.
- Published
- 2013
- Full Text
- View/download PDF
24. Isolation of anticancer drug TAXOL from Pestalotiopsis breviseta with apoptosis and B-Cell lymphoma protein docking studies.
- Author
-
Kathiravan G, Sureban SM, Sree HN, Bhuvaneshwari V, and Kramony E
- Abstract
Background: Extraction and investigation of TAXOL from Pestalotiopsis breviseta (Sacc.) using protein docking, which is a computational technique that samples conformations of small molecules in protein-binding sites. Scoring functions are used to assess which of these conformations best complements the protein binding site and active site prediction., Materials and Methods: Coelomycetous fungi P. breviseta (Sacc.) Steyaert was screened for the production of TAXOL, an anticancer drug., Results: TAXOL PRODUCTION WAS CONFIRMED BY THE FOLLOWING METHODS: Ultraviolet (UV) spectroscopic analysis, Infrared analysis, High performance liquid chromatography analysis (HPLC), and Liquid chromatography mass spectrum (LC-MASS). TAXOL produced by the fungi was compared with authentic TAXOL, and protein docking studies were performed., Conclusion: The BCL2 protein of human origin showed a higher affinity toward the compound paclitaxel. It has the binding energy value of -13.0061 (KJ/Mol) with four hydrogen bonds.
- Published
- 2012
- Full Text
- View/download PDF
25. A word from Journal of Basic and Clinical Pharmacy editor's desk.
- Author
-
Sureban SM and Sree HN
- Published
- 2012
- Full Text
- View/download PDF
26. Journal of Basic and Clinical Pharmacy signs collaboration agreement with Medknow Publications-Wolters Kluwer Health.
- Author
-
Harsha S and Sureban SM
- Published
- 2012
- Full Text
- View/download PDF
27. Epigenetic variants and biomarkers for colon cancer.
- Author
-
Qu D, Sureban SM, and Houchen CW
- Subjects
- Humans, Biomarkers, Tumor biosynthesis, Colonic Neoplasms metabolism, Gene Expression Regulation, Neoplastic physiology, Histones biosynthesis
- Published
- 2012
- Full Text
- View/download PDF
28. Identification of the putative intestinal stem cell marker doublecortin and CaM kinase-like-1 in Barrett's esophagus and esophageal adenocarcinoma.
- Author
-
Vega KJ, May R, Sureban SM, Lightfoot SA, Qu D, Reed A, Weygant N, Ramanujam R, Souza R, Madhoun M, Whorton J, Anant S, Meltzer SJ, and Houchen CW
- Subjects
- Barrett Esophagus pathology, Doublecortin-Like Kinases, Humans, Microarray Analysis, Nerve Tissue Proteins metabolism, RNA, Messenger metabolism, RNA-Binding Proteins metabolism, Receptors, G-Protein-Coupled metabolism, Reverse Transcriptase Polymerase Chain Reaction, Up-Regulation, Adenocarcinoma metabolism, Barrett Esophagus metabolism, Biomarkers, Tumor metabolism, Esophageal Neoplasms metabolism, Esophagus metabolism, Intracellular Signaling Peptides and Proteins metabolism, Protein Serine-Threonine Kinases metabolism, Stem Cells metabolism
- Abstract
Background and Aim: In Barrett's esophagus (BE), the normal esophageal squamous epithelium is replaced with a specialized metaplastic columnar epithelium. BE is a premalignant lesion that can progress to esophageal adenocarcinoma (EAC). Currently, there are no early molecular indicators that would predict progression from BE to EAC. As the only permanent residents of the epithelium, stem cells have been implicated in this metaplastic progression. The aim of the present study was to determine the expression of doublecortin and CaM kinase-like-1 (DCAMKL-1) and other putative gastrointestinal stem cell markers in normal esophageal mucosa (NEM), BE, and EAC., Methods: Human NEM, BE, EAC, and multitissue microarrays were analyzed for DCAMKL-1, and immunohistochemically scored based on staining intensity and tissue involvement, with epithelia and stroma scored separately. Total RNA isolated from BE and paired NEM was subjected to real-time reverse-transcription-polymerase chain reaction analysis for DCAMKL-1, leucine-rich repeat-containing G-protein-coupled receptor (LGR5), and Musashi-1 (Msi-1) mRNA expression., Results: DCAMKL-1 was minimally expressed in squamous NEM, but increased in BE (with and without dysplasia) and EAC tissues. In EAC, we found increased stromal DCAMKL-1 staining compared to adjacent epithelia. Within the submucosa of dysplastic BE tissues, an increase in the endothelial cell expression of DCAMKL-1 was observed. Finally, an upregulation of DCAMKL-1, LGR5, and Msi-1 mRNA was seen in BE compared to squamous NEM., Conclusions: In the present study, we report the progressive increase of DCAMKL-1 expression in BE from dysplasia to EAC. Furthermore, there was an increase in putative stem cell markers DCAMKL-1, LGR5, and Msi-1 mRNA. Taken together, these data suggest that the regulation of resident stem cells might play an important role in the progression of BE to EAC., (© 2011 Journal of Gastroenterology and Hepatology Foundation and Blackwell Publishing Asia Pty Ltd.)
- Published
- 2012
- Full Text
- View/download PDF
29. Hepatitis C virus-induced cancer stem cell-like signatures in cell culture and murine tumor xenografts.
- Author
-
Ali N, Allam H, May R, Sureban SM, Bronze MS, Bader T, Umar S, Anant S, and Houchen CW
- Subjects
- Animals, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Carcinoma, Hepatocellular genetics, Carcinoma, Hepatocellular virology, Cell Line, Tumor, Doublecortin-Like Kinases, Female, Flow Cytometry, Hepacivirus genetics, Hepatitis C genetics, Hepatitis C pathology, Hepatitis C virology, Humans, Immunoenzyme Techniques, Intracellular Signaling Peptides and Proteins antagonists & inhibitors, Intracellular Signaling Peptides and Proteins genetics, Keratin-19 genetics, Keratin-19 metabolism, Liver metabolism, Liver virology, Liver Neoplasms genetics, Liver Neoplasms metabolism, Liver Neoplasms virology, Mice, Mice, Inbred BALB C, Mice, Nude, Oligonucleotide Array Sequence Analysis, Protein Serine-Threonine Kinases antagonists & inhibitors, Protein Serine-Threonine Kinases genetics, RNA, Messenger genetics, RNA, Small Interfering genetics, Real-Time Polymerase Chain Reaction, Replicon, Reverse Transcriptase Polymerase Chain Reaction, Transplantation, Heterologous, Carcinoma, Hepatocellular metabolism, Gene Expression Profiling, Hepacivirus pathogenicity, Intracellular Signaling Peptides and Proteins metabolism, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Protein Serine-Threonine Kinases metabolism, RNA, Viral genetics
- Abstract
Hepatitis C virus (HCV) infection is a prominent risk factor for the development of hepatocellular carcinoma (HCC). Similar to most solid tumors, HCCs are believed to contain poorly differentiated cancer stem cell-like cells (CSCs) that initiate tumorigenesis and confer resistance to chemotherapy. In these studies, we demonstrate that the expression of an HCV subgenomic replicon in cultured cells results in the acquisition of CSC traits. These traits include enhanced expression of doublecortin and CaM kinase-like-1 (DCAMKL-1), Lgr5, CD133, α-fetoprotein, cytokeratin-19 (CK19), Lin28, and c-Myc. Conversely, curing of the replicon from these cells results in diminished expression of these factors. The putative stem cell marker DCAMKL-1 is also elevated in response to the overexpression of a cassette of pluripotency factors. The DCAMKL-1-positive cells isolated from hepatoma cell lines by fluorescence-activated cell sorting (FACS) form spheroids in Matrigel. The HCV RNA abundance and NS5B levels are significantly reduced by the small interfering RNA (siRNA)-led depletion of DCAMKL-1. We further demonstrate that HCV replicon-expressing cells initiate distinct tumor phenotypes compared to the tumors initiated by parent cells lacking the replicon. This HCV-induced phenotype is characterized by high-level expression/coexpression of DCAMKL-1, CK19, α-fetoprotein, and active c-Src. The results obtained by the analysis of liver tissues from HCV-positive patients and liver tissue microarrays reiterate these observations. In conclusion, chronic HCV infection appears to predispose cells toward the path of acquiring cancer stem cell-like traits by inducing DCAMKL-1 and hepatic progenitor and stem cell-related factors. DCAMKL-1 also represents a novel cellular target for combating HCV-induced hepatocarcinogenesis.
- Published
- 2011
- Full Text
- View/download PDF
30. Nanoparticle-based delivery of siDCAMKL-1 increases microRNA-144 and inhibits colorectal cancer tumor growth via a Notch-1 dependent mechanism.
- Author
-
Sureban SM, May R, Mondalek FG, Qu D, Ponnurangam S, Pantazis P, Anant S, Ramanujam RP, and Houchen CW
- Subjects
- Animals, Dipeptides pharmacology, Doublecortin-Like Kinases, Down-Regulation, Gene Expression Regulation, Neoplastic drug effects, HCT116 Cells, Humans, Mice, Mice, Nude, Proto-Oncogene Mas, Proto-Oncogene Proteins c-myc metabolism, Transcription Factors metabolism, Up-Regulation, Xenograft Model Antitumor Assays, Carcinoma drug therapy, Colorectal Neoplasms drug therapy, Intracellular Signaling Peptides and Proteins pharmacology, MicroRNAs metabolism, Nanoparticles administration & dosage, Protein Serine-Threonine Kinases pharmacology, Receptor, Notch1 metabolism
- Abstract
Background: The development of effective drug delivery systems capable of transporting small interfering RNA (siRNA) has been elusive. We have previously reported that colorectal cancer tumor xenograft growth was arrested following treatment with liposomal preparation of siDCAMKL-1. In this report, we have utilized Nanoparticle (NP) technology to deliver DCAMKL-1 specific siRNA to knockdown potential key cancer regulators. In this study, mRNA/miRNA were analyzed using real-time RT-PCR and protein by western blot/immunohistochemistry. siDCAMKL-1 was encapsulated in Poly(lactide-co-glycolide)-based NPs (NP-siDCAMKL-1); Tumor xenografts were generated in nude mice, treated with NP-siDCAMKL-1 and DAPT (γ-secretase inhibitor) alone and in combination. To measure let-7a and miR-144 expression in vitro, HCT116 cells were transfected with plasmids encoding the firefly luciferase gene with let-7a and miR-144 miRNA binding sites in the 3'UTR., Results: Administration of NP-siDCAMKL-1 into HCT116 xenografts resulted in tumor growth arrest, downregulation of proto-oncogene c-Myc and Notch-1 via let-7a and miR-144 miRNA-dependent mechanisms, respectively. A corresponding reduction in let-7a and miR-144 specific luciferase activity was observed in vitro. Moreover, an upregulation of EMT inhibitor miR-200a and downregulation of the EMT-associated transcription factors ZEB1, ZEB2, Snail and Slug were observed in vivo. Lastly, DAPT-mediated inhibition of Notch-1 resulted in HCT116 tumor growth arrest and down regulation of Notch-1 via a miR-144 dependent mechanism., Conclusions: These findings demonstrate that nanoparticle-based delivery of siRNAs directed at critical targets such as DCAMKL-1 may provide a novel approach to treat cancer through the regulation of endogenous miRNAs.
- Published
- 2011
- Full Text
- View/download PDF
31. DCAMKL-1 regulates epithelial-mesenchymal transition in human pancreatic cells through a miR-200a-dependent mechanism.
- Author
-
Sureban SM, May R, Lightfoot SA, Hoskins AB, Lerner M, Brackett DJ, Postier RG, Ramanujam R, Mohammed A, Rao CV, Wyche JH, Anant S, and Houchen CW
- Subjects
- 14-3-3 Proteins metabolism, Animals, Biomarkers, Tumor metabolism, Cell Line, Tumor, Doublecortin-Like Kinases, Exonucleases metabolism, Exoribonucleases, Gene Expression, Homeodomain Proteins genetics, Humans, Immunohistochemistry, Intracellular Signaling Peptides and Proteins metabolism, Mice, Mice, Transgenic, Mutation, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms pathology, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins c-myc metabolism, RNA Interference, Receptor, Notch1 genetics, Receptor, Notch1 metabolism, Repressor Proteins genetics, Reverse Transcriptase Polymerase Chain Reaction, Transcription Factors genetics, Twist-Related Protein 1 genetics, Vimentin metabolism, Zinc Finger E-box Binding Homeobox 2, Zinc Finger E-box-Binding Homeobox 1, ras Proteins genetics, ras Proteins metabolism, Epithelial-Mesenchymal Transition genetics, Intracellular Signaling Peptides and Proteins genetics, MicroRNAs genetics, Pancreatic Neoplasms genetics, Protein Serine-Threonine Kinases genetics
- Abstract
Pancreatic cancer is an exceptionally aggressive disease in great need of more effective therapeutic options. Epithelial-mesenchymal transition (EMT) plays a key role in cancer invasion and metastasis, and there is a gain of stem cell properties during EMT. Here we report increased expression of the putative pancreatic stem cell marker DCAMKL-1 in an established KRAS transgenic mouse model of pancreatic cancer and in human pancreatic adenocarcinoma. Colocalization of DCAMKL-1 with vimentin, a marker of mesenchymal lineage, along with 14-3-3 σ was observed within premalignant PanIN lesions that arise in the mouse model. siRNA-mediated knockdown of DCAMKL-1 in human pancreatic cancer cells induced microRNA miR-200a, an EMT inhibitor, along with downregulation of EMT-associated transcription factors ZEB1, ZEB2, Snail, Slug, and Twist. Furthermore, DCAMKL-1 knockdown resulted in downregulation of c-Myc and KRAS through a let-7a microRNA-dependent mechanism, and downregulation of Notch-1 through a miR-144 microRNA-dependent mechanism. These findings illustrate direct regulatory links between DCAMKL-1, microRNAs, and EMT in pancreatic cancer. Moreover, they demonstrate a functional role for DCAMKL-1 in pancreatic cancer. Together, our results rationalize DCAMKL-1 as a therapeutic target for eradicating pancreatic cancers., (© 2011 AACR.)
- Published
- 2011
- Full Text
- View/download PDF
32. Identification of a novel putative pancreatic stem/progenitor cell marker DCAMKL-1 in normal mouse pancreas.
- Author
-
May R, Sureban SM, Lightfoot SA, Hoskins AB, Brackett DJ, Postier RG, Ramanujam R, Rao CV, Wyche JH, Anant S, and Houchen CW
- Subjects
- Animals, Basic Helix-Loop-Helix Transcription Factors metabolism, Biomarkers metabolism, Cell Differentiation, Cell Separation methods, Doublecortin-Like Kinases, Epithelial Cells cytology, Intermediate Filament Proteins metabolism, Mice, Mice, Inbred C57BL, Mice, Nude, Nerve Tissue Proteins metabolism, Nestin, Spheroids, Cellular cytology, Tissue Distribution, Pancreas cytology, Pancreas metabolism, Protein Serine-Threonine Kinases metabolism, Stem Cells metabolism
- Abstract
Stem cells are critical in maintaining adult homeostasis and have been proposed to be the origin of many solid tumors, including pancreatic cancer. Here we demonstrate the expression patterns of the putative intestinal stem cell marker DCAMKL-1 in the pancreas of uninjured C57BL/6 mice compared with other pancreatic stem/progenitor cell markers. We then determined the viability of isolated pancreatic stem/progenitor cells in isotransplantation assays following DCAMKL-1 antibody-based cell sorting. Sorted cells were grown in suspension culture and injected into the flanks of athymic nude mice. Here we report that DCAMKL-1 is expressed in the main pancreatic duct epithelia and islets, but not within acinar cells. Coexpression was observed with somatostatin, NGN3, and nestin, but not glucagon or insulin. Isolated DCAMKL-1+ cells formed spheroids in suspension culture and induced nodule formation in isotransplantation assays. Analysis of nodules demonstrated markers of early pancreatic development (PDX-1), glandular epithelium (cytokeratin-14 and Ep-CAM), and isletlike structures (somatostatin and secretin). These data taken together suggest that DCAMKL-1 is a novel putative stem/progenitor marker, can be used to isolate normal pancreatic stem/progenitors, and potentially regenerates pancreatic tissues. This may represent a novel tool for regenerative medicine and a target for anti-stem cell-based therapeutics in pancreatic cancer.
- Published
- 2010
- Full Text
- View/download PDF
33. Reg IV regulates normal intestinal and colorectal cancer cell susceptibility to radiation-induced apoptosis.
- Author
-
Bishnupuri KS, Luo Q, Sainathan SK, Kikuchi K, Sureban SM, Sabarinathan M, Gross JH, Aden K, May R, Houchen CW, Anant S, and Dieckgraefe BK
- Subjects
- Adenocarcinoma pathology, Animals, Antibodies, Monoclonal pharmacology, Cell Line, Tumor, Cell Proliferation, Colon pathology, Colorectal Neoplasms pathology, Disease Models, Animal, Female, Humans, Inhibitor of Apoptosis Proteins, Lectins, C-Type antagonists & inhibitors, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Nude, Microtubule-Associated Proteins metabolism, Neoplasm Proteins antagonists & inhibitors, Pancreatitis-Associated Proteins, Proto-Oncogene Proteins c-bcl-2 metabolism, Radiotherapy, Repressor Proteins, Survivin, Transplantation, Heterologous, Treatment Outcome, bcl-X Protein metabolism, Adenocarcinoma radiotherapy, Apoptosis radiation effects, Colon metabolism, Colorectal Neoplasms radiotherapy, Lectins, C-Type metabolism, Neoplasm Proteins metabolism
- Abstract
Background & Aims: Regenerating (Reg) gene IV is predominantly expressed in gastrointestinal cells and highly up-regulated in many gastrointestinal malignancies, including colorectal cancer (CRC). Human CRC cells expressing higher levels of Reg IV gene and its protein product (Reg IV) are resistant to conventional therapies, including irradiation (IR). However, the underlying mechanism is not well defined., Methods: A murine model of IR-induced intestinal injury and in vitro and in vivo models of human CRC were used to determine the role of Reg IV in regulation of normal intestinal and colorectal cancer cell susceptibility to IR-induced apoptosis., Results: Treatments of recombinant human Reg IV (rhR4) protein protected normal intestinal crypt cells from IR-induced apoptosis by increasing the expression of antiapoptotic genes Bcl-2, Bcl-XL, and survivin. However, overexpression of Reg IV in human CRC cells was associated with increased resistance to IR-induced apoptosis. Therefore, we used antagonism of Reg IV as a tool to increase CRC cell susceptibility to IR-induced cell death. Two complementary approaches using specific monoclonal antibodies and small interfering RNAs were tested in both in vitro and in vivo models of human CRC. Both approaches resulted in increased apoptosis and decreased cell proliferation, leading to decreased tumor growth and increased animal survival. Furthermore, these approaches increased CRC cell susceptibility to IR-induced apoptosis., Conclusions: These results implicate Reg IV as an important modulator of gastrointestinal cell susceptibility to IR; hence, it is a potential target for adjunctive treatments for human CRC and other gastrointestinal malignancies.
- Published
- 2010
- Full Text
- View/download PDF
34. Antiviral activity of ancient system of ayurvedic medicinal plant Cissus quadrangularis L. (Vitaceae).
- Author
-
Balasubramanian P, Jayalakshmi K, Vidhya N, Prasad R, Sheriff AK, Kathiravan G, Rajagopal K, and Sureban SM
- Abstract
Partially purified methanolic extract of Cissus quadrangularis (belonging to Vitaceae member, South Indian medicinal plant) have been explored for antiviral activity and their phytochemical characterisation. In vitro antiviral activity against HSV type1 and 2, and Vero cells at non-cytotoxic concentration were determined. HSV1 and HSV2 showed more sensitivity against the partially purified compound. Phytochemical analysis showed the presence of the Steroids and Terpenoids.
- Published
- 2009
35. Effect of taxol from Pestalotiopsis mangiferae on A549 cells-In vitro study.
- Author
-
Kathiravan G and Sureban SM
- Abstract
Pestalotiopsis mangiferae Coelomycete fungi were used to examine the production of taxol. The taxol isolated from this fungus is biologically active against cancer cell lines were investigated for its antiproliferative activity in human Non Small Cell Lung Cancer A549 cells. The results showed that the methylene chloride extraction of Pestalotiopsis mangiferae inhibited the proliferation of A 549 cells as measured by MTT and Trypan blue assay. Flow cytometric analysis showed that methylene chloride extraction of Pestalotiopsis mangiferae blocked cell cycle progression in G0/G1 phase. In addition fungal taxol induced A549 cell apoptosis as determined by propidium iodide staining. Further the percentage of LDH release was increased at increasing concentrations which is a measure of cell death. The levels of sialic acid levels and DNA, RNA and protein levels were decreased after treatment with methylene chloride extraction of Pestalotiopsis mangiferae. We suggests that methylene chloride extraction of Pestalotiopsis mangiferae might be considered for future therapeutic application with further studies against lung cancer.
- Published
- 2009
36. Doublecortin and CaM kinase-like-1 and leucine-rich-repeat-containing G-protein-coupled receptor mark quiescent and cycling intestinal stem cells, respectively.
- Author
-
May R, Sureban SM, Hoang N, Riehl TE, Lightfoot SA, Ramanujam R, Wyche JH, Anant S, and Houchen CW
- Subjects
- Animals, Antigens, Surface analysis, Antigens, Surface metabolism, Biomarkers analysis, Biomarkers metabolism, Bromodeoxyuridine, Cell Cycle physiology, Cell Division physiology, Cell Line, Tumor, Cell Lineage physiology, Cell Proliferation, Doublecortin-Like Kinases, Flow Cytometry, Humans, Intestinal Mucosa cytology, Mice, Mice, Inbred C57BL, Mice, Nude, Protein Serine-Threonine Kinases analysis, Receptors, G-Protein-Coupled analysis, Spheroids, Cellular, Stem Cells cytology, Intestinal Mucosa metabolism, Protein Serine-Threonine Kinases metabolism, Receptors, G-Protein-Coupled metabolism, Stem Cells metabolism
- Abstract
It is thought that small intestinal epithelia (IE) undergo continuous self-renewal primarily due to their population of undifferentiated stem cells. These stem cells give rise to transit amplifying (daughter/progenitor) cells, which can differentiate into all mature cell types required for normal gut function. Identification of stem cells in IE is paramount to fully understanding this renewal process. One major obstacle in gastrointestinal stem cell biology has been the lack of definitive markers that identify small intestinal stem cells (ISCs). Here we demonstrate that the novel putative ISC marker doublecortin and CaM kinase-like-1 (DCAMKL-1) is predominantly expressed in quiescent cells in the lower two-thirds of intestinal crypt epithelium and in occasional crypt-based columnar cells (CBCs). In contrast, the novel putative stem cell marker leucine-rich-repeat-containing G-protein-coupled receptor (LGR5) is observed in rapidly cycling CBCs and in occasional crypt epithelial cells. Furthermore, functionally quiescent DCAMKL-1+ crypt epithelial cells retain bromo-deoxyuridine in a modified label retention assay. Moreover, we demonstrate that DCAMKL-1 is a cell surface expressing protein; DCAMKL-1+ cells, isolated from the adult mouse small intestine by fluorescence activated cell sorting, self-renew and ultimately form spheroids in suspension culture. These spheroids formed glandular epithelial structures in the flanks of athymic nude mice, which expressed multiple markers of gut epithelial lineage. Thus, DCAMKL-1 is a marker of quiescent ISCs and can be distinguished from the cycling stem/progenitors (LGR5+). Moreover, DCAMKL-1 can be used to isolate normal small intestinal stem cells and represents a novel research tool for regenerative medicine and cancer therapy.
- Published
- 2009
- Full Text
- View/download PDF
37. Selective blockade of DCAMKL-1 results in tumor growth arrest by a Let-7a MicroRNA-dependent mechanism.
- Author
-
Sureban SM, May R, Ramalingam S, Subramaniam D, Natarajan G, Anant S, and Houchen CW
- Subjects
- Analysis of Variance, Animals, Blotting, Western, Cell Line, Tumor, Colorectal Neoplasms physiopathology, Disease Models, Animal, Female, Flow Cytometry, Gene Expression Regulation, Neoplastic, HCT116 Cells, Humans, Immunohistochemistry, Lysosomal-Associated Membrane Protein 1 genetics, Mice, Mice, Nude, MicroRNAs biosynthesis, MicroRNAs metabolism, Probability, RNA, Messenger genetics, RNA, Messenger metabolism, Random Allocation, Reverse Transcriptase Polymerase Chain Reaction, Sensitivity and Specificity, Transplantation, Heterologous, Tumor Burden genetics, Colorectal Neoplasms genetics, Lysosomal-Associated Membrane Protein 1 metabolism, MicroRNAs genetics
- Abstract
Background & Aims: MicroRNAs (miRNAs) are non-coding RNAs that regulate gene expression. The tumor suppressor miRNA let-7a has been reported to be inhibited posttranscriptionally in embryonic stem cells and in human cancers. Microtubule-associated kinase DCAMKL-1 is a putative intestinal stem cell marker that is expressed in Apc(Min/+) adenomas. We investigated the role of DCAMKL-1 on expression of let-7a miRNA and the oncogene c-Myc and in tumorigenesis., Methods: Human tissue microassay slides were immunostained for DCAMKL-1. HCT116 and SW480 cells were transfected with DCAMKL-1 small interfering RNA (siRNA) (si-DCAMKL-1) and analyzed for DCAMKL-1, c-Myc (using immunoblot and real-time reverse-transcription polymerase chain reaction [RT-PCR]), and pri-let-7a miRNA (using real-time RT-PCR) levels. A liposomal preparation of si-DCAMKL-1 was administered into HCT116 xenografts in nude mice, and tumor volumes were measured. A luciferase reporter assay, with a plasmid containing a let-7a-binding site at the 3' untranslated region, was utilized to measure let-7a in cell lines. Cells were isolated from normal mouse intestine using DCAMKL-1 and fluorescence-activated cell sorting (FACS) and subjected to pri-let-7a miRNA analysis., Results: Expression of DCAMKL-1 was increased in human colorectal cancers. siRNA-mediated blockade of DCAMKL-1 resulted in H tumor xenograft growth arrest, increased pri-let-7a miRNA, a corresponding decrease in luciferase activity, and decreased expression of the oncogene c-Myc. DCAMKL-1(+) cells isolated by FACS demonstrated a significant decrease in pri-let-7a miRNA, compared with more differentiated cells., Conclusions: DCAMKL-1 is a negative regulator of let-7a miRNA biogenesis in intestinal stem and colorectal cancer cells; it could represent a novel target for anti-cancer stem cell-based strategies.
- Published
- 2009
- Full Text
- View/download PDF
38. Loss of p21Waf1/Cip1/Sdi1 enhances intestinal stem cell survival following radiation injury.
- Author
-
George RJ, Sturmoski MA, May R, Sureban SM, Dieckgraefe BK, Anant S, and Houchen CW
- Subjects
- Animals, Apoptosis radiation effects, Cell Survival radiation effects, Cyclin-Dependent Kinase Inhibitor p21 deficiency, Cyclin-Dependent Kinase Inhibitor p21 genetics, Female, Inhibitor of Apoptosis Proteins, Intestine, Small metabolism, Intestine, Small pathology, Mice, Mice, Inbred C57BL, Mice, Knockout, Microtubule-Associated Proteins metabolism, Mitosis radiation effects, Nerve Tissue Proteins metabolism, RNA, Messenger metabolism, RNA-Binding Proteins metabolism, Repressor Proteins, Stem Cells metabolism, Stem Cells pathology, Survivin, Time Factors, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Intestine, Small radiation effects, Stem Cells radiation effects, Whole-Body Irradiation
- Abstract
The microcolony assay following gamma irradiation (IR) is a functional assay of intestinal stem cell fate. The cyclin-dependent kinase (CDK) inhibitor p21(Waf1/Cip1/Sdi1) (p21) regulates cell cycle arrest following DNA damage. To explore the role of p21 on stem cell fate, we examined the effects of p21 deletion on intestinal crypt survival following IR and expression of the stem/progenitor cell marker Musashi-1 (Msi-1) and the antiapoptotic gene survivin. Intestinal stem cell survival in adult wild-type (WT) and p21(-/-) mice was measured using the microcolony assay. Msi-1, p21, and survivin mRNA were measured using real-time PCR and immunohistochemical analysis. Laser capture microdissection (LCM) was used to isolate mRNA from the crypt stem cell zone. No differences in radiation-induced apoptosis were observed between WT and p21(-/-) mice. However, increased crypt survival (3.0-fold) was observed in p21(-/-) compared with WT mice 3.5 days after 13 Gy. Msi-1 and survivin mRNA were elevated 12- and 7.5-fold, respectively, in LCM-dissected crypts of p21(-/-) compared with WT mice. In conclusion, deletion of p21 results in protection of crypt stem/progenitor cells from IR-induced cell death. Furthermore, the increase in crypt survival is associated with increased numbers of Msi-1- and survivin-expressing cells in regenerative crypts.
- Published
- 2009
- Full Text
- View/download PDF
39. Translation regulatory factor RBM3 is a proto-oncogene that prevents mitotic catastrophe.
- Author
-
Sureban SM, Ramalingam S, Natarajan G, May R, Subramaniam D, Bishnupuri KS, Morrison AR, Dieckgraefe BK, Brackett DJ, Postier RG, Houchen CW, and Anant S
- Subjects
- Animals, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Cell Transformation, Neoplastic genetics, Colonic Neoplasms genetics, Cyclooxygenase 2 genetics, Cyclooxygenase 2 metabolism, Dinoprostone genetics, Dinoprostone metabolism, Female, Fibroblasts metabolism, HeLa Cells, Humans, Interleukin-8 genetics, Interleukin-8 metabolism, Mice, Mice, Nude, NIH 3T3 Cells, Neoplasm Transplantation, Proto-Oncogene Mas, Proto-Oncogene Proteins genetics, RNA, Messenger genetics, RNA, Neoplasm genetics, RNA-Binding Proteins genetics, Spheroids, Cellular metabolism, Vascular Endothelial Growth Factor A genetics, Vascular Endothelial Growth Factor A metabolism, Cell Transformation, Neoplastic metabolism, Colonic Neoplasms metabolism, Mitosis genetics, Protein Biosynthesis genetics, Proto-Oncogene Proteins metabolism, RNA Stability genetics, RNA, Messenger metabolism, RNA, Neoplasm metabolism, RNA-Binding Proteins metabolism
- Abstract
RNA-binding proteins play a key role in post-transcriptional regulation of mRNA stability and translation. We have identified that RBM3, a translation regulatory protein, is significantly upregulated in human tumors, including a stage-dependent increase in colorectal tumors. Forced RBM3 overexpression in NIH3T3 mouse fibroblasts and SW480 human colon epithelial cells increases cell proliferation and development of compact multicellular spheroids in soft agar suggesting the ability to induce anchorage-independent growth. In contrast, downregulating RBM3 in HCT116 colon cancer cells with specific siRNA decreases cell growth in culture, which was partially overcome when treated with prostaglandin E(2), a product of cyclooxygenase (COX)-2 enzyme activity. Knockdown also resulted in the growth arrest of tumor xenografts. We have also identified that RBM3 knockdown increases caspase-mediated apoptosis coupled with nuclear cyclin B1, and phosphorylated Cdc25c, Chk1 and Chk2 kinases, implying that under conditions of RBM3 downregulation, cells undergo mitotic catastrophe. RBM3 enhances COX-2, IL-8 and VEGF mRNA stability and translation. Conversely, RBM3 knockdown results in loss in the translation of these transcripts. These data demonstrate that the RNA stabilizing and translation regulatory protein RBM3 is a novel proto-oncogene that induces transformation when overexpressed and is essential for cells to progress through mitosis.
- Published
- 2008
- Full Text
- View/download PDF
40. Knockdown of RNA binding protein musashi-1 leads to tumor regression in vivo.
- Author
-
Sureban SM, May R, George RJ, Dieckgraefe BK, McLeod HL, Ramalingam S, Bishnupuri KS, Natarajan G, Anant S, and Houchen CW
- Subjects
- Adenocarcinoma genetics, Adenocarcinoma metabolism, Animals, Apoptosis, Biomarkers, Tumor, Blotting, Western, Cell Proliferation, Colorectal Neoplasms genetics, Colorectal Neoplasms metabolism, Disease Progression, Humans, Immunohistochemistry, Mice, Mice, Nude, Neoplasm Transplantation, Neoplasms, Experimental, Nerve Tissue Proteins metabolism, RNA-Binding Proteins metabolism, Reverse Transcriptase Polymerase Chain Reaction, Transplantation, Heterologous, Adenocarcinoma pathology, Colorectal Neoplasms pathology, Gene Expression Regulation, Neoplastic, Nerve Tissue Proteins genetics, RNA, Neoplasm genetics, RNA-Binding Proteins genetics
- Abstract
Background & Aims: In the gut, tumorigenesis is thought to arise from the stem cell population located near the base of intestinal and colonic crypts. The RNA binding protein musashi-1 (Msi-1) is a putative intestinal and progenitor/stem cell marker. Msi-1 expression is increased during rat brain development and in APC(min/+) mice tumors. This study examined a potential role of Msi-1 in tumorigenesis., Methods: Msi-1 small interfering RNA (siRNA) was administered as a liposomal preparation to HCT116 colon adenocarcinoma xenografts in athymic nude mice and tumor volume was measured. Cell proliferation was assessed by hexosaminidase and 3-(4,5-dimethylthiazol 2-yl)-2,5-diphenyltetrazolium bromide MTT assays. siRNA-transfected cells were subjected to 12 Gy gamma-irradiation. Apoptosis was assessed by immunoreactive activated caspase-3 and mitosis was assessed by phosphorylated histone H3 staining. The tumor xenografts were stained similarly for phosphorylated histone H3, activated caspase-3, terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling, Notch-1, and p21(WAF1). Furthermore, siRNA-transfected cells were subjected to cell-cycle analysis and Western blot analyses for Notch-1 and p21(WAF1)., Results: Knockdown of Msi-1 resulted in tumor growth arrest in xenografts, reduced cancer cell proliferation, and increased apoptosis alone and in combination with radiation injury. siRNA-mediated reduction of Msi-1 lead to mitotic catastrophe in tumor cells. Moreover, there was inhibition of Notch-1 and up-regulation of p21(WAF1) after knockdown of Msi-1., Conclusions: Our results show the involvement of Msi-1 in cancer cell proliferation, inhibition of apoptosis, and mitotic catastrophe, suggesting an important potential mechanism for its role in tumorigenesis.
- Published
- 2008
- Full Text
- View/download PDF
41. Diphenyl difluoroketone: a curcumin derivative with potent in vivo anticancer activity.
- Author
-
Subramaniam D, May R, Sureban SM, Lee KB, George R, Kuppusamy P, Ramanujam RP, Hideg K, Dieckgraefe BK, Houchen CW, and Anant S
- Subjects
- Adenocarcinoma genetics, Adenocarcinoma metabolism, Adenocarcinoma pathology, Animals, Apoptosis drug effects, Cell Cycle drug effects, Cell Growth Processes drug effects, Cell Line, Tumor, Colonic Neoplasms genetics, Colonic Neoplasms metabolism, Colonic Neoplasms pathology, Cyclooxygenase 2 biosynthesis, Cyclooxygenase 2 genetics, HCT116 Cells, HT29 Cells, Humans, Interleukin-8 biosynthesis, Interleukin-8 genetics, MAP Kinase Signaling System drug effects, Male, Mice, Mice, Nude, Neovascularization, Pathologic drug therapy, Neovascularization, Pathologic pathology, Stomach Neoplasms genetics, Stomach Neoplasms metabolism, Stomach Neoplasms pathology, Vascular Endothelial Growth Factor A biosynthesis, Vascular Endothelial Growth Factor A genetics, Xenograft Model Antitumor Assays, Adenocarcinoma drug therapy, Benzylidene Compounds pharmacology, Colonic Neoplasms drug therapy, Piperidones pharmacology, Stomach Neoplasms drug therapy
- Abstract
Diphenyl difluoroketone (EF24), a molecule having structural similarity to curcumin, was reported to inhibit proliferation of a variety of cancer cells in vitro. However, the efficacy and in vivo mechanism of action of EF24 in gastrointestinal cancer cells have not been investigated. Here, we assessed the in vivo therapeutic effects of EF24 on colon cancer cells. Using hexosaminidase assay, we determined that EF24 inhibits proliferation of HCT-116 and HT-29 colon and AGS gastric adenocarcinoma cells but not of mouse embryo fibroblasts. Furthermore, the cancer cells showed increased levels of activated caspase-3 and increased Bax to Bcl-2 and Bax to Bcl-xL ratios, suggesting that the cells were undergoing apoptosis. At the same time, cell cycle analysis showed that there was an increased number of cells in the G(2)-M phase. To determine the effects of EF24 in vivo, HCT-116 colon cancer xenografts were established in nude mice and EF24 was given i.p. EF24 significantly suppressed the growth of colon cancer tumor xenografts. Immunostaining for CD31 showed that there was a lower number of microvessels in the EF24-treated animals coupled with decreased cyclooxygenase-2, interleukin-8, and vascular endothelial growth factor mRNA and protein expression. Western blot analyses also showed decreased AKT and extracellular signal-regulated kinase activation in the tumors. Taken together, these data suggest that the novel curcumin-related compound EF24 is a potent antitumor agent that induces caspase-mediated apoptosis during mitosis and has significant therapeutic potential for gastrointestinal cancers.
- Published
- 2008
- Full Text
- View/download PDF
42. Identification of a novel putative gastrointestinal stem cell and adenoma stem cell marker, doublecortin and CaM kinase-like-1, following radiation injury and in adenomatous polyposis coli/multiple intestinal neoplasia mice.
- Author
-
May R, Riehl TE, Hunt C, Sureban SM, Anant S, and Houchen CW
- Subjects
- Animals, Doublecortin-Like Kinases, Gastrointestinal Tract radiation effects, Intestinal Mucosa metabolism, Intestines cytology, Intestines pathology, Intestines radiation effects, Mice, Nerve Tissue Proteins metabolism, Protein Transport radiation effects, RNA-Binding Proteins metabolism, Radiation, Ionizing, Regeneration radiation effects, Stem Cells radiation effects, beta Catenin metabolism, Adenomatous Polyposis Coli metabolism, Biomarkers, Tumor metabolism, Gastrointestinal Tract cytology, Intestinal Neoplasms metabolism, Protein Serine-Threonine Kinases metabolism, Stem Cells cytology
- Abstract
In the gut, tumorigenesis arises from intestinal or colonic crypt stem cells. Currently, no definitive markers exist that reliably identify gut stem cells. Here, we used the putative stem cell marker doublecortin and CaM kinase-like-1 (DCAMKL-1) to examine radiation-induced stem cell apoptosis and adenomatous polyposis coli (APC)/multiple intestinal neoplasia (min) mice to determine the effects of APC mutation on DCAMKL-1 expression. Immunoreactive DCAMKL-1 staining was demonstrated in the intestinal stem cell zone. Furthermore, we observed apoptosis of the cells negative for DCAMKL-1 at 6 hours. We found DNA damage in all the cells in the crypt region, including the DCAMKL-1-positive cells. We also observed stem cell apoptosis and mitotic DCAMKL-1-expressing cells 24 hours after irradiation. Moreover, in APC/min mice, DCAMKL-1-expressing cells were negative for proliferating cell nuclear antigen and nuclear beta-catenin in normal-appearing intestine. However, beta-catenin was nuclear in DCAMKL-1-positive cells in adenomas. Thus, nuclear translocation of beta-catenin distinguishes normal and adenoma stem cells. Targeting DCAMKL-1 may represent a strategy for developing novel chemotherapeutic agents.
- Published
- 2008
- Full Text
- View/download PDF
43. Functional antagonism between RNA binding proteins HuR and CUGBP2 determines the fate of COX-2 mRNA translation.
- Author
-
Sureban SM, Murmu N, Rodriguez P, May R, Maheshwari R, Dieckgraefe BK, Houchen CW, and Anant S
- Subjects
- 3' Untranslated Regions radiation effects, Animals, Antigens, Surface genetics, Binding, Competitive, CELF Proteins, Cell Nucleus metabolism, Cyclooxygenase 2 genetics, Cytoplasm metabolism, Dimerization, ELAV Proteins, ELAV-Like Protein 1, Gamma Rays, HCT116 Cells, HT29 Cells, Humans, Membrane Proteins genetics, Mice, NIH 3T3 Cells, Nerve Tissue Proteins genetics, Protein Binding, Protein Interaction Mapping methods, Protein Transport, RNA Stability, RNA, Messenger radiation effects, RNA-Binding Proteins genetics, Recombinant Fusion Proteins metabolism, Repressor Proteins metabolism, Trans-Activators metabolism, Transfection, 3' Untranslated Regions metabolism, Antigens, Surface metabolism, Cyclooxygenase 2 metabolism, Membrane Proteins metabolism, Nerve Tissue Proteins metabolism, Protein Biosynthesis radiation effects, RNA, Messenger metabolism, RNA-Binding Proteins metabolism
- Abstract
Background and Aims: Cyclooxygenase-2 (COX-2) expression is regulated at the levels of messenger RNA (mRNA) stability and translation by AU-rich elements (ARE) located in its 3' untranslated region (3'UTR). Although structurally homologous RNA binding proteins HuR and CUGBP2 stabilize COX-2 mRNA, HuR induces whereas CUGBP2 inhibits COX-2 mRNA translation. This study aimed to determine the antagonism between these proteins on COX-2 expression., Methods: COX-2 ARE binding activity was determined by nitrocellulose filter binding and UV cross-linking assays using recombinant glutathione S-transferase (GST)/HuR and GST/CUGBP2. Protein:protein interactions were determined by GST pull-down, yeast 2-hybrid, and immunocytochemistry assays. Nucleocytoplasmic shutting was determined by heterokaryon analyses. The effect of CUGBP2 and HuR on COX-2 ARE-dependent translation was shown by a chimeric luciferase mRNA containing COX-2 3'UTR. HT-29 cells were subjected to 12 Gy gamma-irradiation in a cesium irradiator., Results: CUGBP2 and HuR bind with similar affinities to COX-2 ARE, but CUGBP2 competes with HuR for binding. In vitro, HuR and CUGBP2 heterodimerize. Furthermore, FLAG-tagged HuR and myc-tagged CUGBP2 colocalize in the nucleus of HCT-116 cells. Moreover, both proteins shuttled between the nucleus and cytoplasm. In vitro, HuR enhanced whereas CUGBP2 inhibited translation of the chimeric luciferase COX-2 3'UTR mRNA. Furthermore, CUGBP2 competitively inhibited HuR-mediated translation of the transcript. In HT-29 cells transfected with HuR and CUGBP2, a switch in COX-2 mRNA binding from predominantly HuR to CUGBP2 occurred after radiation treatment, which was coupled with increased silencing of the COX-2 mRNA., Conclusions: CUGBP2 overrides HuR and suppresses COX-2 mRNA translation.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.