Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Mol Cancer Res ; 20(3): 387-399, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34753802

ABSTRACT

Regenerating Gene 4 (Reg4) is highly upregulated in gastrointestinal (GI) malignancies including colorectal and pancreatic cancers. Numerous studies demonstrated an association between higher Reg4 expression and tumor aggressiveness, intrinsic resistance to apoptotic death, and poor outcomes from GI malignancies. However, the precise receptor and underlying signaling mechanism have remained unknown. Although we previously reported a Reg4-mediated induction of EGFR activity in colorectal cancer cells, a direct interaction between Reg4 and EGFR was not observed. This study is focused on identifying the cell surface binding partner of Reg4 and dissecting its role in colorectal cancer and pancreatic cancer growth and stem cell survival. In vitro models of human colorectal cancer and pancreatic cancer were used to evaluate the results. Results of this study find: (i) Reg4 interacts with CD44, a transmembrane protein expressed by a population of colorectal cancer and pancreatic cancer cells; (ii) Reg4 activates regulated intramembrane proteolysis of CD44 resulting in γ-secretase-mediated cleavage and release of the CD44 intracytoplasmic domain (CD44ICD) that functions as a transcriptional activator of D-type cyclins involved in the regulation of cancer cell proliferation and Klf4 and Sox2 expression involved in regulating pluripotency of cancer stem cells; and (iii) Reg4 significantly increases colorectal cancer and pancreatic cancer cell proliferation and their clonogenic potential in stem cell assays. IMPLICATIONS: These results suggest that pro-proliferative and pro-stemness effects of Reg4 are mediated through γ-secretase-mediated CD44/CD44ICD signaling, hence strategies to disrupt Reg4-CD44-γ-secretase-CD44ICD signaling axis may increase cancer cell susceptibility to chemo- and radiotherapeutics.


Subject(s)
Colorectal Neoplasms , Pancreatic Neoplasms , Amyloid Precursor Protein Secretases/metabolism , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/pathology , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/pathology , Pancreatitis-Associated Proteins/genetics , Pancreatitis-Associated Proteins/metabolism , Pancreatic Neoplasms
2.
Oncotarget ; 12(4): 278-291, 2021 Feb 16.
Article in English | MEDLINE | ID: mdl-33659040

ABSTRACT

Reg4 is highly expressed in gastrointestinal malignancies and acts as a mitogenic and pro-invasive factor. Our recent works suggest that Reg4 binds with CD44 and induces its proteolytic cleavage to release intra-cytoplasmic domain of CD44 (CD44ICD). The goal of this study is to demonstrate clinical significance of the Reg4-CD44/CD44ICD pathway in stage II/III colon cancer and its association with clinical parameters of aggression. We constructed a tissue microarray (TMA) of 93 stage II/III matched colon adenocarcinoma patients, 23 with recurrent disease. The TMA was immunohistochemically stained for Reg4, CD44, and CD44ICD proteins and analyzed to identify associations with tumor characteristics, recurrence and overall survival. The TMA data analysis showed a significant correlation between Reg4 and CD44 (r2 = 0.23, P = 0.028), CD44 and CD44ICD (r2 = 0.36, p = 0.0004), and Reg4 and CD44ICD (r2 = 0.45, p ≤ 0.0001). Reg4 expression was associated with larger tumor size (r2 = 0.23, p = 0.026). Although, no association was observed between Reg4, CD44, or CD44ICD expression and disease recurrence, Reg4-positive patients had a median survival of 4 years vs. 7 years for Reg4-negative patients (p = 0.04) in patients who recurred. Inhibition of the Reg4-CD44/CD44ICD pathway may be a future therapeutic target for colon cancer patients.

3.
Cancer Res ; 79(6): 1138-1150, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30679179

ABSTRACT

The tryptophan-metabolizing enzyme indoleamine 2,3 dioxygenase 1 (IDO1) is frequently overexpressed in epithelial-derived malignancies, where it plays a recognized role in promoting tumor immune tolerance. We previously demonstrated that the IDO1-kynurenine pathway (KP) also directly supports colorectal cancer growth by promoting activation of ß-catenin and driving neoplastic growth in mice lacking intact adaptive immunity. In this study, we sought to delineate the specific role of epithelial IDO1 in colon tumorigenesis and define how IDO1 and KP metabolites interact with pivotal neoplastic signaling pathways of the colon epithelium. We generated a novel intestinal epithelial-specific IDO1 knockout mouse and utilized established colorectal cancer cell lines containing ß-catenin-stabilizing mutations, human colorectal cancer samples, and human-derived epithelial organoids (colonoids and tumoroids). Mice with intestinal epithelial-specific knockout of IDO1 developed fewer and smaller tumors than wild-type littermates in a model of inflammation-driven colon tumorigenesis. Moreover, their tumors exhibited reduced nuclear ß-catenin and neoplastic proliferation but increased apoptosis. Mechanistically, KP metabolites (except kynurenic acid) rapidly activated PI3K-Akt signaling in the neoplastic epithelium to promote nuclear translocation of ß-catenin, cellular proliferation, and resistance to apoptosis. Together, these data define a novel cell-autonomous function and mechanism by which IDO1 activity promotes colorectal cancer progression. These findings may have implications for the rational design of new clinical trials that exploit a synergy of IDO1 inhibitors with conventional cancer therapies for which Akt activation provides resistance such as radiation.Significance: This study identifies a new mechanistic link between IDO1 activity and PI3K/AKT signaling, both of which are important pathways involved in cancer growth and resistance to cancer therapy.


Subject(s)
Apoptosis , Cell Proliferation , Colonic Neoplasms/pathology , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Kynurenine/metabolism , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Colon/metabolism , Colon/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Tumor Cells, Cultured
4.
Mol Carcinog ; 53 Suppl 1: E169-80, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24151146

ABSTRACT

Upregulation of regenerating gene 4 (Reg4) is observed in many human gastrointestinal malignancies including colorectal cancer (CRC). We previously reported a Reg4-mediated induction of epidermal growth factor receptor-Akt-AP1 signaling regulating CRC cell apoptosis. However, the role of Reg4 in the regulation of CRC cell division is poorly understood. This study tests the hypothesis that Reg4 induces Akt-GSK3ß-ß-Catenin-TCF-4 signaling to regulate CRC cell division. In vitro models of human CRC were used to determine the role of Reg4 in regulation of CRC cell division. Cell cycle studies demonstrated that Reg4 treatment significantly decreased CRC cell number in G1 phase and increased in G2 phase. Subsequently Reg4 significantly increased the mitotic index of CRC cells. As assessed by real-time RT-PCR and Western blot analyses, Reg4 significantly increased the expression of cell cycle regulatory genes Cyclin D1 and D3, and associated Cyclin-dependent kinases (CDK4 and CDK6). Reg4-mediated increase in these genes involved a pathway that included an induced Akt activity by increasing phosphorylation of Thr308 and Ser473, a reduced glycogen synthase kinase 3ß (GSK-3ß) activity by increasing phosphorylation of Ser9, an induced nuclear translocation of ß-Catenin by decreasing phosphorylation of Ser33/37/Thr41, and an increased TCF-4 transcriptional activity. Furthermore, antagonism of Reg4-signaling using Reg4-specific mAbs (2H6 and 3E5) and Akt inhibitor significantly decreased, whereas agonism using GSK-3ß antagonist (SB216763) significantly increased mitotic index and proliferation of CRC cells. These results identify Reg4 as a key regulator of the CRC cell division and proliferation, hence a potential target of human CRC treatment.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Colorectal Neoplasms/pathology , Glycogen Synthase Kinase 3/metabolism , Lectins, C-Type/physiology , Mitosis/physiology , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factors/metabolism , beta Catenin/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Apoptosis , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Blotting, Western , Cell Adhesion , Cell Cycle , Cell Movement , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Humans , Pancreatitis-Associated Proteins , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transcription Factor 4 , Transcription Factors/genetics , Tumor Cells, Cultured , beta Catenin/genetics
5.
Gastroenterology ; 145(2): 416-25.e1-4, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23669411

ABSTRACT

BACKGROUND & AIMS: Indoleamine 2,3 dioxygenase-1 (IDO1) catabolizes tryptophan along the kynurenine pathway. Although IDO1 is expressed in inflamed and neoplastic epithelial cells of the colon, its role in colon tumorigenesis is not well understood. We used genetic and pharmacologic approaches to manipulate IDO1 activity in mice with colitis-associated cancer and human colon cancer cell lines. METHODS: C57Bl6 wild-type (control), IDO1-/-, Rag1-/-, and Rag1/IDO1 double-knockout mice were exposed to azoxymethane and dextran sodium sulfate to induce colitis and tumorigenesis. Colitis severity was assessed by measurements of disease activity, cytokine levels, and histologic analysis. In vitro experiments were conducted using HCT 116 and HT-29 human colon cancer cells. 1-methyl tryptophan and small interfering RNA were used to inhibit IDO1. Kynurenine pathway metabolites were used to simulate IDO1 activity. RESULTS: C57Bl6 mice given pharmacologic inhibitors of IDO1 and IDO1-/- mice had lower tumor burdens and reduced proliferation in the neoplastic epithelium after administration of dextran sodium sulfate and azoxymethane than control mice. These reductions also were observed in Rag1/IDO1 double-knockout mice compared with Rag1-/- mice (which lack mature adaptive immunity). In human colon cancer cells, blockade of IDO1 activity reduced nuclear and activated ß-catenin, transcription of its target genes (cyclin D1 and Axin2), and, ultimately, proliferation. Exogenous administration of IDO1 pathway metabolites kynurenine and quinolinic acid led to activation of ß-catenin and proliferation of human colon cancer cells, and increased tumor growth in mice. CONCLUSIONS: IDO1, which catabolizes tryptophan, promotes colitis-associated tumorigenesis in mice, independent of its ability to limit T-cell-mediated immune surveillance. The epithelial cell-autonomous survival advantage provided by IDO1 to colon epithelial cells indicate its potential as a therapeutic target.


Subject(s)
Colitis/physiopathology , Colonic Neoplasms/etiology , Epithelial Cells/physiology , Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Intestinal Mucosa/physiopathology , beta Catenin/drug effects , Animals , Azoxymethane , Carcinogens , Cell Proliferation/drug effects , Colitis/metabolism , Colitis, Ulcerative , Colonic Neoplasms/chemically induced , Epithelial Cells/drug effects , Epithelial Cells/metabolism , HCT116 Cells , HT29 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Intestinal Mucosa/metabolism , Kynurenine/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Quinolinic Acid/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , beta Catenin/physiology
6.
Inflamm Bowel Dis ; 18(5): 955-67, 2012 May.
Article in English | MEDLINE | ID: mdl-21953855

ABSTRACT

BACKGROUND: The pathogenesis of inflammatory bowel disease (IBD) is associated with a dysregulated mucosal immune response. Certain stimulators of innate immunity (CpG DNA or GM-CSF) are reported to be anti-inflammatory in IBD. Toll-like receptor-7 (TLR7) is an important regulator of innate immunity and its activation plays a key role in induction of type I interferon (IFN). The present study tests the hypothesis that the TLR7 agonists Imiquimod has therapeutic efficacy in IBD. METHODS: Acute colitis was induced in Balb/c mice by giving 5% dextran sodium sulfate (DSS) in drinking water for 7 days. Mice were treated with Imiquimod either orally or topically and its therapeutic effects on disease activity were examined. Isolated mouse CD11c+ dendritic cells and human intestinal epithelial cells (HT29, HCT116) were treated with Imiquimod (10 µg/mL) and their susceptibility to intracellular Salmonella typhimurium infection was assessed by gentamicin protection assay. RESULTS: Oral administration of Imiquimod induced type I IFN expression in the gastrointestinal mucosa and ameliorated DSS-induced acute colitis as assessed by clinical parameters, histology, and mRNA expression of proinflammatory cytokines. Topical administration of Imiquimod also ameliorated DSS colitis by inducing the expression of type I IFN in the colonic mucosa. However, no evidence for a systemic IFN response was observed. Imiquimod treatments to both CD11c+ and intestinal epithelial cells significantly increased expression of antimicrobial peptides (AMPs) and reduced survival of intracellular S. typhimurium. CONCLUSIONS: Imiquimod induces type I IFN and AMP to ameliorate DSS-induced acute colitis and prevents Salmonella survival. Therefore, Imiquimod treatments provide a new therapeutic approach for IBD patients.


Subject(s)
Aminoquinolines/therapeutic use , Antimicrobial Cationic Peptides/metabolism , Colitis/prevention & control , Dextran Sulfate/toxicity , Interferon Inducers/therapeutic use , Interferon Type I/metabolism , Toll-Like Receptor 7/metabolism , Administration, Oral , Administration, Topical , Animals , Biomarkers/metabolism , Colitis/chemically induced , Colitis/metabolism , Cytokines/metabolism , Female , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Gene Expression Profiling , Humans , Imiquimod , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , Peroxidase/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Salmonella Infections/microbiology , Salmonella Infections/mortality , Salmonella Infections/prevention & control , Salmonella typhimurium , Survival Rate
7.
Gastroenterology ; 138(2): 616-26, 626.e1-2, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19900450

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.


Subject(s)
Adenocarcinoma/radiotherapy , Apoptosis/radiation effects , Colon/metabolism , Colorectal Neoplasms/radiotherapy , Lectins, C-Type/metabolism , Neoplasm Proteins/metabolism , 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
8.
Gastroenterology ; 134(5): 1448-58, 2008 May.
Article in English | MEDLINE | ID: mdl-18471519

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.


Subject(s)
Adenocarcinoma/pathology , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Nerve Tissue Proteins/genetics , RNA, Neoplasm/genetics , RNA-Binding Proteins/genetics , 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
9.
Inflamm Bowel Dis ; 14(1): 88-99, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17932977

ABSTRACT

BACKGROUND: Sargramostim, granulocyte macrophage colony-stimulating factor (GM-CSF), a hematopoietic growth factor, stimulates cells of the intestinal innate immune system. Clinical trials show that sargramostim induces clinical response and remission in patients with active Crohn's disease. To study the mechanism, we examined the effects of GM-CSF in the dextran sulfate sodium (DSS)-induced acute colitis model. We hypothesized that GM-CSF may work through effects on dendritic cells (DCs). METHODS: Acute colitis was induced in Balb/c mice by administration of DSS in drinking water. Mice were treated with daily GM-CSF or phosphate-buffered saline (PBS). To probe the role of plasmacytoid DCs (pDCs) in the response to GM-CSF, we further examined the effects of monoclonal antibody 440c, which is specific for a sialic acid-binding immunoglobulin (Ig)-like lectin expressed on pDCs. RESULTS: GM-CSF ameliorates acute DSS-induced colitis, resulting in significantly improved clinical parameters and histology. Microarray analysis showed reduced expression of proinflammatory genes including TNF-alpha and IL1-beta; the results were further confirmed by real-time reverse-transcriptase polymerase chain reaction and serum Bio-plex analysis. GM-CSF treatment significantly expands pDCs and type 1 IFN production. Administration of mAb 440c completely blocked the therapeutic effect of GM-CSF. GM-CSF is also effective in RAG1(-/-) mice, demonstrating activity-independent effects on T and B cells. IFN-beta administration mimics the therapeutic effect of GM-CSF in DSS-treated mice. GM-CSF increases systemic and mucosal type 1 IFN expression and exhibits synergy with pDC activators, such as microbial cytosine-phosphate-guanosine (CpG) DNA. CONCLUSIONS: GM-CSF is effective in the treatment of DSS colitis in a mechanism involving the 440c(+) pDC population.


Subject(s)
Colitis/chemically induced , Colitis/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Animals , Colitis/pathology , Colitis/physiopathology , Colon/pathology , Dendritic Cells/immunology , Dextran Sulfate/toxicity , Female , Homeodomain Proteins/genetics , Interferon Type I/immunology , Interleukin-1beta/genetics , Intestinal Mucosa/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Recombinant Proteins , Tumor Necrosis Factor-alpha/genetics
10.
Cancer Biol Ther ; 5(12): 1714-20, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17106246

ABSTRACT

Expression of anti-apoptotic genes is frequently elevated in tumors, where they increase resistance to chemotherapeutic agents and predict poor patient outcomes. However, key cellular factors regulating anti-apoptotic genes in tumors remain unknown. Increased expression of the regenerating (Reg) genes is commonly observed in gastrointestinal (GI) malignancies including colorectal cancer (CRC). We therefore examined Reg gene expression and associated changes in anti-apoptotic genes in an animal model of GI tumorigenesis. Using real time RT-PCR, we measured expression of Reg genes in human colorectal adenocarcinoma specimens, colon adenocarcinoma cell lines and adenomas from multiple intestinal neoplasia (min) mice heterozygous for a germ-line mutation of the adenomatous polyposis coli (APC) gene. Expression of Reg genes is increased in human colorectal adenocarcinomas and in the intestine of APCmin/+ mice at four weeks of age, a time preceding the spontaneous second mutation in the APC gene. Individual Reg genes exhibited regional expression profiles across the GI tract in mice. Adenomas from 14-week old mice had significant increases in at least one member of the Reg gene family, most commonly Reg IV and an associated increase in expression of the anti-apoptotic gene, Bcl-2. Addition of exogenous recombinant human Reg IV to human colon adenocarcinoma cells significantly increased Bcl-2 and Bcl-xL expression and induced resistance to ionizing radiation. These results show that dysregulation of Reg genes occur early in tumorigenesis. Furthermore, increased expression of Reg genes, specifically Reg IV contribute to adenoma formation and lead to increased resistance to apoptotic cell death in CRC.


Subject(s)
Adenocarcinoma/genetics , Apoptosis/genetics , Colonic Neoplasms/genetics , Colorectal Neoplasms/genetics , Lithostathine/genetics , Animals , Cell Line, Tumor , DNA Primers , Female , Gene Deletion , Gene Expression Regulation, Neoplastic , Genes, APC , Genes, bcl-2 , Humans , Male , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , bcl-X Protein/genetics
11.
Gastroenterology ; 130(1): 137-49, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16401477

ABSTRACT

BACKGROUND & AIMS: Reg IV, a secreted protein and member of the Reg multigene family, is up-regulated in malignancies of the human gastrointestinal tract, including colorectal carcinoma (CRC). However, in vitro signal transduction pathway(s) utilized by Reg IV are not yet known. METHODS: To determine the signaling pathway(s) responsive to Reg IV, we examined the effects of purified recombinant human Reg IV (rhR4) on HCT116 and HT29 colon adenocarcinoma cells. RESULTS: Addition of rhR4 to cultures led to a dose-dependent increase in cell number similar to that observed after treatment with epidermal growth factor (EGF). In addition, rhR4 treatment resulted in rapid phosphorylation of EGF receptor at Tyr992 and Tyr1068 and Akt at Thr308 and Ser473. Using luciferase reporter gene assays, we demonstrated that Reg IV signaling through EGF receptor and Akt results in increased activator protein-1 (AP-1) transcription factor activity. Real-time reverse-transcription polymerase chain reaction and Western blot analyses revealed quantitative increases in c-Jun, JunB, JunD, and FosB expression associated with increased AP-1 activity. Electrophoretic mobility shift assay further revealed significant increases in AP-1 binding activity in rhR4-treated cells, with increased supershift in the presence of antibodies to JunB, JunD, and FosB. Furthermore, rhR4 treatments led to the increased expression of Bcl-2, Bcl-XL, survivin, and matrilysin, genes associated with a poor prognosis in advanced CRC. CONCLUSIONS: Reg IV is a potent activator of the EGF receptor/Akt/AP-1 signaling pathway in CRC. Disruption of Reg signaling may have utility as a therapeutic intervention for human gastrointestinal adenocarcinomas.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/physiopathology , Colonic Neoplasms/genetics , Colonic Neoplasms/physiopathology , Lectins, C-Type/physiology , Apoptosis , ErbB Receptors/metabolism , Humans , Neoplasm Metastasis , Pancreatitis-Associated Proteins , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Transcription Factor AP-1/metabolism , Tumor Cells, Cultured
12.
Protein Expr Purif ; 44(2): 94-103, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16213750

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF) regulates proliferation, differentiation, and function of hematopoietic progenitor cells. Aside from expansion of hematopoietic cells, GM-CSF has shown efficacy in other diseases, including Crohn's disease. While GM-CSF being clinically used in humans, the ability to perform mechanistic studies in murine models is difficult due to the limited availability and rapid clearance of murine GM-CSF in the peripheral blood. To address these issues, we efficiently expressed murine GM-CSF under the control of the AOX1 gene promoter in Pichia pastoris using the Mut(S) strain KM71H. We describe the unique conditions that are required for efficient production by high-density fermentation and purification of mGM-CSF protein. Recombinant mGM-CSF protein was purified by tangential flow ultrafiltration and preparative reverse phase chromatography. To address limited half life or rapid clearance in mice, recombinant murine GM-CSF was modified by lysine-directed polyethylene glycol conjugation (PEGylation). PEG-modified and unmodified proteins were characterized by amino terminus sequence analysis and matrix assisted laser desorption ionization time-of-flight mass spectrometry. Under the mild reaction conditions, the recombinant protein is efficiently modified by PEGylation on an average of 2-3 sites per molecule. In vivo treatment of mice with PEGylated mGM-CSF, but not the unmodified recombinant mGM-CSF, reproduces the potent colony stimulating effects of human GM-CSF in patients on myeloid progenitor populations, as assessed by FACs analysis. This simplified approach for the expression, purification, and modification of a biologically potent form of murine GM-CSF should facilitate the study of central mechanisms of action in murine disease models.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Polyethylene Glycols/chemistry , Amino Acid Sequence , Animals , Blotting, Western , Fermentation , Glycosylation , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacokinetics , Half-Life , Male , Metabolic Clearance Rate , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Molecular Weight , Pichia/genetics , Pichia/metabolism , Polyethylene Glycols/pharmacokinetics , Propionates/chemistry , Protein Sorting Signals/genetics , Recombinant Proteins , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
SELECTION OF CITATIONS
SEARCH DETAIL
...