Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Oncotarget ; 8(25): 40606-40619, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28380450

ABSTRACT

Overexpression of human progastrin increases colonic mucosal proliferation and colorectal cancer progression in mice. The G-protein coupled receptor 56 (GPR56) is known to regulate cell adhesion, migration, proliferation and stem cell biology, but its expression in the gut has not been studied. We hypothesized that the promotion of colorectal cancer by progastrin may be mediated in part through GPR56. Here, we found that GPR56 expresses in rare colonic crypt cells that lineage trace colonic glands consistent with GPR56 marking long-lived colonic stem-progenitor cells. GPR56 was upregulated in transgenic mice overexpressing human progastrin. While recombinant human progastrin promoted the growth and survival of wild-type colonic organoids in vitro, colonic organoids cultured from GPR56-/- mice were resistant to progastrin. We found that progastrin directly bound to, and increased the proliferation of, GPR56-expressing colon cancer cells in vitro, and proliferation was increased in cells that expressed both GPR56 and the cholecystokinin-2 receptor (CCK2R). In vivo, deletion of GPR56 in the mouse germline abrogated progastrin-dependent colonic mucosal proliferation and increased apoptosis. Loss of GPR56 also inhibited progastrin-dependent colonic crypt fission and colorectal carcinogenesis in the azoxymethane (AOM) mouse model of colorectal cancer. Overall, we found that progastrin binds to GPR56 expressing colonic stem cells, which in turn promotes their expansion, and that this GPR56-dependent pathway is an important driver and potential new target in colorectal carcinogenesis.


Subject(s)
Colonic Neoplasms/metabolism , Gastrins/metabolism , Protein Precursors/metabolism , Receptors, G-Protein-Coupled/metabolism , Stem Cells/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Colon/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Gastrins/genetics , Gastrins/pharmacology , Humans , Intestinal Mucosa/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Binding , Protein Precursors/genetics , Protein Precursors/pharmacology , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/metabolism , Receptors, G-Protein-Coupled/genetics
2.
Front Immunol ; 8: 1899, 2017.
Article in English | MEDLINE | ID: mdl-29354121

ABSTRACT

NEO-201 is a novel humanized IgG1 monoclonal antibody that was derived from an immunogenic preparation of tumor-associated antigens from pooled allogeneic colon tumor tissue extracts. It was found to react against a variety of cultured human carcinoma cell lines and was highly reactive against the majority of tumor tissues from many different carcinomas, including colon, pancreatic, stomach, lung, and breast cancers. NEO-201 also exhibited tumor specificity, as the majority of normal tissues were not recognized by this antibody. Functional assays revealed that treatment with NEO-201 is capable of mediating both antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) against tumor cells. Furthermore, the growth of human pancreatic xenograft tumors in vivo was largely attenuated by treatment with NEO-201 both alone and in combination with human peripheral blood mononuclear cells as an effector cell source for ADCC. In vivo biodistribution studies in human tumor xenograft-bearing mice revealed that NEO-201 preferentially accumulates in the tumor but not organ tissue. Finally, a single-dose toxicity study in non-human primates demonstrated safety and tolerability of NEO-201, as a transient decrease in circulating neutrophils was the only related adverse effect observed. These findings indicate that NEO-201 warrants clinical testing as both a novel diagnostic and therapeutic agent for the treatment of a broad variety of carcinomas.

3.
Nat Commun ; 7: 10517, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-26841680

ABSTRACT

CD11b(+)Gr-1(+) myeloid-derived suppressor cells (MDSCs) expand in the spleen during cancer and promote progression through suppression of cytotoxic T cells. An anti-inflammatory reflex arc involving the vagus nerve and memory T cells is necessary for resolution of acute inflammation. Failure of this neural circuit could promote procarcinogenic inflammation and altered tumour immunity. Here we show that splenic TFF2, a secreted anti-inflammatory peptide, is released by vagally modulated memory T cells to suppress the expansion of MDSCs through CXCR4. Splenic denervation interrupts the anti-inflammatory neural arc, resulting in the expansion of MDSCs and colorectal cancer. Deletion of Tff2 recapitulates splenic denervation to promote carcinogenesis. Colorectal carcinogenesis could be suppressed through transgenic overexpression of TFF2, adenoviral transfer of TFF2 or transplantation of TFF2-expressing bone marrow. TFF2 is important to the anti-inflammatory reflex arc and plays an essential role in arresting MDSC proliferation. TFF2 offers a potential approach to prevent and to treat cancer.


Subject(s)
Cell Proliferation/genetics , Colitis/genetics , Colorectal Neoplasms/genetics , Mucins/genetics , Muscle Proteins/genetics , Myeloid Cells/immunology , Peptides/genetics , Receptors, CXCR4/immunology , Spleen/immunology , T-Lymphocytes, Cytotoxic/immunology , Vagus Nerve , Adoptive Transfer , Animals , Blotting, Western , Bone Marrow Transplantation , Colitis/immunology , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/immunology , Cytokines/immunology , Denervation , Disease Progression , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunohistochemistry , Inflammation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucins/immunology , Mucins/metabolism , Muscle Proteins/immunology , Muscle Proteins/metabolism , Neoplasms/genetics , Neoplasms/immunology , Peptides/immunology , Peptides/metabolism , Permeability , Reverse Transcriptase Polymerase Chain Reaction , Spleen/innervation , T-Lymphocyte Subsets/immunology , Trefoil Factor-2 , Vagotomy, Truncal , Vagus Nerve Stimulation
4.
Gastroenterology ; 144(1): 155-66, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23041326

ABSTRACT

BACKGROUND & AIMS: Interleukin (IL)-8 has an important role in initiating inflammation in humans, attracting immune cells such as neutrophils through their receptors CXCR1 and CXCR2. IL-8 has been proposed to contribute to chronic inflammation and cancer. However, mice do not have the IL-8 gene, so human cancer cell lines and xenograft studies have been used to study the role of IL-8 in colon and gastric carcinogenesis. We generated mice that carry a bacterial artificial chromosome that encompasses the entire human IL-8 gene, including its regulatory elements (IL-8Tg mice). METHODS: We studied the effects of IL-8 expression in APCmin(+/-) mice and IL-8Tg mice given azoxymethane and dextran sodium sulfate (DSS). We also examined the effects of IL-8 expression in gastric cancer in INS-GAS mice that overexpress gastrin and IL-8Tg mice infected with Helicobacter felis. RESULTS: In IL-8Tg mice, expression of human IL-8 was controlled by its own regulatory elements, with virtually no messenger RNA or protein detectable under basal conditions. IL-8 was strongly up-regulated on systemic or local inflammatory stimulation, increasing mobilization of immature CD11b(+)Gr-1(+) myeloid cells (IMCs) with thioglycolate-induced peritonitis, DSS-induced colitis, and H. felis-induced gastritis. IL-8 was increased in colorectal tumors from patients and IL-8Tg mice compared with nontumor tissues. IL-8Tg mice developed more tumors than wild-type mice following administration of azoxymethane and DSS. Expression of IL-8 increased tumorigenesis in APCmin(+/-) mice compared with APCmin(+/-) mice that lack IL-8; this was associated with increased numbers of IMCs and angiogenesis in the tumors. CONCLUSIONS: IL-8 contributes to gastrointestinal carcinogenesis by mobilizing IMCs and might be a therapeutic target for gastrointestinal cancers.


Subject(s)
Cell Movement/drug effects , Cell Transformation, Neoplastic/metabolism , Colitis/metabolism , Colonic Neoplasms/metabolism , Gastritis/metabolism , Interleukin-8/metabolism , Myeloid Cells/drug effects , RNA, Messenger/metabolism , Animals , Azoxymethane , Cell Line, Tumor , Colitis/chemically induced , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Dendritic Cells/metabolism , Dextran Sulfate , Gastritis/microbiology , Helicobacter Infections/complications , Helicobacter felis , Humans , Interleukin-8/genetics , Interleukin-8/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , Mice, Transgenic , Myeloid Cells/metabolism , Primary Cell Culture , Tumor Burden , Up-Regulation/drug effects
5.
Cancer Invest ; 30(4): 275-86, 2012 May.
Article in English | MEDLINE | ID: mdl-22480191

ABSTRACT

Transgenic mice overexpressing human progastrin (hGAS) show colonic crypt hyper-proliferation and elevated susceptibility to colon carcinogenesis. We aimed to investigate effects of p53 mutation on colon carcinogenesis in hGAS mice. We show that introducing a p53 gene mutation further increases progastrin dependent BrdU labeling and results in markedly elevated number of aberrant crypt foci (ACF) and colonic tumors. We demonstrate that hGAS/Lgr5-GFP mice have higher number of Lgr5+ colonic stem cells per crypt when compared to Lgr5-GFP mice indicating that progastrin changes crypt biology through increased stem cell numbers and additional p53 mutation leads to more aggressive phenotype in this murine colon cancer model.


Subject(s)
Cell Transformation, Neoplastic/genetics , Colonic Neoplasms/genetics , Gastrins/metabolism , Protein Precursors/metabolism , Tumor Suppressor Protein p53/genetics , Aberrant Crypt Foci/chemically induced , Aberrant Crypt Foci/genetics , Aberrant Crypt Foci/metabolism , Animals , Azoxymethane/toxicity , Carcinogens/toxicity , Cell Proliferation , Colonic Neoplasms/chemically induced , Colonic Neoplasms/metabolism , Disease Models, Animal , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation
6.
J Biol Chem ; 284(6): 3650-62, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-19064997

ABSTRACT

The secreted trefoil factor family 2 (TFF2) protein contributes to the protection of the gastrointestinal mucosa from injury by strengthening and stabilizing mucin gels, stimulating epithelial restitution, and restraining the associated inflammation. Although trefoil factors have been shown to activate signaling pathways, no cell surface receptor has been directly linked to trefoil peptide signaling. Here we demonstrate the ability of TFF2 peptide to activate signaling via the CXCR4 chemokine receptor in cancer cell lines. We found that both mouse and human TFF2 proteins (at approximately 0.5 microm) activate Ca2+ signaling in lymphoblastic Jurkat cells that could be abrogated by receptor desensitization (with SDF-1alpha) or pretreatment with the specific antagonist AMD3100 or an anti-CXCR4 antibody. TFF2 pretreatment of Jurkat cells decreased Ca2+ rise and chemotactic response to SDF-1alpha. In addition, the CXCR4-negative gastric epithelial cell line AGS became highly responsive to TFF2 treatment upon expression of the CXCR4 receptor. TFF2-induced activation of mitogen-activated protein kinases in gastric and pancreatic cancer cells, KATO III and AsPC-1, respectively, was also dependent on the presence of the CXCR4 receptor. Finally we demonstrate a distinct proliferative effect of TFF2 protein on an AGS gastric cancer cell line that expresses CXCR4. Overall these data identify CXCR4 as a bona fide signaling receptor for TFF2 and suggest a mechanism through which TFF2 may modulate immune and tumorigenic responses in vivo.


Subject(s)
Mucins/metabolism , Muscle Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Peptides/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Receptors, CXCR4/metabolism , Animals , Anti-HIV Agents/pharmacology , Benzylamines , Calcium Signaling/drug effects , Chemokine CXCL12/pharmacology , Cyclams , Gene Expression Regulation, Neoplastic/drug effects , Heterocyclic Compounds/pharmacology , Humans , Jurkat Cells , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Mucins/pharmacology , Muscle Proteins/pharmacology , NIH 3T3 Cells , Neoplasm Proteins/agonists , Neoplasm Proteins/pharmacology , Neoplasms, Glandular and Epithelial/pathology , Peptides/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, CXCR4/agonists , Trefoil Factor-2
7.
Regul Pept ; 151(1-3): 106-14, 2008 Nov 29.
Article in English | MEDLINE | ID: mdl-18674570

ABSTRACT

The unprocessed gastrin precursor, progastrin (PG), is often overexpressed in colon cancer and other malignancies where it appears to stimulate colonic growth. Overexpression of progastrin also stimulates proliferation of normal colonic mucosa, but the receptors mediating these effects have not been identified. Here we report the development of a non-radioactive assay for assessment of PG binding to normal and transformed cells. Progastrin was labeled using biotinylation, and binding of biotinylated PG to cells was assessed using flow cytometry. Using this approach, we show strong and specific binding of PG to some cell lines (IEC-6, IEC-18, HT-29, COLO320) and minimal binding to others (HeLa, DC2.4, Jurkat). We also found PG binding to several non-gut epithelial lines, such as CHO-K1, COS-6 and HEK293 cells. The specificity of binding was confirmed by competition with cold, unlabeled PG but not with glycine-extended gastrin or amidated gastrin-17. Binding was not influenced by the presence of the classical CCK-2 receptor, but was partially dependent on the charged glycosaminoglycans (GAG). The analysis of primary colonic tissues isolated from wild type C57BL/6 mouse, revealed a small epithelial subpopulation of non-hematopoietic (CD45-negative) cells that strongly interacted with PG. Surprisingly, this population was greatly expanded in gastrin knockout mice. This non-radioactive, FACS-based assay should prove useful for further characterization of cells expressing the progastrin receptor.


Subject(s)
Gastrins/metabolism , Gastrointestinal Tract/metabolism , Protein Precursors/metabolism , Animals , Annexin A2/metabolism , CHO Cells , COS Cells , Cell Line , Chlorocebus aethiops , Colon/cytology , Colon/metabolism , Cricetinae , Cricetulus , Epithelial Cells/metabolism , Flow Cytometry , Gastrins/deficiency , Gastrins/genetics , Gastrointestinal Tract/cytology , Glycosaminoglycans/metabolism , HeLa Cells , Humans , In Vitro Techniques , Jurkat Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Cholecystokinin B/metabolism
8.
Cell Immunol ; 240(2): 86-95, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16904090

ABSTRACT

Heterotrimeric G-proteins control diverse biological processes by conveying signals from seven-transmembrane receptors to intracellular effectors. Although their signaling roles were originally ascribed to their GTP-bound alpha-subunits, more recent evidence points to the equally active roles played by their betagamma-dimers. To elucidate the individual contributions of their gamma-subtypes, we used a gene targeting approach to show that mice lacking the gamma3-subtype display a defective T-cell dependent immune response. To identify the cellular basis for this defect, we demonstrated that gamma3-mRNA is strongly induced in activated CD4+ T-cells. To determine the mechanism for this regulated expression, we used several strategies to identify the importance of a Runx consensus sequence element in the first intron of the gamma3 gene and the Runx1 protein. Overall, these data provide the first genetic evidence for the tight regulation and involvement of the G protein gamma3-subtype in mounting an effective immune response in mice.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , GTP-Binding Protein gamma Subunits/genetics , Gene Expression Regulation/genetics , Animals , Antibody Formation/genetics , Antibody Formation/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bone Marrow/metabolism , CD4-Positive T-Lymphocytes/immunology , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , Female , Gene Expression Regulation/immunology , Humans , Introns/genetics , Jurkat Cells , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Mutation/immunology , Oligonucleotides/genetics , Oligonucleotides/metabolism , Protein Binding , Spleen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymus Gland/metabolism , Vaccination
9.
Biochem Biophys Res Commun ; 336(1): 190-6, 2005 Oct 14.
Article in English | MEDLINE | ID: mdl-16139800

ABSTRACT

While Wnt and Ras signaling pathways are activated during progression of colorectal cancers, many of their important downstream targets remain to be elucidated. The gastrin gene encodes for a family of peptide growth factors that are commonly upregulated in colorectal neoplasia. Previously, we showed that the Wnt signaling pathway moderately stimulates the gastrin promoter. To determine whether Ras signaling can cooperate with Wnt signaling in transcriptional regulation of gastrin gene expression, we have analyzed the response of murine gastrin promoter-reporter gene constructs to combinations of oncogenic stimulation in transient transfection assays. We found a strong (25- to 40-fold) synergistic stimulation of the gastrin promoter by the combination of oncogenic beta-catenin and K-ras overexpression. Deletion analysis localized the response element to an area between -140 and -110bp upstream in the murine gastrin promoter. Electrophoretic mobility shift assays detected a complex containing beta-catenin/TCF, AP1, and SMAD3/4 transcription factors that bound to a DNA element through AP1 and SMAD binding sites. Gastrin promoter activation could be further enhanced or suppressed by the co-expression of wild type SMAD4 or dominant negative mutant of SMAD4, respectively, and abrogated by the PI3K inhibitor, LY20004, but not by the MEK inhibitor, PD98059. Taken together, our data strongly suggest that oncogenic Wnt and Ras signaling pathways can synergistically induce gastrin expression, possibly contributing to neoplastic progression.


Subject(s)
Cytoskeletal Proteins/physiology , DNA-Binding Proteins/metabolism , Gastrins/genetics , Oncogene Protein p21(ras)/physiology , Promoter Regions, Genetic , Trans-Activators/metabolism , Trans-Activators/physiology , Animals , Genes, Reporter , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Luciferases/genetics , Mice , Phosphatidylinositol 3-Kinases/metabolism , Smad Proteins , Transfection , Wnt Proteins , beta Catenin
10.
J Biol Chem ; 279(41): 42492-502, 2004 Oct 08.
Article in English | MEDLINE | ID: mdl-15292219

ABSTRACT

The transforming growth factor-beta (TGF-beta) and Wnt/wingless pathways play critical roles in the specification of cell fate during development and also contribute to cancer formation and progression. Whereas Wnt signaling is clearly pro-oncogenic, TGF-beta signaling is cell- and context-dependent, manifesting both inhibitory and proliferative effects. The growth factor, gastrin, has previously been shown to be a downstream target of the Wnt pathway and a promoter of gastrointestinal cancer. In this study, we show that the mouse gastrin promoter is regulated synergistically by TGF-beta/Smads and beta-catenin/T-cell factor (TCF). Co-transfection of Smad3/Smad4 and beta-catenin expression constructs synergistically activated mouse gastrin promoter activity 30-60-fold in AGS cells with minimal effect seen with either construct alone. This activation was further potentiated by TGF-beta1 treatment. Mutating either the TCF binding site or the Smad-binding element (SBE) diminished the activation of gastrin expression by Smad3/Smad4 and beta-catenin and led to a loss of gastrin promoter responsiveness to TGF-beta1 treatment. Wnt and TGF-beta regulated endogenous gastrin mRNA levels in AGS cells in a similar fashion, as revealed by small interference RNA studies or overexpression of Smads and TCF4/beta-catenin. Electrophoretic mobility shift assays and DNA affinity precipitation assays showed that the putative SBE and T-cell factor (TCF) sites were able to bind a complex containing Smads and beta-catenin/TCF4. In addition, the synergy between Smads and beta-catenin/TCF4 was dependent on CREB-binding protein (CBP)/P300, as demonstrated by overexpression of CBP or E1A. Moreover, by using a heterogeneous promoter reporter system, we showed that this complex containing Smads/TCF4/beta-catenin complex was able to up-regulate transcription at isolated SBE or TCF sites. Thus, the Wnt signaling pathway is able to activate some target genes through its actions as a co-activator at non-TCF sites and has the potential to profoundly alter transcriptional responses to TGF-beta signaling.


Subject(s)
Gastrins/genetics , Gene Expression Regulation , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Base Sequence , Binding Sites , Blotting, Western , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoskeletal Proteins/metabolism , DNA/metabolism , DNA-Binding Proteins/metabolism , Gastrins/biosynthesis , Gastrins/metabolism , Genes, Reporter , Humans , Immunoprecipitation , Luciferases/metabolism , Mice , Models, Biological , Molecular Sequence Data , Mutation , Oligonucleotides/chemistry , Plasmids/metabolism , Protein Binding , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Smad3 Protein , Smad4 Protein , Trans-Activators/metabolism , Transcriptional Activation , Transfection , Up-Regulation , Wnt Proteins , beta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...