Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Nat Commun ; 12(1): 1482, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33674596

ABSTRACT

Immune evasion is a hallmark of KRAS-driven cancers, but the underlying causes remain unresolved. Here, we use a mouse model of pancreatic ductal adenocarcinoma to inactivate KRAS by CRISPR-mediated genome editing. We demonstrate that at an advanced tumor stage, dependence on KRAS for tumor growth is reduced and is manifested in the suppression of antitumor immunity. KRAS-deficient cells retain the ability to form tumors in immunodeficient mice. However, they fail to evade the host immune system in syngeneic wild-type mice, triggering strong antitumor response. We uncover changes both in tumor cells and host immune cells attributable to oncogenic KRAS expression. We identify BRAF and MYC as key mediators of KRAS-driven tumor immune suppression and show that loss of BRAF effectively blocks tumor growth in mice. Applying our results to human PDAC we show that lowering KRAS activity is likewise associated with a more vigorous immune environment.


Subject(s)
Immune Evasion/physiology , Models, Genetic , Pancreatic Neoplasms/immunology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cytokines/metabolism , Disease Models, Animal , Female , Gene Editing , Gene Expression Profiling , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Pancreatic Ducts/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/immunology , Transcriptome , Pancreatic Neoplasms
2.
Oncotarget ; 8(11): 17995-18009, 2017 Mar 14.
Article in English | MEDLINE | ID: mdl-28152508

ABSTRACT

KRAS is the most commonly mutated oncogene, frequently associated with some of the deadliest forms of cancer. However, the need for potent and specific KRAS inhibitors remains unmet. Here, we evaluated the effects of selected cytotoxic agents on oncogenic KRAS signaling and drug response. The data provided new insights into the functional interaction between the KRAS and MYC pathways and revealed key differences between WT and mutant KRAS expressing cells. Systematic investigation of non-small cell lung cancer cell lines revealed that KRAS mutation can paradoxically increase the sensitivity of cells to cytotoxic agents. We identify MYC as a key regulator of the cellular stress responses and tumor cell viability as MYC expression was suppressed in drug-sensitive but not resistant cells. Furthermore, this suppression was driven by hyperactive KRAS/MAPK signaling. Our findings support a direct link between MYC and cancer cell viability, and raise the possibility that inactivation of MYC may be an effective therapeutic strategy for KRAS mutant tumors across various cancer types.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Antineoplastic Agents/pharmacology , Blotting, Western , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cytotoxins/pharmacology , Gene Expression Regulation, Neoplastic/genetics , Humans , Lung Neoplasms/metabolism , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction/drug effects
3.
Oncotarget ; 6(18): 15814-27, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26158412

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive, metastatic disease with limited treatment options. Factors contributing to the metastatic predisposition and therapy resistance in pancreatic cancer are not well understood. Here, we used a mouse model of KRAS-driven pancreatic carcinogenesis to define distinct subtypes of PDAC metastasis: epithelial, mesenchymal and quasi-mesenchymal. We examined pro-survival signals in these cells and the therapeutic response differences between them. Our data indicate that the initiation and maintenance of the transformed state are separable, and that KRAS dependency is not a fundamental constant of KRAS-initiated tumors. Moreover, some cancer cells can shuttle between the KRAS dependent (drug-sensitive) and independent (drug-tolerant) states and thus escape extinction. We further demonstrate that inhibition of KRAS signaling alone via co-targeting the MAPK and PI3K pathways fails to induce extensive tumor cell death and, therefore, has limited efficacy against PDAC. However, the addition of histone deacetylase (HDAC) inhibitors greatly improves outcomes, reduces the self-renewal of cancer cells, and blocks cancer metastasis in vivo. Our results suggest that targeting HDACs in combination with KRAS or its effector pathways provides an effective strategy for the treatment of PDAC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Histone Deacetylase Inhibitors/pharmacology , MAP Kinase Kinase Kinases/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Animals , Carcinoma, Pancreatic Ductal/enzymology , Carcinoma, Pancreatic Ductal/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm , Genes, ras , Histone Deacetylase Inhibitors/administration & dosage , Humans , MAP Kinase Signaling System/drug effects , Mice , Mice, Nude , Mutation , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Protein Kinase Inhibitors/administration & dosage , Xenograft Model Antitumor Assays
4.
Proc Natl Acad Sci U S A ; 111(9): 3466-71, 2014 Mar 04.
Article in English | MEDLINE | ID: mdl-24550494

ABSTRACT

Pancreatic cancer is one of the deadliest human malignancies. A striking feature of pancreatic cancer is that activating Kras mutations are found in ∼90% of cases. However, apart from a restricted population of cells expressing pancreatic and duodenal homeobox 1 (PDX1), most pancreatic cells are refractory to Kras-driven transformation. In the present study, we sought to determine which subsets of PDX1+ cells may be responsible for tumor growth. Using the Lox-Stop-Lox-KrasG12D genetic mouse model of pancreatic carcinogenesis, we isolated a population of KrasG12D-expressing PDX1+ cells with an inherent capacity to metastasize. This population of cells bears the surface phenotype of EpCAM+CD24+CD44+CD133-SCA1- and is closer in its properties to stem-like cells than to more mature cell types. We further demonstrate that the tumorigenic capacity of PDX1+ cells is limited, becoming progressively lost as the cells acquire a mature phenotype. These data are consistent with the hypothesis that the adult pancreas harbors a dormant progenitor cell population that is capable of initiating tumor growth under conditions of oncogenic stimulation. We present evidence that constitutive activation of the mitogen-activated protein kinase (MAPK/ERK) signaling and stabilization of the MYC protein are the two main driving forces behind the development of pancreatic cancer cells with stem-cell-like properties and high metastatic potential. Our results suggest that pancreatic cells bearing Kras mutation can be induced to differentiate into quasi-normal cells with suppressed tumorigenicity by selective inhibition of the MAPK/ERK/MYC signaling cascade.


Subject(s)
Homeodomain Proteins/metabolism , Lung Neoplasms/secondary , MAP Kinase Signaling System/physiology , Neoplasm Metastasis/physiopathology , Pancreatic Neoplasms/physiopathology , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Trans-Activators/metabolism , Animals , Blotting, Western , Flow Cytometry , Lung Neoplasms/pathology , Mice , Mice, Knockout , Mice, Nude , Mutation, Missense/genetics , Pancreatic Neoplasms/metabolism , Polymerase Chain Reaction , Tumor Suppressor Protein p53/genetics
5.
Proc Natl Acad Sci U S A ; 110(10): 3937-42, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23431158

ABSTRACT

Genetically or epigenetically defined reprogramming is a hallmark of cancer cells. However, a causal association between genome reprogramming and cancer has not yet been conclusively established. In particular, little is known about the mechanisms that underlie metastasis of cancer, and even less is known about the identity of metastasizing cancer cells. In this study, we used a model of conditional expression of oncogenic KrasG12D allele in primary mouse cells to show that reprogramming and dedifferentiation is a fundamental early step in malignant transformation and cancer initiation. Our data indicate that stable expression of activated KrasG12D confers on cells a large degree of phenotypic plasticity that predisposes them to neoplastic transformation and acquisition of stem cell characteristics. We have developed a genetically tractable model system to investigate the origins and evolution of metastatic pancreatic cancer cells. We show that metastatic conversion of KrasG12D-expressing cells that exhibit different degrees of differentiation and malignancy can be reconstructed in cell culture, and that the proto-oncogene c-Myc controls the generation of self-renewing metastatic cancer cells. Collectively, our results support a model wherein non-stem cancer cells have the potential to dedifferentiate and acquire stem cell properties as a direct consequence of oncogene-induced plasticity. Moreover, the disturbance in the normally existing dynamic equilibrium between cancer stem cells and non-stem cancer cells allows the formation of cancer stem cells with high metastatic capacity at any time during cancer progression.


Subject(s)
Cell Transdifferentiation/genetics , Cell Transformation, Neoplastic/genetics , Genes, myc , Genes, ras , Animals , Cell Transformation, Neoplastic/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Genes, p53 , Mice , Mice, Knockout , Models, Genetic , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phenotype , Point Mutation
6.
Am J Physiol Gastrointest Liver Physiol ; 292(5): G1347-58, 2007 May.
Article in English | MEDLINE | ID: mdl-17272518

ABSTRACT

Diseases of intestinal inflammation like necrotizing enterocolitis (NEC) are associated with impaired epithelial barrier integrity and the sustained release of intestinal nitric oxide (NO). NO modifies the cytoskeletal regulator RhoA-GTPase, suggesting that NO could affect barrier healing by inhibiting intestinal restitution. We now hypothesize that NO inhibits enterocyte migration through RhoA-GTPase and sought to determine the pathways involved. The induction of NEC was associated with increased enterocyte NO release and impaired migration of bromodeoxyuridine-labeled enterocytes from terminal ileal crypts to villus tips. In IEC-6 enterocytes, NO significantly inhibited enterocyte migration and activated RhoA-GTPase while increasing the formation of stress fibers. In parallel, exposure of IEC-6 cells to NO increased the phosphorylation of focal adhesion kinase (pFAK) and caused a striking increase in cell-matrix adhesiveness, suggesting a mechanism by which NO could impair enterocyte migration. NEC was associated with increased expression of pFAK in the terminal ileal mucosa of wild-type mice and a corresponding increase in disease severity compared with inducible NO synthase knockout mice, confirming the dependence of NO for FAK phosphorylation in vivo and its role in the pathogenesis of NEC. Strikingly, inhibition of the protein tyrosine phosphatase SHP-2 in IEC-6 cells prevented the activation of RhoA by NO, restored focal adhesions, and reversed the inhibitory effects of NO on enterocyte migration. These data indicate that NO impairs mucosal healing by inhibiting enterocyte migration through activation of RhoA in a SHP-2-dependent manner and support a possible role for SHP-2 as a therapeutic target in diseases of intestinal inflammation like NEC.


Subject(s)
Cell Migration Inhibition , Enterocytes/physiology , Nitric Oxide/physiology , rhoA GTP-Binding Protein/metabolism , Animals , Animals, Newborn , Cell Line , Enterocolitis, Necrotizing/pathology , Enterocolitis, Necrotizing/physiopathology , Enterocytes/drug effects , Enzyme Activation , Focal Adhesion Protein-Tyrosine Kinases/biosynthesis , Intestinal Mucosa/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Mice , Nitric Oxide/metabolism , Nitroso Compounds/pharmacology , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatases/physiology , Rats , SH2 Domain-Containing Protein Tyrosine Phosphatases , src Homology Domains/physiology
7.
J Biol Chem ; 280(10): 8893-900, 2005 Mar 11.
Article in English | MEDLINE | ID: mdl-15632155

ABSTRACT

RON is a receptor tyrosine kinase in the MET family. We have expressed and purified active RON using the Sf9/baculovirus system. The constructs used in this study comprise the kinase domain alone and the kinase domain plus the C-terminal region. The construct containing the kinase domain alone has a higher specific activity than the construct containing the kinase and C-terminal domains. Purified RON undergoes autophosphorylation, and the exogenous RON C terminus serves as a substrate. Peptides containing a dityrosine motif derived from the C-terminal tail inhibit RON in vitro or when delivered into intact cells, consistent with an autoinhibitory mechanism. Phenylalanine substitutions within these peptides increase the inhibitory potency. Moreover, introduction of these Phe residues into the dityrosine motif of the RON kinase leads to a decrease in kinase activity. Taken together, our data suggest a model in which the C-terminal tail of RON regulates kinase activity via an interaction with the kinase catalytic domain.


Subject(s)
Receptor Protein-Tyrosine Kinases/metabolism , 3T3 Cells , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , Cell Line , Conserved Sequence , DNA Primers , Kinetics , Mice , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphorylation , Polymerase Chain Reaction , Receptor Protein-Tyrosine Kinases/chemistry , Spodoptera , Tyrosine
8.
Oncogene ; 22(44): 6909-18, 2003 Oct 09.
Article in English | MEDLINE | ID: mdl-14534538

ABSTRACT

Receptor tyrosine kinases (RTKs) such as the fibroblast growth factor receptor (FGFR) and the epidermal growth factor receptor are overexpressed in a variety of cancers. In addition to overexpression, the FGFRs are found mutated in some cancers. The Src homology 2 domain-containing phosphotyrosine phosphatase (SHP2) is a critical mediator of RTK signaling, but its role in oncogenic RTK-induced cell transformation and cancer development is largely unknown. In the current report, we demonstrate that constitutively activated FGFR3 (K/E-FR3) transforms NIH-3T3 cells, and that SHP2 is a critical mediator of this transformation. Infection of K/E-FR3-transformed 3T3 cells with a retrovirus carrying a dominant-negative mutant of SHP2 (C/S-SHP2) retarded cell growth, reversed the transformation phenotype and inhibited focus-forming ability. Furthermore, treatment of K/E-FR3-transformed NIH-3T3 cells with PD98059 or LY294002, specific inhibitors of MEK and PI3K, respectively, inhibited focus formation. Biochemical analysis showed that K/E-FR3 activates the Ras-ERK and the PI3K signaling pathways, and that the C/S SHP2 mutant suppressed this effect via competitive displacement of interaction of the endogenous SHP2 with FRS2. However, the C/S SHP2 protein did not show any effect on receptor autophosphorylation, FRS2 tyrosine phosphorylation or interaction of Grb2 with K/E-FR3 or FRS2. Together, the results show that K/E-FR3 is transforming and that the Ras-ERK and the PI3K-Akt signaling pathways, which are positively regulated by SHP2, are important for K/E-FR3-induced transformation.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , Cell Transformation, Viral , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases , Receptors, Fibroblast Growth Factor/metabolism , 3T3 Cells , Animals , COS Cells , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Carrier Proteins/genetics , Cell Line, Transformed , Cell Transformation, Neoplastic , Cells, Cultured , Chlorocebus aethiops , Chromones/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factors/metabolism , Flavonoids/pharmacology , Gene Expression Regulation , Membrane Glycoproteins , Membrane Proteins/genetics , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Morpholines/pharmacology , Mutagenesis, Site-Directed , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptor, Fibroblast Growth Factor, Type 3 , Receptors, Fibroblast Growth Factor/genetics , Retroviridae/genetics , Signal Transduction/drug effects , ras Proteins/metabolism
9.
Oncogene ; 22(41): 6311-8, 2003 Sep 25.
Article in English | MEDLINE | ID: mdl-14508511

ABSTRACT

Transformation of fibroblasts by V-SEA involves activation of the ERK and phosphatidylinositol 3-kinase (PI3K) pathways. Effector proteins that are key mediators of the ERK and PI3K pathways, namely Grb2, the tyrosine phosphatase, SHP2 and PI3K, interact with the two phosphotyrosines found in the bidentate motif in the carboxy-terminal region of V-SEA. Genetic analysis demonstrated that while Y557 was a primary binding site and thus activator of the PI3K-Akt pathway, Y564 also contributed to the activation of this pathway. Y564 was located within a Grb2-binding motif, this raised the possibility that a protein that associated with Grb2 might be important for this PI3K activation. The scaffolding proteins Gab1 and/or Gab2 were candidates for this role. In this report, we demonstrate that V-SEA preferentially interacts with Gab2. Furthermore by using Gab2 null fibroblasts, we demonstrate that Gab2 is essential for fibroblast transformation by V-SEA. Using mutant forms of Gab2, we show that activation of the PI3K-Akt pathway via Gab2 is required for V-SEA-induced transformation. However, efficient fibroblast transformation also requires the SHP2 interaction site on Gab2.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Fibroblasts/metabolism , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Adaptor Proteins, Signal Transducing , Animals , Mice , Phosphatidylinositol 3-Kinases/metabolism
10.
Oncogene ; 21(5): 697-707, 2002 Jan 24.
Article in English | MEDLINE | ID: mdl-11850798

ABSTRACT

V-SEA is the transforming component of S13 Avian Erythroblastosis Retrovirus that causes erythroblastosis and anemia in chicken. Like all members in the family (MET, RON, SEA), its cytosolic domain possesses two tyrosine autophosphorylation sites in the tandemly arranged bidentate motif that serve as docking sites for SH2 domain-containing proteins. Here, we investigated phosphotyrosine-dependent activation of signaling pathways and their significance in V-SEA-induced transformation and/or proliferation. We demonstrated that V-SEA activates the PI3K-Akt signaling pathway primarily in Y557- and secondarily in Y564-dependent manner. V-SEA was also shown to induce the tyrosine phosphorylation of the Gab2 protein, leading to PI3K association and thus providing an alternative route for PI3K activation. On the other hand, activation of the Ras-ERK pathway is primarily via Y564 and secondarily via Y557. A dominant-negative form of Ras inhibited V-SEA-induced ERK phosphorylation in concentration dependent manner suggesting the importance of the Grb2-Ras signaling axis in V-SEA-induced ERK activation. The biological significance of activation of the PI3K-Akt and the Ras-ERK pathways in V-SEA-induced transformation was analysed in the V-SEA-RAT1 and V-SEA-3T3 cell lines by employing specific inhibitors, LY294002 and PD98059 compounds. Both the PD and LY compounds inhibited cell growth, but only the PD compound caused reversion of the transformed phenotype. In addition, both compounds inhibited focal colony formation by the transformants in soft agar. Thus, transformation by the V-SEA oncogene is a function of the concomitant activation of, at least, the PI3K-Akt and Ras-ERK signaling pathways that regulate cell growth and morphology.


Subject(s)
Cell Transformation, Viral , MAP Kinase Signaling System , Oncogene Proteins, Viral/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cell Division , Cell Line , Cell Line, Transformed , Cell Size , Chromones/pharmacology , Colony-Forming Units Assay , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphoproteins/physiology , Protein-Tyrosine Kinases/pharmacology , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins p21(ras)/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...