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 ; 7(22): 31681-8, 2016 May 31.
Article in English | MEDLINE | ID: mdl-27213588

ABSTRACT

Recurrent fusion of the v-myb avian myelobastosis viral oncogene homolog (MYB) and nuclear factor I/B (NFIB) generates the MYB-NFIB transcription factor, which has been detected in a high percentage of individuals with adenoid cystic carcinoma (ACC). To understand the functional role of this fusion protein in carcinogenesis, we generated a conditional mutant transgenic mouse that expresses MYB-NFIB along with p53 mutation in tissues that give rise to ACC: mammary tissue, salivary glands, or systemically in the whole body. Expression of the oncogene in mammary tissue resulted in hyperplastic glands that developed into adenocarcinoma in 27.3% of animals. Systemic expression of the MYB-NFIB fusion caused more rapid development of this breast phenotype, but mice died due to abnormal proliferation in the glomerular compartment of the kidney, which led to development of glomerulonephritis. These findings suggest the MYB-NFIB fusion is oncogenic and treatments targeting this transcription factor may lead to therapeutic responses in ACC patients.


Subject(s)
Adenocarcinoma/genetics , Cell Transformation, Neoplastic/genetics , Gene Fusion , Mammary Neoplasms, Experimental/genetics , Oncogene Proteins, Fusion/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Female , Gene Expression Regulation, Neoplastic , Genes, p53 , Genetic Predisposition to Disease , Glomerulonephritis/genetics , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Hyperplasia , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice, Inbred C57BL , Mice, Transgenic , Oncogene Proteins, Fusion/metabolism , Phenotype , Proto-Oncogene Mas , Salivary Glands/metabolism , Salivary Glands/pathology , Time Factors
2.
Cancer Res ; 74(17): 4676-84, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25035393

ABSTRACT

Somatic mutations in FGFR2 are present in 4% to 5% of patients diagnosed with non-small cell lung cancer (NSCLC). Amplification and mutations in FGFR genes have been identified in patients with NSCLCs, and clinical trials are testing the efficacy of anti-FGFR therapies. FGFR2 and other FGFR kinase family gene alterations have been found in both lung squamous cell carcinoma and lung adenocarcinoma, although mouse models of FGFR-driven lung cancers have not been reported. Here, we generated a genetically engineered mouse model (GEMM) of NSCLC driven by a kinase domain mutation in FGFR2. Combined with p53 ablation, primary grade 3/4 adenocarcinoma was induced in the lung epithelial compartment exhibiting locally invasive and pleiotropic tendencies largely made up of multinucleated cells. Tumors were acutely sensitive to pan-FGFR inhibition. This is the first FGFR2-driven lung cancer GEMM, which can be applied across different cancer indications in a preclinical setting.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Protein Kinase Inhibitors/pharmacology , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Disease Models, Animal , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutation/drug effects , Mutation/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
3.
Cancer Cell ; 25(5): 590-604, 2014 May 12.
Article in English | MEDLINE | ID: mdl-24794706

ABSTRACT

Lung squamous cell carcinoma (SCC) is a deadly disease for which current treatments are inadequate. We demonstrate that biallelic inactivation of Lkb1 and Pten in the mouse lung leads to SCC that recapitulates the histology, gene expression, and microenvironment found in human disease. Lkb1;Pten null (LP) tumors expressed the squamous markers KRT5, p63 and SOX2, and transcriptionally resembled the basal subtype of human SCC. In contrast to mouse adenocarcinomas, the LP tumors contained immune populations enriched for tumor-associated neutrophils. SCA1(+)NGFR(+) fractions were enriched for tumor-propagating cells (TPCs) that could serially transplant the disease in orthotopic assays. TPCs in the LP model and NGFR(+) cells in human SCCs highly expressed Pd-ligand-1 (PD-L1), suggesting a mechanism of immune escape for TPCs.


Subject(s)
B7-H1 Antigen/biosynthesis , Carcinoma, Squamous Cell/immunology , Lung Neoplasms/immunology , PTEN Phosphohydrolase/genetics , Protein Serine-Threonine Kinases/genetics , Tumor Escape/immunology , AMP-Activated Protein Kinases , Animals , Antigens, Ly/biosynthesis , B-Lymphocytes/immunology , Carcinoma, Squamous Cell/genetics , Disease Models, Animal , Immune Tolerance/immunology , Keratin-15 , Keratin-5/biosynthesis , Killer Cells, Natural/immunology , Lung/metabolism , Lung Neoplasms/genetics , Lymphocyte Activation/immunology , Macrophages/immunology , Membrane Proteins/biosynthesis , Metabolome , Mice , Neutrophils/immunology , Phosphoproteins/biosynthesis , Receptor, Nerve Growth Factor/biosynthesis , SOXB1 Transcription Factors/biosynthesis , T-Lymphocytes/immunology , Trans-Activators/biosynthesis , Transcription, Genetic , Tumor Cells, Cultured
4.
Genes Dev ; 28(5): 479-90, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24589777

ABSTRACT

Mutations in isocitrate dehydrogenase 1 and 2 (IDH1/2) have been discovered in several cancer types and cause the neurometabolic syndrome D2-hydroxyglutaric aciduria (D2HGA). The mutant enzymes exhibit neomorphic activity resulting in production of D2-hydroxyglutaric acid (D-2HG). To study the pathophysiological consequences of the accumulation of D-2HG, we generated transgenic mice with conditionally activated IDH2(R140Q) and IDH2(R172K) alleles. Global induction of mutant IDH2 expression in adults resulted in dilated cardiomyopathy, white matter abnormalities throughout the central nervous system (CNS), and muscular dystrophy. Embryonic activation of mutant IDH2 resulted in more pronounced phenotypes, including runting, hydrocephalus, and shortened life span, recapitulating the abnormalities observed in D2HGA patients. The diseased hearts exhibited mitochondrial damage and glycogen accumulation with a concordant up-regulation of genes involved in glycogen biosynthesis. Notably, mild cardiac hypertrophy was also observed in nude mice implanted with IDH2(R140Q)-expressing xenografts, suggesting that 2HG may potentially act in a paracrine fashion. Finally, we show that silencing of IDH2(R140Q) in mice with an inducible transgene restores heart function by lowering 2HG levels. Together, these findings indicate that inhibitors of mutant IDH2 may be beneficial in the treatment of D2HGA and suggest that 2HG produced by IDH mutant tumors has the potential to provoke a paraneoplastic condition.


Subject(s)
Cardiomyopathies/genetics , Glutarates/metabolism , Isocitrate Dehydrogenase/genetics , Mutation , Neurodegenerative Diseases/genetics , Animals , Cardiomyopathies/enzymology , Cardiomyopathies/pathology , Cell Line , Gene Expression Profiling , Gene Expression Regulation, Developmental , Heart/physiopathology , Humans , Isocitrate Dehydrogenase/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/pathology
5.
Cancer Discov ; 3(12): 1355-63, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24078774

ABSTRACT

UNLABELLED: The success in lung cancer therapy with programmed death (PD)-1 blockade suggests that immune escape mechanisms contribute to lung tumor pathogenesis. We identified a correlation between EGF receptor (EGFR) pathway activation and a signature of immunosuppression manifested by upregulation of PD-1, PD-L1, CTL antigen-4 (CTLA-4), and multiple tumor-promoting inflammatory cytokines. We observed decreased CTLs and increased markers of T-cell exhaustion in mouse models of EGFR-driven lung cancer. PD-1 antibody blockade improved the survival of mice with EGFR-driven adenocarcinomas by enhancing effector T-cell function and lowering the levels of tumor-promoting cytokines. Expression of mutant EGFR in bronchial epithelial cells induced PD-L1, and PD-L1 expression was reduced by EGFR inhibitors in non-small cell lung cancer cell lines with activated EGFR. These data suggest that oncogenic EGFR signaling remodels the tumor microenvironment to trigger immune escape and mechanistically link treatment response to PD-1 inhibition. SIGNIFICANCE: We show that autochthonous EGFR-driven lung tumors inhibit antitumor immunity by activating the PD-1/PD-L1 pathway to suppress T-cell function and increase levels of proinflammatory cytokines. These findings indicate that EGFR functions as an oncogene through non-cell-autonomous mechanisms and raise the possibility that other oncogenes may drive immune escape.


Subject(s)
B7-H1 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/immunology , Cytokines/metabolism , ErbB Receptors/metabolism , Lung Neoplasms/immunology , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/immunology , Tumor Escape , Animals , B7-H1 Antigen/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oncogenes , Programmed Cell Death 1 Receptor/genetics , Signal Transduction , Tumor Microenvironment
6.
Clin Cancer Res ; 19(22): 6183-92, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24045185

ABSTRACT

PURPOSE: Amplification of MYC is one of the most common genetic alterations in lung cancer, contributing to a myriad of phenotypes associated with growth, invasion, and drug resistance. Murine genetics has established both the centrality of somatic alterations of Kras in lung cancer, as well as the dependency of mutant Kras tumors on MYC function. Unfortunately, drug-like small-molecule inhibitors of KRAS and MYC have yet to be realized. The recent discovery, in hematologic malignancies, that bromodomain and extra-terminal (BET) bromodomain inhibition impairs MYC expression and MYC transcriptional function established the rationale of targeting KRAS-driven non-small cell lung cancer (NSCLC) with BET inhibition. EXPERIMENTAL DESIGN: We performed functional assays to evaluate the effects of JQ1 in genetically defined NSCLC cell lines harboring KRAS and/or LKB1 mutations. Furthermore, we evaluated JQ1 in transgenic mouse lung cancer models expressing mutant kras or concurrent mutant kras and lkb1. Effects of bromodomain inhibition on transcriptional pathways were explored and validated by expression analysis. RESULTS: Although JQ1 is broadly active in NSCLC cells, activity of JQ1 in mutant KRAS NSCLC is abrogated by concurrent alteration or genetic knockdown of LKB1. In sensitive NSCLC models, JQ1 treatment results in the coordinate downregulation of the MYC-dependent transcriptional program. We found that JQ1 treatment produces significant tumor regression in mutant kras mice. As predicted, tumors from mutant kras and lkb1 mice did not respond to JQ1. CONCLUSION: Bromodomain inhibition comprises a promising therapeutic strategy for KRAS-mutant NSCLC with wild-type LKB1, via inhibition of MYC function. Clinical studies of BET bromodomain inhibitors in aggressive NSCLC will be actively pursued. Clin Cancer Res; 19(22); 6183-92. ©2013 AACR.


Subject(s)
Azepines/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Nuclear Proteins/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Transcription Factors/antagonists & inhibitors , Triazoles/pharmacology , AMP-Activated Protein Kinases , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Lung Neoplasms/genetics , Mice , Mice, Knockout , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , RNA Interference , RNA, Small Interfering , Signal Transduction/drug effects , Transcription Factors/genetics
7.
Cancer Discov ; 3(8): 870-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23715154

ABSTRACT

The LKB1/STK11 tumor suppressor encodes a serine/threonine kinase, which coordinates cell growth, polarity, motility, and metabolism. In non-small cell lung carcinoma, LKB1 is somatically inactivated in 25% to 30% of cases, often concurrently with activating KRAS mutations. Here, we used an integrative approach to define novel therapeutic targets in KRAS-driven LKB1-mutant lung cancers. High-throughput RNA interference screens in lung cancer cell lines from genetically engineered mouse models driven by activated KRAS with or without coincident Lkb1 deletion led to the identification of Dtymk, encoding deoxythymidylate kinase (DTYMK), which catalyzes dTTP biosynthesis, as synthetically lethal with Lkb1 deficiency in mouse and human lung cancer lines. Global metabolite profiling showed that Lkb1-null cells had a striking decrease in multiple nucleotide metabolites as compared with the Lkb1-wild-type cells. Thus, LKB1-mutant lung cancers have deficits in nucleotide metabolism that confer hypersensitivity to DTYMK inhibition, suggesting that DTYMK is a potential therapeutic target in this aggressive subset of tumors.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Nucleoside-Phosphate Kinase/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Death , Cell Line, Tumor , DNA Damage , DNA Replication , Gene Knockdown Techniques , Genomics , High-Throughput Screening Assays , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Metabolomics , Mice , Models, Genetic , Molecular Targeted Therapy , Nucleoside-Phosphate Kinase/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , RNA Interference , Thymine Nucleotides/metabolism
8.
Mol Biol Cell ; 24(4): 474-82, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23283986

ABSTRACT

The malignant brain cancer glioblastoma multiforme (GBM) displays invasive growth behaviors that are regulated by extracellular cues within the neural microenvironment. The adhesion and signaling pathways that drive GBM cell invasion remain largely uncharacterized. Here we use human GBM cell lines, primary patient samples, and preclinical mouse models to demonstrate that integrin αvß8 is a major driver of GBM cell invasion. ß8 integrin is overexpressed in many human GBM cells, with higher integrin expression correlating with increased invasion and diminished patient survival. Silencing ß8 integrin in human GBM cells leads to impaired tumor cell invasion due to hyperactivation of the Rho GTPases Rac1 and Cdc42. ß8 integrin coimmunoprecipitates with Rho-GDP dissociation inhibitor 1 (RhoGDI1), an intracellular signaling effector that sequesters Rho GTPases in their inactive GDP-bound states. Silencing RhoGDI1 expression or uncoupling αvß8 integrin-RhoGDI1 protein interactions blocks GBM cell invasion due to Rho GTPase hyperactivation. These data reveal for the first time that αvß8 integrin, via interactions with RhoGDI1, regulates activation of Rho proteins to promote GBM cell invasiveness. Hence targeting the αvß8 integrin-RhoGDI1 signaling axis might be an effective strategy for blocking GBM cell invasion.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Integrins/genetics , cdc42 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/genetics , rho Guanine Nucleotide Dissociation Inhibitor alpha/genetics , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Adhesion/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Integrins/antagonists & inhibitors , Integrins/metabolism , Male , Mice , Mice, Nude , Neoplasm Invasiveness/genetics , Neoplasm Transplantation , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , Signal Transduction , Stereotaxic Techniques , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rho Guanine Nucleotide Dissociation Inhibitor alpha/metabolism
9.
Cancer Res ; 71(20): 6371-81, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21859829

ABSTRACT

Glioblastoma multiforme (GBM) is a highly invasive brain tumor that develops florid microvascular proliferation and hemorrhage. However, mechanisms that favor invasion versus angiogenesis in this setting remain largely uncharacterized. Here, we show that integrin ß8 is an essential regulator of both GBM-induced angiogenesis and tumor cell invasiveness. Highly angiogenic and poorly invasive tumors expressed low levels of ß8 integrin, whereas highly invasive tumors with limited neovascularization expressed high levels of ß8 integrin. Manipulating ß8 integrin protein levels altered the angiogenic and invasive growth properties of GBMs, in part, reflected by a diminished activation of latent TGFßs, which are extracellular matrix protein ligands for ß8 integrin. Taken together, these results establish a role for ß8 integrin in differential control of angiogenesis versus tumor cell invasion in GBM. Our findings suggest that inhibiting ß8 integrin or TGFß signaling may diminish tumor cell invasiveness during malignant progression and following antivascular therapies.


Subject(s)
Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Glioblastoma/blood supply , Glioblastoma/pathology , Integrin beta Chains/metabolism , Neovascularization, Pathologic/metabolism , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Glioblastoma/metabolism , Humans , Male , Mice , Mice, Nude , Neoplasm Invasiveness , Transforming Growth Factor beta/metabolism
10.
J Cell Sci ; 122(Pt 11): 1842-51, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19461074

ABSTRACT

Central nervous system (CNS) neurovascular units are multicellular complexes consisting of neural cells, blood vessels and a milieu of extracellular matrix (ECM) proteins. ECM-mediated adhesion and signaling events within neurovascular units probably contribute to proper CNS development and physiology; however, the molecular mechanisms that control these events remain largely undetermined. Previous studies from our group and others showed that ablation of the ECM receptor, alphavbeta8 integrin, in neural progenitor cells (NPCs) of the embryonic mouse brain results in severe developmental neurovascular pathologies and premature death. Here, we have investigated the functions for this integrin in the adult brain by studying mice harboring a homozygous-null beta8 gene mutation generated on an outbred background that permits survival for several months. We show that adult beta8-/- mice display widespread defects in neurovascular unit homeostasis, including increased numbers of intracerebral blood vessels with pronounced perivascular astrogliosis. Furthermore, in neurogenic regions of the adult brain, where NPCs cluster around blood vessels in neurovascular niches, beta8 integrin is essential for normal control of NPC proliferation and survival. Analysis of NPCs cultured ex vivo reveals that the growth and survival defects correlate, in part, with diminished integrin-mediated activation of latent transforming growth factor beta1 (TGFbeta1), which is an ECM protein ligand for alphavbeta8 integrin. Collectively, these data identify essential functions for beta8 integrin in regulating neurovascular unit physiology in the post-natal mouse brain.


Subject(s)
Brain , Homeostasis , Integrins/metabolism , Neurogenesis/physiology , Animals , Blood-Brain Barrier/physiology , Brain/blood supply , Brain/cytology , Brain/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Extracellular Matrix/metabolism , Integrins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Neurons/metabolism , Olfactory Bulb/abnormalities , Olfactory Bulb/anatomy & histology , Phenotype , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...