Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Exp Med ; 217(6)2020 06 01.
Article in English | MEDLINE | ID: mdl-32271879

ABSTRACT

We have generated mouse models of malignant mesothelioma (MM) based upon disruption of the Bap1, Nf2, and Cdkn2ab tumor suppressor loci in various combinations as also frequently observed in human MM. Inactivation of all three loci in the mesothelial lining of the thoracic cavity leads to a highly aggressive MM that recapitulates the histological features and gene expression profile observed in human patients. The tumors also show a similar inflammatory phenotype. Bap1 deletion alone does not cause MM but dramatically accelerates MM development when combined with Nf2 and Cdkn2ab (hereafter BNC) disruption. The accelerated tumor development is accompanied by increased Polycomb repression and EZH2-mediated redistribution of H3K27me3 toward promoter sites with concomitant activation of PI3K and MAPK pathways. Treatment of BNC tumor-bearing mice with cisplatin and pemetrexed, the current frontline treatment, prolongs survival. This makes the autochthonous mouse model described here very well suited to explore the pathogenesis of MM and validate new treatment regimens for MM, including immunotherapy.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p15/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Gene Deletion , Mesothelioma, Malignant/metabolism , Neurofibromin 2/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Animals , Disease Models, Animal , Disease Progression , Humans , Immunophenotyping , MAP Kinase Signaling System/drug effects , Mesothelioma, Malignant/genetics , Mesothelioma, Malignant/pathology , Mice , Phosphatidylinositol 3-Kinases/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Transcription, Genetic/drug effects , Tumor Microenvironment/drug effects
2.
Cell Rep ; 30(11): 3837-3850.e3, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32187553

ABSTRACT

Fibroblast growth factor receptor 1 (FGFR1) is frequently amplified in human small-cell lung cancer (SCLC), but its contribution to SCLC and other lung tumors has remained elusive. Here, we assess the tumorigenic capacity of constitutive-active FGFR1 (FGFR1K656E) with concomitant RB and P53 depletion in mouse lung. Our results reveal a context-dependent effect of FGFR1K656E: it impairs SCLC development from CGRPPOS neuroendocrine (NE) cells, which are considered the major cell of origin of SCLC, whereas it promotes SCLC and low-grade NE bronchial lesions from tracheobronchial-basal cells. Moreover, FGFR1K656E induces lung adenocarcinoma (LADC) from most lung cell compartments. However, its expression is not sustained in LADC originating from CGRPPOS cells. Therefore, cell context and tumor stage should be taken into account when considering FGFR1 inhibition as a therapeutic option.


Subject(s)
Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Oncogenes , Receptor, Fibroblast Growth Factor, Type 1/genetics , Retinoblastoma Protein/metabolism , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Tumor Suppressor Protein p53/metabolism , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Animals , Bronchi/pathology , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic , Humans , Integrases/metabolism , Keratins/metabolism , Lung Neoplasms/genetics , Mice , Mutation/genetics , Nasal Cavity/pathology , Neurosecretory Systems/pathology , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Small Cell Lung Carcinoma/genetics
3.
Cell Rep ; 27(11): 3345-3358.e4, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31189116

ABSTRACT

Small-cell lung cancer is the most aggressive type of lung cancer, characterized by a remarkable response to chemotherapy followed by development of resistance. Here, we describe SCLC subtypes in Mycl- and Nfib-driven GEMM that include CDH1-high peripheral primary tumor lesions and CDH1-negative, aggressive intrapulmonary metastases. Cisplatin treatment preferentially eliminates the latter, thus revealing a striking differential response. Using a combined transcriptomic and proteomic approach, we find a marked reduction in proliferation and metabolic rewiring following cisplatin treatment and present evidence for a distinctive metabolic and structural profile defining intrinsically resistant populations. This offers perspectives for effective combination therapies that might also hold promise for treating human SCLC, given the very similar response of both mouse and human SCLC to cisplatin.


Subject(s)
Carcinoma, Small Cell/genetics , Drug Resistance, Neoplasm , Genetic Heterogeneity , Lung Neoplasms/genetics , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Small Cell/drug therapy , Cell Line, Tumor , Cisplatin/therapeutic use , Female , Humans , Lung Neoplasms/drug therapy , Male , Mice , Proteome/genetics , Proteome/metabolism , Transcriptome
4.
Mol Cancer Ther ; 18(4): 762-770, 2019 04.
Article in English | MEDLINE | ID: mdl-30872379

ABSTRACT

Small cell lung cancer (SCLC) is generally regarded as very difficult to treat, mostly due to the development of metastases early in the disease and a quick relapse with resistant disease. SCLC patients initially show a good response to treatment with the DNA damaging agents cisplatin and etoposide. This is, however, quickly followed by the development of resistant disease, which urges the development of novel therapies for this type of cancer. In this study, we set out to compile a comprehensive overview of the vulnerabilities of SCLC. A functional genome-wide screen where all individual genes were knocked out was performed to identify novel vulnerabilities of SCLC. By analysis of the knockouts that were lethal to these cancer cells, we identified several processes to be synthetic vulnerabilities in SCLC. We were able to validate the vulnerability to inhibition of the replication stress response machinery by use of Chk1 and ATR inhibitors. Strikingly, SCLC cells were more sensitive to these inhibitors than nontransformed cells. In addition, these inhibitors work synergistically with either etoposide and cisplatin, where the interaction is largest with the latter. ATR inhibition by VE-822 treatment in combination with cisplatin also outperforms the combination of cisplatin with etoposide in vivo Altogether, our study uncovered a critical dependence of SCLC on the replication stress response and urges the validation of ATR inhibitors in combination with cisplatin in a clinical setting.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cisplatin/therapeutic use , Isoxazoles/therapeutic use , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrazines/therapeutic use , Small Cell Lung Carcinoma/drug therapy , A549 Cells , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , CRISPR-Associated Protein 9/genetics , Cell Survival/drug effects , Checkpoint Kinase 1/antagonists & inhibitors , Cisplatin/administration & dosage , DNA Damage/drug effects , Drug Synergism , Etoposide/administration & dosage , Etoposide/therapeutic use , Humans , Isoxazoles/administration & dosage , Isoxazoles/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Pyrazines/administration & dosage , Pyrazines/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
5.
Cell Rep ; 16(3): 631-43, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27373156

ABSTRACT

Small cell lung cancer (SCLC) is an aggressive neuroendocrine tumor, and no effective treatment is available to date. Mouse models of SCLC based on the inactivation of Rb1 and Trp53 show frequent amplifications of the Nfib and Mycl genes. Here, we report that, although overexpression of either transcription factor accelerates tumor growth, NFIB specifically promotes metastatic spread. High NFIB levels are associated with expansive growth of a poorly differentiated and almost exclusively E-cadherin (CDH1)-negative invasive tumor cell population. Consistent with the mouse data, we find that NFIB is overexpressed in almost all tested human metastatic high-grade neuroendocrine lung tumors, warranting further assessment of NFIB as a tumor progression marker in a clinical setting.


Subject(s)
Biomarkers, Tumor/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , NFI Transcription Factors/metabolism , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Animals , Cadherins/metabolism , Cell Proliferation/physiology , Disease Models, Animal , Disease Progression , Gene Expression Regulation, Neoplastic/physiology , Humans , Mice , Neoplasm Metastasis/pathology , Proto-Oncogene Proteins c-myc/metabolism , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism
6.
EMBO Mol Med ; 6(2): 212-25, 2014 02.
Article in English | MEDLINE | ID: mdl-24401838

ABSTRACT

Human cancers modeled in Genetically Engineered Mouse Models (GEMMs) can provide important mechanistic insights into the molecular basis of tumor development and enable testing of new intervention strategies. The inherent complexity of these models, with often multiple modified tumor suppressor genes and oncogenes, has hampered their use as preclinical models for validating cancer genes and drug targets. In our newly developed approach for the fast generation of tumor cohorts we have overcome this obstacle, as exemplified for three GEMMs; two lung cancer models and one mesothelioma model. Three elements are central for this system; (i) The efficient derivation of authentic Embryonic Stem Cells (ESCs) from established GEMMs, (ii) the routine introduction of transgenes of choice in these GEMM-ESCs by Flp recombinase-mediated integration and (iii) the direct use of the chimeric animals in tumor cohorts. By applying stringent quality controls, the GEMM-ESC approach proofs to be a reliable and effective method to speed up cancer gene assessment and target validation. As proof-of-principle, we demonstrate that MycL1 is a key driver gene in Small Cell Lung Cancer.


Subject(s)
Embryonic Stem Cells/cytology , Gene Transfer Techniques , Lung Neoplasms/pathology , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Proliferation , Cells, Cultured , Chimera , Clone Cells , DNA Nucleotidyltransferases/metabolism , Disease Models, Animal , Embryonic Stem Cells/metabolism , Genes, Reporter , Genomic Instability , Genotype , Germ Cells/metabolism , Humans , Luciferases/metabolism , Lung Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Oncogenes , Phenotype , Pluripotent Stem Cells/metabolism , Quality Control , Reproducibility of Results , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology
7.
Exp Biol Med (Maywood) ; 238(7): 798-802, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23788175

ABSTRACT

During the past years, evidence accumulated showing that glycine comprises anti-inflammatory activities. These effects occur, at least in part, via the activation of glycine-gated chloride channels (GlyR). Glycine is one of the major structural units of collagen, making up about 30% of the amino acids. This study aims to investigate the anti-inflammatory potential of collagen hydrolysate (CH) using the zymosan-induced ear-skin inflammation mouse model. After oral intake of 12.5, 25 or 50 mg CH the plasma levels of glycine increased in a concentration-dependent manner. CH was able to counteract zymosan-induced ear-skin inflammation locally (ear swelling) as well as systemically (IL-6 production by lipopolysaccharide (LPS)-stimulated whole blood cells). The LPS-stimulated IL-6 production in whole blood correlated positively with the ear swelling response. This correlation was abolished by strychnine (a glycine receptor antagonist), indicating the involvement of GlyR. Collectively, these data show that CH is able to modulate inflammatory responses both locally as well as systemically. This effect might be constituted by inhibiting pro-inflammatory cytokine production via GlyR.


Subject(s)
Collagen/pharmacology , Inflammation/pathology , Protein Hydrolysates/pharmacology , Zymosan/antagonists & inhibitors , Animals , Chromatography, High Pressure Liquid , Ear/pathology , Glycine/blood , Hydrolysis/drug effects , Inflammation/blood , Inflammation/chemically induced , Interleukin-6/blood , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred BALB C , Skin/drug effects , Skin/pathology , Zymosan/pharmacology
8.
Nature ; 449(7165): 1003-7, 2007 Oct 25.
Article in English | MEDLINE | ID: mdl-17934449

ABSTRACT

The intestinal epithelium is the most rapidly self-renewing tissue in adult mammals. It is currently believed that four to six crypt stem cells reside at the +4 position immediately above the Paneth cells in the small intestine; colon stem cells remain undefined. Lgr5 (leucine-rich-repeat-containing G-protein-coupled receptor 5, also known as Gpr49) was selected from a panel of intestinal Wnt target genes for its restricted crypt expression. Here, using two knock-in alleles, we reveal exclusive expression of Lgr5 in cycling columnar cells at the crypt base. In addition, Lgr5 was expressed in rare cells in several other tissues. Using an inducible Cre knock-in allele and the Rosa26-lacZ reporter strain, lineage-tracing experiments were performed in adult mice. The Lgr5-positive crypt base columnar cell generated all epithelial lineages over a 60-day period, suggesting that it represents the stem cell of the small intestine and colon. The expression pattern of Lgr5 suggests that it marks stem cells in multiple adult tissues and cancers.


Subject(s)
Colon/cytology , Intestine, Small/cytology , Receptors, G-Protein-Coupled/metabolism , Stem Cells/metabolism , Alleles , Animals , Biomarkers , Cell Line, Tumor , Gene Expression Profiling , Genes, Reporter , Humans , Mice , Paneth Cells/metabolism , Receptors, G-Protein-Coupled/genetics
9.
Nature ; 435(7044): 959-63, 2005 Jun 16.
Article in English | MEDLINE | ID: mdl-15959515

ABSTRACT

The self-renewing epithelium of the small intestine is ordered into stem/progenitor crypt compartments and differentiated villus compartments. Recent evidence indicates that the Wnt cascade is the dominant force in controlling cell fate along the crypt-villus axis. Here we show a rapid, massive conversion of proliferative crypt cells into post-mitotic goblet cells after conditional removal of the common Notch pathway transcription factor CSL/RBP-J. We obtained a similar phenotype by blocking the Notch cascade with a gamma-secretase inhibitor. The inhibitor also induced goblet cell differentiation in adenomas in mice carrying a mutation of the Apc tumour suppressor gene. Thus, maintenance of undifferentiated, proliferative cells in crypts and adenomas requires the concerted activation of the Notch and Wnt cascades. Our data indicate that gamma-secretase inhibitors, developed for Alzheimer's disease, might be of therapeutic benefit in colorectal neoplastic disease.


Subject(s)
Adenoma/pathology , Cell Proliferation/drug effects , Endopeptidases/metabolism , Goblet Cells/cytology , Intestine, Small/cytology , Membrane Proteins/antagonists & inhibitors , Protease Inhibitors/pharmacology , Adenoma/enzymology , Adenoma/genetics , Adenoma/metabolism , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases , Cell Differentiation/drug effects , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dibenzazepines/pharmacology , Female , Genes, APC , Goblet Cells/drug effects , Goblet Cells/metabolism , Goblet Cells/pathology , Immunoglobulin J Recombination Signal Sequence-Binding Protein , Intestine, Small/drug effects , Intestine, Small/enzymology , Intestine, Small/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phenotype , Receptors, Notch , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...