Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
Add more filters











Publication year range
2.
Oncoimmunology ; 10(1): 1885778, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33643696

ABSTRACT

Non-small cell lung cancers (NSCLCs) establish a highly immunosuppressive tumor microenvironment supporting cancer growth. To interfere with cancer-mediated immunosuppression, selective immune-checkpoint inhibitors (ICIs) have been approved as a standard-of-care treatment for NSCLCs. However, the majority of patients poorly respond to ICI-based immunotherapies. Oncolytic viruses are amongst the many promising immunomodulatory treatments tested as standalone therapy or in combination with ICIs to improve therapeutic outcome. Previously, we demonstrated the oncolytic and immunomodulatory efficacy of low-pathogenic influenza Aviruses (IAVs) against NSCLCs in immunocompetent transgenic mice with alung-specific overexpression of active Raf kinase (Raf-BxB). IAV infection not only resulted in significant primary virus-induced oncolysis, but also caused afunctional reversion of tumor-associated macrophages (TAMs) comprising additional anti-cancer activity. Here we show that NSCLCs as well as TAMs and cytotoxic immune cells overexpress IC molecules of the PD-L2/PD-1 and B7-H3 signaling axes. Thus, we aimed to combine oncolytic IAV-infection with ICIs to exploit the benefits of both anti-cancer approaches. Strikingly, IAV infection combined with the novel B7-H3 ICI led to increased levels of M1-polarized alveolar macrophages and increased lung infiltration by cytotoxic Tlymphocytes, which finally resulted in significantly improved oncolysis of about 80% of existing tumors. In contrast, application of clinically approved α-PD-1 IC antibodies alone or in combination with oncolytic IAV did not provide additional oncolytic or immunomodulatory efficacy. Thus, individualized therapy with synergistically acting oncolytic IAV and B7-H3 ICI might be an innovative future approach to target NSCLCs that are resistant to approved ICIs in patients.


Subject(s)
Influenza, Human , Lung Neoplasms , Oncolytic Viruses , Orthomyxoviridae , Animals , Humans , Immune Checkpoint Inhibitors , Lung , Lung Neoplasms/therapy , Mice , Oncolytic Viruses/genetics , Tumor Microenvironment
3.
Cancer Res ; 80(19): 4199-4211, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32816854

ABSTRACT

Although NF-κB is known to play a pivotal role in lung cancer, contributing to tumor growth, microenvironmental changes, and metastasis, the epigenetic regulation of NF-κB in tumor context is largely unknown. Here we report that the IKK2/NF-κB signaling pathway modulates metastasis-associated protein 2 (MTA2), a component of the nucleosome remodeling and deacetylase complex (NuRD). In triple transgenic mice, downregulation of IKK2 (Sftpc-cRaf-IKK2DN) in cRaf-induced tumors in alveolar epithelial type II cells restricted tumor formation, whereas activation of IKK2 (Sftpc-cRaf-IKK2CA) supported tumor growth; both effects were accompanied by altered expression of MTA2. Further studies employing genetic inhibition of MTA2 suggested that in primary tumor growth, independent of IKK2, MTA2/NuRD corepressor complex negatively regulates NF-κB signaling and tumor growth, whereas later dissociation of MTA2/NuRD complex from the promoter of NF-κB target genes and IKK2-dependent positive regulation of MTA2 leads to activation of NF-κB signaling, epithelial-mesenchymal transition, and lung tumor metastasis. These findings reveal a previously unrecognized biphasic role of MTA2 in IKK2/NF-κB-driven primary-to-metastatic lung tumor progression. Addressing the interaction between MTA2 and NF-κB would provide potential targets for intervention of tumor growth and metastasis. SIGNIFICANCE: These findings strongly suggest a prominent role of MTA2 in primary tumor growth, lung metastasis, and NF-κB signaling modulatory functions.


Subject(s)
Histone Deacetylases/metabolism , Lung Neoplasms/pathology , NF-kappa B/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Animals , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , HEK293 Cells , Histone Deacetylases/genetics , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Inflammation/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/genetics , Repressor Proteins/genetics , Signal Transduction , Trans-Activators/genetics , Tumor Microenvironment
4.
Oncotarget ; 9(5): 6518-6535, 2018 Jan 19.
Article in English | MEDLINE | ID: mdl-29464089

ABSTRACT

Identifying metastatic tumor growth at an early stage has been one of the biggest challenges in the treatment of lung cancer. By genetic lineage tracing approach in a conditional model of Non-Small Cell Lung Cancer (NSCLC) in mice, we demonstrate that cystic lesions represent an early stage of metastatic invasion. We generated a mouse model for NSCLC which incorporated a heritable DsRed fluorescent tag driven by the ubiquitous CAG promoter in the alveolar type II cells of the lung. We found early cystic lesions in a secondary organ (liver) that lacked the expression of bona fide lung makers namely Scgb1a1 and surfactant protein C Sftpc and were DsRed positive hence identifying lung as their source of origin. This demonstrates the significant potential of alveolar type II cells in orchestrating the process of metastasis, rendering it as one of the target cell types of the lung of therapeutic importance in human NSCLC.

5.
Sci Transl Med ; 9(416)2017 Nov 15.
Article in English | MEDLINE | ID: mdl-29141888

ABSTRACT

Dyspnea is a frequent, devastating, and poorly understood symptom of advanced lung cancer. In our cohort, among 519 patients who underwent a computed tomography scan for the diagnosis of lung cancer, 250 had a mean pulmonary artery diameter of >28 mm, indicating pulmonary hypertension (PH). In human lung cancer tissue, we consistently observed increased vascular remodeling and perivascular inflammatory cell accumulation (macrophages/lymphocytes). Vascular remodeling, PH, and perivascular inflammatory cell accumulation were mimicked in three mouse models of lung cancer (LLC1, KRasLA2 , and cRaf-BxB). In contrast, NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ immunodeficient xenograft and dominant-negative IKK2 mutant triple transgenic (Sftpc-rtTA/Tet-O-Ikk2DN) mice did not develop PH. Coculturing human lung cancer cells with macrophages and lymphocytes strongly up-regulated cytokine release, provoking enhanced migration, apoptosis resistance, and phosphodiesterase 5 (PDE5)-mediated up-regulation of human lung vascular cells, which are typical features of PH. The PDE5 inhibitor sildenafil largely suppressed PH in the LLC1 model. We conclude that lung cancer-associated PH represents a distinct PH category; targeting inflammation in the microenvironment and PDE5 offers a potential therapeutic option.


Subject(s)
Dyspnea/physiopathology , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/physiopathology , Lung Neoplasms/immunology , Lung Neoplasms/physiopathology , Animals , Apoptosis/physiology , Cell Line, Tumor , Cells, Cultured , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Dendritic Cells , Dyspnea/immunology , Echocardiography , Humans , Hypertension, Pulmonary/etiology , Immunohistochemistry , In Vitro Techniques , Inflammation/immunology , Inflammation/physiopathology , Lung Neoplasms/complications , Macrophages/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , T-Lymphocytes/metabolism
6.
EMBO Mol Med ; 8(12): 1380-1389, 2016 12.
Article in English | MEDLINE | ID: mdl-27821429

ABSTRACT

Lung cancer (LC) is the leading cause of cancer-related deaths worldwide. Early LC diagnosis is crucial to reduce the high case fatality rate of this disease. In this case-control study, we developed an accurate LC diagnosis test using retrospectively collected formalin-fixed paraffin-embedded (FFPE) human lung tissues and prospectively collected exhaled breath condensates (EBCs). Following international guidelines for diagnostic methods with clinical application, reproducible standard operating procedures (SOP) were established for every step comprising our LC diagnosis method. We analyzed the expression of distinct mRNAs expressed from GATA6 and NKX2-1, key regulators of lung development. The Em/Ad expression ratios of GATA6 and NKX2-1 detected in EBCs were combined using linear kernel support vector machines (SVM) into the LC score, which can be used for LC detection. LC score-based diagnosis achieved a high performance in an independent validation cohort. We propose our method as a non-invasive, accurate, and low-price option to complement the success of computed tomography imaging (CT) and chest X-ray (CXR) for LC diagnosis.


Subject(s)
Breath Tests/methods , GATA6 Transcription Factor/analysis , Lung Neoplasms/diagnosis , Nuclear Proteins/analysis , Transcription Factors/analysis , Case-Control Studies , Decision Support Techniques , Humans , Lung Neoplasms/pathology , Prospective Studies , Protein Isoforms/analysis , Retrospective Studies , Thyroid Nuclear Factor 1
7.
Nature ; 528(7583): 570-4, 2015 Dec 24.
Article in English | MEDLINE | ID: mdl-26675719

ABSTRACT

Variant rs351855-G/A is a commonly occurring single-nucleotide polymorphism of coding regions in exon 9 of the fibroblast growth factor receptor FGFR4 (CD334) gene (c.1162G>A). It results in an amino-acid change at codon 388 from glycine to arginine (p.Gly388Arg) in the transmembrane domain of the receptor. Despite compelling genetic evidence for the association of this common variant with cancers of the bone, breast, colon, prostate, skin, lung, head and neck, as well as soft-tissue sarcomas and non-Hodgkin lymphoma, the underlying biological mechanism has remained elusive. Here we show that substitution of the conserved glycine 388 residue to a charged arginine residue alters the transmembrane spanning segment and exposes a membrane-proximal cytoplasmic signal transducer and activator of transcription 3 (STAT3) binding site Y(390)-(P)XXQ(393). We demonstrate that such membrane-proximal STAT3 binding motifs in the germline of type I membrane receptors enhance STAT3 tyrosine phosphorylation by recruiting STAT3 proteins to the inner cell membrane. Remarkably, such germline variants frequently co-localize with somatic mutations in the Catalogue of Somatic Mutations in Cancer (COSMIC) database. Using Fgfr4 single nucleotide polymorphism knock-in mice and transgenic mouse models for breast and lung cancers, we validate the enhanced STAT3 signalling induced by the FGFR4 Arg388-variant in vivo. Thus, our findings elucidate the molecular mechanism behind the genetic association of rs351855 with accelerated cancer progression and suggest that germline variants of cell-surface molecules that recruit STAT3 to the inner cell membrane are a significant risk for cancer prognosis and disease progression.


Subject(s)
Cell Membrane/metabolism , Germ-Line Mutation , Receptor, Fibroblast Growth Factor, Type 4/genetics , Receptor, Fibroblast Growth Factor, Type 4/metabolism , STAT3 Transcription Factor/metabolism , Amino Acid Motifs/genetics , Amino Acid Sequence , Animals , Binding Sites/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line , Disease Models, Animal , Disease Progression , Exons/genetics , Female , Gene Knock-In Techniques , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Phosphorylation , Phosphotyrosine/metabolism , Polymorphism, Single Nucleotide/genetics , Receptor, Fibroblast Growth Factor, Type 4/chemistry , Signal Transduction
8.
Cell Mol Life Sci ; 72(20): 3983-98, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25975225

ABSTRACT

The family of RAF kinases transduces extracellular information to the nucleus, and their activation is crucial for cellular regulation on many levels, ranging from embryonic development to carcinogenesis. B-RAF and C-RAF modulate neurogenesis and neuritogenesis during chicken inner ear development. C-RAF deficiency in humans is associated with deafness in the rare genetic insulin-like growth factor 1 (IGF-1), Noonan and Leopard syndromes. In this study, we show that RAF kinases are expressed in the developing inner ear and in adult mouse cochlea. A homozygous C-Raf deletion in mice caused profound deafness with no evident cellular aberrations except for a remarkable reduction of the K(+) channel Kir4.1 expression, a trait that suffices as a cause of deafness. To explore the role of C-Raf in cellular protection and repair, heterozygous C-Raf (+/-) mice were exposed to noise. A reduced C-RAF level negatively affected hearing preservation in response to noise through mechanisms involving the activation of JNK and an exacerbated apoptotic response. Taken together, these results strongly support a role for C-RAF in hearing protection.


Subject(s)
Ear, Inner/metabolism , Hearing Loss/genetics , Noise , Proto-Oncogene Proteins c-raf/physiology , Animals , Apoptosis/genetics , Cochlea/metabolism , Ear, Inner/embryology , Female , Hearing Loss/metabolism , Male , Mice , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , Signal Transduction
9.
PLoS One ; 10(2): e0117794, 2015.
Article in English | MEDLINE | ID: mdl-25674792

ABSTRACT

Lung cancer is the most deadly type of cancer in humans, with non-small-cell lung cancer (NSCLC) being the most frequent and aggressive type of lung cancer showing high resistance to radiation and chemotherapy. Despite the outstanding progress made in anti-tumor therapy, discovering effective anti-tumor drugs is still a challenging task. Here we describe a new semisynthetic derivative of cucurbitacin B (DACE) as a potent inhibitor of NSCLC cell proliferation. DACE arrested the cell cycle of lung epithelial cells at the G2/M phase and induced cell apoptosis by interfering with EGFR activation and its downstream signaling, including AKT, ERK, and STAT3. Consistent with our in vitro studies, intraperitoneal application of DACE significantly suppressed the growth of mouse NSCLC that arises from type II alveolar pneumocytes due to constitutive expression of a human oncogenic c-RAF kinase (c-RAF-1-BxB) transgene in these cells. Taken together, these findings suggest that DACE is a promising lead compound for the development of an anti-lung-cancer drug.


Subject(s)
Antineoplastic Agents/pharmacology , Triterpenes/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Caspase 3/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytoskeleton/metabolism , Disease Models, Animal , ErbB Receptors/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, Transgenic , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Triterpenes/administration & dosage , Triterpenes/chemical synthesis , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays , raf Kinases/genetics , raf Kinases/metabolism
10.
J Biol Chem ; 289(39): 26804-26816, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25096573

ABSTRACT

Tumors are often greatly dependent on signaling cascades promoting cell growth or survival and may become hypersensitive to inactivation of key components within these signaling pathways. Ras and RAF mutations found in human cancer confer constitutive activity to these signaling molecules thereby converting them into an oncogenic state. RAF dimerization is required for normal Ras-dependent RAF activation and is required for the oncogenic potential of mutant RAFs. Here we describe a new mouse model for lung tumor development to investigate the role of B-RAF in oncogenic C-RAF-mediated adenoma initiation and growth. Conditional elimination of B-RAF in C-RAF BxB-expressing embryonic alveolar epithelial type II cells did not block adenoma formation. However, loss of B-RAF led to significantly reduced tumor growth. The diminished tumor growth upon B-RAF inactivation was due to reduced cell proliferation in absence of senescence and increased apoptosis. Furthermore, B-RAF elimination inhibited C-RAF BxB-mediated activation of the mitogenic cascade. In line with these data, mutation of Ser-621 in C-RAF BxB abrogated in vitro the dimerization with B-RAF and blocked the ability to activate the MAPK cascade. Taken together these data indicate that B-RAF is an important factor in oncogenic C-RAF-mediated tumorigenesis.


Subject(s)
Adenoma/enzymology , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/enzymology , Lung Neoplasms/enzymology , MAP Kinase Signaling System , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Pulmonary Alveoli/enzymology , Respiratory Mucosa/enzymology , Adenoma/genetics , Adenoma/pathology , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Epithelial Cells/pathology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Transgenic , Mutation , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-raf/genetics , Pulmonary Alveoli/pathology , Respiratory Mucosa/pathology
11.
J Biol Chem ; 288(49): 35237-52, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24158441

ABSTRACT

In metazoans, the highly conserved MAPK signaling pathway regulates cell fate decision. Aberrant activation of this pathway has been implicated in multiple human cancers and some developmental disorders. KSR1 functions as an essential scaffold that binds the individual components of the cascade and coordinates their assembly into multiprotein signaling platforms. The mechanism of KSR1 regulation is highly complex and not completely understood. In this study, we identified Tyr(728) as a novel regulatory phosphorylation site in KSR1. We show that Tyr(728) is phosphorylated by LCK, uncovering an additional and unexpected link between Src kinases and MAPK signaling. To understand how phosphorylation of Tyr(728) may regulate the role of KSR1 in signal transduction, we integrated structural modeling and biochemical studies. We demonstrate that Tyr(728) is involved in maintaining the conformation of the KSR1 kinase domain required for binding to MEK. It also affects phosphorylation and activation of MEK by RAF kinases and consequently influences cell proliferation. Moreover, our studies suggest that phosphorylation of Tyr(728) may affect the intrinsic kinase activity of KSR1. Together, we propose that phosphorylation of Tyr(728) may regulate the transition between the scaffolding and the catalytic function of KSR1 serving as a control point used to fine-tune cellular responses.


Subject(s)
Mitogen-Activated Protein Kinase Kinases/metabolism , Protein Kinases/chemistry , Protein Kinases/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Proliferation , Enzyme Activation , Humans , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , MAP Kinase Signaling System , Mice , Models, Molecular , Molecular Dynamics Simulation , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Protein Binding , Protein Conformation , Protein Kinases/genetics , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Tyrosine/chemistry
12.
PLoS One ; 8(3): e58259, 2013.
Article in English | MEDLINE | ID: mdl-23505473

ABSTRACT

This study focuses on the role of the kinase BRaf in postnatal brain development. Mice expressing truncated, non-functional BRaf in neural stem cell-derived brain tissue demonstrate alterations in the cerebellum, with decreased sizes and fuzzy borders of the glomeruli in the granule cell layer. In addition we observed reduced numbers and misplaced ectopic Purkinje cells that showed an altered structure of their dendritic arborizations in the hippocampus, while the overall cornus ammonis architecture appeared to be unchanged. In male mice lacking BRaf in the hippocampus the size of the granule cell layer was normal at postnatal day 12 (P12) but diminished at P21, as compared to control littermates. This defect was caused by a reduced ability of dentate gyrus progenitor cells to differentiate into NeuN positive granule cell neurons. In vitro cell culture of P0/P1 hippocampal cells revealed that BRaf deficient cells were impaired in their ability to form microtubule-associated protein 2 positive neurons. Together with the alterations in behaviour, such as autoaggression and loss of balance fitness, these observations indicate that in the absence of BRaf all neuronal cellular structures develop, but neuronal circuits in the cerebellum and hippocampus are partially disturbed besides impaired neuronal generation in both structures.


Subject(s)
Cell Differentiation/genetics , Cerebellum/metabolism , Hippocampus/metabolism , Neurons/cytology , Neurons/metabolism , Proto-Oncogene Proteins B-raf/genetics , Animals , Animals, Newborn , Behavior, Animal , Cerebellum/growth & development , Dentate Gyrus/growth & development , Dentate Gyrus/metabolism , Gene Deletion , Hippocampus/growth & development , Male , Mice , Mice, Transgenic , Phenotype
13.
Mol Cancer Res ; 11(2): 161-72, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23239811

ABSTRACT

Human lung cancer is a disease with high incidence and accounts for most cancer-related deaths in both men and women. Metastasis is a common event in non-small cell lung carcinoma (NSCLC), diminishing the survival chance of the patients with this type of tumor. It has been shown that MYC is involved in the development of metastasis from NSCLC, but the mechanism underlying this switch remained to be identified. Here, we focus on GATA4 as a MYC target in the development of metastasis with origin in lung adenocarcinoma, the most common type of NSCLC. Epigenetic alterations at the GATA4 promoter level were observed after MYC expression in lung adenocarcinoma in vivo and in vitro. Such alterations include site-specific demethylation that accompanies the displacement of the MYC-associated zinc finger protein (MAZ) from the GATA4 promoter, which leads to GATA4 expression. Histone modification analysis of the GATA4 promoter revealed a switch from repressive histone marks to active histone marks after MYC binding, which corresponds to active GATA4 expression. Our results thus identify a novel epigenetic mechanism by which MYC activates GATA4 leading to metastasis in lung adenocarcinoma, suggesting novel potential targets for the development of antimetastatic therapy.


Subject(s)
Adenocarcinoma/genetics , GATA4 Transcription Factor/genetics , Genes, myc , Lung Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Cell Adhesion/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , DNA Methylation , DNA-Binding Proteins/genetics , Epigenesis, Genetic , Female , GATA4 Transcription Factor/biosynthesis , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mucin-2/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins p21(ras) , Transcription Factors/genetics , ras Proteins/genetics
14.
Epigenetics Chromatin ; 5(1): 12, 2012 Aug 07.
Article in English | MEDLINE | ID: mdl-22870894

ABSTRACT

BACKGROUND: Gene-environment interactions are mediated by epigenetic mechanisms. Polycomb Group proteins constitute part of an epigenetic cellular transcriptional memory system that is subject to dynamic modulation during differentiation. Molecular insight in processes that control dynamic chromatin association and dissociation of Polycomb repressive complexes during and beyond development is limited. We recently showed that MK3 interacts with Polycomb repressive complex 1 (PRC1). The functional relevance of this interaction, however, remained poorly understood. MK3 is activated downstream of mitogen- and stress-activated protein kinases (M/SAPKs), all of which fulfill crucial roles during development. We here use activation of the immediate-early response gene ATF3, a bona fide PRC1 target gene, as a model to study how MK3 and its effector kinases MAPK/ERK and SAPK/P38 are involved in regulation of PRC1-dependent ATF3 transcription. RESULTS: Our current data show that mitogenic signaling through ERK, P38 and MK3 regulates ATF3 expression by PRC1/chromatin dissociation and epigenetic modulation. Mitogenic stimulation results in transient P38-dependent H3S28 phosphorylation and ERK-driven PRC1/chromatin dissociation at PRC1 targets. H3S28 phosphorylation by itself appears not sufficient to induce PRC1/chromatin dissociation, nor ATF3 transcription, as inhibition of MEK/ERK signaling blocks BMI1/chromatin dissociation and ATF3 expression, despite induced H3S28 phosphorylation. In addition, we establish that concomitant loss of local H3K27me3 promoter marking is not required for ATF3 activation. We identify pERK as a novel signaling-induced binding partner of PRC1, and provide evidence that MK3 controls ATF3 expression in cultured cells via negative regulatory feedback on M/SAPKs. Dramatically increased ectopic wing vein formation in the absence of Drosophila MK in a Drosophila ERK gain-of-function wing vein patterning model, supports the existence of MK-mediated negative feedback regulation on pERK. CONCLUSION: We here identify and characterize important actors in a PRC1-dependent epigenetic signal/response mechanism, some of which appear to be nonspecific global responses, whereas others provide modular specificity. Our findings provide novel insight into a Polycomb-mediated epigenetic mechanism that dynamically controls gene transcription and support a direct link between PRC1 and cellular responses to changes in the microenvironment.

15.
J Biol Chem ; 287(34): 28445-55, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22711539

ABSTRACT

Inhibitor of apoptosis (IAPs) proteins are characterized by the presence of evolutionarily conserved baculoviral inhibitor of apoptosis repeat (BIR) domains, predominantly known for their role in inhibiting caspases and, thereby, apoptosis. We have shown previously that multi-BIR domain-containing IAPs, cellular IAPs, and X-linked IAP can control tumor cell migration by directly regulating the protein stability of C-RAF kinase. Here, we extend our observations to a single BIR domain containing IAP family member melanoma-IAP (ML-IAP). We show that ML-IAP can directly bind to C-RAF and that ML-IAP depletion leads to an increase in C-RAF protein levels, MAPK activation, and cell migration in melanoma cells. Thus, our results unveil a thus far unknown role for ML-IAP in controlling C-RAF stability and cell migration.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Movement , Inhibitor of Apoptosis Proteins/metabolism , Melanoma/metabolism , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Line, Tumor , Enzyme Stability/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Inhibitor of Apoptosis Proteins/genetics , Melanoma/genetics , Melanoma/pathology , Neoplasm Proteins/genetics , Protein Binding/genetics , Proto-Oncogene Proteins c-raf/genetics
16.
J Biol Chem ; 287(27): 23128-40, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22605333

ABSTRACT

The maternally imprinted Ras-related tumor suppressor gene DiRas3 is lost or down-regulated in more than 60% of ovarian and breast cancers. The anti-tumorigenic effect of DiRas3 is achieved through several mechanisms, including inhibition of cell proliferation, motility, and invasion, as well as induction of apoptosis and autophagy. Re-expression of DiRas3 in cancer cells interferes with the signaling through Ras/MAPK and PI3K. Despite intensive research, the mode of interference of DiRas3 with the Ras/RAF/MEK/ERK signal transduction is still a matter of speculation. In this study, we show that DiRas3 associates with the H-Ras oncogene and that activation of H-Ras enforces this interaction. Furthermore, while associated with DiRas3, H-Ras is able to bind to its effector protein C-RAF. The resulting multimeric complex consisting of DiRas3, C-RAF, and active H-Ras is more stable than the two protein complexes H-Ras·C-RAF or H-Ras·DiRas3, respectively. The consequence of this complex formation is a DiRas3-mediated recruitment and anchorage of C-RAF to components of the membrane skeleton, suppression of C-RAF/B-RAF heterodimerization, and inhibition of C-RAF kinase activity.


Subject(s)
MAP Kinase Signaling System/physiology , Proto-Oncogene Proteins c-raf/chemistry , Proto-Oncogene Proteins c-raf/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , rho GTP-Binding Proteins/metabolism , Animals , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Cytoskeleton/metabolism , Dimerization , Genes, Tumor Suppressor/physiology , Humans , Multiprotein Complexes/metabolism , Prenylation/physiology , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , rho GTP-Binding Proteins/genetics
17.
Int J Cancer ; 131(12): 2808-19, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-22511343

ABSTRACT

A major obstacle in the successful treatment of cancer is the occurrence of chemoresistance. Cancer cells surviving chemotherapy and giving rise to a recurrence of the tumor are termed cancer stem cells and can be identified by elevated levels of certain stem cell markers. Eradication of this cell population is a priority objective in cancer therapy. Here, we report elevated levels of stem cell markers in MCF-7 mammospheres. Likewise, an upregulation of HER2 and its differential expression within individual cells of mammospheres was observed. Sorting for HER2(high) and HER2(low) cells revealed an upregulation of stem cell markers NANOG, OCT4 and SOX2 in the HER2(low) cell fraction. Accordingly, HER2(low) cells also showed reduced proliferation, ductal-like outgrowths and an increased number of colonies in matrigel. Xenografts from subcutaneously injected HER2(low) sorted cells exihibited earlier onset but slower growth of tumors and an increase in stem cell markers compared to tumors developed from the HER2(high) fraction. Treatment of mammospheres with salinomycin reduced the expression of SOX2 indicating a selective targeting of cancer stem cells. Trastuzumab however, did not reduce the expression of SOX2 in mammospheres. Furthermore, a combinatorial treatment of mammospheres with trastuzumab and salinomycin was superior to single treatment with each drug. Thus, targeting HER2 expressing tumors with anti-HER2 therapies will not necessarily eliminate cancer stem cells and may lead to a more aggressive cancer cell phenotype. Our study demonstrates efficient killing of both HER2 positive cells and cancer stem cells, hence opening a possibility for a new combinatorial treatment strategy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Receptor, ErbB-2/metabolism , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Base Sequence , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , DNA Primers , Female , Humans , Pyrans/administration & dosage , Pyrans/pharmacology , Real-Time Polymerase Chain Reaction , Trastuzumab
18.
Neoplasia ; 14(12): 1164-77, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23308049

ABSTRACT

Although progenitor cells of the conducting airway have been spatially localized and some insights have been gained regarding their molecular phenotype, relatively little is known about the mechanisms regulating their maintenance, activation, and differentiation. This study investigates the potential roles of E-cadherin in mouse Clara cells, as these cells were shown to represent the progenitor/stem cells of the conducting airways and have been implicated as the cell of origin of human non-small cell lung cancer. Postnatal inactivation of E-cadherin affected Clara cell differentiation and compromised airway regeneration under injury conditions. In steady-state adult lung, overexpression of the dominant negative E-cadherin led to an expansion of the bronchiolar stem cells and decreased differentiation concomitant with canonical Wnt signaling activation. Expansion of the bronchiolar stem cell pool was associated with an incessant proliferation of neuroepithelial body.associated Clara cells that ultimately gave rise to bronchiolar hyperplasia. Despite progressive hyperplasia, only a minority of the mice developed pulmonary solid tumors, suggesting that the loss of E-cadherin function leads to tumor formation when additional mutations are sustained. The present study reveals that E-cadherin plays a critical role in the regulation of proliferation and homeostasis of the epithelial cells lining the conducting airways.


Subject(s)
Bronchioles/metabolism , Bronchioles/pathology , Cadherins/metabolism , Lung Neoplasms/metabolism , Precancerous Conditions/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Stem Cells/metabolism , Animals , Cadherins/genetics , Cell Differentiation , Cell Proliferation , Homeostasis , Hyperplasia , Lung Neoplasms/pathology , Mice , Mice, Transgenic , Precancerous Conditions/pathology , Regeneration , Stem Cells/pathology , Uteroglobin/metabolism , Wnt Signaling Pathway
19.
PLoS One ; 6(12): e29093, 2011.
Article in English | MEDLINE | ID: mdl-22194995

ABSTRACT

Growth factor induced signaling cascades are key regulatory elements in tissue development, maintenance and regeneration. Perturbations of these cascades have severe consequences, leading to developmental disorders and neoplastic diseases. As a major function in signal transduction, activating mutations in RAF family kinases are the cause of human tumorigenesis, where B-RAF V600E has been identified as the prevalent mutant. In order to address the oncogenic function of B-RAF V600E, we have generated transgenic mice expressing the activated oncogene specifically in lung alveolar epithelial type II cells. Constitutive expression of B-RAF V600E caused abnormalities in alveolar epithelium formation that led to airspace enlargements. These lung lesions showed signs of tissue remodeling and were often associated with chronic inflammation and low incidence of lung tumors. The inflammatory cell infiltration did not precede the formation of the lung lesions but was rather accompanied with late tumor development. These data support a model where the continuous regenerative process initiated by oncogenic B-RAF-driven alveolar disruption provides a tumor-promoting environment associated with chronic inflammation.


Subject(s)
Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Cell Transformation, Neoplastic/pathology , Lung Neoplasms/pathology , Mutation/genetics , Proto-Oncogene Proteins B-raf/genetics , Airway Remodeling , Animals , Cell Death , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Collagen/metabolism , Epithelial-Mesenchymal Transition , Goblet Cells/metabolism , Goblet Cells/pathology , Humans , Hyperplasia , Immunohistochemistry , Inflammation/pathology , Lung Neoplasms/metabolism , Macrophages/metabolism , Mice , Mice, Transgenic , STAT3 Transcription Factor/metabolism , Signal Transduction
20.
Neoplasia ; 13(11): 1005-18, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22131876

ABSTRACT

Here we describe a novel conditional mouse lung tumor model for investigation of the pathogenesis of human lung cancer. On the basis of the frequent involvement of the Ras-RAF-MEK-ERK signaling pathway in human non-small cell lung carcinoma (NSCLC), we have explored the target cell availability, reversibility, and cell type specificity of transformation by oncogenic C-RAF. Targeting expression to alveolar type II cells or to Clara cells, the two likely precursors of human NSCLC, revealed differential tumorigenicity between these cells. Whereas expression of oncogenic C-RAF in alveolar type II cells readily induced multifocal macroscopic lung tumors independent of the developmental state, few tumors with type II pneumocytes features and incomplete penetrance were found when targeted to Clara cells. Induced tumors did not progress and were strictly dependent on the initiating oncogene. Deinduction of mice resulted in tumor regression due to autophagy rather than apoptosis. Induction of autophagic cell death in regressing lung tumors suggests the use of autophagy enhancers as a treatment choice for patients with NSCLC.


Subject(s)
Autophagy/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Transformation, Neoplastic/genetics , Lung Neoplasms/pathology , Proto-Oncogene Proteins c-raf/genetics , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/physiology , Gene Expression/physiology , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Mice , Mice, Transgenic , Transfection , Transplantation, Heterologous , Tumor Burden/genetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL