Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 15(6)2023 Mar 20.
Article in English | MEDLINE | ID: mdl-36980752

ABSTRACT

Kirsten rat sarcoma virus (KRAS)-mutant cancers are frequent, metastatic, lethal, and largely undruggable. While interleukin (IL)-1ß and nuclear factor (NF)-κB inhibition hold promise against cancer, untargeted treatments are not effective. Here, we show that human KRAS-mutant cancers are addicted to IL-1ß via inflammatory versican signaling to macrophage inhibitor of NF-κB kinase (IKK) ß. Human pan-cancer and experimental NF-κB reporter, transcriptome, and proteome screens reveal that KRAS-mutant tumors trigger macrophage IKKß activation and IL-1ß release via secretory versican. Tumor-specific versican silencing and macrophage-restricted IKKß deletion prevents myeloid NF-κB activation and metastasis. Versican and IKKß are mutually addicted and/or overexpressed in human cancers and possess diagnostic and prognostic power. Non-oncogene KRAS/IL-1ß addiction is abolished by IL-1ß and TLR1/2 inhibition, indicating cardinal and actionable roles for versican and IKKß in metastasis.

2.
EMBO Mol Med ; 14(2): e13631, 2022 02 07.
Article in English | MEDLINE | ID: mdl-34898002

ABSTRACT

Malignant pleural mesothelioma (MPM) arises from mesothelial cells lining the pleural cavity of asbestos-exposed individuals and rapidly leads to death. MPM harbors loss-of-function mutations in BAP1, NF2, CDKN2A, and TP53, but isolated deletion of these genes alone in mice does not cause MPM and mouse models of the disease are sparse. Here, we show that a proportion of human MPM harbor point mutations, copy number alterations, and overexpression of KRAS with or without TP53 changes. These are likely pathogenic, since ectopic expression of mutant KRASG12D in the pleural mesothelium of conditional mice causes epithelioid MPM and cooperates with TP53 deletion to drive a more aggressive disease form with biphasic features and pleural effusions. Murine MPM cell lines derived from these tumors carry the initiating KRASG12D lesions, secondary Bap1 alterations, and human MPM-like gene expression profiles. Moreover, they are transplantable and actionable by KRAS inhibition. Our results indicate that KRAS alterations alone or in accomplice with TP53 alterations likely play an important and underestimated role in a proportion of patients with MPM, which warrants further exploration.


Subject(s)
Lung Neoplasms , Mesothelioma, Malignant , Mesothelioma , Pleural Neoplasms , Proto-Oncogene Proteins p21(ras) , Animals , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mesothelioma/genetics , Mesothelioma/pathology , Mesothelioma, Malignant/genetics , Mesothelioma, Malignant/pathology , Mice , Pleural Neoplasms/genetics , Pleural Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
3.
Adv Ther (Weinh) ; 3(7)2020 Jul.
Article in English | MEDLINE | ID: mdl-33884290

ABSTRACT

Nanoparticle-based targeted drug delivery holds promise for treatment of cancers. However, most approaches fail to be translated into clinical success due to ineffective tumor targeting in vivo. Here, the delivery potential of mesoporous silica nanoparticles (MSN) functionalized with targeting ligands for EGFR and CCR2 is explored in lung tumors. The addition of active targeting ligands on MSNs enhances their uptake in vitro but fails to promote specific delivery to tumors in vivo, when administered systemically via the blood or locally to the lung into immunocompetent murine lung cancer models. Ineffective tumor targeting is due to efficient clearance of the MSNs by the phagocytic cells of the liver, spleen, and lung. These limitations, however, are successfully overcome using a novel organ-restricted vascular delivery (ORVD) approach. ORVD in isolated and perfused mouse lungs of Kras-mutant mice enables effective nanoparticle extravasation from the tumor vasculature into the core of solid lung tumors. In this study, ORVD promotes tumor cell-specific uptake of nanoparticles at cellular resolution independent of their functionalization with targeting ligands. Organ-restricted vascular delivery thus opens new avenues for optimized nanoparticles for lung cancer therapy and may have broad applications for other vascularized tumor types.

4.
Elife ; 82019 05 29.
Article in English | MEDLINE | ID: mdl-31140976

ABSTRACT

Lung cancer and chronic lung diseases impose major disease burdens worldwide and are caused by inhaled noxious agents including tobacco smoke. The cellular origins of environmental-induced lung tumors and of the dysfunctional airway and alveolar epithelial turnover observed with chronic lung diseases are unknown. To address this, we combined mouse models of genetic labeling and ablation of airway (club) and alveolar cells with exposure to environmental noxious and carcinogenic agents. Club cells are shown to survive KRAS mutations and to form lung tumors after tobacco carcinogen exposure. Increasing numbers of club cells are found in the alveoli with aging and after lung injury, but go undetected since they express alveolar proteins. Ablation of club cells prevents chemical lung tumors and causes alveolar destruction in adult mice. Hence club cells are important in alveolar maintenance and carcinogenesis and may be a therapeutic target against premalignancy and chronic lung disease.


Subject(s)
Adenocarcinoma of Lung/pathology , Carcinogens/metabolism , Environmental Exposure , Epithelial Cells/pathology , Epithelial Cells/physiology , Animals , Cell Proliferation , Cell Survival , Disease Models, Animal , Epithelial Cells/drug effects , Mice , Pulmonary Alveoli/cytology , Respiratory Mucosa/cytology , Tobacco Smoking/adverse effects
5.
Cancer Res ; 78(11): 2939-2951, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29588349

ABSTRACT

Although oncogenic activation of NFκB has been identified in various tumors, the NFκB-activating kinases (inhibitor of NFκB kinases, IKK) responsible for this are elusive. In this study, we determined the role of IKKα and IKKß in KRAS-mutant lung adenocarcinomas induced by the carcinogen urethane and by respiratory epithelial expression of oncogenic KRASG12D Using NFκB reporter mice and conditional deletions of IKKα and IKKß, we identified two distinct early and late activation phases of NFκB during chemical and genetic lung adenocarcinoma development, which were characterized by nuclear translocation of RelB, IκBß, and IKKα in tumor-initiated cells. IKKα was a cardinal tumor promoter in chemical and genetic KRAS-mutant lung adenocarcinoma, and respiratory epithelial IKKα-deficient mice were markedly protected from the disease. IKKα specifically cooperated with mutant KRAS for tumor induction in a cell-autonomous fashion, providing mutant cells with a survival advantage in vitro and in vivo IKKα was highly expressed in human lung adenocarcinoma, and a heat shock protein 90 inhibitor that blocks IKK function delivered superior effects against KRAS-mutant lung adenocarcinoma compared with a specific IKKß inhibitor. These results demonstrate an actionable requirement for IKKα in KRAS-mutant lung adenocarcinoma, marking the kinase as a therapeutic target against this disease.Significance: These findings report a novel requirement for IKKα in mutant KRAS lung tumor formation, with potential therapeutic applications. Cancer Res; 78(11); 2939-51. ©2018 AACR.


Subject(s)
Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , I-kappa B Kinase/genetics , Lung Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , A549 Cells , Animals , Cell Line , Cell Line, Tumor , Disease Progression , HEK293 Cells , Humans , Lung Neoplasms/genetics , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , NF-kappaB-Inducing Kinase
6.
Nat Commun ; 9(1): 672, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29445180

ABSTRACT

Malignant pleural effusion (MPE) is a frequent metastatic manifestation of human cancers. While we previously identified KRAS mutations as molecular culprits of MPE formation, the underlying mechanism remained unknown. Here, we determine that non-canonical IKKα-RelB pathway activation of KRAS-mutant tumor cells mediates MPE development and this is fueled by host-provided interleukin IL-1ß. Indeed, IKKα is required for the MPE-competence of KRAS-mutant tumor cells by activating non-canonical NF-κB signaling. IL-1ß fuels addiction of mutant KRAS to IKKα resulting in increased CXCL1 secretion that fosters MPE-associated inflammation. Importantly, IL-1ß-mediated NF-κB induction in KRAS-mutant tumor cells, as well as their resulting MPE-competence, can only be blocked by co-inhibition of both KRAS and IKKα, a strategy that overcomes drug resistance to individual treatments. Hence we show that mutant KRAS facilitates IKKα-mediated responsiveness of tumor cells to host IL-1ß, thereby establishing a host-to-tumor signaling circuit that culminates in inflammatory MPE development and drug resistance.


Subject(s)
Genes, ras , Interleukin-1beta/metabolism , Myeloid Cells/metabolism , NF-kappa B/metabolism , Pleural Effusion, Malignant/metabolism , Animals , Cell Line, Tumor , Chemokine CXCL1/metabolism , Female , Humans , I-kappa B Kinase/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation , Receptors, Interleukin-1/metabolism
7.
Nat Commun ; 8: 15205, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28508873

ABSTRACT

Malignant pleural effusion (MPE) is the lethal consequence of various human cancers metastatic to the pleural cavity. However, the mechanisms responsible for the development of MPE are still obscure. Here we show that mutant KRAS is important for MPE induction in mice. Pleural disseminated, mutant KRAS bearing tumour cells upregulate and systemically release chemokine ligand 2 (CCL2) into the bloodstream to mobilize myeloid cells from the host bone marrow to the pleural space via the spleen. These cells promote MPE formation, as indicated by splenectomy and splenocyte restoration experiments. In addition, KRAS mutations are frequently detected in human MPE and cell lines isolated thereof, but are often lost during automated analyses, as indicated by manual versus automated examination of Sanger sequencing traces. Finally, the novel KRAS inhibitor deltarasin and a monoclonal antibody directed against CCL2 are equally effective against an experimental mouse model of MPE, a result that holds promise for future efficient therapies against the human condition.


Subject(s)
Adenocarcinoma/genetics , Antineoplastic Agents/pharmacology , Lung Neoplasms/genetics , Myeloid Cells/pathology , Pleural Effusion, Malignant/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Antineoplastic Agents/therapeutic use , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Cell Line, Tumor , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/metabolism , Chickens , Chorioallantoic Membrane , Female , HEK293 Cells , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Pleural Cavity/cytology , Pleural Cavity/pathology , Pleural Effusion, Malignant/drug therapy , Pleural Effusion, Malignant/pathology , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/metabolism , RNA, Small Interfering/metabolism , Spleen/cytology , Spleen/pathology , Up-Regulation , Xenograft Model Antitumor Assays
8.
Neuropharmacology ; 121: 140-157, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28461162

ABSTRACT

Neurotrophic factors are among the most promising treatments aiming at slowing or stopping and even reversing Parkinson's disease (PD). However, in most cases, they cannot readily cross the human blood-brain-barrier (BBB). Herein, we propose as a therapeutic for PD the small molecule 17-beta-spiro-[5-androsten-17,2'-oxiran]-3beta-ol (BNN-20), a synthetic analogue of DHEA, which crosses the BBB and is deprived of endocrine side-effects. Using the "weaver" mouse, a genetic model of PD, which exhibits progressive dopaminergic neurodegeneration in the Substantia Nigra (SN), we have shown that long-term administration (P1-P21) of BNN-20 almost fully protected the dopaminergic neurons and their terminals, via i) a strong anti-apoptotic effect, probably mediated through the Tropomyosin receptor kinase B (TrkB) neurotrophin receptor's PI3K-Akt-NF-κB signaling pathway, ii) by exerting an efficient antioxidant effect, iii) by inducing significant anti-inflammatory activity and iv) by restoring Brain-Derived Neurotrophic Factor (BDNF) levels. By intercrossing "weaver" with NGL mice (dual GFP/luciferase-NF-κΒ reporter mice, NF-κΒ.GFP.Luc), we obtained Weaver/NGL mice that express the NF-κB reporter in all somatic cells. Acute BNN-20 administration to Weaver/NGL mice induced a strong NF-κB-dependent transcriptional response in the brain as detected by bioluminescence imaging, which was abolished by co-administration of the TrkB inhibitor ANA-12. This indicates that BNN-20 exerts its beneficial action (at least in part) through the TrkB-PI3K-Akt-NF-κB signaling pathway. These results could be of clinical relevance, as they suggest BNN-20 as an important neuroprotective agent acting through the TrkB neurotrophin receptor pathway, mimicking the action of the endogenous neurotrophin BDNF. Thus BNN-20 could be proposed for treatment of PD.


Subject(s)
Dehydroepiandrosterone/analogs & derivatives , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Mesencephalon/cytology , Receptor, trkB/metabolism , Adjuvants, Immunologic/pharmacology , Animals , Animals, Newborn , Antigens, CD1/metabolism , Azepines/pharmacology , Benzamides/pharmacology , CHO Cells , Cricetulus , Dehydroepiandrosterone/pharmacology , Dopamine Plasma Membrane Transport Proteins/metabolism , Female , Male , Mesencephalon/drug effects , Mesencephalon/metabolism , Mice , Mice, Neurologic Mutants , Models, Genetic , Signal Transduction/drug effects , Signal Transduction/physiology , Tubulin/metabolism , Tyrosine 3-Monooxygenase/metabolism
9.
EMBO Mol Med ; 9(5): 672-686, 2017 05.
Article in English | MEDLINE | ID: mdl-28341702

ABSTRACT

The lungs are frequently affected by cancer metastasis. Although NRAS mutations have been associated with metastatic potential, their exact role in lung homing is incompletely understood. We cross-examined the genotype of various tumor cells with their ability for automatic pulmonary dissemination, modulated NRAS expression using RNA interference and NRAS overexpression, identified NRAS signaling partners by microarray, and validated them using Cxcr1- and Cxcr2-deficient mice. Mouse models of spontaneous lung metastasis revealed that mutant or overexpressed NRAS promotes lung colonization by regulating interleukin-8-related chemokine expression, thereby initiating interactions between tumor cells, the pulmonary vasculature, and myeloid cells. Our results support a model where NRAS-mutant, chemokine-expressing circulating tumor cells target the CXCR1-expressing lung vasculature and recruit CXCR2-expressing myeloid cells to initiate metastasis. We further describe a clinically relevant approach to prevent NRAS-driven pulmonary metastasis by inhibiting chemokine signaling. In conclusion, NRAS promotes the colonization of the lungs by various tumor types in mouse models. IL-8-related chemokines, NRAS signaling partners in this process, may constitute an important therapeutic target against pulmonary involvement by cancers of other organs.


Subject(s)
GTP Phosphohydrolases/genetics , Lung Neoplasms/blood supply , Lung Neoplasms/secondary , Lung/blood supply , Membrane Proteins/genetics , Up-Regulation , Animals , Cell Line, Tumor , GTP Phosphohydrolases/immunology , Gene Expression Regulation, Neoplastic , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-8/immunology , Lung/immunology , Lung/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Membrane Proteins/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Monomeric GTP-Binding Proteins , Mutation , Signal Transduction
10.
PLoS One ; 10(7): e0132527, 2015.
Article in English | MEDLINE | ID: mdl-26147201

ABSTRACT

Nuclear factor (NF)-κB signalling is required for lung adenocarcinoma development in mice, and both of its subunits RelA and RelB were independently reported to be highly expressed in human non-small cell lung cancer (NSCLC). To comprehensively examine NF-κB expression in NSCLC, we analyzed serial sections of primary tumor samples from 77 well-documented patients (36 adenocarcinomas, 40 squamous cell carcinomas and 3 large cell carcinomas) for immunoreactivity of RelA, RelB, P50, and P52/P100. Tumor and intratumoral stroma areas were discriminated based on proliferating cell nuclear antigen immunoreactivity and inflammatory infiltration was assessed in intratumoral stroma areas. NF-κB immunoreactivity was quantified by intensity, extent, and nuclear localization and was cross-examined with tumor cell proliferation, inflammatory infiltration, and clinical-pathologic data. We found that the expression of the different NF-κB subunits was not concordant, warranting our integral approach. Overall, RelA, RelB, and P50 were expressed at higher levels compared with P52/P100. However, RelA and P50 were predominantly expressed in intratumoral stroma, but RelB in tumor cells. Importantly, tumor area RelA expression was correlated with the intensity of inflammatory infiltration, whereas RelB expression was identified in proliferating tumor cells. Using multiple logistic regression, we identified that tumor RelB expression was an independent predictor of lymph node metastasis, and tumor P50 was an independent predictor of TNM6 stage IIB or higher, whereas tumor RelA was an independent predictor of inflammatory infiltration. We conclude that pathologic studies of NF-κB expression in cancer should include multiple pathway components. Utilizing such an approach, we identified intriguing associations between distinct NF-κB subunits and clinical and pathologic features of NSCLC.


Subject(s)
Adenocarcinoma/metabolism , Carcinoma, Large Cell/metabolism , Carcinoma, Squamous Cell/metabolism , Lung Neoplasms/metabolism , Lung/metabolism , NF-kappa B/metabolism , Adenocarcinoma/pathology , Aged , Aged, 80 and over , Animals , Carcinoma, Large Cell/pathology , Carcinoma, Squamous Cell/pathology , Cell Nucleus/metabolism , Disease Models, Animal , Female , Humans , Lung/pathology , Lung Neoplasms/pathology , Male , Mice , Middle Aged , NF-kappa B p50 Subunit/metabolism , NF-kappa B p52 Subunit/metabolism , Transcription Factor RelA/metabolism , Transcription Factor RelB/metabolism
11.
J Clin Invest ; 125(6): 2317-34, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25915587

ABSTRACT

Mast cells (MCs) have been identified in various tumors; however, the role of these cells in tumorigenesis remains controversial. Here, we quantified MCs in human and murine malignant pleural effusions (MPEs) and evaluated the fate and function of these cells in MPE development. Evaluation of murine MPE-competent lung and colon adenocarcinomas revealed that these tumors actively attract and subsequently degranulate MCs in the pleural space by elaborating CCL2 and osteopontin. MCs were required for effusion development, as MPEs did not form in mice lacking MCs, and pleural infusion of MCs with MPE-incompetent cells promoted MPE formation. Once homed to the pleural space, MCs released tryptase AB1 and IL-1ß, which in turn induced pleural vasculature leakiness and triggered NF-κB activation in pleural tumor cells, thereby fostering pleural fluid accumulation and tumor growth. Evaluation of human effusions revealed that MCs are elevated in MPEs compared with benign effusions. Moreover, MC abundance correlated with MPE formation in a human cancer cell-induced effusion model. Treatment of mice with the c-KIT inhibitor imatinib mesylate limited effusion precipitation by mouse and human adenocarcinoma cells. Together, the results of this study indicate that MCs are required for MPE formation and suggest that MC-dependent effusion formation is therapeutically addressable.


Subject(s)
Mast Cells/metabolism , Pleural Effusion, Malignant/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Benzamides/pharmacology , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Humans , Imatinib Mesylate , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lung Neoplasms/diet therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mast Cells/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Piperazines/pharmacology , Pleural Cavity/metabolism , Pleural Cavity/pathology , Pleural Effusion, Malignant/drug therapy , Pleural Effusion, Malignant/genetics , Pleural Effusion, Malignant/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Pyrimidines/pharmacology , Tryptases/genetics , Tryptases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL