Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cancer Immunol Immunother ; 72(12): 4457-4470, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37796299

ABSTRACT

BACKGROUND: The inducible Kras/p53 lung adenocarcinoma mouse model, which faithfully recapitulates human disease, is routinely initiated by the intratracheal instillation of a virus-based Cre recombinase delivery system. Handling virus-based delivery systems requires elevated biosafety levels, e.g., biosafety level 2 (BSL-2). However, in experimental animal research facilities, following exposure to viral vectors in a BSL-2 environment, rodents may not be reclassified to BSL-1 according to standard practice, preventing access to small animal micro-computed tomography (micro-CT) scanners that are typically housed in general access areas such as BSL-1 rooms. Therefore, our goal was to adapt the protocol so that the Cre-induced KP mouse model could be handled under BSL-1 conditions during the entire procedure. RESULTS: The Kras-Lox-STOP-Lox-G12D/p53 flox/flox (KP)-based lung adenocarcinoma mouse model was activated by intratracheal instillation of either an adenoviral-based or a gutless, adeno-associated viral-based Cre delivery system. Tumor growth was monitored over time by micro-CT. We have successfully substituted the virus-based Cre delivery system with a commercially available, gutless, adeno-associated, Cre-expressing vector that allows the KP mouse model to be handled and imaged in a BSL-1 facility. By optimizing the anesthesia protocol and switching to a microscope-guided vector instillation procedure, productivity was increased and procedure-related complications were significantly reduced. In addition, repeated micro-CT analysis of individual animals allowed us to monitor tumor growth longitudinally, dramatically reducing the number of animals required per experiment. Finally, we documented the evolution of tumor volume for different doses, which revealed that individual tumor nodules induced by low-titer AAV-Cre transductions can be monitored over time by micro-CT. CONCLUSION: Modifications to the anesthesia and instillation protocols increased the productivity of the original KP protocol. In addition, the switch to a gutless, adeno-associated, Cre-expressing vector allowed longitudinal monitoring of tumor growth under BSL-1 conditions, significantly reducing the number of animals required for an experiment, in line with the 3R principles.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Mice , Animals , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Dependovirus/genetics , X-Ray Microtomography , Tumor Suppressor Protein p53 , Containment of Biohazards , Disease Models, Animal , Genetic Vectors/genetics
2.
Int J Mol Sci ; 24(2)2023 Jan 16.
Article in English | MEDLINE | ID: mdl-36675307

ABSTRACT

KRAS is one of the most commonly mutated genes, an event that leads to development of highly aggressive and resistant to any type of available therapy tumors. Mutated KRAS drives a complex network of lipid metabolic rearrangements to support the adaptation of cancer cells to harsh environmental conditions and ensure their survival. Because there has been only a little success in the continuous efforts of effectively targeting KRAS-driven tumors, it is of outmost importance to delineate the exact mechanisms of how they get rewired, leading to this distinctive phenotype. Therefore, the aim of this review is to summarize the available data acquired over the last years with regard to the lipid metabolic regulation of KRAS-driven tumors and elucidate their specific characteristics in an attempt to unravel novel therapeutic targets.


Subject(s)
Neoplasms , Proto-Oncogene Proteins p21(ras) , Humans , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Lipids , Mutation
3.
Int J Mol Sci ; 25(1)2023 Dec 23.
Article in English | MEDLINE | ID: mdl-38203404

ABSTRACT

Hematopoietic stem cell transplantation-associated thrombotic microangiopathy (HSCT-TMA) and graft-versus-host disease (GvHD) represent life-threatening syndromes after allogeneic hematopoietic stem cell transplantation (allo-HSCT). In both conditions, endothelial dysfunction is a common denominator, and development of relevant biomarkers is of high importance for both diagnosis and prognosis. Despite the fact that soluble urokinase plasminogen activator receptor (suPAR) and growth differentiation factor-15 (GDF-15) have been determined as endothelial injury indices in various clinical settings, their role in HSCT-related complications remains unexplored. In this context, we used immunoenzymatic methods to measure suPAR and GDF-15 levels in HSCT-TMA, acute and/or chronic GVHD, control HSCT recipients, and apparently healthy individuals of similar age and gender. We found considerably greater SuPAR and GDF-15 levels in HSCT-TMA and GVHD patients compared to allo-HSCT and healthy patients. Both GDF-15 and suPAR concentrations were linked to EASIX at day 100 and last follow-up. SuPAR was associated with creatinine and platelets at day 100 and last follow-up, while GDF-15 was associated only with platelets, suggesting that laboratory values do not drive EASIX. SuPAR, but not GDF-15, was related to soluble C5b-9 levels, a sign of increased HSCT-TMA risk. Our study shows for the first time that suPAR and GDF-15 indicate endothelial damage in allo-HSCT recipients. Rigorous validation of these biomarkers in many cohorts may provide utility for their usefulness in identifying and stratifying allo-HSCT recipients with endothelial cell impairment.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Thrombotic Microangiopathies , Adult , Humans , Receptors, Urokinase Plasminogen Activator , Growth Differentiation Factor 15 , Hematopoietic Stem Cell Transplantation/adverse effects , Graft vs Host Disease/etiology , Biomarkers
4.
J Exp Clin Cancer Res ; 41(1): 16, 2022 Jan 08.
Article in English | MEDLINE | ID: mdl-34998392

ABSTRACT

BACKGROUND: KRAS is the predominant oncogene mutated in pancreatic ductal adenocarcinoma (PDAC), the fourth cause of cancer-related deaths worldwide. Mutant KRAS-driven tumors are metabolically programmed to support their growth and survival, which can be used to identify metabolic vulnerabilities. In the present study, we aimed to understand the role of extracellularly derived fatty acids in KRAS-driven pancreatic cancer. METHODS: To assess the dependence of PDAC cells on extracellular fatty acids we employed delipidated serum or RNAi-mediated suppression of ACSL3 (to inhibit the activation and cellular retention of extracellular fatty acids) followed by cell proliferation assays, qPCR, apoptosis assays, immunoblots and fluorescence microscopy experiments. To assess autophagy in vivo, we employed the KrasG12D/+;p53flox/flox;Pdx1-CreERT2 (KPC) mice crossed with Acsl3 knockout mice, and to assess the efficacy of the combination therapy of ACSL3 and autophagy inhibition we used xenografted human cancer cell-derived tumors in immunocompromised mice. RESULTS: Here we show that depletion of extracellularly derived lipids either by serum lipid restriction or suppression of ACSL3, triggers autophagy, a process that protects PDAC cells from the reduction of bioenergetic intermediates. Combined extracellular lipid deprivation and autophagy inhibition exhibits anti-proliferative and pro-apoptotic effects against PDAC cell lines in vitro and promotes suppression of xenografted human pancreatic cancer cell-derived tumors in mice. Therefore, we propose lipid deprivation and autophagy blockade as a potential co-targeting strategy for PDAC treatment. CONCLUSIONS: Our work unravels a central role of extracellular lipid supply in ensuring fatty acid provision in cancer cells, unmasking a previously unappreciated metabolic vulnerability of PDAC cells.


Subject(s)
Autophagy/immunology , Lipid Metabolism/immunology , Pancreatic Neoplasms/therapy , Animals , Cell Proliferation , Disease Models, Animal , Humans , Mice , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms
5.
EMBO Mol Med ; 13(9): e13193, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34369083

ABSTRACT

KRAS oncoprotein is commonly mutated in human cancer, but effective therapies specifically targeting KRAS-driven tumors remain elusive. Here, we show that combined treatment with fibroblast growth factor receptor 1 (FGFR1) and polo-like kinase 1 (PLK1) inhibitors evoke synergistic cytotoxicity in KRAS-mutant tumor models in vitro and in vivo. Pharmacological and genetic suppression of FGFR1 and PLK1 synergizes to enhance anti-proliferative effects and cell death in KRAS-mutant lung and pancreatic but not colon nor KRAS wild-type cancer cells. Mechanistically, co-targeting FGFR1 and PLK1 upregulates reactive oxygen species (ROS), leading to oxidative stress-activated c-Jun N-terminal kinase (JNK)/p38 pathway and E2F1-induced apoptosis. We further delineate that autophagy protects from PLK1/FGFR1 inhibitor cytotoxicity and that antagonizing the compensation mechanism by clinically approved chloroquine fully realizes the therapeutic potential of PLK1 and FGFR1 targeting therapy, producing potent and durable responses in KRAS-mutant patient-derived xenografts and a genetically engineered mouse model of Kras-induced lung adenocarcinoma. These results suggest a previously unappreciated role for FGFR1 and PLK1 in the surveillance of metabolic stress and demonstrate a synergistic drug combination for treating KRAS-mutant cancer.


Subject(s)
Lung Neoplasms , Receptor, Fibroblast Growth Factor, Type 1 , Animals , Cell Cycle Proteins , Cell Line, Tumor , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mice , Mutation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins , Proto-Oncogene Proteins p21(ras)/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Polo-Like Kinase 1
6.
Cancers (Basel) ; 13(4)2021 Feb 14.
Article in English | MEDLINE | ID: mdl-33672917

ABSTRACT

Cytotoxic chemotherapy remains the only treatment option for most pancreatic ductal adenocarcinoma patients. Currently, the median overall survival of patients with advanced disease rarely exceeds 1 year. The complex network of pancreatic cancer composed of immune cells, endothelial cells, and cancer-associated fibroblasts confers intratumoral and intertumoral heterogeneity with distinct proliferative and metastatic propensity. This heterogeneity can explain why tumors do not behave uniformly and are able to escape therapy. The advance in technology of whole-genome sequencing has now provided the possibility of identifying every somatic mutation, copy-number change, and structural variant in a given cancer, giving rise to personalized targeted therapies. In this review, we provide an overview of the current and emerging treatment strategies in pancreatic cancer. By highlighting new paradigms in pancreatic ductal adenocarcinoma treatment, we hope to stimulate new thoughts for clinical trials aimed at improving patient outcomes.

7.
Nat Cell Biol ; 22(11): 1382-1395, 2020 11.
Article in English | MEDLINE | ID: mdl-33077911

ABSTRACT

Mutant KRAS modulates the metabolic plasticity of cancer cells to confer a growth advantage during hypoxia, but the molecular underpinnings are largely unknown. Using a lipidomic screen, we found that PLCγ1 is suppressed during hypoxia in KRAS-mutant human lung adenocarcinoma cancer cell lines. Suppression of PLCγ1 in hypoxia promotes a less oxidative cancer cell metabolism state, reduces the formation of mitochondrial reactive oxygen species and switches tumour bioenergetics towards glycolysis by impairing Ca2+ entry into the mitochondria. This event prevents lipid peroxidation, antagonizes apoptosis and increases cancer cell proliferation. Accordingly, loss of function of Plcg1 in a mouse model of KrasG12D-driven lung adenocarcinoma increased the expression of glycolytic genes, boosted tumour growth and reduced survival. In patients with KRAS-mutant lung adenocarcinomas, low PLCγ1 expression correlates with increased expression of hypoxia markers and predicts poor patient survival. Thus, our work reveals a mechanism of cancer cell adaptation to hypoxia with potential therapeutic value.


Subject(s)
Adenocarcinoma of Lung/enzymology , Lung Neoplasms/enzymology , Mutation , Phospholipase C gamma/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Hypoxia , A549 Cells , Adaptation, Physiological , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Animals , Cell Proliferation , Cell Survival , Energy Metabolism , Female , Humans , Lipid Peroxidation , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice, Inbred NOD , Mice, Transgenic , Mitochondria/enzymology , Mitochondria/pathology , Phospholipase C gamma/genetics , Signal Transduction
8.
Sci Adv ; 6(44)2020 10.
Article in English | MEDLINE | ID: mdl-33127675

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is characterized by marked fibrosis and low immunogenicity, features that are linked to treatment resistance and poor clinical outcomes. Therefore, understanding how PDAC regulates the desmoplastic and immune stromal components is of great clinical importance. We found that acyl-CoA synthetase long-chain 3 (ACSL3) is up-regulated in PDAC and correlates with increased fibrosis. Our in vivo results show that Acsl3 knockout hinders PDAC progression, markedly reduces tumor fibrosis and tumor-infiltrating immunosuppressive cells, and increases cytotoxic T cell infiltration. This effect is, at least in part, due to decreased plasminogen activator inhibitor-1 (PAI-1) secretion from tumor cells. Accordingly, PAI-1 expression in PDAC positively correlates with markers of fibrosis and immunosuppression and predicts poor patient survival. We found that PAI-1 pharmacological inhibition strongly enhances chemo- and immunotherapeutic response against PDAC, increasing survival of mice. Thus, our results unveil ACSL3-PAI-1 signaling as a requirement for PDAC progression with druggable attributes.


Subject(s)
Carcinoma, Pancreatic Ductal , Coenzyme A Ligases , Pancreatic Neoplasms , Animals , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Coenzyme A Ligases/genetics , Fibrosis , Mice , Pancreatic Neoplasms/pathology , Plasminogen Activator Inhibitor 1/genetics , Serpin E2
10.
Cell Rep ; 30(11): 3851-3863.e6, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32187554

ABSTRACT

Cancer therapy is limited, in part, by lack of specificity. Thus, identifying molecules that are selectively expressed by, and relevant for, cancer cells is of paramount medical importance. Here, we show that peptidyl-prolyl-cis-trans-isomerase (PPIase) FK506-binding protein 10 (FKBP10)-positive cells are present in cancer lesions but absent in the healthy parenchyma of human lung. FKBP10 expression negatively correlates with survival of lung cancer patients, and its downregulation causes a dramatic diminution of lung tumor burden in mice. Mechanistically, our results from gain- and loss-of-function assays show that FKBP10 boosts cancer growth and stemness via its PPIase activity. Also, FKBP10 interacts with ribosomes, and its downregulation leads to reduction of translation elongation at the beginning of open reading frames (ORFs), particularly upon insertion of proline residues. Thus, our data unveil FKBP10 as a cancer-selective molecule with a key role in translational reprogramming, stem-like traits, and growth of lung cancer.


Subject(s)
Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Protein Biosynthesis , Tacrolimus Binding Proteins/metabolism , Animals , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Peptidylprolyl Isomerase/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Ribosomes/metabolism
11.
Oncogene ; 39(14): 2948-2960, 2020 04.
Article in English | MEDLINE | ID: mdl-32034305

ABSTRACT

Enhanced prostaglandin production promotes the development and progression of cancer. Prostaglandins are generated from arachidonic acid (AA) by the action of cyclooxygenase (COX) isoenzymes. However, how cancer cells are able to maintain an elevated supply of AA for prostaglandin production remains unclear. Here, by using lung cancer cell lines and clinically relevant KrasG12D-driven mouse models, we show that the long-chain acyl-CoA synthetase (ACSL3) channels AA into phosphatidylinositols to provide the lysophosphatidylinositol-acyltransferase 1 (LPIAT1) with a pool of AA to sustain high prostaglandin synthesis. LPIAT1 knockdown suppresses proliferation and anchorage-independent growth of lung cancer cell lines, and hinders in vivo tumorigenesis. In primary human lung tumors, the expression of LPIAT1 is elevated compared with healthy tissue, and predicts poor patient survival. This study uncovers the ACSL3-LPIAT1 axis as a requirement for the sustained prostaglandin synthesis in lung cancer with potential therapeutic value.


Subject(s)
Acyltransferases/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Coenzyme A Ligases/metabolism , Prostaglandins/metabolism , Signal Transduction/physiology , A549 Cells , Animals , Carcinogenesis/metabolism , Cell Line, Tumor , Female , Humans , Lung/metabolism , Lung Neoplasms , Male , Mice , Mice, Inbred NOD
12.
Int J Mol Sci ; 20(15)2019 Jul 24.
Article in English | MEDLINE | ID: mdl-31344914

ABSTRACT

The deregulation of cancer cell metabolic networks is now recognized as one of the hallmarks of cancer. Abnormal lipid synthesis and extracellular lipid uptake are advantageous modifications fueling the needs of uncontrolled cancer cell proliferation. Fatty acids are placed at the crossroads of anabolic and catabolic pathways, as they are implicated in the synthesis of phospholipids and triacylglycerols, or they can undergo ß-oxidation. Key players to these decisions are the long-chain acyl-CoA synthetases, which are enzymes that catalyze the activation of long-chain fatty acids of 12-22 carbons. Importantly, the long-chain acyl-CoA synthetases are deregulated in many types of tumors, providing a rationale for anti-tumor therapeutic opportunities. The purpose of this review is to summarize the last up-to-date findings regarding their role in cancer, and to discuss the related emerging tumor targeting opportunities.


Subject(s)
Coenzyme A Ligases/metabolism , Lipids/genetics , Neoplasms/genetics , Animals , Coenzyme A Ligases/genetics , Humans , Lipids/biosynthesis , Neoplasms/enzymology , Neoplasms/pathology , Phospholipids/biosynthesis , Phospholipids/metabolism , Triglycerides/biosynthesis , Triglycerides/metabolism
13.
Cell Rep ; 18(8): 1858-1868, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28228253

ABSTRACT

Cancer stem cells (CSCs) have high tumorigenic capacity. Here, we show that stem-like traits of specific human cancer cells are reduced by overexpression of the histone deacetylase sirtuin 6 (SIRT6). SIRT6-sensitive cancer cells bear mutations that activate phosphatidylinositol-3-kinase (PI3K) signaling, and overexpression of SIRT6 reduces growth, progression, and grade of breast cancer in a mouse model with PI3K activation. Tumor metabolomic and transcriptomic analyses reveal that SIRT6 overexpression dampens PI3K signaling and stem-like characteristics and causes metabolic rearrangements in this cancer model. Ablation of a PI3K activating mutation in otherwise isogenic cancer cells is sufficient to convert SIRT6-sensitive into SIRT6-insensitive cells. SIRT6 overexpression suppresses PI3K signaling at the transcriptional level and antagonizes tumor sphere formation independent of its histone deacetylase activity. Our data identify SIRT6 as a putative molecular target that hinders stemness of tumors with PI3K activation.


Subject(s)
Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Sirtuins/metabolism , Acetylation , Animals , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Mutation/physiology , Signal Transduction/physiology , Transcription, Genetic/physiology
14.
Cell Rep ; 16(6): 1614-1628, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27477280

ABSTRACT

KRAS is one of the most commonly mutated oncogenes in human cancer. Mutant KRAS aberrantly regulates metabolic networks. However, the contribution of cellular metabolism to mutant KRAS tumorigenesis is not completely understood. We report that mutant KRAS regulates intracellular fatty acid metabolism through Acyl-coenzyme A (CoA) synthetase long-chain family member 3 (ACSL3), which converts fatty acids into fatty Acyl-CoA esters, the substrates for lipid synthesis and ß-oxidation. ACSL3 suppression is associated with depletion of cellular ATP and causes the death of lung cancer cells. Furthermore, mutant KRAS promotes the cellular uptake, retention, accumulation, and ß-oxidation of fatty acids in lung cancer cells in an ACSL3-dependent manner. Finally, ACSL3 is essential for mutant KRAS lung cancer tumorigenesis in vivo and is highly expressed in human lung cancer. Our data demonstrate that mutant KRAS reprograms lipid homeostasis, establishing a metabolic requirement that could be exploited for therapeutic gain.


Subject(s)
Acyl Coenzyme A/metabolism , Coenzyme A Ligases/genetics , Lung Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Carcinogenesis/metabolism , Cell Transformation, Neoplastic/metabolism , Coenzyme A Ligases/metabolism , Fatty Acids/metabolism , Humans , Ligases/metabolism , Lipid Metabolism/genetics , Lipogenesis/physiology , Lung Neoplasms/metabolism , Mice, Knockout , Oxidation-Reduction
15.
Mol Cell ; 58(6): 977-88, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26028537

ABSTRACT

Target of Rapamycin (TOR) plays central roles in the regulation of eukaryote growth as the hub of two essential multiprotein complexes: TORC1, which is rapamycin-sensitive, and the lesser characterized TORC2, which is not. TORC2 is a key regulator of lipid biosynthesis and Akt-mediated survival signaling. In spite of its importance, its structure and the molecular basis of its rapamycin insensitivity are unknown. Using crosslinking-mass spectrometry and electron microscopy, we determined the architecture of TORC2. TORC2 displays a rhomboid shape with pseudo-2-fold symmetry and a prominent central cavity. Our data indicate that the C-terminal part of Avo3, a subunit unique to TORC2, is close to the FKBP12-rapamycin-binding domain of Tor2. Removal of this sequence generated a FKBP12-rapamycin-sensitive TORC2 variant, which provides a powerful tool for deciphering TORC2 function in vivo. Using this variant, we demonstrate a role for TORC2 in G2/M cell-cycle progression.


Subject(s)
Multiprotein Complexes/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/chemistry , Antifungal Agents/metabolism , Antifungal Agents/pharmacology , Binding Sites/genetics , Biocatalysis/drug effects , Blotting, Western , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Drug Resistance/genetics , Mass Spectrometry/methods , Mechanistic Target of Rapamycin Complex 2 , Microscopy, Electron , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutation , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Structure, Tertiary , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/ultrastructure , Sirolimus/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
16.
Cell Metab ; 21(1): 117-25, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25533479

ABSTRACT

Dietary effects on tumor biology can be exploited to unravel cancer vulnerabilities. Here, we present surprising evidence for anti-proliferative action of high-calorie-diet (HCD) feeding on KRAS-driven lung tumors. Tumors of mice that commenced HCD feeding before tumor onset displayed defective unfolded protein response (UPR) and unresolved endoplasmic reticulum (ER) stress. Unresolved ER stress and reduced proliferation are reversed by chemical chaperone treatment. Whole-genome transcriptional analyses revealed FKBP10 as one of the most downregulated chaperones in tumors of the HCD-pre-tumor-onset group. FKBP10 downregulation dampens tumor growth in vitro and in vivo. Providing translational value to these results, we report that FKBP10 is expressed in human KRAS-positive and -negative lung cancers, but not in healthy parenchyma. Collectively, our data shed light on an unexpected anti-tumor action of HCD imposed before tumor onset and identify FKBP10 as a putative therapeutic target to selectively hinder lung cancer.


Subject(s)
Diet , Lung Neoplasms/pathology , Phenylbutyrates/toxicity , ras Proteins/metabolism , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Down-Regulation , Doxycycline/toxicity , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/metabolism , Male , Mice , Mice, SCID , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Tacrolimus Binding Proteins/antagonists & inhibitors , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism , Transplantation, Heterologous , Unfolded Protein Response/drug effects , ras Proteins/genetics
17.
Cancer Discov ; 3(4): 444-57, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23358651

ABSTRACT

UNLABELLED: Non-small cell lung cancer (NSCLC) often expresses mutant KRAS together with tumor-associated mutations of the CDKN2A locus, which are associated with aggressive, therapy-resistant tumors. Here, we unravel specific requirements for the maintenance of NSCLC that carries this genotype. We establish that the extracellular signal-regulated kinase (ERK)/RHOA/focal adhesion kinase (FAK) network is deregulated in high-grade lung tumors. Suppression of RHOA or FAK induces cell death selectively in mutant KRAS;INK4A/ARF-deficient lung cancer cells. Furthermore, pharmacologic inhibition of FAK caused tumor regression specifically in the high-grade lung cancer that developed in mutant Kras;Cdkn2a-null mice. These findings provide a rationale for the rapid implementation of genotype-specific targeted therapies using FAK inhibitors in patients with cancer. SIGNIFICANCE: Targeted therapies are effective for only a small fraction of patients with cancer. We report that FAK inhibitors exert potent antitumor effects in NSCLCs that express mutant KRAS in association with INK4A/ARF deficiency. These results reveal a novel genotype-specific vulnerability of cancer cells that can be exploited for therapeutic purposes.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Genes, ras/genetics , rhoA GTP-Binding Protein/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Nude , Mice, Transgenic , Protein Kinase Inhibitors/therapeutic use , Signal Transduction , Xenograft Model Antitumor Assays
18.
Breast Cancer Res ; 14(5): R137, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-23098208

ABSTRACT

INTRODUCTION: Intrinsic plasticity of breast carcinoma cells allows them to undergo a transient and reversible conversion into mesenchymal cells to disseminate into distant organs, where they can re-differentiate to an epithelial-like status to form a cohesive secondary mass. The p130Cas scaffold protein is overexpressed in human ER+ and HER2+ breast cancer where it contributes to cancer progression, invasion and resistance to therapy. However, its role in regulating mesenchymal aggressive breast cancer cells remains to be determined. The aim of this study was to investigate the molecular and functional involvement of this adaptor protein in breast cancer cell plasticity. METHODS: We used silencing strategies and rescue experiments to evaluate phenotypic and biochemical changes from mesenchymal to epithelial traits in breast tumor cell lines. In the mouse A17 cell model previously related to mesenchymal cancer stem cells and basal-like breast cancer, we biochemically dissected the signaling pathways involved and performed functional in vivo tumor growth ability assays. The significance of the signaling platform was assessed in a human setting through the use of specific inhibitors in aggressive MDA-MB-231 subpopulation LM2-4175 cells. To evaluate the clinical relevance of the results, we analyzed publicly available microarray data from the Netherlands Cancer Institute and from the Koo Foundation Sun Yat-Sen Cancer Center. RESULTS: We show that p130Cas silencing induces loss of mesenchymal features, by downregulating Vimentin, Snail, Slug and Twist transcriptional factors, resulting in the acquirement of epithelial-like traits. Mechanistically, p130Cas controls Cyclooxygenase-2 transcriptional expression, which in turn contributes to p130Cas-dependent maintenance of mesenchymal phenotype. This cascade of events also compromises in vivo tumor growth through inhibition of cell signaling controlling cell cycle progression. c-Src and JNK kinases are sequential players in p130Cas/ Cyclooxygenase-2 axis and their pharmacological inhibition is sufficient to downregulate Cyclooxygenase-2 leading to an epithelial phenotype. Finally, in silico microarray data analysis indicates that p130Cas and Cyclooxygenase-2 concomitant overexpression predicts poor survival and high probability of breast tumor recurrence. CONCLUSIONS: Overall, these data identify a new p130Cas/Cyclooxygenase-2 axis as a crucial element in the control of breast tumor plasticity, opening new therapeutic strategies leading to inhibition of these pathways in aggressive breast carcinoma.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Crk-Associated Substrate Protein/metabolism , Cyclooxygenase 2/metabolism , Animals , Breast Neoplasms/genetics , CSK Tyrosine-Protein Kinase , Cell Line, Tumor , Crk-Associated Substrate Protein/genetics , Cyclooxygenase 2/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Models, Biological , Phenotype , Quantitative Trait, Heritable , src-Family Kinases/metabolism
19.
Cancer Res ; 72(9): 2275-84, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22406621

ABSTRACT

The ubiquitin-like SUMO proteins covalently modify protein substrates and regulate their functional properties. In a broad spectrum of cancers, the tumor suppressor PML undergoes ubiquitin-mediated degradation primed by CK2 phosphorylation. Here, we report that the SUMO E3-ligase inhibitor PIAS1 regulates oncogenic signaling through its ability to sumoylate PML and the PML-RARA oncoprotein of acute promyelocytic leukemia (APL). PIAS1-mediated SUMOylation of PML promoted CK2 interaction and ubiquitin/proteasome-mediated degradation of PML, attenuating its tumor suppressor functions. In addition, PIAS1-mediated SUMOylation of PML-RARA was essential for induction of its degradation by arsenic trioxide, an effective APL treatment. Moreover, PIAS1 suppression abrogated the ability of arsenic trioxide to trigger apoptosis in APL cells. Lastly, PIAS1 was also essential for PML degradation in non-small cell lung carcinoma (NSCLC) cells, and PML and PIAS1 were inversely correlated in NSCLC cell lines and primary specimens. Together, our findings reveal novel roles for PIAS1 and the SUMOylation machinery in regulating oncogenic networks and the response to leukemia therapy.


Subject(s)
Nuclear Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Protein Inhibitors of Activated STAT/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Arsenic Trioxide , Arsenicals/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , HEK293 Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Nuclear Proteins/genetics , Oncogene Proteins, Fusion/genetics , Oxides/pharmacology , Polymorphism, Single Nucleotide , Promyelocytic Leukemia Protein , Protein Inhibitors of Activated STAT/genetics , Rats , Small Ubiquitin-Related Modifier Proteins/genetics , Transcription Factors/genetics , Transfection , Tumor Suppressor Proteins/genetics , Ubiquitin/metabolism
20.
EMBO Mol Med ; 4(7): 594-602, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22359342

ABSTRACT

Expression of oncogenic K-RAS in primary cells elicits oncogene-induced cellular senescence (OIS), a form of growth arrest that potently opposes tumourigenesis. This effect has been largely attributed to transcriptional mechanisms that depend on the p53 tumour suppressor protein. The PML tumour suppressor was initially identified as a component of the PML-RARα oncoprotein of acute promyelocytic leukaemia (APL). PML, a critical OIS mediator, is upregulated by oncogenic K-RAS in vivo and in vitro. We demonstrate here that oncogenic K-RAS induces PML protein upregulation by activating the RAS/MEK1/mTOR/eIF4E pathway even in the absence of p53. Under these circumstances, PML mRNA is selectively associated to polysomes. Importantly, we find that the PML 5' untranslated mRNA region plays a key role in mediating PML protein upregulation and that its presence is essential for an efficient OIS response. These findings demonstrate that upregulation of PML translation plays a central role in oncogenic K-RAS-induced OIS. Thus, selective translation initiation plays a critical role in tumour suppression with important therapeutic implications for the treatment of solid tumours and APL.


Subject(s)
Cellular Senescence , Leukemia, Promyelocytic, Acute/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , ras Proteins/metabolism , 5' Untranslated Regions , Animals , Cell Line , DNA-Binding Proteins/metabolism , Fibroblasts/metabolism , Humans , Leukemia, Promyelocytic, Acute/pathology , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...