Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunother Cancer ; 11(3)2023 03.
Article in English | MEDLINE | ID: mdl-36868570

ABSTRACT

BACKGROUND: Loss of Ambra1 (autophagy and beclin 1 regulator 1), a multifunctional scaffold protein, promotes the formation of nevi and contributes to several phases of melanoma development. The suppressive functions of Ambra1 in melanoma are mediated by negative regulation of cell proliferation and invasion; however, evidence suggests that loss of Ambra1 may also affect the melanoma microenvironment. Here, we investigate the possible impact of Ambra1 on antitumor immunity and response to immunotherapy. METHODS: This study was performed using an Ambra1-depleted BrafV600E /Pten-/ - genetically engineered mouse (GEM) model of melanoma, as well as GEM-derived allografts of BrafV600E /Pten-/ - and BrafV600E /Pten-/ -/Cdkn2a-/ - tumors with Ambra1 knockdown. The effects of Ambra1 loss on the tumor immune microenvironment (TIME) were analyzed using NanoString technology, multiplex immunohistochemistry, and flow cytometry. Transcriptome and CIBERSORT digital cytometry analyses of murine melanoma samples and human melanoma patients (The Cancer Genome Atlas) were applied to determine the immune cell populations in null or low-expressing AMBRA1 melanoma. The contribution of Ambra1 on T-cell migration was evaluated using a cytokine array and flow cytometry. Tumor growth kinetics and overall survival analysis in BrafV600E /Pten-/ -/Cdkn2a-/ - mice with Ambra1 knockdown were evaluated prior to and after administration of a programmed cell death protein-1 (PD-1) inhibitor. RESULTS: Loss of Ambra1 was associated with altered expression of a wide range of cytokines and chemokines as well as decreased infiltration of tumors by regulatory T cells, a subpopulation of T cells with potent immune-suppressive properties. These changes in TIME composition were associated with the autophagic function of Ambra1. In the BrafV600E /Pten-/ -/Cdkn2a-/ - model inherently resistant to immune checkpoint blockade, knockdown of Ambra1 led to accelerated tumor growth and reduced overall survival, but at the same time conferred sensitivity to anti-PD-1 treatment. CONCLUSIONS: This study shows that loss of Ambra1 affects the TIME and the antitumor immune response in melanoma, highlighting new functions of Ambra1 in the regulation of melanoma biology.


Subject(s)
Melanoma , Proto-Oncogene Proteins B-raf , Humans , Animals , Mice , Autophagy , Cell Movement , Cell Proliferation , Cytokines , Tumor Microenvironment , Adaptor Proteins, Signal Transducing
2.
Oncoimmunology ; 12(1): 2158610, 2023.
Article in English | MEDLINE | ID: mdl-36545256

ABSTRACT

Immune-checkpoint inhibitors (ICI) are highly effective in reinvigorating T cells to attack cancer. Nevertheless, a large subset of patients fails to benefit from ICI, partly due to lack of the cancer neoepitopes necessary to trigger an immune response. In this study, we used the thiopurine 6-thioguanine (6TG) to induce random mutations and thus increase the level of neoepitopes presented by tumor cells. Thiopurines are prodrugs which are converted into thioguanine nucleotides that are incorporated into DNA (DNA-TG), where they can induce mutation through single nucleotide mismatching. In a pre-clinical mouse model of a mutation-low melanoma cell line, we demonstrated that 6TG induced clinical-grade DNA-TG integration resulting in an improved tumor control that was strongly T cell dependent. 6TG exposure increased the tumor mutational burden, without affecting tumor cell proliferation and cell death. Moreover, 6TG treatment re-shaped the tumor microenvironment by increasing T and NK immune cells, making the tumors more responsive to immune-checkpoint blockade. We further validated that 6TG exposure improved tumor control in additional mouse models of melanoma. These findings have paved the way for a phase I/II clinical trial that explores whether treatment with thiopurines can increase the proportion of otherwise treatment-resistant cancer patients who may benefit from ICI therapy (NCT05276284).


Subject(s)
Melanoma , Thioguanine , Animals , Mice , Immune Checkpoint Inhibitors , Melanoma/drug therapy , Melanoma/genetics , Mutation , Thioguanine/pharmacology , Thioguanine/therapeutic use , Tumor Microenvironment , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic
3.
Nat Commun ; 12(1): 2550, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33953176

ABSTRACT

Melanoma is the deadliest skin cancer. Despite improvements in the understanding of the molecular mechanisms underlying melanoma biology and in defining new curative strategies, the therapeutic needs for this disease have not yet been fulfilled. Herein, we provide evidence that the Activating Molecule in Beclin-1-Regulated Autophagy (Ambra1) contributes to melanoma development. Indeed, we show that Ambra1 deficiency confers accelerated tumor growth and decreased overall survival in Braf/Pten-mutated mouse models of melanoma. Also, we demonstrate that Ambra1 deletion promotes melanoma aggressiveness and metastasis by increasing cell motility/invasion and activating an EMT-like process. Moreover, we show that Ambra1 deficiency in melanoma impacts extracellular matrix remodeling and induces hyperactivation of the focal adhesion kinase 1 (FAK1) signaling, whose inhibition is able to reduce cell invasion and melanoma growth. Overall, our findings identify a function for AMBRA1 as tumor suppressor in melanoma, proposing FAK1 inhibition as a therapeutic strategy for AMBRA1 low-expressing melanoma.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Melanoma/genetics , Melanoma/metabolism , Animals , Autophagy/physiology , Beclin-1/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Models, Animal , Female , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation, Neoplastic , Humans , Male , Melanoma/pathology , Mice , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phenotype , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Signal Transduction , Transcriptome
4.
Nat Commun ; 11(1): 4709, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32948765

ABSTRACT

Glioblastoma cancer-stem like cells (GSCs) display marked resistance to ionizing radiation (IR), a standard of care for glioblastoma patients. Mechanisms underpinning radio-resistance of GSCs remain largely unknown. Chromatin state and the accessibility of DNA lesions to DNA repair machineries are crucial for the maintenance of genomic stability. Understanding the functional impact of chromatin remodeling on DNA repair in GSCs may lay the foundation for advancing the efficacy of radio-sensitizing therapies. Here, we present the results of a high-content siRNA microscopy screen, revealing the transcriptional elongation factor SPT6 to be critical for the genomic stability and self-renewal of GSCs. Mechanistically, SPT6 transcriptionally up-regulates BRCA1 and thereby drives an error-free DNA repair in GSCs. SPT6 loss impairs the self-renewal, genomic stability and tumor initiating capacity of GSCs. Collectively, our results provide mechanistic insights into how SPT6 regulates DNA repair and identify SPT6 as a putative therapeutic target in glioblastoma.


Subject(s)
DNA Repair , Genomic Instability , Glioblastoma/genetics , Neoplastic Stem Cells , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Apoptosis , BRCA1 Protein , Brain Neoplasms/genetics , Cell Cycle Checkpoints , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Silencing , Glioblastoma/pathology , HEK293 Cells , Heterografts , Humans , Mice , Mice, Inbred BALB C , Neoplastic Stem Cells/pathology , RNA, Small Interfering/genetics , Radiation Tolerance , Radiation, Ionizing , Transcriptome
5.
EBioMedicine ; 29: 112-127, 2018 03.
Article in English | MEDLINE | ID: mdl-29433983

ABSTRACT

Epidermal growth factor receptor (EGFR)-mutation-positive non-smallcell lung cancer (NSCLC) is incurable, despite high rates of response to EGFR tyrosine kinase inhibitors (TKIs). We investigated receptor tyrosine kinases (RTKs), Src family kinases and focal adhesion kinase (FAK) as genetic modifiers of innate resistance in EGFR-mutation-positive NSCLC. We performed gene expression analysis in two cohorts (Cohort 1 and Cohort 2) of EGFR-mutation-positive NSCLC patients treated with EGFR TKI. We evaluated the efficacy of gefitinib or osimertinib with the Src/FAK/Janus kinase 2 (JAK2) inhibitor, TPX0005 in vitro and in vivo. In Cohort 1, CUB domain-containing protein-1 (CDCP1) was an independent negative prognostic factor for progression-free survival (hazard ratio of 1.79, p=0.0407) and overall survival (hazard ratio of 2.23, p=0.0192). A two-gene model based on AXL and CDCP1 expression was strongly associated with the clinical outcome to EGFR TKIs, in both cohorts of patients. Our preclinical experiments revealed that several RTKs and non-RTKs, were up-regulated at baseline or after treatment with gefitinib or osimertinib. TPX-0005 plus EGFR TKI suppressed expression and activation of RTKs and downstream signaling intermediates. Co-expression of CDCP1 and AXL is often observed in EGFR-mutation-positive tumors, limiting the efficacy of EGFR TKIs. Co-treatment with EGFR TKI and TPX-0005 warrants testing.


Subject(s)
Antigens, CD/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Cell Adhesion Molecules/genetics , ErbB Receptors/genetics , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Mutation , Neoplasm Proteins/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Adult , Aged , Aged, 80 and over , Animals , Antigens, Neoplasm , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Adhesion Molecules/agonists , Cell Survival , Disease Models, Animal , Drug Resistance, Neoplasm , Enzyme Activation , ErbB Receptors/antagonists & inhibitors , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Male , Mice , Middle Aged , Models, Biological , Neoplasm Proteins/agonists , Proteomics/methods , Proto-Oncogene Proteins/agonists , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor Protein-Tyrosine Kinases/agonists , Survival Analysis , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
6.
Mol Cell Biol ; 36(6): 923-40, 2015 Dec 28.
Article in English | MEDLINE | ID: mdl-26711268

ABSTRACT

Notch activation in aortic endothelial cells (ECs) takes place at embryonic stages during cardiac valve formation and induces endothelial-to-mesenchymal transition (EndMT). Using aortic ECs, we show here that active Notch expression promotes EndMT, resulting in downregulation of vascular endothelial cadherin (VE-cadherin) and upregulation of mesenchymal genes such as those for fibronectin and Snail1/2. In these cells, transforming growth factor ß1 exacerbates Notch effects by increasing Snail1 and fibronectin activation. When Notch-downstream pathways were analyzed, we detected an increase in glycogen synthase kinase 3ß (GSK-3ß) phosphorylation and inactivation that facilitates Snail1 nuclear retention and protein stabilization. However, the total activity of Akt was downregulated. The discrepancy between Akt activity and GSK-3ß phosphorylation is explained by a Notch-induced switch in the Akt isoforms, whereby Akt1, the predominant isoform expressed in ECs, is decreased and Akt2 transcription is upregulated. Mechanistically, Akt2 induction requires the stimulation of the ß-catenin/TCF4 transcriptional complex, which activates the Akt2 promoter. Active, phosphorylated Akt2 translocates to the nucleus in Notch-expressing cells, resulting in GSK-3ß inactivation in this compartment. Akt2, but not Akt1, colocalizes in the nucleus with lamin B in the nuclear envelope. In addition to promoting GSK-3ß inactivation, Notch downregulates Forkhead box O1 (FoxO1), another Akt2 nuclear substrate. Moreover, Notch protects ECs from oxidative stress-induced apoptosis through an Akt2- and Snail1-dependent mechanism.


Subject(s)
Cell Death , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Notch/metabolism , Transcription Factors/genetics , Animals , Aorta/cytology , Cell Line , Endothelial Cells/cytology , Endothelial Cells/metabolism , Epithelial-Mesenchymal Transition , Gene Expression Regulation , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Mice , Oxidative Stress , Protein Isoforms/analysis , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Stability , Proto-Oncogene Proteins c-akt/analysis , Proto-Oncogene Proteins c-akt/genetics , Snail Family Transcription Factors , Swine , Transcription Factors/analysis , Transcription Factors/metabolism , Up-Regulation , beta Catenin/metabolism
7.
Nucleic Acids Res ; 42(2): 1079-94, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24157836

ABSTRACT

The zinc finger transcription factor Snail1 regulates epithelial to mesenchymal transition, repressing epithelial markers and activating mesenchymal genes. Snail1 is an extremely labile protein degraded by the cytoplasmic ubiquitin-ligases ß-TrCP1/FBXW1 and Ppa/FBXL14. Using a short hairpin RNA screening, we have identified FBXL5 as a novel Snail1 ubiquitin ligase. FBXL5 is located in the nucleus where it interacts with Snail1 promoting its polyubiquitination and affecting Snail1 protein stability and function by impairing DNA binding. Snail1 downregulation by FBXL5 is prevented by Lats2, a protein kinase that phosphorylates Snail1 precluding its nuclear export but not its polyubiquitination. Actually, although polyubiquitination by FBXL5 takes place in the nucleus, Snail1 is degraded in the cytosol. Finally, FBXL5 is highly sensitive to stress conditions and is downregulated by iron depletion and γ-irradiation, explaining Snail1 stabilization in these conditions. These results characterize a novel nuclear ubiquitin ligase controlling Snail1 protein stability and provide the molecular basis for understanding how radiotherapy upregulates the epithelial to mesenchymal transition-inducer Snail1.


Subject(s)
Cell Nucleus/enzymology , F-Box Proteins/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Cell Line, Tumor , Cell Nucleus/metabolism , DNA/metabolism , Gamma Rays , Humans , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Stability , RNA, Small Interfering , Snail Family Transcription Factors , Ubiquitin-Protein Ligase Complexes
8.
J Biol Chem ; 286(14): 12024-32, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21317430

ABSTRACT

Snail1 and Zeb1 are E-cadherin-transcriptional repressors induced during epithelial mesenchymal transition (EMT). In this article we have analyzed the factors controlling Zeb1 expression during EMT. In NMuMG cells treated with TGF-ß, Snail1 RNA and protein are induced 1 h after addition of the cytokine preceding Zeb1 up-regulation that requires 6-8 h. Zeb1 gene expression is caused by increased RNA levels but also by enhanced protein stability and is markedly dependent on Snail1 because depletion of this protein prevents Zeb1 protein and RNA up-regulation. In addition to Snail1, depletion of the Twist transcriptional factor retards Zeb1 stimulation by TGF-ß or decreases Zeb1 expression in other cellular models indicating that this factor is also required for Zeb1 expression. Accordingly, Snail1 and Twist cooperate in the induction of Zeb1: co-transfection of both cDNAs is required for the maximal expression of ZEB1 mRNA. Unexpectedly, the expression of Snail1 and Twist shows a mutual dependence although to a different extent; whereas Twist depletion retards Snail1 up-regulation by TGF-ß, Snail1 is necessary for the rapid increase in Twist protein and later up-regulation of Twist1 mRNA induced by the cytokine. Besides this effect on Twist, Snail1 also induces the nuclear translocation of Ets1, another factor required for Zeb1 expression. Both Twist and Ets1 bind to the ZEB1 promoter although to different elements: whereas Ets1 interacts with the proximal promoter, Twist does it with a 700-bp sequence upstream of the transcription start site. These results indicate that Snail1 controls Zeb1 expression at multiple levels and acts cooperatively with Twist in the ZEB1 gene transcription induction.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Twist-Related Protein 1/metabolism , Animals , Cells, Cultured , Chromatin Immunoprecipitation , Epithelial-Mesenchymal Transition/genetics , Homeodomain Proteins/genetics , Humans , Mice , Protein Binding/drug effects , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-ets-1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Snail Family Transcription Factors , Transcription Factors/genetics , Transforming Growth Factor beta/pharmacology , Twist-Related Protein 1/genetics , Zinc Finger E-box-Binding Homeobox 1
SELECTION OF CITATIONS
SEARCH DETAIL
...