Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Nature ; 626(7998): 401-410, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38297129

ABSTRACT

Ferroptosis is a form of cell death that has received considerable attention not only as a means to eradicate defined tumour entities but also because it provides unforeseen insights into the metabolic adaptation that tumours exploit to counteract phospholipid oxidation1,2. Here, we identify proferroptotic activity of 7-dehydrocholesterol reductase (DHCR7) and an unexpected prosurvival function of its substrate, 7-dehydrocholesterol (7-DHC). Although previous studies suggested that high concentrations of 7-DHC are cytotoxic to developing neurons by favouring lipid peroxidation3, we now show that 7-DHC accumulation confers a robust prosurvival function in cancer cells. Because of its far superior reactivity towards peroxyl radicals, 7-DHC effectively shields (phospho)lipids from autoxidation and subsequent fragmentation. We provide validation in neuroblastoma and Burkitt's lymphoma xenografts where we demonstrate that the accumulation of 7-DHC is capable of inducing a shift towards a ferroptosis-resistant state in these tumours ultimately resulting in a more aggressive phenotype. Conclusively, our findings provide compelling evidence of a yet-unrecognized antiferroptotic activity of 7-DHC as a cell-intrinsic mechanism that could be exploited by cancer cells to escape ferroptosis.


Subject(s)
Burkitt Lymphoma , Dehydrocholesterols , Ferroptosis , Neuroblastoma , Animals , Humans , Burkitt Lymphoma/metabolism , Burkitt Lymphoma/pathology , Cell Survival , Dehydrocholesterols/metabolism , Lipid Peroxidation , Neoplasm Transplantation , Neuroblastoma/metabolism , Neuroblastoma/pathology , Oxidation-Reduction , Phenotype , Reproducibility of Results
2.
Semin Cancer Biol ; 81: 232-240, 2022 06.
Article in English | MEDLINE | ID: mdl-33610722

ABSTRACT

Melanomas and their precursors, the melanocytes, are frequently exposed to UV due to their anatomic location, leading to DNA damage and reactive oxygen stress related harm. Such damage can result in multinucleation or polyploidy, in particularly in presence of mitotic or cell division failure. As a consequence, the cell encounters either of two fates: mitotic catastrophe, resulting in cell death, or survival and recovery, the latter occurring less frequently. However, when cells manage to recover in an polyploid state, they have often acquired new features, which allow them to tolerate and adapt to oncogene- or therapy induced stress. This review focuses on polyploidy inducers in melanoma and their effects on transcriptional reprogramming and phenotypic adaptation as well as the relevance of polyploid melanoma cells for therapy resistance.


Subject(s)
Melanoma , Polyploidy , DNA Damage , Humans , Melanocytes , Melanoma/genetics , Melanoma/therapy , Mitosis , Oncogenes
3.
Eur Arch Otorhinolaryngol ; 280(9): 4215-4223, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37272953

ABSTRACT

PURPOSE: Local failure and distant metastases occur frequently in sinonasal mucosal melanoma (SNMM). Response rates to chemotherapy are low and targetable mutations are rarely detected. However, there is increasing data indicating efficacy of immune checkpoint inhibition (ICI). The aim of this retrospective monocenter study was to assess the mutational landscape and to evaluate the outcome of surgical treatment and ICI in SNMM in a real-world setting. METHODS: Thirty-eight SNMM patients being treated between 1999 and 2020 at our institution were retrospectively reviewed. Survival curves were generated according to Kaplan-Meier and compared by the log-rank test. RESULTS: Local failure was seen in 60% of patients treated in a curative intent. Overall, 24% of all patients suffered from regional and 66% from distant metastases. Next generation sequencing revealed mutations of BRAF, NRAS and KRAS. One out of three patients treated with a primary ICI showed a complete response (CR) and two showed progressive disease. Eleven patients received ICI as a palliative treatment. CR could be observed in three patients and stable disease in one patient. In the whole study population, the 5-year overall survival rate (OS) was 26%. OS was better for patients who received ICI during the course of disease. CONCLUSIONS: Recurrences and distant metastases are frequent in SNMM. Durable CR could be observed after primary and palliative ICI. Therefore, ICI in a palliative, adjuvant or even neoadjuvant setting might play a promising role in SNMM therapy while targetable mutations are rarely detected.


Subject(s)
Melanoma , Paranasal Sinus Neoplasms , Humans , Immune Checkpoint Inhibitors/therapeutic use , Retrospective Studies , Melanoma/drug therapy , Melanoma/genetics , Paranasal Sinus Neoplasms/drug therapy , Paranasal Sinus Neoplasms/genetics , Combined Modality Therapy
4.
Anal Chem ; 94(41): 14214-14222, 2022 10 18.
Article in English | MEDLINE | ID: mdl-36194871

ABSTRACT

Mass spectrometry-based immunopeptidomics enables the comprehensive identification of major histocompatibility complex (MHC) peptides from a cell culture as well as from tissue or tumor samples and is applied for the identification of tumor-specific and viral T-cell epitopes. Although mass spectrometry is generally considered an "unbiased" method for MHC peptide identification, the physicochemical properties of MHC peptides can greatly influence their detectability. Here, we demonstrate that highly hydrophobic peptides are lost during sample preparation when C18 solid-phase extraction (SPE) is used for separating MHC peptides from proteins. To overcome this limitation, we established an optimized protocol involving restricted access material (RAM). Compared to C18-SPE, RAM-SPE improved the overall MHC peptide recovery and extended the landscape of mass spectrometry-detectable MHC peptides toward more hydrophobic peptides.


Subject(s)
Epitopes, T-Lymphocyte , Major Histocompatibility Complex , Mass Spectrometry/methods , Peptides/chemistry , Solid Phase Extraction/methods
5.
Int J Mol Sci ; 22(8)2021 Apr 07.
Article in English | MEDLINE | ID: mdl-33916908

ABSTRACT

Receptor tyrosine kinases (RTK) are rarely mutated in cutaneous melanoma, but the expression and activation of several RTK family members are associated with a proinvasive phenotype and therapy resistance. Epidermal growth factor receptor (EGFR) is a member of the RTK family and is only expressed in a subgroup of melanomas with poor prognosis. The insight into regulators of EGFR expression and activation is important for the understanding of the development of this malignant melanoma phenotype. Here, we describe that the transcription factor NRF2, the master regulator of the oxidative and electrophilic stress response, mediates the expression and activation of EGFR in melanoma by elevating the levels of EGFR as well as its ligands EGF and TGFα. ChIP sequencing data show that NRF2 directly binds to the promoter of EGF, which contains a canonical antioxidant response element. Accordingly, EGF is induced by oxidative stress and is also increased in lung adenocarcinoma and head and neck carcinoma with mutationally activated NRF2. In contrast, regulation of EGFR and TGFA occurs by an indirect mechanism, which is enabled by the ability of NRF2 to block the activity of the melanocytic lineage factor MITF in melanoma. MITF effectively suppresses EGFR and TGFA expression and therefore serves as link between NRF2 and EGFR. As EGFR was previously described to stimulate NRF2 activity, the mutual activation of NRF2 and EGFR pathways was investigated. The presence of NRF2 was necessary for full EGFR pathway activation, as NRF2-knockout cells showed reduced AKT activation in response to EGF stimulation compared to controls. Conversely, EGF led to the nuclear localization and activation of NRF2, thereby demonstrating that NRF2 and EGFR are connected in a positive feedback loop in melanoma. In summary, our data show that the EGFR-positive melanoma phenotype is strongly supported by NRF2, thus revealing a novel maintenance mechanism for this clinically challenging melanoma subpopulation.


Subject(s)
Melanoma/metabolism , NF-E2-Related Factor 2/metabolism , Signal Transduction , Antioxidant Response Elements , Base Sequence , Binding Sites , Biomarkers, Tumor , Carrier Proteins , Cell Line, Tumor , ErbB Receptors/metabolism , Humans , Kaplan-Meier Estimate , Ligands , Melanoma/etiology , Melanoma/mortality , Melanoma/pathology , Models, Biological , Nucleotide Motifs , Protein Binding
6.
Biochem Soc Trans ; 48(5): 2253-2259, 2020 10 30.
Article in English | MEDLINE | ID: mdl-33125483

ABSTRACT

Lipid peroxidation has been associated with a wide array of (patho)physiological conditions. Remarkably, in the last few years, a novel cell death modality termed ferroptosis was recognized as a process initiated by iron-dependent oxidation of lipids. The sensitivity to ferroptosis is determined by the activity of antioxidant systems working on the repair of oxidized phospholipids and also metabolic pathways controlling the availability of substrates susceptible to lipid peroxidation. Non-enzymatic antioxidants such as vitamin E, which has long been acknowledged as an efficient inhibitor of lipid peroxidation, play an important and often neglected role in subverting ferroptosis. Recent works dissecting the mechanisms that determine ferroptosis sensitivity have provided further insights into the contribution of alternative metabolic pathways able to suppress lipid peroxidation. Specifically, the role of ubiquinone and tetrahydrobiopterin (BH4) has been brought forth, with the identification of specific enzymatic systems responsible for their regeneration, as critical factors suppressing ferroptosis. Therefore, in the present manuscript, we address these emerging concepts and propose that the characterization of these antioxidant repair mechanisms will not only open a new understanding of disease conditions where ferroptosis plays a role but also offer opportunities to identify and sensitize cells to ferroptosis in the context of cancer treatment.


Subject(s)
Antioxidants/chemistry , Ferroptosis , Gene Expression Regulation , Oxygen/chemistry , Animals , Biopterins/analogs & derivatives , Biopterins/chemistry , Cell Death/drug effects , Humans , Iron , Lipid Peroxidation , Lipids/chemistry , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Ubiquinone/chemistry , Vitamin E/metabolism
7.
Cancer ; 125(4): 586-600, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30561760

ABSTRACT

BACKGROUND: Increasing knowledge of cancer genomes has triggered the development of specific targeted inhibitors, thus providing a valuable therapeutic pool. METHODS: In this report, the authors analyze the presence of targetable alterations in 136 tumor samples from 92 patients with melanoma using a comprehensive approach based on targeted DNA sequencing and supported by RNA and protein analysis. Three topics of high clinical relevance are addressed: the identification of rare, activating alterations; the detection of patient-specific, co-occurring single nucleotide variants (SNVs) and copy number variations (CNVs) in parallel pathways; and the presence of cancer-relevant germline mutations. RESULTS: The analysis of patient-matched blood and tumor samples was done with a custom-designed gene panel that was enriched for genes from clinically targetable pathways. To detect alterations with high therapeutic relevance for patients with unknown driver mutations, genes that are untypical for melanoma also were included. Among all patients, CNVs were identified in one-third of samples and contained amplifications of druggable kinases, such as CDK4, ERBB2, and KIT. Considering SNVs and CNVs, 60% of patients with metastases exhibited co-occurring activations of at least 2 pathways, thus providing a rationale for individualized combination therapies. Unexpectedly, 9% of patients carry potentially protumorigenic germline mutations frequently affecting receptor tyrosine kinases. Remarkably two-thirds of BRAF/NRAS wild-type melanomas harbor activating mutations or CNVs in receptor tyrosine kinases. CONCLUSIONS: The results indicate that the integrated analysis of SNVs, CNVs, and germline mutations reveals new druggable targets for combination tumor therapy.


Subject(s)
Biomarkers, Tumor/genetics , GTP Phosphohydrolases/genetics , Gene Expression Regulation, Neoplastic , Melanoma/pathology , Membrane Proteins/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/pathology , Case-Control Studies , Cyclin-Dependent Kinase 4/genetics , DNA Copy Number Variations , Follow-Up Studies , High-Throughput Nucleotide Sequencing , Humans , Melanoma/genetics , Prognosis , Proto-Oncogene Proteins c-kit/genetics , Receptor, ErbB-2/genetics , Skin Neoplasms/genetics
8.
Cancer Metastasis Rev ; 36(1): 23-33, 2017 03.
Article in English | MEDLINE | ID: mdl-28197745

ABSTRACT

Inhibition of the BRAF/MAPK pathway belongs to the standard therapies for patients with activating BRAFV600E/K mutations. However, even in well-responding tumors, anti-tumorigenic effect and clinical benefit are only transient, and the original tumors often relapse. This demonstrates that there are remaining residual tumors, which have withstood therapy-induced apoptosis and which have the potential to resume growth. Although BRAF mutant melanoma cells seem to depend on BRAF/MAPK signaling, the inhibition of this pathway triggers several events, which modulate the tumor as well as the tumor niche. After a certain adaptation period, this can turn out to be beneficial for tumor growth and metastasis-even in cases of good initial tumor response. This review sheds light on the biology of BRAF/MEK inhibitor-sensitive melanoma cells, which survive targeted therapy and will address the crosstalk signaling events occurring in BRAF mutant melanomas when the BRAF/MAPK pathway is fully blocked. The knowledge of these events is important for potential future drug combinations, which enhance the inhibitory effect of BRAF/MEK inhibition, particularly in patients not eligible for immune therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , MAP Kinase Signaling System/drug effects , Melanoma/drug therapy , Melanoma/enzymology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Humans , Molecular Targeted Therapy , Proto-Oncogene Proteins B-raf/metabolism
9.
Biochem J ; 471(2): 267-79, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26285655

ABSTRACT

Tumour cells are reported to display an imbalance in the levels of ROS (reactive oxygen species). Frequently, elevated ROS production goes along with compensatory up-regulation of antioxidant enzymes. Accordingly, we found in a previous study that protein levels of several peroxiredoxins, including PRDX6 (peroxiredoxin 6), are highly elevated in experimentally induced melanomas. In the present study, we investigated the functional role of PRDX6 in human melanoma cells. PRDX6 is a bifunctional enzyme, which harbours iPLA2 (Ca(2+)-independent phospholipase A2) activity in addition to its peroxidase function. Our results show that PRDX6 is strongly expressed in most melanoma cells and its expression levels are maintained in a post-transcriptional manner, particularly by EGFR (epidermal growth factor receptor)-dependent signalling. PRDX6 enhances cell viability mainly by enhancing proliferation, which goes along with activation of Src family kinases. Interestingly, we were able to show that the phospholipase activity of the enzyme mediates the pro-proliferative effect of PRDX6. We identified AA (arachidonic acid) as a crucial effector of PRDX6-dependent proliferation and inducer of Src family kinase activation. These results support further the biological importance of the emerging field of lipid signalling in melanoma and highlight the particular functional relevance of PRDX6-dependent phospholipase activity.


Subject(s)
Arachidonic Acid/metabolism , Melanoma/enzymology , Neoplasm Proteins/metabolism , Peroxiredoxin VI/metabolism , Signal Transduction , Animals , Arachidonic Acid/genetics , Cell Line, Tumor , Cell Survival/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Melanoma/genetics , Mice , Neoplasm Proteins/genetics , Peroxiredoxin VI/genetics , src-Family Kinases/genetics , src-Family Kinases/metabolism
10.
J Cell Sci ; 126(Pt 17): 3862-72, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23843609

ABSTRACT

Melanomas have a high angiogenic potential, but respond poorly to medical treatment and metastasize very early. To understand the early events in tumor angiogenesis, animal models with high tumor resolution and blood vessel resolution are required, which provide the opportunity to test the ability of small molecule inhibitors to modulate the angiogenic tumor program. We have established a transgenic melanoma angiogenesis model in the small laboratory fish species Japanese medaka. Here, pigment cells are transformed by an oncogenic receptor tyrosine kinase in fish expressing GFP throughout their vasculature. We show that angiogenesis occurs in a reactive oxygen species (ROS)- and NF-κB-dependent, but hypoxia-independent manner. Intriguingly, we observed that blood vessel sprouting is induced even by single transformed pigment cells. The oncogenic receptor as well as human melanoma cells harboring other oncogenes caused the production of pro-angiogenic factors, most prominently angiogenin, through NF-κB signaling. Inhibiting NF-κB prevented tumor angiogenesis and led to the regression of existing tumor blood vessels. In conclusion, our high-resolution medaka melanoma model discloses that ROS and NF-κB signaling from single tumor cells causes hypoxia-independent angiogenesis, thus, demonstrating that the intrinsic malignant tumor cell features are sufficient to initiate and maintain a pro-angiogenic signaling threshold.


Subject(s)
Melanoma/blood supply , Neovascularization, Pathologic/metabolism , Reactive Oxygen Species/metabolism , Transcription Factor RelA/metabolism , Animals , Cell Hypoxia , Cell Line, Tumor , Cell Survival , Fish Proteins/pharmacology , Gene Expression Profiling , Green Fluorescent Proteins/genetics , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Melanoma/metabolism , Mice , Oryzias , RNA Interference , RNA, Small Interfering , Receptor Protein-Tyrosine Kinases/pharmacology , Ribonuclease, Pancreatic/biosynthesis , Ribonuclease, Pancreatic/genetics , Ribonuclease, Pancreatic/metabolism , Signal Transduction , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism
12.
bioRxiv ; 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38895428

ABSTRACT

Occurrence of degenerative interactions is thought to serve as a mechanism underlying hybrid unfitness. However, the molecular mechanisms underpinning the genetic interaction and how they contribute to overall hybrid incompatibilities are limited to only a handful of examples. A vertebrate model organism, Xiphophorus , is used to study hybrid dysfunction and it has been shown from this model that diseases, such as melanoma, can occur in certain interspecies hybrids. Melanoma development is due to hybrid inheritance of an oncogene, xmrk , and loss of a co-evolved tumor modifier. It was recently found that adgre5 , a G protein-coupled receptor involved in cell adhesion, is a tumor regulator gene in naturally hybridizing Xiphophorus species X. birchmanni and X. malinche . We hypothesized that one of the two parental alleles of adgre5 is involved in regulation of cell proliferation, migration and melanomagenesis. Accordingly, we assessed the function of adgre5 alleles from each parental species of the melanoma-bearing hybrids using in vitro cell proliferation and migration assays. In addition, we expressed each adgre5 allele with the xmrk oncogene in transgenic medaka. We found that cells transfected with the X. birchmanni adgre5 exhibited decreased proliferation and migration compared to those with the X. malinche allele. Moreover, X. birchmanni allele of adgre5 completely inhibited melanoma development in xmrk transgenic medaka, while X. malinche adgre5 expression did not exhibit melanoma suppressive activity in medaka. These findings showed that adgre5 is a natural melanoma suppressor and provide new insight in melanoma etiology.

13.
Redox Biol ; 70: 103011, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38219574

ABSTRACT

The cystine/glutamate antiporter xCT is an important source of cysteine for cancer cells. Once taken up, cystine is reduced to cysteine and serves as a building block for the synthesis of glutathione, which efficiently protects cells from oxidative damage and prevents ferroptosis. As melanomas are particularly exposed to several sources of oxidative stress, we investigated the biological role of cysteine and glutathione supply by xCT in melanoma. xCT activity was abolished by genetic depletion in the Tyr::CreER; BrafCA; Ptenlox/+ melanoma model and by acute cystine withdrawal in melanoma cell lines. Both interventions profoundly impacted melanoma glutathione levels, but they were surprisingly well tolerated by murine melanomas in vivo and by most human melanoma cell lines in vitro. RNA sequencing of human melanoma cells revealed a strong adaptive upregulation of NRF2 and ATF4 pathways, which orchestrated the compensatory upregulation of genes involved in antioxidant defence and de novo cysteine biosynthesis. In addition, the joint activation of ATF4 and NRF2 triggered a phenotypic switch characterized by a reduction of differentiation genes and induction of pro-invasive features, which was also observed after erastin treatment or the inhibition of glutathione synthesis. NRF2 alone was capable of inducing the phenotypic switch in a transient manner. Together, our data show that cystine or glutathione levels regulate the phenotypic plasticity of melanoma cells by elevating ATF4 and NRF2.


Subject(s)
Cysteine , Melanoma , Mice , Animals , Humans , Cysteine/metabolism , Cystine , Sulfhydryl Compounds , Melanoma/genetics , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Glutathione/metabolism , Oxidative Stress , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism
14.
Mol Oncol ; 2024 May 28.
Article in English | MEDLINE | ID: mdl-38807304

ABSTRACT

Immunotherapy has achieved tremendous success in melanoma. However, only around 50% of advanced melanoma patients benefit from immunotherapy. Cyclin-dependent kinase inhibitor 2A (CDKN2A), encoding the two tumor-suppressor proteins p14ARF and p16INK4a, belongs to the most frequently inactivated gene loci in melanoma and leads to decreased T cell infiltration. While the role of p16INK4a has been extensively investigated, knowledge about p14ARF in melanoma is scarce. In this study, we elucidate the impact of reduced p14ARF expression on melanoma immunogenicity. Knockdown of p14ARF in melanoma cell lines diminished their recognition and killing by melanoma differentiation antigen (MDA)-specific T cells. Resistance was caused by a reduction of the peptide surface density of presented MDAs. Immunopeptidomic analyses revealed that antigen presentation via human leukocyte antigen class I (HLA-I) molecules was enhanced upon p14ARF downregulation in general, but absolute and relative expression of cognate peptides was decreased. However, this phenotype is associated with a favorable outcome for melanoma patients. Limiting Wnt5a signaling reverted this phenotype, suggesting an involvement of non-canonical Wnt signaling. Taken together, our data indicate a new mechanism limiting MDA-specific T cell responses by decreasing both absolute and relative MDA-peptide presentation in melanoma.

15.
Cell Rep ; 42(7): 112724, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37410595

ABSTRACT

The redox regulator NRF2 becomes activated upon oxidative and electrophilic stress and orchestrates a response program associated with redox regulation, metabolism, tumor therapy resistance, and immune suppression. Here, we describe an unrecognized link between the integrated stress response (ISR) and NRF2 mediated by the ISR effector ATF4. The ISR is commonly activated after starvation or ER stress and plays a central role in tissue homeostasis and cancer plasticity. ATF4 increases NRF2 transcription and induces the glutathione-degrading enzyme CHAC1, which we now show to be critically important for maintaining NRF2 activation. In-depth analyses reveal that NRF2 supports ATF4-induced cells by increasing cystine uptake via the glutamate-cystine antiporter xCT. In addition, NRF2 upregulates genes mediating thioredoxin usage and regeneration, thus balancing the glutathione decrease. In conclusion, we demonstrate that the NRF2 response serves as second layer of the ISR, an observation highly relevant for the understanding of cellular resilience in health and disease.


Subject(s)
Activating Transcription Factor 4 , NF-E2-Related Factor 2 , Neoplasms , Humans , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Cystine/metabolism , Glutathione/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress
16.
Cancers (Basel) ; 15(2)2023 Jan 06.
Article in English | MEDLINE | ID: mdl-36672341

ABSTRACT

(1) Background: C-X-C Motif Chemokine Receptor 4 (CXCR4) and Fibroblast Activation Protein Alpha (FAP) are promising theranostic targets. However, it is unclear whether CXCR4 and FAP positivity mark distinct microenvironments, especially in solid tumors. (2) Methods: Using Random Forest (RF) analysis, we searched for entity-independent mRNA and microRNA signatures related to CXCR4 and FAP overexpression in our pan-cancer cohort from The Cancer Genome Atlas (TCGA) database-representing n = 9242 specimens from 29 tumor entities. CXCR4- and FAP-positive samples were assessed via StringDB cluster analysis, EnrichR, Metascape, and Gene Set Enrichment Analysis (GSEA). Findings were validated via correlation analyses in n = 1541 tumor samples. TIMER2.0 analyzed the association of CXCR4 / FAP expression and infiltration levels of immune-related cells. (3) Results: We identified entity-independent CXCR4 and FAP gene signatures representative for the majority of solid cancers. While CXCR4 positivity marked an immune-related microenvironment, FAP overexpression highlighted an angiogenesis-associated niche. TIMER2.0 analysis confirmed characteristic infiltration levels of CD8+ cells for CXCR4-positive tumors and endothelial cells for FAP-positive tumors. (4) Conclusions: CXCR4- and FAP-directed PET imaging could provide a non-invasive decision aid for entity-agnostic treatment of microenvironment in solid malignancies. Moreover, this machine learning workflow can easily be transferred towards other theranostic targets.

17.
EMBO Mol Med ; 15(8): e18014, 2023 08 07.
Article in English | MEDLINE | ID: mdl-37435859

ABSTRACT

Ferroptosis has emerged as an attractive strategy in cancer therapy. Understanding the operational networks regulating ferroptosis may unravel vulnerabilities that could be harnessed for therapeutic benefit. Using CRISPR-activation screens in ferroptosis hypersensitive cells, we identify the selenoprotein P (SELENOP) receptor, LRP8, as a key determinant protecting MYCN-amplified neuroblastoma cells from ferroptosis. Genetic deletion of LRP8 leads to ferroptosis as a result of an insufficient supply of selenocysteine, which is required for the translation of the antiferroptotic selenoprotein GPX4. This dependency is caused by low expression of alternative selenium uptake pathways such as system Xc- . The identification of LRP8 as a specific vulnerability of MYCN-amplified neuroblastoma cells was confirmed in constitutive and inducible LRP8 knockout orthotopic xenografts. These findings disclose a yet-unaccounted mechanism of selective ferroptosis induction that might be explored as a therapeutic strategy for high-risk neuroblastoma and potentially other MYCN-amplified entities.


Subject(s)
Ferroptosis , Neuroblastoma , Humans , Cell Line, Tumor , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/genetics , Neuroblastoma/drug therapy , Selenocysteine/therapeutic use , Animals
18.
Int J Cancer ; 130(12): 2801-11, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-21732343

ABSTRACT

Recently, we discovered that the loss of E-cadherin induces c-Jun protein expression, which is a member of the AP-1 transcription factor family and a key player in the processes of cell proliferation and tumor development and also found in elevated levels in melanomas. Notably, the mRNA level of c-Jun was not affected, suggesting that c-Jun is regulated at post-transcriptional level. Here, we present data that suggest that the dynamic cytoskeletal network, linked to E-cadherin, is involved in the regulation of the c-Jun protein and transcriptional activity. In a signaling cascade, the loss of E-cadherin activates the transcriptional regulator ETS-1 and consequently leads to the induction of RhoC expression that stabilizes c-Jun in melanoma. The link between RhoC and c-Jun seems to be indirect via the cytoskeleton. We conclude that the loss of E-cadherin mediated cell-adhesion induces c-Jun protein expression in a multistep process, offering several possibilities for therapeutic intervention.


Subject(s)
Melanoma/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , Proto-Oncogene Proteins c-jun/metabolism , rho GTP-Binding Proteins/metabolism , Cadherins/metabolism , Cell Line, Tumor , Cytoskeleton/metabolism , Humans , Nocodazole/pharmacology , Promoter Regions, Genetic , Proto-Oncogene Proteins c-jun/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/genetics , Transcription, Genetic , Transcriptional Activation , rho GTP-Binding Proteins/biosynthesis , rhoA GTP-Binding Protein/biosynthesis , rhoB GTP-Binding Protein/biosynthesis , rhoC GTP-Binding Protein
19.
J Hepatol ; 56(2): 419-25, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21888874

ABSTRACT

BACKGROUND & AIMS: Liver cancer, mainly hepatocellular carcinoma, is a major malignancy and currently there are no effective treatment protocols due to insufficient understanding of hepatocarcinogenesis. As a potentially high-throughput and cost-effective experimental model, the zebrafish is increasingly recognized for disease studies. Here, we aim at using the zebrafish to generate a convenient hepatocellular carcinoma model. METHODS: Using the Tet-on system for liver-specific expression of fish oncogene xmrk, a hyperactive version of epidermal growth factor receptor homolog, we have generated transgenic zebrafish with inducible development of liver cancer. RESULTS: Liver tumors were rapidly induced with 100% penetrance in both juvenile and adult xmrk transgenic fish. Histological examination indicated that they all showed features of hepatocellular carcinoma. The induced liver tumors regressed rapidly upon inducer withdrawal. During the tumor induction stage, we detected increased cell proliferation and activation of Xmrk downstream targets Erk and Stat5, which were important for liver tumorigenesis as proved by inhibition experiments. When tumors regressed, there were decreased phosphorylated Erk and Stat5 accompanied with an increase in apoptosis. CONCLUSIONS: Our zebrafish model demonstrates the potential of a hyperactivated epidermal growth factor receptor pathway in initiating heptocarcinogenesis. It provides clear evidence for the requirement of only a single oncogene for HCC initiation and maintenance and is thus a convenient model for further investigation of oncogene addiction and future anti-cancer drug screening.


Subject(s)
Fish Proteins/genetics , Liver Neoplasms, Experimental/genetics , Oncogenes , Receptor Protein-Tyrosine Kinases/genetics , Zebrafish/genetics , Animals , Animals, Genetically Modified , Apoptosis/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Doxycycline/administration & dosage , Female , Gene Expression , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , MAP Kinase Signaling System , Male , Oncogenes/drug effects , Recombinant Proteins/genetics , STAT5 Transcription Factor/metabolism
20.
Pigment Cell Melanoma Res ; 34(2): 268-279, 2021 03.
Article in English | MEDLINE | ID: mdl-33205526

ABSTRACT

The transcription factor NRF2 is known as the master regulator of the oxidative stress response. Tumor entities presenting oncogenic activation of NRF2, such as lung adenocarcinoma, are associated with drug resistance, and accumulating evidence demonstrates its involvement in immune evasion. In other cancer types, the KEAP1/NRF2 pathway is not commonly mutated, but NRF2 is activated by other means such as radiation, oncogenic activity, cytokines, or other pro-oxidant triggers characteristic of the tumor niche. The obvious effect of stress-activated NRF2 is the protection from oxidative or electrophilic damage and the adaptation of the tumor metabolism to changing conditions. However, data from melanoma also reveal a role of NRF2 in modulating differentiation and suppressing anti-tumor immunity. This review summarizes the function of NRF2 in this tumor entity and discusses the implications for current tumor therapies.


Subject(s)
Antioxidants/pharmacology , Gene Expression Regulation, Neoplastic , Immune Evasion , NF-E2-Related Factor 2/metabolism , Neoplasms/pathology , Oxidative Stress , Animals , Humans , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL