Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 66
1.
Nature ; 626(7998): 401-410, 2024 Feb.
Article En | MEDLINE | ID: mdl-38297129

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.


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.
Sci Signal ; 14(682)2021 05 11.
Article En | MEDLINE | ID: mdl-33975980

Members of the RAF family of serine-threonine kinases are intermediates in the mitogen-activated protein kinase and extracellular signal-regulated kinase (MAPK-ERK) signaling pathway, which controls key differentiation processes in B cells. By analyzing mice with B cell-specific deletion of Raf1, Braf, or both, we showed that Raf-1 and B-Raf acted together in mediating the positive selection of pre-B and transitional B cells as well as in initiating plasma cell differentiation. However, genetic or chemical inactivation of RAFs led to increased ERK phosphorylation in mature B cells. ERK activation in the absence of Raf-1 and B-Raf was mediated by multiple RAF-independent pathways, with phosphoinositide 3-kinase (PI3K) playing an important role. Furthermore, we found that ERK phosphorylation strongly increased during the transition from activated B cells to pre-plasmablasts. This increase in ERK phosphorylation did not occur in B cells lacking both Raf-1 and B-Raf, which most likely explains the partial block of plasma cell differentiation in mice lacking both RAFs. Collectively, our data indicate that B-Raf and Raf-1 are not necessary to mediate ERK phosphorylation in naïve or activated B cells but are essential for mediating the marked increase in ERK phosphorylation during the transition from activated B cells to pre-plasmablasts.


B-Lymphocytes/cytology , Extracellular Signal-Regulated MAP Kinases , Plasma Cells/cytology , Proto-Oncogene Proteins c-raf , Animals , Cell Differentiation , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Phosphatidylinositol 3-Kinases , Phosphorylation , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism
3.
Haematologica ; 105(4): 937-950, 2020 04.
Article En | MEDLINE | ID: mdl-31248967

Glutathione peroxidase 4 (GPX4) is unique as it is the only enzyme that can prevent detrimental lipid peroxidation in vivo by reducing lipid peroxides to the respective alcohols thereby stabilizing oxidation products of unsaturated fatty acids. During reticulocyte maturation, lipid peroxidation mediated by 15-lipoxygenase in humans and rabbits and by 12/15-lipoxygenase (ALOX15) in mice was considered the initiating event for the elimination of mitochondria but is now known to occur through mitophagy. Yet, genetic ablation of the Alox15 gene in mice failed to provide evidence for this hypothesis. We designed a different genetic approach to tackle this open conundrum. Since either other lipoxygenases or non-enzymatic autooxidative mechanisms may compensate for the loss of Alox15, we asked whether ablation of Gpx4 in the hematopoietic system would result in the perturbation of reticulocyte maturation. Quantitative assessment of erythropoiesis indices in the blood, bone marrow (BM) and spleen of chimeric mice with Gpx4 ablated in hematopoietic cells revealed anemia with an increase in the fraction of erythroid precursor cells and reticulocytes. Additional dietary vitamin E depletion strongly aggravated the anemic phenotype. Despite strong extramedullary erythropoiesis reticulocytes failed to mature and accumulated large autophagosomes with engulfed mitochondria. Gpx4-deficiency in hematopoietic cells led to systemic hepatic iron overload and simultaneous severe iron demand in the erythroid system. Despite extremely high erythropoietin and erythroferrone levels in the plasma, hepcidin expression remained unchanged. Conclusively, perturbed reticulocyte maturation in response to Gpx4 loss in hematopoietic cells thus causes ineffective erythropoiesis, a phenotype partially masked by dietary vitamin E supplementation.


Erythropoiesis , Iron , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Reticulocytes , Vitamin E , Animals , Homeostasis , Mice , Rabbits
4.
Cell Rep ; 29(9): 2731-2744.e4, 2019 Nov 26.
Article En | MEDLINE | ID: mdl-31775041

Aerobic organisms need to maintain cellular redox homeostasis. Glutathione peroxidase-4 (Gpx4) has the unique ability to protect cells against lipid peroxidation. Here, we show that Gpx4 is absolutely required to prevent ferroptosis during development, maintenance, and responses of innate-like B cells, namely, the B1 and marginal zone (MZ) B cells. In contrast, Gpx4 is dispensable for the development, germinal center reactions, and antibody responses of follicular B2 cells. Mechanistically, we show increased lipid metabolism and sensitivity to lipid peroxidation and ferroptosis in B1 and MZ B cells compared to follicular B2 cells, consistent with the requirement of Gpx4 in innate-like B cells. This high sensitivity to ferroptosis of innate-like B cells may be used to therapeutically target Gpx4 in certain forms of B cell malignancies involving B1 cells.


B-Lymphocytes/metabolism , Cytoskeletal Proteins/metabolism , Ferroptosis/drug effects , Glutathione Peroxidase/therapeutic use , Lipid Peroxidation/drug effects , Humans
5.
Eur J Immunol ; 49(5): 709-723, 2019 05.
Article En | MEDLINE | ID: mdl-30802940

Antioxidant systems maintain cellular redox homeostasis. The thioredoxin-1 (Trx1) and the glutathione (GSH)/glutaredoxin-1 (Grx1) systems are key players in preserving cytosolic redox balance. In fact, T lymphocytes critically rely on reducing equivalents from the Trx1 system for DNA biosynthesis during metabolic reprogramming upon activation. We here show that the Trx1 system is also indispensable for development and functionality of marginal zone (MZ) B cells and B1 cells in mice. In contrast, development of conventional B cells, follicular B-cell homeostasis, germinal center reactions, and antibody responses are redundantly sustained by both antioxidant pathways. Proliferating B2 cells lacking Txnrd1 have increased glutathione (GSH) levels and upregulated cytosolic Grx1, which is barely detectable in expanding thymocytes. These results suggest that the redox capacity driving proliferation is more robust and flexible in B cells than in T cells, which may have profound implications for the therapy of B and T-cell neoplasms.


B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Glutaredoxins/genetics , Thioredoxins/genetics , Animals , B-Lymphocytes/cytology , Biomarkers , Cell Proliferation/genetics , Germinal Center/immunology , Germinal Center/metabolism , Glutaredoxins/metabolism , Mice , Mice, Transgenic , Thioredoxins/metabolism
6.
Blood ; 127(1): 139-48, 2016 Jan 07.
Article En | MEDLINE | ID: mdl-26463424

Maintaining cellular redox balance is vital for cell survival and tissue homoeostasis because imbalanced production of reactive oxygen species (ROS) may lead to oxidative stress and cell death. The antioxidant enzyme glutathione peroxidase 4 (Gpx4) is a key regulator of oxidative stress-induced cell death. We show that mice with deletion of Gpx4 in hematopoietic cells develop anemia and that Gpx4 is essential for preventing receptor-interacting protein 3 (RIP3)-dependent necroptosis in erythroid precursor cells. Absence of Gpx4 leads to functional inactivation of caspase 8 by glutathionylation, resulting in necroptosis, which occurs independently of tumor necrosis factor α activation. Although genetic ablation of Rip3 normalizes reticulocyte maturation and prevents anemia, ROS accumulation and lipid peroxidation in Gpx4-deficient cells remain high. Our results demonstrate that ROS and lipid hydroperoxides function as not-yet-recognized unconventional upstream signaling activators of RIP3-dependent necroptosis.


Apoptosis , Erythroid Cells/pathology , Glutathione Peroxidase/physiology , Necrosis , Oxidative Stress , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Animals , Blotting, Western , Cell Differentiation , Cell Proliferation , Cells, Cultured , Erythroid Cells/metabolism , Flow Cytometry , Humans , Immunoenzyme Techniques , Mice , Mice, Knockout , Phospholipid Hydroperoxide Glutathione Peroxidase , Reactive Oxygen Species/metabolism
7.
J Exp Med ; 212(4): 555-68, 2015 Apr 06.
Article En | MEDLINE | ID: mdl-25824823

The selenoenzyme glutathione peroxidase 4 (Gpx4) is a major scavenger of phospholipid hydroperoxides. Although Gpx4 represents a key component of the reactive oxygen species-scavenging network, its relevance in the immune system is yet to be defined. Here, we investigated the importance of Gpx4 for physiological T cell responses by using T cell-specific Gpx4-deficient mice. Our results revealed that, despite normal thymic T cell development, CD8(+) T cells from T(ΔGpx4/ΔGpx4) mice had an intrinsic defect in maintaining homeostatic balance in the periphery. Moreover, both antigen-specific CD8(+) and CD4(+) T cells lacking Gpx4 failed to expand and to protect from acute lymphocytic choriomeningitis virus and Leishmania major parasite infections, which were rescued with diet supplementation of high dosage of vitamin E. Notably, depletion of the Gpx4 gene in the memory phase of viral infection did not affect T cell recall responses upon secondary infection. Ex vivo, Gpx4-deficient T cells rapidly accumulated membrane lipid peroxides and concomitantly underwent cell death driven by ferroptosis but not necroptosis. These studies unveil an essential role of Gpx4 for T cell immunity.


CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Leishmania major/immunology , Leishmaniasis, Cutaneous/immunology , Lipid Peroxidation/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Animals , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Cell Death/genetics , Cell Death/immunology , Cell Membrane/genetics , Cell Membrane/immunology , Glutathione Peroxidase/genetics , Glutathione Peroxidase/immunology , Immunologic Memory/drug effects , Immunologic Memory/genetics , Leishmaniasis, Cutaneous/genetics , Leishmaniasis, Cutaneous/pathology , Lipid Peroxidation/genetics , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/pathology , Mice , Mice, Knockout , Phospholipid Hydroperoxide Glutathione Peroxidase , Thymus Gland/immunology , Thymus Gland/pathology , Vitamin E/pharmacology , Vitamins/pharmacology
8.
J Immunol ; 194(4): 1434-45, 2015 Feb 15.
Article En | MEDLINE | ID: mdl-25595783

Elevated levels of the transcription factor c-myc are strongly associated with various cancers, and in particular B cell lymphomas. Although many of c-MYC's functions have been elucidated, its effect on the presentation of Ag through the HLA class II pathway has not been reported previously. This is an issue of considerable importance, given the low immunogenicity of many c-MYC-positive tumors. We report in this paper that increased c-MYC expression has a negative effect on the ability of B cell lymphomas to functionally present Ags/peptides to CD4(+) T cells. This defect was associated with alterations in the expression of distinct cofactors as well as interactions of antigenic peptides with class II molecules required for the presentation of class II-peptide complexes and T cell engagement. Using early passage Burkitt's lymphoma (BL) tumors and transformed cells, we show that compared with B lymphoblasts, BL cells express decreased levels of the class II editor HLA-DM, lysosomal thiol-reductase GILT, and a 47-kDa enolase-like protein. Functional Ag presentation was partially restored in BL cells treated with a c-MYC inhibitor, demonstrating the impact of this oncogene on Ag recognition. This restoration of HLA class II-mediated Ag presentation in early passage BL tumors/cells was linked to enhanced HLA-DM expression and a concurrent decrease in HLA-DO in BL cells. Taken together, these results reveal c-MYC exerts suppressive effects at several critical checkpoints in Ag presentation, which contribute to the immunoevasive properties of BL tumors.


Antigen Presentation/immunology , Histocompatibility Antigens Class II/immunology , Lymphoma, B-Cell/immunology , Proto-Oncogene Proteins c-myc/immunology , Tumor Escape/immunology , Blotting, Western , Flow Cytometry , Humans , Mass Spectrometry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tumor Cells, Cultured
9.
Nat Cell Biol ; 16(12): 1180-91, 2014 Dec.
Article En | MEDLINE | ID: mdl-25402683

Ferroptosis is a non-apoptotic form of cell death induced by small molecules in specific tumour types, and in engineered cells overexpressing oncogenic RAS. Yet, its relevance in non-transformed cells and tissues is unexplored and remains enigmatic. Here, we provide direct genetic evidence that the knockout of glutathione peroxidase 4 (Gpx4) causes cell death in a pathologically relevant form of ferroptosis. Using inducible Gpx4(-/-) mice, we elucidate an essential role for the glutathione/Gpx4 axis in preventing lipid-oxidation-induced acute renal failure and associated death. We furthermore systematically evaluated a library of small molecules for possible ferroptosis inhibitors, leading to the discovery of a potent spiroquinoxalinamine derivative called Liproxstatin-1, which is able to suppress ferroptosis in cells, in Gpx4(-/-) mice, and in a pre-clinical model of ischaemia/reperfusion-induced hepatic damage. In sum, we demonstrate that ferroptosis is a pervasive and dynamic form of cell death, which, when impeded, promises substantial cytoprotection.


Acute Kidney Injury/pathology , Apoptosis , Glutathione Peroxidase/genetics , Quinoxalines/pharmacology , Reperfusion Injury/pathology , Spiro Compounds/pharmacology , Animals , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Cardiolipins/metabolism , Cell Line , Humans , Imidazoles/pharmacology , In Situ Nick-End Labeling , Indoles/pharmacology , Kidney/metabolism , Kidney/pathology , Lipid Peroxidation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Peroxidases/pharmacology , Phosphatidylcholines/metabolism , Phosphatidylethanolamines/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase
10.
PLoS Pathog ; 10(5): e1004068, 2014 May.
Article En | MEDLINE | ID: mdl-24853673

Polyclonal Epstein-Barr virus (EBV)-infected B cell line (lymphoblastoid cell lines; LCL)-stimulated T-cell preparations have been successfully used to treat EBV-positive post-transplant lymphoproliferative disorders (PTLD) in transplant recipients, but function and specificity of the CD4+ component are still poorly defined. Here, we assessed the tumor-protective potential of different CD4+ T-cell specificities in a PTLD-SCID mouse model. Injection of different virus-specific CD4+ T-cell clones showed that single specificities were capable of prolonging mouse survival and that the degree of tumor protection directly correlated with recognition of target cells in vitro. Surprisingly, some CD4+ T-cell clones promoted tumor development, suggesting that besides antigen recognition, still elusive functional differences exist among virus-specific T cells. Of several EBV-specific CD4+ T-cell clones tested, those directed against virion antigens proved most tumor-protective. However, enriching these specificities in LCL-stimulated preparations conferred no additional survival benefit. Instead, CD4+ T cells specific for unknown, probably self-antigens were identified as principal antitumoral effectors in LCL-stimulated T-cell lines. These results indicate that virion and still unidentified cellular antigens are crucial targets of the CD4+ T-cell response in this preclinical PTLD-model and that enriching the corresponding T-cell specificities in therapeutic preparations may enhance their clinical efficacy. Moreover, the expression in several EBV-negative B-cell lymphoma cell lines implies that these putative autoantigen(s) might also qualify as targets for T-cell-based immunotherapy of virus-negative B cell malignancies.


Autoantigens/immunology , CD4-Positive T-Lymphocytes/physiology , Herpesvirus 4, Human/immunology , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/virology , Peripheral Blood Stem Cell Transplantation/adverse effects , Animals , Cells, Cultured , Disease Models, Animal , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/transmission , Herpesvirus 4, Human/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, SCID , Postoperative Complications/immunology , Transplantation, Homologous/adverse effects
11.
Int J Cancer ; 135(5): 1153-64, 2014 Sep 01.
Article En | MEDLINE | ID: mdl-24500882

Adoptive T cell therapy is an important additional treatment option for malignant diseases resistant to chemotherapy. Using a murine high-grade B cell lymphoma model, we have addressed the question whether the B cell differentiation antigen CD19 can act as rejection antigen. CD19(-/-) mice inoculated with CD19(+) B cell lymphoma cells showed higher survival rates than WT mice and were protected against additional tumor challenge. T cell depletion prior to tumor transfer completely abolished the protective response. By heterotypic vaccination of CD19(-/-) mice against murine CD19, survival after tumor challenge was significantly increased. To define protective epitopes within the CD19 molecule, T cells collected from mice that had survived the tumor transfer were analyzed for IFNγ secretion in response to CD19-derived peptides. The majority of mice exhibited a CD4(+) T cell response to CD19 peptide 27, which was the most dominant epitope after CD19 vaccination. A peptide 27-specific CD4(+) T cell line protected CD19(-/-) mice against challenge with CD19(+) lymphoma and also cured a significant proportion of WT mice from recurrent disease in a model of minimal residual disease after chemotherapy. In conclusion, our data highlight CD19-specific CD4(+) T cells for adoptive T cell therapy of B cell lymphomas.


Antigens, CD19/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Lymphoma, B-Cell/therapy , Animals , Antigens, CD19/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lymphocyte Depletion , Lymphoma, B-Cell/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Tumor Cells, Cultured
12.
PLoS One ; 8(10): e77375, 2013.
Article En | MEDLINE | ID: mdl-24130880

Over-expression of the proto-oncogene c-MYC is frequently observed in a variety of tumors and is a hallmark of Burkitt´s lymphoma. The fact that many tumors are oncogene-addicted to c-MYC, renders c-MYC a powerful target for anti-tumor therapy. Using a xenogenic vaccination strategy by immunizing C57BL/6 mice with human c-MYC protein or non-homologous peptides, we show that the human c-MYC protein, despite its high homology between mouse and man, contains several immunogenic epitopes presented in the context of murine H2(b) haplotype. We identified an MHC class II-restricted CD4⁺ T-cell epitope and therein an MHC class I-restricted CD8⁺ T-cell epitope (SSPQGSPEPL) that, after prime/boost immunization, protected up to 25% of mice against a lethal lymphoma challenge. Lymphoma-rejecting animals contained MHC multimer-binding CD8⁺ cell within the peripheral blood and displayed in vivo cytolytic activity with specificity for SSPQGSPEPL. Taken together these data suggest that oncogenic c-MYC can be targeted with specific T-cells.


CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Lymphoma/prevention & control , Proto-Oncogene Proteins c-myc/immunology , Amino Acid Sequence , Animals , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/chemistry , Humans , Interferon-gamma/immunology , Lymphoma/immunology , Mice , Mice, Inbred C57BL , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/administration & dosage , Proto-Oncogene Proteins c-myc/chemistry , Vaccination
13.
Circ Res ; 113(4): 408-17, 2013 Aug 02.
Article En | MEDLINE | ID: mdl-23770613

RATIONALE: Growing evidence indicates that oxidative stress contributes markedly to endothelial dysfunction. The selenoenzyme glutathione peroxidase 4 (Gpx4) is an intracellular antioxidant enzyme important for the protection of membranes by its unique activity to reduce complex hydroperoxides in membrane bilayers and lipoprotein particles. Yet a role of Gpx4 in endothelial cell function has remained enigmatic. OBJECTIVE: To investigate the role of Gpx4 ablation and subsequent lipid peroxidation in the vascular compartment in vivo. METHODS AND RESULTS: Endothelium-specific deletion of Gpx4 had no obvious impact on normal vascular homeostasis, nor did it impair tumor-derived angiogenesis in mice maintained on a normal diet. In stark contrast, aortic explants from endothelium-specific Gpx4 knockout mice showed a markedly reduced number of endothelial branches in sprouting assays. To shed light onto this apparent discrepancy between the in vivo and ex vivo results, we depleted mice of a second antioxidant, vitamin E, which is normally absent under ex vivo conditions. Therefore, mice were fed a vitamin E-depleted diet for 6 weeks before endothelial deletion of Gpx4 was induced by 4-hydroxytamoxifen. Surprisingly, ≈80% of the knockout mice died. Histopathological analysis revealed detachment of endothelial cells from the basement membrane and endothelial cell death in multiple organs, which triggered thrombus formation. Thromboembolic events were the likely cause of various clinical pathologies, including heart failure, renal and splenic microinfarctions, and paraplegia. CONCLUSIONS: Here, we show for the first time that in the absence of Gpx4, sufficient vitamin E supplementation is crucial for endothelial viability.


Glutathione Peroxidase/deficiency , Glutathione Peroxidase/genetics , Thrombosis/etiology , Thrombosis/mortality , Vitamin E Deficiency/complications , Vitamin E/genetics , Animals , Apoptosis/physiology , Blood Pressure/physiology , Disease Models, Animal , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Female , Glutathione Peroxidase/metabolism , Heart Rate/physiology , Lipid Peroxidation/physiology , Male , Mice , Mice, Knockout , Mice, Transgenic , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/physiopathology , Oxidative Stress/physiology , Phospholipid Hydroperoxide Glutathione Peroxidase , Thrombosis/physiopathology , Vitamin E/metabolism , Vitamin E Deficiency/metabolism , Vitamin E Deficiency/physiopathology
14.
J Exp Med ; 210(7): 1331-50, 2013 Jul 01.
Article En | MEDLINE | ID: mdl-23752226

The basic helix-loop-helix transcription factor AP4/TFAP4/AP-4 is encoded by a c-MYC target gene and displays up-regulation concomitantly with c-MYC in colorectal cancer (CRC) and numerous other tumor types. Here a genome-wide characterization of AP4 DNA binding and mRNA expression was performed using a combination of microarray, genome-wide chromatin immunoprecipitation, next-generation sequencing, and bioinformatic analyses. Thereby, hundreds of induced and repressed AP4 target genes were identified. Besides many genes involved in the control of proliferation, the AP4 target genes included markers of stemness (LGR5 and CD44) and epithelial-mesenchymal transition (EMT) such as SNAIL, E-cadherin/CDH1, OCLN, VIM, FN1, and the Claudins 1, 4, and 7. Accordingly, activation of AP4 induced EMT and enhanced migration and invasion of CRC cells. Conversely, down-regulation of AP4 resulted in mesenchymal-epithelial transition and inhibited migration and invasion. In addition, AP4 induction was required for EMT, migration, and invasion caused by ectopic expression of c-MYC. Inhibition of AP4 in CRC cells resulted in decreased lung metastasis in mice. Elevated AP4 expression in primary CRC significantly correlated with liver metastasis and poor patient survival. These findings imply AP4 as a new regulator of EMT that contributes to metastatic processes in CRC and presumably other carcinomas.


Colorectal Neoplasms/pathology , DNA-Binding Proteins/physiology , Epithelial-Mesenchymal Transition/physiology , Transcription Factors/physiology , Animals , Caco-2 Cells , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/physiopathology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/biosynthesis , Epithelial-Mesenchymal Transition/genetics , HT29 Cells , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , RNA, Small Interfering/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/biosynthesis , Transplantation, Heterologous , Up-Regulation/genetics
15.
PLoS One ; 7(7): e42021, 2012.
Article En | MEDLINE | ID: mdl-22860051

BACKGROUND: A given tumor is usually dependent on the oncogene that is activated in the respective tumor entity. This phenomenon called oncogene addiction provides the rationale for attempts to target oncogene products in a therapeutic manner, be it by small molecules, by small interfering RNAs (siRNA) or by antigen-specific T cells. As the proto-oncogene product is required also for the function of normal cells, this raises the question whether there is a therapeutic window between the adverse effects of specific inhibitors or T cells to normal tissue that may limit their application, and their beneficial tumor-specific therapeutic action. To address this crucial question, suitable mouse strains need to be developed, that enable expression of the human proto-oncogene not only in tumor but also in normal cells. The aim of this work is to provide such a mouse strain for the human proto-oncogene product c-MYC. PRINCIPAL FINDINGS: We generated C57BL/6-derived embryonic stem cells that are transgenic for a humanized c-Myc gene and established a mouse strain (hc-Myc) that expresses human c-MYC instead of the murine ortholog. These transgenic animals harbor the humanized c-Myc gene integrated into the endogenous murine c-Myc locus. Despite the lack of the endogenous murine c-Myc gene, homozygous mice show a normal phenotype indicating that human c-MYC can replace its murine ortholog. CONCLUSIONS: The newly established hc-Myc mouse strain provides a model system to study in detail the adverse effects of therapies that target the human c-MYC protein. To mimic the clinical situation, hc-Myc mice may be cross-bred to mice that develop tumors due to overexpression of human c-MYC. With these double transgenic mice it will be possible to study simultaneously the therapeutic efficiency and adverse side effects of MYC-specific therapies in the same mouse.


Genes, myc , Proto-Oncogene Proteins c-myc/genetics , Animals , Blotting, Southern , Embryonic Stem Cells , Flow Cytometry , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Polymerase Chain Reaction , Proto-Oncogene Mas
16.
PLoS One ; 7(3): e34552, 2012.
Article En | MEDLINE | ID: mdl-22479645

To study mechanisms of T cell-mediated rejection of B cell lymphomas, we developed a murine lymphoma model wherein three potential rejection antigens, human c-MYC, chicken ovalbumin (OVA), and GFP are expressed. After transfer into wild-type mice 60-70% of systemically growing lymphomas expressing all three antigens were rejected; lymphomas expressing only human c-MYC protein were not rejected. OVA expressing lymphomas were infiltrated by T cells, showed MHC class I and II upregulation, and lost antigen expression, indicating immune escape. In contrast to wild-type recipients, 80-100% of STAT1-, IFN-γ-, or IFN-γ receptor-deficient recipients died of lymphoma, indicating that host IFN-γ signaling is critical for rejection. Lymphomas arising in IFN-γ- and IFN-γ-receptor-deficient mice had invariably lost antigen expression, suggesting that poor overall survival of these recipients was due to inefficient elimination of antigen-negative lymphoma variants. Antigen-dependent eradication of lymphoma cells in wild-type animals was dependent on cross-presentation of antigen by cells of the tumor stroma. These findings provide first evidence for an important role of the tumor stroma in T cell-mediated control of hematologic neoplasias and highlight the importance of incorporating stroma-targeting strategies into future immunotherapeutic approaches.


Antigens/immunology , Interferon-gamma/immunology , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Animals , Antigens/genetics , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Chickens , Gene Expression Regulation, Neoplastic , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , Humans , Lymphoma, B-Cell/genetics , Mice , Mice, Inbred C57BL , Ovalbumin/genetics , Ovalbumin/immunology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/immunology , Signal Transduction
18.
PLoS Genet ; 8(3): e1002573, 2012.
Article En | MEDLINE | ID: mdl-22438825

c-Myc (hereafter called Myc) belongs to a family of transcription factors that regulates cell growth, cell proliferation, and differentiation. Myc initiates the transcription of a large cast of genes involved in cell growth by stimulating metabolism and protein synthesis. Some of these, like those involved in glycolysis, may be part of the Warburg effect, which is defined as increased glucose uptake and lactate production in the presence of adequate oxygen supply. In this study, we have taken a mouse-genetics approach to challenge the role of select Myc-regulated metabolic enzymes in tumorigenesis in vivo. By breeding λ-Myc transgenic mice, Apc(Min) mice, and p53 knockout mice with mouse models carrying inactivating alleles of Lactate dehydrogenase A (Ldha), 3-Phosphoglycerate dehydrogenase (Phgdh) and Serine hydroxymethyltransferase 1 (Shmt1), we obtained offspring that were monitored for tumor development. Very surprisingly, we found that these genes are dispensable for tumorigenesis in these genetic settings. However, experiments in fibroblasts and colon carcinoma cells expressing oncogenic Ras show that these cells are sensitive to Ldha knockdown. Our genetic models reveal cell context dependency and a remarkable ability of tumor cells to adapt to alterations in critical metabolic pathways. Thus, to achieve clinical success, it will be of importance to correctly stratify patients and to find synthetic lethal combinations of inhibitors targeting metabolic enzymes.


Glycine Hydroxymethyltransferase , L-Lactate Dehydrogenase , Lymphoma, B-Cell/metabolism , Phosphoglycerate Dehydrogenase , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Adenomatous Polyposis Coli Protein/genetics , Animals , Cell Transformation, Neoplastic , Disease Models, Animal , Fibroblasts , Gene Expression Regulation, Neoplastic , Genes, ras/genetics , Glycine Hydroxymethyltransferase/genetics , Glycine Hydroxymethyltransferase/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Lactate Dehydrogenase 5 , Lymphoma, B-Cell/genetics , Metabolic Networks and Pathways , Mice , Mice, Inbred C57BL , Mice, Transgenic , NIH 3T3 Cells , Oligonucleotide Array Sequence Analysis , Phosphoglycerate Dehydrogenase/genetics , Phosphoglycerate Dehydrogenase/metabolism , Tumor Suppressor Protein p53/genetics
19.
Eur J Cell Biol ; 91(1): 31-5, 2012 Jan.
Article En | MEDLINE | ID: mdl-21458882

Epstein-Barr virus (EBV) establishes lifelong persistent infections in humans and has been implicated in the pathogenesis of several human malignancies. Protective immunity against EBV is mediated by T cells, as indicated by an increased incidence of EBV-associated malignancies in immunocompromised patients, and by the successful treatment of EBV-associated post-transplant lymphoproliferative disease (PTLD) in transplant recipients by the infusion of polyclonal EBV-specific T cell lines. To implement this treatment modality as a conventional therapeutic option, and to extend this protocol to other EBV-associated diseases, generic and more direct approaches for the generation of EBV-specific T cell lines enriched in disease-relevant specificities need to be developed. To this aim, we studied the poorly defined EBV-specific CD4+ T cell response during acute and chronic infection.


Adaptive Immunity , Adoptive Transfer/methods , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Epstein-Barr Virus Infections/immunology , Genome, Viral , Herpesvirus 4, Human/immunology , Lymphoproliferative Disorders/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/virology , CD4-Positive T-Lymphocytes/transplantation , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/virology , Cell Line , Cloning, Molecular , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/therapy , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Nuclear Antigens/immunology , Genomic Library , Humans , Lymphocyte Transfusion , Lymphoproliferative Disorders/etiology , Lymphoproliferative Disorders/therapy , Lymphoproliferative Disorders/virology , Mice , Recombinant Proteins/immunology , Virion/immunology
20.
Eur Heart J ; 32(9): 1121-33, 2011 May.
Article En | MEDLINE | ID: mdl-21247928

AIMS: Cardiac energy requirement is met to a large extent by oxidative phosphorylation in mitochondria that are highly abundant in cardiac myocytes. Human mitochondrial thioredoxin reductase (TXNRD2) is a selenocysteine-containing enzyme essential for mitochondrial oxygen radical scavenging. Cardiac-specific deletion of Txnrd2 in mice results in dilated cardiomyopathy (DCM). The aim of this study was to investigate whether TXNRD2 mutations explain a fraction of monogenic DCM cases. METHODS AND RESULTS: Sequencing and subsequent genotyping of TXNRD2 in patients diagnosed with DCM (n = 227) and in DCM-free (n = 683) individuals from the general population sample KORA S4 was performed. The functional impact of observed mutations on Txnrd2 function was tested in mouse fibroblasts. We identified two novel amino acid residue-altering TXNRD2 mutations [175G > A (Ala59Thr) and 1124G > A (Gly375Arg)] in three heterozygous carriers among 227 patients that were not observed in the 683 DCM-free individuals. Both DCM-associated mutations result in amino acid substitutions of highly conserved residues in helices contributing to the flavin-adenine dinucleotide (FAD)-binding domain of TXNRD2. Functional analysis of both mutations in Txnrd2(-/-) mouse fibroblasts revealed that contrasting to wild-type (wt) Txnrd2, neither mutant did restore Txnrd2 function. Mutants even impaired the survival of Txnrd2 wt cells under oxidative stress by a dominant-negative mechanism. CONCLUSION: For the first time, we describe mutations in DCM patients in a gene involved in the regulation of cellular redox state. TXNRD2 mutations may explain a fraction of human DCM disease burden.


Cardiomyopathy, Dilated/genetics , Mutation/genetics , Thioredoxin Reductase 2/genetics , Aged , Amino Acid Substitution/genetics , Animals , Cardiomyopathy, Dilated/enzymology , Cells, Cultured , Female , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Genotype , Heterozygote , Homeostasis/physiology , Humans , Immunoblotting , Male , Mice , Microscopy, Electron , Middle Aged , Mitochondria/enzymology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Protein Conformation , Reactive Oxygen Species/metabolism
...