99 results on '"Meierjohann S"'
Search Results
2. Cystathionase mediates senescence evasion in melanocytes and melanoma cells
- Author
-
Leikam, C, Hufnagel, A, Walz, S, Kneitz, S, Fekete, A, Müller, M J, Eilers, M, Schartl, M, and Meierjohann, S
- Published
- 2014
- Full Text
- View/download PDF
3. Hyperactivation of constitutively dimerized oncogenic EGF receptors by autocrine loops
- Author
-
Laisney, J A G C, Mueller, T D, Schartl, M, and Meierjohann, S
- Published
- 2013
- Full Text
- View/download PDF
4. Oncogene activation in melanocytes links reactive oxygen to multinucleated phenotype and senescence
- Author
-
Leikam, C, Hufnagel, A, Schartl, M, and Meierjohann, S
- Published
- 2008
- Full Text
- View/download PDF
5. MEK Inhibition Can Protect Melanoma Cell Lines from Apoptosis: P-085
- Author
-
Haydn, J., Maurus, K., Schartl, M., and Meierjohann, S.
- Published
- 2013
6. Multicentre study on standardisation of melanoma cell culture - an initiative of the German Melanoma Research Network
- Author
-
Eberle, J., Spangler, B., Becker, J. C., Heinemann, S. H., Klein, C. A., Kunz, M., Kuphal, S., Langer, P., Mauch, C., Meierjohann, S., Paschen, A., Schadendorf, D., Schartl, M., Schittek, B., Schönherr, R., Tüting, T., Zigrino, P., and Bosserhoff, A. K.
- Published
- 2010
- Full Text
- View/download PDF
7. In vitro evidence for senescent multinucleated melanocytes as a source for tumor-initiating cells
- Author
-
Leikam, C, primary, Hufnagel, A L, additional, Otto, C, additional, Murphy, D J, additional, Mühling, B, additional, Kneitz, S, additional, Nanda, I, additional, Schmid, M, additional, Wagner, T U, additional, Haferkamp, S, additional, Bröcker, E-B, additional, Schartl, M, additional, and Meierjohann, S, additional
- Published
- 2015
- Full Text
- View/download PDF
8. MMP13 mediates cell cycle progression in melanocytes and melanoma cells: in vitro studies of migration and proliferation.
- Author
-
Meierjohann, S., Hufnagel, A., Wende, E., Kleinschmidt, M.A., Wolf, K. van der, Friedl, P.H.A., Gaubatz, S., Schartl, M., Meierjohann, S., Hufnagel, A., Wende, E., Kleinschmidt, M.A., Wolf, K. van der, Friedl, P.H.A., Gaubatz, S., and Schartl, M.
- Abstract
Contains fulltext : 87376.pdf (publisher's version ) (Open Access), BACKGROUND: Melanoma cells are usually characterized by a strong proliferative potential and efficient invasive migration. Among the multiple molecular changes that are recorded during progression of this disease, aberrant activation of receptor tyrosine kinases (RTK) is often observed. Activation of matrix metalloproteases goes along with RTK activation and usually enhances RTK-driven migration. The purpose of this study was to examine RTK-driven three-dimensional migration of melanocytes and the pro-tumorigenic role of matrix metalloproteases for melanocytes and melanoma cells. RESULTS: Using experimental melanocyte dedifferentiation as a model for early melanomagenesis we show that an activated EGF receptor variant potentiates migration through three-dimensional fibrillar collagen. EGFR stimulation also resulted in a strong induction of matrix metalloproteases in a MAPK-dependent manner. However, neither MAPK nor MMP activity were required for migration, as the cells migrated in an entirely amoeboid mode. Instead, MMPs fulfilled a function in cell cycle regulation, as their inhibition resulted in strong growth inhibition of melanocytes. The same effect was observed in the human melanoma cell line A375 after stimulation with FCS. Using sh- and siRNA techniques, we could show that MMP13 is the protease responsible for this effect. Along with decreased proliferation, knockdown of MMP13 strongly enhanced pigmentation of melanocytes. CONCLUSIONS: Our data show for the first time that growth stimuli are mediated via MMP13 in melanocytes and melanoma, suggesting an autocrine MMP13-driven loop. Given that MMP13-specific inhibitors are already developed, these results support the evaluation of these inhibitors in the treatment of melanoma.
- Published
- 2010
9. Cystathionase mediates senescence evasion in melanocytes and melanoma cells
- Author
-
Leikam, C, primary, Hufnagel, A, additional, Walz, S, additional, Kneitz, S, additional, Fekete, A, additional, Müller, M J, additional, Eilers, M, additional, Schartl, M, additional, and Meierjohann, S, additional
- Published
- 2013
- Full Text
- View/download PDF
10. Hyperactivation of constitutively dimerized oncogenic EGF receptors by autocrine loops
- Author
-
Laisney, J A G C, primary, Mueller, T D, additional, Schartl, M, additional, and Meierjohann, S, additional
- Published
- 2012
- Full Text
- View/download PDF
11. From Mendelian to molecular genetics: the Xiphophorus melanoma model
- Author
-
MEIERJOHANN, S, primary and SCHARTL, M, additional
- Published
- 2006
- Full Text
- View/download PDF
12. The generation of melanoma model in zebrafish
- Author
-
Werner, M., primary, Schartl, M., additional, and Meierjohann, S., additional
- Published
- 2006
- Full Text
- View/download PDF
13. Amoeboid migration in transformed melanocytes
- Author
-
Meierjohann, S., primary, Wende, E., additional, Kraiss, A., additional, Wolf, K., additional, Friedl, P., additional, and Schartl, M., additional
- Published
- 2006
- Full Text
- View/download PDF
14. Thioredoxin and glutathione system of malaria parasitePlasmodium falciparum
- Author
-
M�ller, S., primary, Gilberger, T. W., additional, Krnajski, Z., additional, L�ersen, K., additional, Meierjohann, S., additional, and Walter, R. D., additional
- Published
- 2001
- Full Text
- View/download PDF
15. MMP13 mediates cell cycle progression in melanocytes and melanoma cells: in vitro studies of migration and proliferation
- Author
-
Friedl Peter, Wolf Katarina, Kleinschmidt Markus A, Wende Elisabeth, Hufnagel Anita, Meierjohann Svenja, Gaubatz Stefan, and Schartl Manfred
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Melanoma cells are usually characterized by a strong proliferative potential and efficient invasive migration. Among the multiple molecular changes that are recorded during progression of this disease, aberrant activation of receptor tyrosine kinases (RTK) is often observed. Activation of matrix metalloproteases goes along with RTK activation and usually enhances RTK-driven migration. The purpose of this study was to examine RTK-driven three-dimensional migration of melanocytes and the pro-tumorigenic role of matrix metalloproteases for melanocytes and melanoma cells. Results Using experimental melanocyte dedifferentiation as a model for early melanomagenesis we show that an activated EGF receptor variant potentiates migration through three-dimensional fibrillar collagen. EGFR stimulation also resulted in a strong induction of matrix metalloproteases in a MAPK-dependent manner. However, neither MAPK nor MMP activity were required for migration, as the cells migrated in an entirely amoeboid mode. Instead, MMPs fulfilled a function in cell cycle regulation, as their inhibition resulted in strong growth inhibition of melanocytes. The same effect was observed in the human melanoma cell line A375 after stimulation with FCS. Using sh- and siRNA techniques, we could show that MMP13 is the protease responsible for this effect. Along with decreased proliferation, knockdown of MMP13 strongly enhanced pigmentation of melanocytes. Conclusions Our data show for the first time that growth stimuli are mediated via MMP13 in melanocytes and melanoma, suggesting an autocrine MMP13-driven loop. Given that MMP13-specific inhibitors are already developed, these results support the evaluation of these inhibitors in the treatment of melanoma.
- Published
- 2010
- Full Text
- View/download PDF
16. Gene expression analysis after receptor tyrosine kinase activation reveals new potential melanoma proteins
- Author
-
Krause Michael, Samans Birgit, Haydn Johannes M, Teutschbein Janka, Eilers Martin, Schartl Manfred, and Meierjohann Svenja
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Melanoma is an aggressive tumor with increasing incidence. To develop accurate prognostic markers and targeted therapies, changes leading to malignant transformation of melanocytes need to be understood. In the Xiphophorus melanoma model system, a mutated version of the EGF receptor Xmrk (Xiphophorus melanoma receptor kinase) triggers melanomagenesis. Cellular events downstream of Xmrk, such as the activation of Akt, Ras, B-Raf or Stat5, were also shown to play a role in human melanomagenesis. This makes the elucidation of Xmrk downstream targets a useful method for identifying processes involved in melanoma formation. Methods Here, we analyzed Xmrk-induced gene expression using a microarray approach. Several highly expressed genes were confirmed by realtime PCR, and pathways responsible for their induction were revealed using small molecule inhibitors. The expression of these genes was also monitored in human melanoma cell lines, and the target gene FOSL1 was knocked down by siRNA. Proliferation and migration of siRNA-treated melanoma cell lines were then investigated. Results Genes with the strongest upregulation after receptor activation were FOS-like antigen 1 (Fosl1), early growth response 1 (Egr1), osteopontin (Opn), insulin-like growth factor binding protein 3 (Igfbp3), dual-specificity phosphatase 4 (Dusp4), and tumor-associated antigen L6 (Taal6). Interestingly, most genes were blocked in presence of a SRC kinase inhibitor. Importantly, we found that FOSL1, OPN, IGFBP3, DUSP4, and TAAL6 also exhibited increased expression levels in human melanoma cell lines compared to human melanocytes. Knockdown of FOSL1 in human melanoma cell lines reduced their proliferation and migration. Conclusion Altogether, the data show that the receptor tyrosine kinase Xmrk is a useful tool in the identification of target genes that are commonly expressed in Xmrk-transgenic melanocytes and melanoma cell lines. The identified molecules constitute new possible molecular players in melanoma development. Specifically, a role of FOSL1 in melanomagenic processes is demonstrated. These data are the basis for future detailed analyses of the investigated target genes.
- Published
- 2010
- Full Text
- View/download PDF
17. Lineage-specific co-evolution of the Egf receptor/ligand signaling system
- Author
-
Meierjohann Svenja, Walter Ronald B, Braasch Ingo, Laisney Juliette AGC, and Schartl Manfred
- Subjects
Evolution ,QH359-425 - Abstract
Abstract Background The epidermal growth factor receptor (Egfr) with its numerous ligands has fundamental roles in development, cell differentiation and physiology. Dysfunction of the receptor-ligand system contributes to many human malignancies. Consistent with such various tasks, the Egfr gene family has expanded during vertebrate evolution as a consequence of several rounds of whole genome duplication. Of particular interest is the effect of the fish-specific whole genome duplication (FSGD) on the ligand-receptor system, as it has supplied this largest group of vertebrates with additional opportunities for sub- and/or neofunctionalization in this signaling system. Results We identified the predicted components of the Egf receptor-ligand signaling system in teleost fishes (medaka, platyfish, stickleback, pufferfishes and zebrafish). We found two duplicated egfr genes, egfra and egfrb, in all available teleost genomes. Surprisingly only one copy for each of the seven Egfr ligands could be identified in most fishes, with zebrafish hbegf being the only exception. Special focus was put on medaka, for which we more closely investigated all Egf receptors and Egfr ligands. The different expression patterns of egfra, egfrb and their ligands in medaka tissues and embryo stages suggest differences in role and function. Preferential co-expression of different subsets of Egfr ligands corroborates the possible subfunctionalization and specialization of the two receptors in adult tissues. Bioinformatic analyses of the ligand-receptor interface between Egfr and its ligands show a very weak evolutionary conservation within this region. Using in vitro analyses of medaka Egfra, we could show that this receptor is only activated by medaka ligands, but not by human EGF. Altogether, our data suggest a lineage-specific Egfr/Egfr ligand co-evolution. Conclusions Our data indicate that medaka Egfr signaling occurs via its two copies, Egfra and Egfrb, each of them being preferentially coexpressed with different subsets of Egfr ligands. This fish-specific occurrence of Egf receptor specialization offers unique opportunities to study the functions of different Egf receptor-ligand combinations and their biological outputs in vertebrates. Furthermore, our results strongly support the use of homologous ligands in future studies, as sufficient cross-specificity is very unlikely for this ligand/receptor system.
- Published
- 2010
- Full Text
- View/download PDF
18. Gene expression analysis after receptor tyrosine kinase activation reveals new potential melanoma proteins.
- Author
-
Teutschbein J, Haydn JM, Samans B, Krause M, Eilers M, Schartl M, Meierjohann S, Teutschbein, Janka, Haydn, Johannes M, Samans, Birgit, Krause, Michael, Eilers, Martin, Schartl, Manfred, and Meierjohann, Svenja
- Abstract
Background: Melanoma is an aggressive tumor with increasing incidence. To develop accurate prognostic markers and targeted therapies, changes leading to malignant transformation of melanocytes need to be understood. In the Xiphophorus melanoma model system, a mutated version of the EGF receptor Xmrk (Xiphophorus melanoma receptor kinase) triggers melanomagenesis. Cellular events downstream of Xmrk, such as the activation of Akt, Ras, B-Raf or Stat5, were also shown to play a role in human melanomagenesis. This makes the elucidation of Xmrk downstream targets a useful method for identifying processes involved in melanoma formation.Methods: Here, we analyzed Xmrk-induced gene expression using a microarray approach. Several highly expressed genes were confirmed by realtime PCR, and pathways responsible for their induction were revealed using small molecule inhibitors. The expression of these genes was also monitored in human melanoma cell lines, and the target gene FOSL1 was knocked down by siRNA. Proliferation and migration of siRNA-treated melanoma cell lines were then investigated.Results: Genes with the strongest upregulation after receptor activation were FOS-like antigen 1 (Fosl1), early growth response 1 (Egr1), osteopontin (Opn), insulin-like growth factor binding protein 3 (Igfbp3), dual-specificity phosphatase 4 (Dusp4), and tumor-associated antigen L6 (Taal6). Interestingly, most genes were blocked in presence of a SRC kinase inhibitor. Importantly, we found that FOSL1, OPN, IGFBP3, DUSP4, and TAAL6 also exhibited increased expression levels in human melanoma cell lines compared to human melanocytes. Knockdown of FOSL1 in human melanoma cell lines reduced their proliferation and migration.Conclusion: Altogether, the data show that the receptor tyrosine kinase Xmrk is a useful tool in the identification of target genes that are commonly expressed in Xmrk-transgenic melanocytes and melanoma cell lines. The identified molecules constitute new possible molecular players in melanoma development. Specifically, a role of FOSL1 in melanomagenic processes is demonstrated. These data are the basis for future detailed analyses of the investigated target genes. [ABSTRACT FROM AUTHOR]- Published
- 2010
- Full Text
- View/download PDF
19. Functional test of a naturally occurred tumor modifier gene provides insights to melanoma development.
- Author
-
Garcia-Olazabal M, Adolfi MC, Wilde B, Hufnagel A, Paudel R, Lu Y, Meierjohann S, Rosenthal GG, and Schartl M
- Abstract
Occurrence of degenerative interactions is thought to serve as a mechanism underlying hybrid unfitness in most animal systems. 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 Xiphophorus birchmanni (X. birchmanni) and Xiphophorus malinche (X. malinche). We hypothesized that 1 of the 2 parental alleles of adgre5 is involved in regulation of cell growth, migration, and melanomagenesis. Accordingly, we assessed the function of adgre5 alleles from each parental species of the melanoma-bearing hybrids using in vitro cell growth 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 growth 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 provide evidence that adgre5 is a natural melanoma suppressor and provide new insight in melanoma etiology., Competing Interests: Conflicts of interest: The author(s) declare no conflicts of interest., (© The Author(s) 2025. Published by Oxford University Press on behalf of The Genetics Society of America.)
- Published
- 2025
- Full Text
- View/download PDF
20. Loss of p14 diminishes immunogenicity in melanoma via non-canonical Wnt signaling by reducing the peptide surface density.
- Author
-
Wohlfarth J, Kosnopfel C, Faber D, Berthold M, Siedel C, Bernhardt M, Schlosser A, Aprati T, Liu D, Schrama D, Houben R, Schadendorf D, Goebeler M, Meierjohann S, and Schilling B
- Subjects
- Humans, Cell Line, Tumor, Peptides immunology, Peptides metabolism, Histocompatibility Antigens Class I metabolism, Histocompatibility Antigens Class I immunology, Histocompatibility Antigens Class I genetics, Wnt-5a Protein metabolism, Wnt-5a Protein genetics, Wnt-5a Protein immunology, Antigen Presentation immunology, T-Lymphocytes immunology, T-Lymphocytes metabolism, Antigens, Neoplasm immunology, Antigens, Neoplasm metabolism, Antigens, Neoplasm genetics, Melanoma immunology, Melanoma pathology, Melanoma metabolism, Melanoma genetics, Wnt Signaling Pathway immunology, Tumor Suppressor Protein p14ARF metabolism, Tumor Suppressor Protein p14ARF genetics
- 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 p14
ARF 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., (© 2024 The Authors. Molecular Oncology published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)- Published
- 2024
- Full Text
- View/download PDF
21. Functional Test of a Naturally Occurred Tumor Modifier Gene Provides Insights to Melanoma Development.
- Author
-
Garcia-Olazabal M, Adolfi MC, Wilde B, Hufnagel A, Paudel R, Lu Y, Meierjohann S, Rosenthal GG, and Schartl M
- 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.
- Published
- 2024
- Full Text
- View/download PDF
22. Messing with cancer therapy: how the melanoma phenotype predicts checkpoint inhibitor response.
- Author
-
Meierjohann S and Bertolotto C
- Subjects
- Humans, Antibodies, Monoclonal, Phenotype, Melanoma drug therapy, Melanoma genetics, Skin Neoplasms
- Published
- 2024
- Full Text
- View/download PDF
23. Thiol starvation triggers melanoma state switching in an ATF4 and NRF2-dependent manner.
- Author
-
Meinert M, Jessen C, Hufnagel A, Kreß JKC, Burnworth M, Däubler T, Gallasch T, Xavier da Silva TN, Dos Santos AF, Ade CP, Schmitz W, Kneitz S, Friedmann Angeli JP, and Meierjohann S
- Subjects
- Mice, Animals, Humans, Cystine, Sulfhydryl Compounds, 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, Cysteine metabolism, Melanoma genetics
- 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; Braf
CA ; 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., Competing Interests: Declaration of competing interest The authors report no declarations of interest., (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
24. 7-Dehydrocholesterol is an endogenous suppressor of ferroptosis.
- Author
-
Freitas FP, Alborzinia H, Dos Santos AF, Nepachalovich P, Pedrera L, Zilka O, Inague A, Klein C, Aroua N, Kaushal K, Kast B, Lorenz SM, Kunz V, Nehring H, Xavier da Silva TN, Chen Z, Atici S, Doll SG, Schaefer EL, Ekpo I, Schmitz W, Horling A, Imming P, Miyamoto S, Wehman AM, Genaro-Mattos TC, Mirnics K, Kumar L, Klein-Seetharaman J, Meierjohann S, Weigand I, Kroiss M, Bornkamm GW, Gomes F, Netto LES, Sathian MB, Konrad DB, Covey DF, Michalke B, Bommert K, Bargou RC, Garcia-Saez A, Pratt DA, Fedorova M, Trumpp A, Conrad M, and Friedmann Angeli JP
- Subjects
- Animals, Humans, Cell Survival, Lipid Peroxidation, Neoplasm Transplantation, Oxidation-Reduction, Phenotype, Reproducibility of Results, Burkitt Lymphoma metabolism, Burkitt Lymphoma pathology, Dehydrocholesterols metabolism, Ferroptosis, Neuroblastoma metabolism, Neuroblastoma pathology
- 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 oxidation
1,2 . 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., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2024
- Full Text
- View/download PDF
25. Multimodal treatment and immune checkpoint inhibition in sinonasal mucosal melanoma: real-world data of a retrospective, single-center study.
- Author
-
Scherzad A, Stöth M, Meyer TJ, Haug L, Gehrke T, Schilling B, Meierjohann S, Scheich M, Hagen R, Gesierich A, and Hackenberg S
- Subjects
- Humans, Immune Checkpoint Inhibitors therapeutic use, Retrospective Studies, Combined Modality Therapy, Melanoma drug therapy, Melanoma genetics, Paranasal Sinus Neoplasms drug therapy, Paranasal Sinus Neoplasms genetics
- 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., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
26. LRP8-mediated selenocysteine uptake is a targetable vulnerability in MYCN-amplified neuroblastoma.
- Author
-
Alborzinia H, Chen Z, Yildiz U, Freitas FP, Vogel FCE, Varga JP, Batani J, Bartenhagen C, Schmitz W, Büchel G, Michalke B, Zheng J, Meierjohann S, Girardi E, Espinet E, Flórez AF, Dos Santos AF, Aroua N, Cheytan T, Haenlin J, Schlicker L, Xavier da Silva TN, Przybylla A, Zeisberger P, Superti-Furga G, Eilers M, Conrad M, Fabiano M, Schweizer U, Fischer M, Schulze A, Trumpp A, and Friedmann Angeli JP
- Subjects
- Humans, Cell Line, Tumor, N-Myc Proto-Oncogene Protein genetics, N-Myc Proto-Oncogene Protein metabolism, Selenocysteine therapeutic use, Animals, Ferroptosis, Neuroblastoma genetics, Neuroblastoma drug therapy
- 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., (© 2023 The Authors. Published under the terms of the CC BY 4.0 license.)- Published
- 2023
- Full Text
- View/download PDF
27. The integrated stress response effector ATF4 is an obligatory metabolic activator of NRF2.
- Author
-
Kreß JKC, Jessen C, Hufnagel A, Schmitz W, Xavier da Silva TN, Ferreira Dos Santos A, Mosteo L, Goding CR, Friedmann Angeli JP, and Meierjohann S
- Subjects
- Humans, Cystine metabolism, Glutathione metabolism, Oxidative Stress, Activating Transcription Factor 4 genetics, Activating Transcription Factor 4 metabolism, Neoplasms, NF-E2-Related Factor 2 metabolism
- 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., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
28. Predicting Microenvironment in CXCR4- and FAP-Positive Solid Tumors-A Pan-Cancer Machine Learning Workflow for Theranostic Target Structures.
- Author
-
Marquardt A, Hartrampf P, Kollmannsberger P, Solimando AG, Meierjohann S, Kübler H, Bargou R, Schilling B, Serfling SE, Buck A, Werner RA, Lapa C, and Krebs M
- 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.
- Published
- 2023
- Full Text
- View/download PDF
29. Extending the Mass Spectrometry-Detectable Landscape of MHC Peptides by Use of Restricted Access Material.
- Author
-
Bernhardt M, Cruz-Garcia Y, Rech A, Meierjohann S, Erhard F, Schilling B, and Schlosser A
- Subjects
- Mass Spectrometry methods, Peptides chemistry, Solid Phase Extraction methods, Epitopes, T-Lymphocyte, Major Histocompatibility Complex
- 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.
- Published
- 2022
- Full Text
- View/download PDF
30. Effect of stress-induced polyploidy on melanoma reprogramming and therapy resistance.
- Author
-
Meierjohann S
- Subjects
- DNA Damage, Humans, Melanocytes, Mitosis, Oncogenes, Melanoma genetics, Melanoma therapy, Polyploidy
- 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., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
31. NRF2 Enables EGFR Signaling in Melanoma Cells.
- Author
-
Kreß JKC, Jessen C, Marquardt A, Hufnagel A, and Meierjohann S
- Subjects
- 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, Melanoma metabolism, NF-E2-Related Factor 2 metabolism, Signal Transduction
- 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.
- Published
- 2021
- Full Text
- View/download PDF
32. Subgroup-Independent Mapping of Renal Cell Carcinoma-Machine Learning Reveals Prognostic Mitochondrial Gene Signature Beyond Histopathologic Boundaries.
- Author
-
Marquardt A, Solimando AG, Kerscher A, Bittrich M, Kalogirou C, Kübler H, Rosenwald A, Bargou R, Kollmannsberger P, Schilling B, Meierjohann S, and Krebs M
- Abstract
Background: Renal cell carcinoma (RCC) is divided into three major histopathologic groups-clear cell (ccRCC), papillary (pRCC) and chromophobe RCC (chRCC). We performed a comprehensive re-analysis of publicly available RCC datasets from the TCGA (The Cancer Genome Atlas) database, thereby combining samples from all three subgroups, for an exploratory transcriptome profiling of RCC subgroups. Materials and Methods: We used FPKM (fragments per kilobase per million) files derived from the ccRCC, pRCC and chRCC cohorts of the TCGA database, representing transcriptomic data of 891 patients. Using principal component analysis, we visualized datasets as t-SNE plot for cluster detection. Clusters were characterized by machine learning, resulting gene signatures were validated by correlation analyses in the TCGA dataset and three external datasets (ICGC RECA-EU, CPTAC-3-Kidney, and GSE157256). Results: Many RCC samples co-clustered according to histopathology. However, a substantial number of samples clustered independently from histopathologic origin ( mixed subgroup )-demonstrating divergence between histopathology and transcriptomic data. Further analyses of mixed subgroup via machine learning revealed a predominant mitochondrial gene signature-a trait previously known for chRCC-across all histopathologic subgroups. Additionally, ccRCC samples from mixed subgroup presented an inverse correlation of mitochondrial and angiogenesis-related genes in the TCGA and in three external validation cohorts. Moreover, mixed subgroup affiliation was associated with a highly significant shorter overall survival for patients with ccRCC-and a highly significant longer overall survival for chRCC patients. Conclusions: Pan-RCC clustering according to RNA-sequencing data revealed a distinct histology-independent subgroup characterized by strengthened mitochondrial and weakened angiogenesis-related gene signatures. Moreover, affiliation to mixed subgroup went along with a significantly shorter overall survival for ccRCC and a longer overall survival for chRCC patients. Further research could offer a therapy stratification by specifically addressing the mitochondrial metabolism of such tumors and its microenvironment., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Marquardt, Solimando, Kerscher, Bittrich, Kalogirou, Kübler, Rosenwald, Bargou, Kollmannsberger, Schilling, Meierjohann and Krebs.)
- Published
- 2021
- Full Text
- View/download PDF
33. NRF2-dependent stress defense in tumor antioxidant control and immune evasion.
- Author
-
Friedmann Angeli JP and Meierjohann S
- Subjects
- Animals, Humans, Neoplasms drug therapy, Neoplasms immunology, Neoplasms metabolism, Signal Transduction, Antioxidants pharmacology, Gene Expression Regulation, Neoplastic, Immune Evasion, NF-E2-Related Factor 2 metabolism, Neoplasms pathology, Oxidative Stress
- 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., (© 2020 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
34. Correction: The transcription factor NRF2 enhances melanoma malignancy by blocking differentiation and inducing COX2 expression.
- Author
-
Jessen C, Kreß JKC, Baluapuri A, Hufnagel A, Schmitz W, Kneitz S, Roth S, Marquardt A, Appenzeller S, Ade CP, Glutsch V, Wobser M, Friedmann-Angeli JP, Mosteo L, Goding CR, Schilling B, Geissinger E, Wolf E, and Meierjohann S
- Published
- 2021
- Full Text
- View/download PDF
35. Correction: Emerging aspects in the regulation of ferroptosis.
- Author
-
Nehring H, Meierjohann S, and Angeli JPF
- Published
- 2020
- Full Text
- View/download PDF
36. Emerging aspects in the regulation of ferroptosis.
- Author
-
Nehring H, Meierjohann S, and Friedmann Angeli JP
- Subjects
- 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, Antioxidants chemistry, Ferroptosis, Gene Expression Regulation, Oxygen chemistry
- 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., (© 2020 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society.)
- Published
- 2020
- Full Text
- View/download PDF
37. The transcription factor NRF2 enhances melanoma malignancy by blocking differentiation and inducing COX2 expression.
- Author
-
Jessen C, Kreß JKC, Baluapuri A, Hufnagel A, Schmitz W, Kneitz S, Roth S, Marquardt A, Appenzeller S, Ade CP, Glutsch V, Wobser M, Friedmann-Angeli JP, Mosteo L, Goding CR, Schilling B, Geissinger E, Wolf E, and Meierjohann S
- Subjects
- Activating Transcription Factor 4 metabolism, Adenocarcinoma of Lung genetics, Adenocarcinoma of Lung immunology, Adenocarcinoma of Lung pathology, Animals, Cell Differentiation genetics, Cell Line, Tumor, Datasets as Topic, Dinoprostone metabolism, Disease Models, Animal, Female, Gene Expression Regulation, Neoplastic immunology, Gene Knockdown Techniques, Gene Knockout Techniques, Humans, Immunity, Innate genetics, Lung Neoplasms genetics, Lung Neoplasms immunology, Lung Neoplasms pathology, Male, Melanoma genetics, Melanoma immunology, Mice, Microphthalmia-Associated Transcription Factor metabolism, NF-E2-Related Factor 2 genetics, Skin Neoplasms genetics, Skin Neoplasms immunology, Tumor Escape genetics, Cyclooxygenase 2 metabolism, Melanoma pathology, NF-E2-Related Factor 2 metabolism, Skin Neoplasms pathology
- Abstract
The transcription factor NRF2 is the major mediator of oxidative stress responses and is closely connected to therapy resistance in tumors harboring activating mutations in the NRF2 pathway. In melanoma, such mutations are rare, and it is unclear to what extent melanomas rely on NRF2. Here we show that NRF2 suppresses the activity of the melanocyte lineage marker MITF in melanoma, thereby reducing the expression of pigmentation markers. Intriguingly, we furthermore identified NRF2 as key regulator of immune-modulating genes, linking oxidative stress with the induction of cyclooxygenase 2 (COX2) in an ATF4-dependent manner. COX2 is critical for the secretion of prostaglandin E2 and was strongly induced by H
2 O2 or TNFα only in presence of NRF2. Induction of MITF and depletion of COX2 and PGE2 were also observed in NRF2-deleted melanoma cells in vivo. Furthermore, genes corresponding to the innate immune response such as RSAD2 and IFIH1 were strongly elevated in absence of NRF2 and coincided with immune evasion parameters in human melanoma datasets. Even in vitro, NRF2 activation or prostaglandin E2 supplementation blunted the induction of the innate immune response in melanoma cells. Transcriptome analyses from lung adenocarcinomas indicate that the observed link between NRF2 and the innate immune response is not restricted to melanoma.- Published
- 2020
- Full Text
- View/download PDF
38. Role of melanoma inhibitory activity in melanocyte senescence.
- Author
-
Feuerer L, Lamm S, Henz I, Kappelmann-Fenzl M, Haferkamp S, Meierjohann S, Hellerbrand C, Kuphal S, and Bosserhoff AK
- Subjects
- Animals, Cell Cycle, Cell Line, Tumor, Disease Models, Animal, HEK293 Cells, Humans, Mice, Knockout, Mice, Transgenic, Cellular Senescence, Extracellular Matrix Proteins metabolism, Melanocytes metabolism, Melanocytes pathology, Neoplasm Proteins metabolism
- Abstract
The protein melanoma inhibitory activity (MIA) is known to be expressed in melanoma and to support melanoma progression. Interestingly, previous studies also observed the expression of MIA in nevi. Concentrating on these findings, we revealed that MIA expression is correlated with a senescent state in melanocytes. Induction of replicative or oncogene-induced senescence resulted in increased MIA expression in vitro. Notably, MIA knockdown in senescent melanocytes reduced the percentage of senescence-associated beta-Gal-positive cells and enhanced proliferation. Using the melanoma mouse model Tg(Grm1), MIA-deficient mice supported the impact of MIA on senescence by showing a significantly earlier tumor onset compared to controls. In melanocytes, MIA knockdown led to a downregulation of the cell cycle inhibitor p21 in vitro and in vivo. In contrast, after induction of hTERT in human melanoma cells, p21 regulation by MIA was lost. In summary, our data show for the first time that MIA is a regulator of cellular senescence in human and murine melanocytes., (© 2019 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
39. IFN-gamma-induced PD-L1 expression in melanoma depends on p53 expression.
- Author
-
Thiem A, Hesbacher S, Kneitz H, di Primio T, Heppt MV, Hermanns HM, Goebeler M, Meierjohann S, Houben R, and Schrama D
- Subjects
- Biomarkers, Tumor, Cell Line, Tumor, Gene Editing, Gene Targeting, Humans, Interferon-gamma pharmacology, Melanoma immunology, RNA, Small Interfering genetics, Signal Transduction, B7-H1 Antigen genetics, Gene Expression Regulation, Neoplastic drug effects, Interferon-gamma metabolism, Melanoma genetics, Melanoma metabolism, Tumor Suppressor Protein p53 genetics
- Abstract
Background: Immune checkpoint inhibition and in particular anti-PD-1 immunotherapy have revolutionized the treatment of advanced melanoma. In this regard, higher tumoral PD-L1 protein (gene name: CD274) expression is associated with better clinical response and increased survival to anti-PD-1 therapy. Moreover, there is increasing evidence that tumor suppressor proteins are involved in immune regulation and are capable of modulating the expression of immune checkpoint proteins. Here, we determined the role of p53 protein (gene name: TP53) in the regulation of PD-L1 expression in melanoma., Methods: We analyzed publicly available mRNA and protein expression data from the cancer genome/proteome atlas and performed immunohistochemistry on tumors with known TP53 status. Constitutive and IFN-ɣ-induced PD-L1 expression upon p53 knockdown in wildtype, TP53-mutated or JAK2-overexpressing melanoma cells or in cells, in which p53 was rendered transcriptionally inactive by CRISPR/Cas9, was determined by immunoblot or flow cytometry. Similarly, PD-L1 expression was investigated after overexpression of a transcriptionally-impaired p53 (L22Q, W23S) in TP53-wt or a TP53-knockout melanoma cell line. Immunoblot was applied to analyze the IFN-ɣ signaling pathway., Results: For TP53-mutated tumors, an increased CD274 mRNA expression and a higher frequency of PD-L1 positivity was observed. Interestingly, positive correlations of IFNG mRNA and PD-L1 protein in both TP53-wt and -mutated samples and of p53 and PD-L1 protein suggest a non-transcriptional mode of action of p53. Indeed, cell line experiments revealed a diminished IFN-ɣ-induced PD-L1 expression upon p53 knockdown in both wildtype and TP53-mutated melanoma cells, which was not the case when p53 wildtype protein was rendered transcriptionally inactive or by ectopic expression of p53
L22Q,W23S , a transcriptionally-impaired variant, in TP53-wt cells. Accordingly, expression of p53L22Q,W23S in a TP53-knockout melanoma cell line boosted IFN-ɣ-induced PD-L1 expression. The impaired PD-L1-inducibility after p53 knockdown was associated with a reduced JAK2 expression in the cells and was almost abrogated by JAK2 overexpression., Conclusions: While having only a small impact on basal PD-L1 expression, both wildtype and mutated p53 play an important positive role for IFN-ɣ-induced PD-L1 expression in melanoma cells by supporting JAK2 expression. Future studies should address, whether p53 expression levels might influence response to anti-PD-1 immunotherapy.- Published
- 2019
- Full Text
- View/download PDF
40. The identification of patient-specific mutations reveals dual pathway activation in most patients with melanoma and activated receptor tyrosine kinases in BRAF/NRAS wild-type melanomas.
- Author
-
Appenzeller S, Gesierich A, Thiem A, Hufnagel A, Jessen C, Kneitz H, Regensburger M, Schmidt C, Zirkenbach V, Bischler T, Schilling B, Siedel C, Goebeler ME, Houben R, Schrama D, Gehrig A, Rost S, Maurus K, Bargou R, Rosenwald A, Schartl M, Goebeler M, and Meierjohann S
- Subjects
- 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, 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
- 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., (© 2018 American Cancer Society.)
- Published
- 2019
- Full Text
- View/download PDF
41. RNA-seq analysis identifies different transcriptomic types and developmental trajectories of primary melanomas.
- Author
-
Kunz M, Löffler-Wirth H, Dannemann M, Willscher E, Doose G, Kelso J, Kottek T, Nickel B, Hopp L, Landsberg J, Hoffmann S, Tüting T, Zigrino P, Mauch C, Utikal J, Ziemer M, Schulze HJ, Hölzel M, Roesch A, Kneitz S, Meierjohann S, Bosserhoff A, Binder H, and Schartl M
- Subjects
- Adult, Aged, Aged, 80 and over, Down-Regulation genetics, Female, Gene Expression genetics, Gene Expression Profiling methods, Humans, Male, Microphthalmia-Associated Transcription Factor genetics, Middle Aged, Mutation genetics, Nevus, Pigmented genetics, Sequence Analysis, RNA methods, Transcription, Genetic genetics, Melanoma genetics, Transcriptome genetics
- Abstract
Recent studies revealed trajectories of mutational events in early melanomagenesis, but the accompanying changes in gene expression are far less understood. Therefore, we performed a comprehensive RNA-seq analysis of laser-microdissected melanocytic nevi (n = 23) and primary melanoma samples (n = 57) and characterized the molecular mechanisms of early melanoma development. Using self-organizing maps, unsupervised clustering, and analysis of pseudotime (PT) dynamics to identify evolutionary trajectories, we describe here two transcriptomic types of melanocytic nevi (N1 and N2) and primary melanomas (M1 and M2). N1/M1 lesions are characterized by pigmentation-type and MITF gene signatures, and a high prevalence of NRAS mutations in M1 melanomas. N2/M2 lesions are characterized by inflammatory-type and AXL gene signatures with an equal distribution of wild-type and mutated BRAF and low prevalence of NRAS mutations in M2 melanomas. Interestingly, N1 nevi and M1 melanomas and N2 nevi and M2 melanomas, respectively, cluster together, but there is no clustering in a stage-dependent manner. Transcriptional signatures of M1 melanomas harbor signatures of BRAF/MEK inhibitor resistance and M2 melanomas harbor signatures of anti-PD-1 antibody treatment resistance. Pseudotime dynamics of nevus and melanoma samples are suggestive for a switch-like immune-escape mechanism in melanoma development with downregulation of immune genes paralleled by an increasing expression of a cell cycle signature in late-stage melanomas. Taken together, the transcriptome analysis identifies gene signatures and mechanisms underlying development of melanoma in early and late stages with relevance for diagnostics and therapy.
- Published
- 2018
- Full Text
- View/download PDF
42. Early onset of diffuse melanosis cutis under pembrolizumab therapy illustrates the limitations of anti-PD-1 checkpoint inhibitors.
- Author
-
Thiem A, Schummer P, Ueberschaar S, Kerstan A, Kneitz H, Schrama D, Appenzeller S, Meierjohann S, Schilling B, Goebeler M, and Gesierich A
- Subjects
- Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized pharmacology, Antineoplastic Agents, Immunological pharmacology, Female, Humans, Male, Melanosis pathology, Skin Neoplasms pathology, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents, Immunological therapeutic use, Melanosis drug therapy, Skin Neoplasms drug therapy
- Abstract
Anti-PD-1 targeted immunotherapies have revolutionized the treatment of advanced melanoma and other tumor entities, and long disease-free intervals have been reported in responding patients. However, a considerable number of patients still progress rapidly after the start of anti-PD-1 antibodies. Here, we document two patients, 78 and 85-year old, who suffered from advanced BRAF-V600 wild-type melanoma and received pembrolizumab 2 mg/kg every 3 weeks as the first systemic treatment. After only one, respectively, two infusions of pembrolizumab, both patients developed melanuria and diffuse melanosis cutis (DMC) on sun-exposed areas of their skin. Both also had liver metastases, which have been reported to be associated with DMC before. Pembrolizumab was stopped because of rapid tumor progression and both patients died within 2 months after manifestation of DMC. We conclude that DMC is a condition that may be a negative predictor of response to anti-PD-1 treatment. With respect to the very short survival, which is even shorter than that reported after conventional therapies, it needs to be evaluated whether immunotherapy may be a (relative) contraindication in DMC patients when another treatment option, that is targeted therapy, is available.
- Published
- 2018
- Full Text
- View/download PDF
43. BRAF inhibition causes resilience of melanoma cell lines by inducing the secretion of FGF1.
- Author
-
Grimm J, Hufnagel A, Wobser M, Borst A, Haferkamp S, Houben R, and Meierjohann S
- Abstract
Approximately half of all melanoma patients harbour activating mutations in the serine/threonine kinase BRAF. This is the basis for one of the main treatment strategies for this tumor type, the targeted therapy with BRAF and MEK inhibitors. While the initial responsiveness to these drugs is high, resistance develops after several months, frequently at sites of the previously responding tumor. This indicates that tumor response is incomplete and that a certain tumor fraction survives even in drug-sensitive patients, e.g., in a therapy-induced senescence-like state. Here, we show in several melanoma cell lines that BRAF inhibition induces a secretome with stimulating effect on fibroblasts and naive melanoma cells. Several senescence-associated factors were found to be transcribed and secreted in response to BRAF or MEK inhibition, among them members of the fibroblast growth factor family. We identified the growth factor FGF1 as mediator of resilience towards BRAF inhibition, which limits the pro-apoptotic effects of the drug and activates fibroblasts to secrete HGF. FGF1 regulation was mediated by the PI3K pathway and by FRA1, a direct target gene of the MAPK pathway. When FGFR inhibitors were applied in parallel to BRAF inhibitors, resilience was broken, thus providing a rationale for combined therapeutical application.
- Published
- 2018
- Full Text
- View/download PDF
44. Panel Sequencing Shows Recurrent Genetic FAS Alterations in Primary Cutaneous Marginal Zone Lymphoma.
- Author
-
Maurus K, Appenzeller S, Roth S, Kuper J, Rost S, Meierjohann S, Arampatzi P, Goebeler M, Rosenwald A, Geissinger E, and Wobser M
- Subjects
- Death Domain genetics, Diagnosis, Differential, High-Throughput Nucleotide Sequencing, Humans, Lymphoma, B-Cell, Marginal Zone diagnosis, Mutation, Pseudolymphoma diagnosis, Skin Neoplasms diagnosis, Biomarkers, Tumor genetics, Lymphoma, B-Cell, Marginal Zone genetics, Skin Neoplasms genetics, fas Receptor genetics
- Abstract
Primary cutaneous marginal zone lymphoma (PCMZL) represents an indolent subtype of non-Hodgkin lymphoma that is clinically characterized by slowly growing skin tumors with a very low propensity for systemic dissemination. The underlying genetic basis of PCMZL has not been comprehensively elucidated. To gain deeper insight into the molecular pathogenesis of PCMZL, we performed hybridization-based panel sequencing of 38 patients with well-characterized PCMZL. In 32 of the 38 patients, we identified genetic alterations within 39 selected target genes. The most frequently detected alterations (24/38 patients, 63.2%) affected the FAS gene, of which 22 patients harbored alterations, which affect the functionally relevant death domain of the apoptosis-regulating FAS/CD95 protein in a dominant-negative manner. In addition, we identified highly recurrent mutations in three other genes, namely SLAMF1, SPEN, and NCOR2. Our molecular data suggest that apoptosis defects provide the molecular basis of the observed clinical features of PCMZL, which commonly presents with only slowly growing skin tumors, reflecting its invariably indolent behavior. From a diagnostic point of view, highly recurrent FAS mutations in PCMZL presumably separate this indolent lymphoma entity from pseudolymphoma, and this adds adjunctive discriminatory features at a molecular level., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
45. Targeting the Senescence-Overriding Cooperative Activity of Structurally Unrelated H3K9 Demethylases in Melanoma.
- Author
-
Yu Y, Schleich K, Yue B, Ji S, Lohneis P, Kemper K, Silvis MR, Qutob N, van Rooijen E, Werner-Klein M, Li L, Dhawan D, Meierjohann S, Reimann M, Elkahloun A, Treitschke S, Dörken B, Speck C, Mallette FA, Zon LI, Holmen SL, Peeper DS, Samuels Y, Schmitt CA, and Lee S
- Published
- 2018
- Full Text
- View/download PDF
46. BIK is involved in BRAF/MEK inhibitor induced apoptosis in melanoma cell lines.
- Author
-
Borst A, Haferkamp S, Grimm J, Rösch M, Zhu G, Guo S, Li C, Gao T, Meierjohann S, Schrama D, and Houben R
- Subjects
- Cell Line, Tumor, Cell Proliferation drug effects, Gene Expression Regulation, Neoplastic drug effects, Humans, Indoles, Melanoma metabolism, Melanoma pathology, Mitochondrial Proteins, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Proto-Oncogene Proteins B-raf metabolism, Pyridones, Pyrimidinones, RNA, Messenger metabolism, Sulfonamides, Vemurafenib, Antineoplastic Agents pharmacology, Apoptosis drug effects, Apoptosis Regulatory Proteins metabolism, Enzyme Inhibitors pharmacology, Melanoma drug therapy, Membrane Proteins metabolism, Protein Kinase Inhibitors pharmacology
- Abstract
In patients with BRAF-mutated melanoma specific inhibitors of BRAF
V600E and MEK1/2 frequently induce initial tumor reduction, frequently followed by relapse. As demonstrated previously, BRAFV600E -inhibition induces apoptosis only in a fraction of treated cells, while the remaining arrest and survive providing a source or a niche for relapse. To identify factors contributing to the differential initial response towards BRAF/MEK inhibition, we established M14 melanoma cell line-derived single cell clones responding to treatment with BRAF inhibitor vemurafenib and MEK inhibitor trametinib predominantly with either cell cycle arrest (CCA-cells) or apoptosis (A-cells). Screening for differentially expressed apoptosis-related genes revealed loss of BCL2-Interacting Killer (BIK) mRNA in CCA-cells. Importantly, ectopic expression of BIK in CCA-cells resulted in increased apoptosis rates following vemurafenib/trametinib treatment, while knockdown/knockout of BIK in A-cells attenuated the apoptotic response. Furthermore, we demonstrate reversible epigenetic silencing of BIK mRNA expression in CCA-cells. Importantly, HDAC inhibitor treatment associated with re-expression of BIK augmented sensitivity of CCA-cells towards vemurafenib/trametinib treatment both in vitro and in vivo. In conclusion, our results suggest that BIK can be a critical mediator of melanoma cell fate determination in response to MAPK pathway inhibition., (Copyright © 2017 Elsevier B.V. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
47. Crosstalk signaling in targeted melanoma therapy.
- Author
-
Meierjohann S
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Humans, Molecular Targeted Therapy, Proto-Oncogene Proteins B-raf metabolism, Antineoplastic Combined Chemotherapy Protocols pharmacology, MAP Kinase Signaling System drug effects, Melanoma drug therapy, Melanoma enzymology, Proto-Oncogene Proteins B-raf antagonists & inhibitors
- Abstract
Inhibition of the BRAF/MAPK pathway belongs to the standard therapies for patients with activating BRAF
V600E/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.- Published
- 2017
- Full Text
- View/download PDF
48. Peroxiredoxin 6 triggers melanoma cell growth by increasing arachidonic acid-dependent lipid signalling.
- Author
-
Schmitt A, Schmitz W, Hufnagel A, Schartl M, and Meierjohann S
- Subjects
- 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, Arachidonic Acid metabolism, Melanoma enzymology, Neoplasm Proteins metabolism, Peroxiredoxin VI metabolism, Signal Transduction
- 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., (© 2015 Authors; published by Portland Press Limited.)
- Published
- 2015
- Full Text
- View/download PDF
49. Hypoxia-independent drivers of melanoma angiogenesis.
- Author
-
Meierjohann S
- Abstract
Tumor angiogenesis is a process which is traditionally regarded as the tumor's response to low nutrient supply occurring under hypoxic conditions. However, hypoxia is not a pre-requisite for angiogenesis. The fact that even single tumor cells or small tumor cell aggregates are capable of attracting blood vessels reveals the early metastatic capability of tumor cells. This review sheds light on the hypoxia-independent mechanisms of tumor angiogenesis in melanoma.
- Published
- 2015
- Full Text
- View/download PDF
50. The MAPK pathway as an apoptosis enhancer in melanoma.
- Author
-
Haydn JM, Hufnagel A, Grimm J, Maurus K, Schartl M, and Meierjohann S
- Subjects
- Adaptor Proteins, Signal Transducing, Antineoplastic Agents pharmacology, Apoptosis genetics, Blotting, Western, Butadienes pharmacology, Cell Line, Tumor, Cisplatin pharmacology, Enzyme Inhibitors pharmacology, Gene Expression Regulation, Neoplastic drug effects, Humans, Indazoles pharmacology, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, MAP Kinase Signaling System genetics, Melanoma genetics, Melanoma metabolism, Melanoma pathology, Membrane Proteins genetics, Membrane Proteins metabolism, Mitogen-Activated Protein Kinase 1 genetics, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3 genetics, Mitogen-Activated Protein Kinase 3 metabolism, Mutation, Missense, Nitriles pharmacology, Phosphatidylinositol 3-Kinases metabolism, Phosphoinositide-3 Kinase Inhibitors, Phosphorylation drug effects, Proto-Oncogene Proteins B-raf genetics, Proto-Oncogene Proteins B-raf metabolism, Proto-Oncogene Proteins c-akt genetics, Proto-Oncogene Proteins c-akt metabolism, RNA Interference, Repressor Proteins genetics, Repressor Proteins metabolism, Reverse Transcriptase Polymerase Chain Reaction, Sulfonamides pharmacology, Apoptosis drug effects, Benzamides pharmacology, Cell Proliferation drug effects, MAP Kinase Signaling System drug effects
- Abstract
Inhibition of RAF/MEK/ERK signaling is beneficial for many patients with BRAF(V600E)-mutated melanoma. However, primary and secondary resistances restrict long-lasting therapy success. Combination therapies are therefore urgently needed. Here, we evaluate the cellular effect of combining a MEK inhibitor with a genotoxic apoptosis inducer. Strikingly, we observed that an activated MAPK pathway promotes in several melanoma cell lines the pro-apoptotic response to genotoxic stress, and MEK inhibition reduces intrinsic apoptosis. This goes along with MEK inhibitor induced increased RAS and P-AKT levels. The protective effect of the MEK inhibitor depends on PI3K signaling, which prevents the induction of pro-apoptotic PUMA that mediates apoptosis after DNA damage. We could show that the MEK inhibitor dependent feedback loop is enabled by several factors, including EGF receptor and members of the SPRED family. The simultaneous knockdown of SPRED1 and SPRED2 mimicked the effects of MEK inhibitor such as PUMA repression and protection from apoptosis. Our data demonstrate that MEK inhibition of BRAF(V600E)-positive melanoma cells can protect from genotoxic stress, thereby achieving the opposite of the intended anti-tumorigenic effect of the combination of MEK inhibitor with inducers of intrinsic apoptosis.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.