225 results on '"Stiewe T"'
Search Results
2. Phosphorylation control of p53 DNA binding cooperativity balances tumorigensis and aging
- Author
-
Timofeev, O., Koch, L., Niederau, C., Tscherne, A., Schneikert, J., Klimovich, M., Elmshäuser, S., Zeitlinger, M., Mernberger, M., Nist, A., Osterburg, C., Dötsch, V., Hrabě de Angelis, M., Stiewe, T., German Mouse Clinic Consortium (Aguilar-Pimentel, J.A., Schmidt-Weber, C.B., Becker, L., Klopstock, T., Cho, Y.-L., Spielmann, N., Amarie, O.V., Garrett, L., Hölter, S.M., Wurst, W., Calzada-Wack, J., Sanz-Moreno, A., Klein-Rodewald, T., Rathkolb, B., Wolf, E., Östereicher, M.A., Miller, G., Maier, H., Stöger, C., Leuchtenberger, S., Gailus-Durner, V., and Fuchs, H.)
- Subjects
0301 basic medicine ,Aging ,Cancer Research ,Carcinogenesis ,DNA damage ,Longevity ,medicine.disease_cause ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Neoplasms ,medicine ,Animals ,Phosphorylation ,Transcription factor ,Chemistry ,Wild type ,DNA ,Cell biology ,Haematopoiesis ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Tumor Suppressor Protein p53 ,Stem cell ,DNA Damage - Abstract
Posttranslational modifications are essential for regulating the transcription factor p53, which binds DNA in a highly cooperative manner to control expression of a plethora of tumor-suppressive programs. Here we show at the biochemical, cellular, and organismal level that the cooperative nature of DNA binding is reduced by phosphorylation of highly conserved serine residues (human S183/S185, mouse S180) in the DNA-binding domain. To explore the role of this inhibitory phosphorylation in vivo, new phosphorylation-deficient p53-S180A knock-in mice were generated. Chromatin immunoprecipitation sequencing and RNA sequencing studies of S180A knock-in cells demonstrated enhanced DNA binding and increased target gene expression. In vivo, this translated into a tissue-specific vulnerability of the bone marrow that caused depletion of hematopoietic stem cells and impaired proper regeneration of hematopoiesis after DNA damage. Median lifespan was significantly reduced by 20% from 709 days in wild type to only 568 days in S180A littermates. Importantly, lifespan was reduced by a loss of general fitness and increased susceptibility to age-related diseases, not by increased cancer incidence as often seen in other p53-mutant mouse models. For example, S180A knock-in mice showed markedly reduced spontaneous tumorigenesis and increased resistance to Myc-driven lymphoma and Eml4–Alk-driven lung cancer. Preventing phosphorylation of S183/S185 in human cells boosted p53 activity and allowed tumor cells to be killed more efficiently. Together, our data identify p53 DNA-binding domain phosphorylation as a druggable mechanism that balances tumorigenesis and aging. Significance: These findings demonstrate that p53 tumor suppressor activity is reduced by DNA-binding domain phosphorylation to prevent aging and identify this phosphorylation as a potential target for cancer therapy. See related commentary by Horikawa, p. 5164
- Published
- 2020
3. Regulation of telomerase activity by the p53 family member p73
- Author
-
Beitzinger, M, Oswald, C, Beinoraviciute-Kellner, R, and Stiewe, T
- Published
- 2006
- Full Text
- View/download PDF
4. Distinct IL-1 alpha-responsive enhancers promote acute and coordinated changes in chromatin topology in a hierarchical manner
- Author
-
Weiterer, SS, Meier-Soelch, J, Georgomanolis, T, Mizi, A, Beyerlein, A, Weiser, H, Brant, L, Mayr-Buro, C, Jurida, L, Beuerlein, K, Muller, H, Weber, A, Tenekeci, U, Dittrich-Breiholz, O, Bartkuhn, M, Nist, A, Stiewe, T, van Ijcken, Wilfred, Riedlinger, T, Schmitz, ML, Papantonis, A, Kracht, M, Weiterer, SS, Meier-Soelch, J, Georgomanolis, T, Mizi, A, Beyerlein, A, Weiser, H, Brant, L, Mayr-Buro, C, Jurida, L, Beuerlein, K, Muller, H, Weber, A, Tenekeci, U, Dittrich-Breiholz, O, Bartkuhn, M, Nist, A, Stiewe, T, van Ijcken, Wilfred, Riedlinger, T, Schmitz, ML, Papantonis, A, and Kracht, M
- Published
- 2020
5. p73 in apoptosis
- Author
-
Stiewe, T. and Pützer, B. M.
- Published
- 2001
- Full Text
- View/download PDF
6. Improved safety through tamoxifen-regulated induction of cytotoxic genes delivered by Ad vectors for cancer gene therapy
- Author
-
Pützer, BM, Stiewe, T, Crespo, F, and Esche, H
- Published
- 2000
- Full Text
- View/download PDF
7. Oncogenic RAS enables DNA damage- and p53-dependent differentiation of acute myeloid leukemia cells in response to chemotherapy: V571
- Author
-
Meyer, M., Rübsamen, D., Slany, R., Illmer, T., Stabla, K., Roth, P., Stiewe, T., Eilers, M., and Neubauer, A.
- Published
- 2009
8. Genomic Location of PRMT6-Dependent H3R2 Methylation Is Linked to the Transcriptional Outcome of Associated Genes
- Author
-
Bouchard, C. (Caroline), Sahu, P. (Peeyush), Meixner, M. (Marion), Nötzold, R.R. (René Reiner), Rust, M.B. (Marco B.), Kremmer, E. (Elisabeth), Feederle, R. (Regina), Hart-Smith, G. (Gene), Finkernagel, F. (Florian), Bartkuhn, M. (Marek), Savai Pullamsetti, S. (Soni), Nist, A. (Andrea), Stiewe, T. (Thorsten), Philipsen, J.N.J. (Sjaak), Bauer, U.M. (Uta-Maria), Bouchard, C. (Caroline), Sahu, P. (Peeyush), Meixner, M. (Marion), Nötzold, R.R. (René Reiner), Rust, M.B. (Marco B.), Kremmer, E. (Elisabeth), Feederle, R. (Regina), Hart-Smith, G. (Gene), Finkernagel, F. (Florian), Bartkuhn, M. (Marek), Savai Pullamsetti, S. (Soni), Nist, A. (Andrea), Stiewe, T. (Thorsten), Philipsen, J.N.J. (Sjaak), and Bauer, U.M. (Uta-Maria)
- Abstract
Protein arginine methyltransferase 6 (PRMT6) catalyzes asymmetric dimethylation of histone H3 at arginine 2 (H3R2me2a). This mark has been reported to associate with silent genes. Here, we use a cell model of neural differentiation, which upon PRMT6 knockout exhibits proliferation and differentiation defects. Strikingly, we detect PRMT6-dependent H3R2me2a at active genes, both at promoter and enhancer sites. Loss of H3R2me2a from promoter sites leads to enhanced KMT2A binding and H3K4me3 deposition together with increased target gene transcription, supporting a repressive nature of H3R2me2a. At enhancers, H3R2me2a peaks co-localize with the active enhancer marks H3K4me1 and H3K27ac. Here, loss of H3R2me2a results in reduced KMT2D binding and H3K4me1/H3K27ac deposition together with decreased transcription of associated genes, indicating that H3R2me2a also exerts activation functions. Our work suggests that PRMT6 via H3R2me2a interferes with the deposition of adjacent histone marks and modulates the activity of important differentiation-associated genes by opposing transcriptional effects. Bouchard et al. identify the genome-wide, PRMT6-dependent occurrence of H3R2me2a in a cell model of neural differentiation. H3R2me2a is localized at promoters and enhancers of active genes and influences the chromatin recruitment of histone lysine methyltransferases. Thereby, H3R2me2a modulates the deposition of adjacent histone H3 marks and regulates the transcriptional output of genes relevant for pluripotency and differentiation.
- Published
- 2018
- Full Text
- View/download PDF
9. Genomic Location of PRMT6-Dependent H3R2 Methylation Is Linked to the Transcriptional Outcome of Associated Genes
- Author
-
Bouchard, C, Sahu, P, Meixner, M, Notzold, RR, Rust, MB, Kremmer, E, Feederle, R, Hart-Smith, G, Finkernagel, F, Bartkuhn, M, Pullamsetti, SS, Nist, A, Stiewe, T, Philipsen, Sjaak, Bauer, UM, Bouchard, C, Sahu, P, Meixner, M, Notzold, RR, Rust, MB, Kremmer, E, Feederle, R, Hart-Smith, G, Finkernagel, F, Bartkuhn, M, Pullamsetti, SS, Nist, A, Stiewe, T, Philipsen, Sjaak, and Bauer, UM
- Published
- 2018
10. Induktion des Autophagie-assoziierten Zelltodes durch Histon-Deacetylase Inhibition in pankreatischen neuroendokrinen Tumorzellsphäroiden
- Author
-
Matrood, S, additional, Wissniowski, T, additional, Wiese, D, additional, Griesmann, H, additional, Egidi, M, additional, Wanzel, M, additional, Stiewe, T, additional, Buchholz, M, additional, Gress, T, additional, Bartsch, D, additional, and Di Fazio, P, additional
- Published
- 2018
- Full Text
- View/download PDF
11. PO-233 Chemoresistant NSCLC cells are hypersensitive to metabolic drugs due to mTOR-mediated inhibition of autophagy
- Author
-
Wanzel, M., primary, Gremke, N., additional, Schmoll, L., additional, Pagenstecher, A., additional, Schneikert, J., additional, and Stiewe, T., additional
- Published
- 2018
- Full Text
- View/download PDF
12. Autophagie in einem NAFL/NASH- Maus und humanem in vitro Modell
- Author
-
Polte, SCH, additional, Okamoto, K, additional, Matono, T, additional, Stiewe, T, additional, Wanzel, M, additional, Bartsch, DK, additional, Gress, TM, additional, Wissniowski, TT, additional, and Di Fazio, P, additional
- Published
- 2017
- Full Text
- View/download PDF
13. 792 CK1α ablation in keratinocytes induces p53-dependent, sunburn-protective, skin hyperpigmentation
- Author
-
Chang, C., primary, Ito, T., additional, Stiewe, T., additional, Wakamatsu, K., additional, and Ben-Neriah, Y., additional
- Published
- 2017
- Full Text
- View/download PDF
14. CRISPR-Cas9-based target validation for p53-reactivating model compounds
- Author
-
Wanzel, M., primary, Vischedyk, J., additional, Gittler, M., additional, Gremke, N., additional, Mernberger, M., additional, Charles, J., additional, Schneikert, J., additional, Bretz, A.C., additional, Nist, A., additional, and Stiewe, T., additional
- Published
- 2016
- Full Text
- View/download PDF
15. Monitoring the dynamics of clonal tumor evolution in vivo using secreted luciferases
- Author
-
Charles, J.P., primary, Fuchs, J., additional, Hefter, M., additional, Vischedyk, J.B., additional, Kleint, M., additional, Vogiatzi, F., additional, Nist, A., additional, Timofeev, O., additional, Wanzel, M., additional, and Stiewe, T., additional
- Published
- 2016
- Full Text
- View/download PDF
16. Mutant p53 promotes tumor progression by the endoplasmic reticulum UDPase ENTPD5
- Author
-
Vogiatzi, F., primary, Brandt, D.T., additional, Fuchs, J., additional, Grikscheit, K., additional, Timofeev, O., additional, Nist, A., additional, Mernberger, M., additional, Grosse, R., additional, and Stiewe, T., additional
- Published
- 2016
- Full Text
- View/download PDF
17. Widespread epigenetic activation of ΔNp73 in small cell lung cancer causes vulnerability to Tip60-p400 inhibition
- Author
-
Nist, A., primary, Krampitz, A.M., additional, Schlereth, K., additional, Mernberger, M., additional, Moßner, M.C., additional, Muley, T., additional, Dammann, R., additional, and Stiewe, T., additional
- Published
- 2016
- Full Text
- View/download PDF
18. Immune and Inflammatory Cell Composition of Human Lung Cancer Stroma
- Author
-
Banat, GA, primary, Tretyn, A, additional, Pullamsetti, SS, additional, Wilhelm, J, additional, Weigert, A, additional, Ebel, K, additional, Stiewe, T, additional, Grimminger, F, additional, Seeger, W, additional, Fink, L, additional, and Savai, R, additional
- Published
- 2015
- Full Text
- View/download PDF
19. 73: Parainflammation in cancer
- Author
-
Lasry, A., primary, Hamza, H., additional, Kadosh, E., additional, Elyada, E., additional, Pribluda, A., additional, Alitalo, K., additional, Stiewe, T., additional, Oren, M., additional, Pikarsky, E., additional, and Ben-Neriah, Y., additional
- Published
- 2014
- Full Text
- View/download PDF
20. 562 - CRISPR-Cas9-based target validation for p53-reactivating model compounds
- Author
-
Wanzel, M., Vischedyk, J., Gittler, M., Gremke, N., Mernberger, M., Charles, J., Schneikert, J., Bretz, A.C., Nist, A., and Stiewe, T.
- Published
- 2016
- Full Text
- View/download PDF
21. Adaptation of cancer cells from different entities to the MDM2 inhibitor nutlin-3 results in the emergence of p53-mutated multi-drug-resistant cancer cells
- Author
-
Michaelis, M, primary, Rothweiler, F, additional, Barth, S, additional, Cinatl, J, additional, van Rikxoort, M, additional, Löschmann, N, additional, Voges, Y, additional, Breitling, R, additional, von Deimling, A, additional, Rödel, F, additional, Weber, K, additional, Fehse, B, additional, Mack, E, additional, Stiewe, T, additional, Doerr, H W, additional, and Speidel, D, additional
- Published
- 2011
- Full Text
- View/download PDF
22. C-terminal diversity within the p53 family accounts for differences in DNA binding and transcriptional activity
- Author
-
Sauer, M., primary, Bretz, A. C., additional, Beinoraviciute-Kellner, R., additional, Beitzinger, M., additional, Burek, C., additional, Rosenwald, A., additional, Harms, G. S., additional, and Stiewe, T., additional
- Published
- 2008
- Full Text
- View/download PDF
23. The p53 family inhibitor ΔNp73 interferes with multiple developmental programs
- Author
-
Hüttinger-Kirchhof, N, primary, Cam, H, additional, Griesmann, H, additional, Hofmann, L, additional, Beitzinger, M, additional, and Stiewe, T, additional
- Published
- 2005
- Full Text
- View/download PDF
24. Regulation of telomerase activity by the p53 family member p73
- Author
-
Beitzinger, M, primary, Oswald, C, additional, Beinoraviciute-Kellner, R, additional, and Stiewe, T, additional
- Published
- 2005
- Full Text
- View/download PDF
25. Mechanism of E2F1-induced apoptosis in primary vascular smooth muscle cells
- Author
-
STANELLE, J, primary, STIEWE, T, additional, RODICKER, F, additional, KOHLER, K, additional, THESELING, C, additional, and PUTZER, B, additional
- Published
- 2003
- Full Text
- View/download PDF
26. Antitumor Capacity of a Dominant-NegativeRETProto-Oncogene Mutant in a Medullary Thyroid Carcinoma Model
- Author
-
Drosten, M., primary, Stiewe, T., additional, and Pützer, B.M., additional
- Published
- 2003
- Full Text
- View/download PDF
27. Increased ΔN-p73 expression in tumors by upregulation of the E2F1-regulated, TA-promoter-derived ΔN′-p73 transcript
- Author
-
Pützer, B M, primary, Tuve, S, additional, Tannapfel, A, additional, and Stiewe, T, additional
- Published
- 2003
- Full Text
- View/download PDF
28. Role of p73 in malignancy: tumor suppressor or oncogene?
- Author
-
Stiewe, T, primary and Pützer, B M, additional
- Published
- 2002
- Full Text
- View/download PDF
29. Large Nontransplanted Hepatocellular Carcinoma in Woodchucks: Treatment With Adenovirus-Mediated Delivery of Interleukin 12/B7.1 Genes
- Author
-
Putzer, B. M., primary, Stiewe, T., additional, Rodicker, F., additional, Schildgen, O., additional, Ruhm, S., additional, Dirsch, O., additional, Fiedler, M., additional, Damen, U., additional, Tennant, B., additional, Scherer, C., additional, Graham, F. L., additional, and Roggendorf, M., additional
- Published
- 2001
- Full Text
- View/download PDF
30. E1A is sufficient by itself to induce apoptosis independent of p53 and other adenoviral gene products
- Author
-
Pützer, B M, primary, Stiewe, T, additional, Parssanedjad, K, additional, Rega, S, additional, and Esche, H, additional
- Published
- 2000
- Full Text
- View/download PDF
31. The p53 family inhibitor ΔNp73 interferes with multiple developmental programs.
- Author
-
Hüttinger-Kirchhof, N., Cam, H., Griesmann, H., Hofmann, L., Beitzinger, M., and Stiewe, T.
- Subjects
LETTERS to the editor ,CELL differentiation - Abstract
The article presents a letter to the editor regarding the role of the p53 family inhibitor in cellular differentiation and embryogenesis.
- Published
- 2006
- Full Text
- View/download PDF
32. Antitumor Capacity of a Dominant-Negative RET Proto-Oncogene Mutant in a Medullary Thyroid Carcinoma Model
- Author
-
Drosten, M., Stiewe, T., and Pützer, B.M.
- Abstract
Gain-of-function mutations in the RET proto-oncogene resulting in a constitutively active receptor tyrosine kinase have been identified as responsible for three subtypes of multiple endocrine neoplasia type 2 (MEN-2) and the development of sporadic medullary and papillary thyroid carcinoma. An important strategy in cancer gene therapy is the inhibition of oncogenic signal transduction by interfering with the molecular mechanisms of activation. In the present study, we tested the therapeutic capacity of an adenovirus expressing a dominant-negative (dn) RET mutant, RET51.flag, under the control of a synthetic C cell-selective calcitonin promoter (TSE2.CP1) against human medullary thyroid cancer (MTC). Infection of human MTC-derived TT cells with Ad-TSE2.CP1-dn-RET51.flag resulted in the accumulation of immature RET protein in the endoplasmic reticulum and a strong reduction of oncogenic RET receptor on the cell surface, indicating that RET51.flag exhibits a dominant-negative effect over endogenous oncogenic protein. Analysis of potential downstream mechanisms associated with the inhibition of oncogenic RET signaling by overexpression of mutant RET51.flag revealed a significant loss of cell viability in TT cells due to the induction of apoptosis. Finally, we examined the antitumor activity of the dominant-negative RET approach in vivo. Inoculation of Ad-TSE2.CP1- dn-RET51.flag-expressing MTC cells into nude mice led to complete suppression of tumor growth. Moreover, a single intratumoral injection of Ad-TSE2.CP1-dn-RET51.flag into established thyroid tumors resulted in prolonged survival of treated mice compared with the controls. Our data suggest that adenoviral delivery of dn-RET51.flag may be a reliable strategy of effective molecular intervention for RET oncogene-related MTC.
- Published
- 2003
- Full Text
- View/download PDF
33. Increased ?N-p73 expression in tumors by upregulation of the E2F1-regulated, TA-promoter-derived ?N'-p73 transcript.
- Author
-
Putzer, B M, Tuve, S, Tannapfel, A, and Stiewe, T
- Subjects
TUMOR suppressor proteins ,TUMORS ,CELL death ,TRANSCRIPTION factors - Abstract
Cell Death and Differentiation (2003) 10, 612-614. doi:10.1038/sj.cdd.4401205 [ABSTRACT FROM AUTHOR]
- Published
- 2003
34. Deregulation of PPARβ/δ target genes in tumor-associated macrophages by fatty acid ligands in the ovarian cancer microenvironment
- Author
-
Schumann T, Adhikary T, Wortmann A, Finkernagel F, Lieber S, Schnitzer E, Legrand N, Schober Y, Wa, Nockher, Pm, Toth, We, Diederich, Nist A, Stiewe T, Wagner U, Reinartz S, Müller-Brüsselbach S, and Rolf Müller
35. Malignant transformation in a defined genetic background: proteome changes displayed by 2D-PAGE
- Author
-
Vogiatzi Fotini, Pütz Stephanie M, Stiewe Thorsten, and Sickmann Albert
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Cancer arises from normal cells through the stepwise accumulation of genetic alterations. Cancer development can be studied by direct genetic manipulation within experimental models of tumorigenesis. Thereby, confusion by the genetic heterogeneity of patients can be circumvented. Moreover, identification of the critical changes that convert a pre-malignant cell into a metastatic, therapy resistant tumor cell, however, is one necessary step to develop effective and selective anti-cancer drugs. Thus, for the current study a cell culture model for malignant transformation was used: Primary human fibroblasts of the BJ strain were sequentially transduced with retroviral vectors encoding the genes for hTERT (cell line BJ-T), simian virus 40 early region (SV40 ER, cell line BJ-TE) and H-Ras V12 (cell line BJ-TER). Results The stepwise malignant transformation of human fibroblasts was analyzed on the protein level by differential proteome analysis. We observed 39 regulated protein spots and therein identified 67 different proteins. The strongest change of spot patterns was detected due to integration of SV40 ER. Among the proteins being significantly regulated during the malignant transformation process well known proliferating cell nuclear antigen (PCNA) as well as the chaperones mitochondrial heat shock protein 75 kDa (TRAP-1) and heat shock protein HSP90 were identified. Moreover, we find out, that TRAP-1 is already up-regulated by means of SV40 ER expression instead of H-Ras V12. Furthermore Peroxiredoxin-6 (PRDX6), Annexin A2 (p36), Plasminogen activator inhibitor 2 (PAI-2) and Keratin type II cytoskeletal 7 (CK-7) were identified to be regulated. For some protein candidates we confirmed our 2D-PAGE results by Western Blot. Conclusion These findings give further hints for intriguing interactions between the p16-RB pathway, the mitochondrial chaperone network and the cytoskeleton. In summary, using a cell culture model for malignant transformation analyzed with 2D-PAGE, proteome and cellular changes can be related to defined steps of tumorigenesis.
- Published
- 2010
- Full Text
- View/download PDF
36. 330 - Mutant p53 promotes tumor progression by the endoplasmic reticulum UDPase ENTPD5.
- Author
-
Vogiatzi, F., Brandt, D.T., Fuchs, J., Grikscheit, K., Timofeev, O., Nist, A., Mernberger, M., Grosse, R., and Stiewe, T.
- Published
- 2016
- Full Text
- View/download PDF
37. 350 - Monitoring the dynamics of clonal tumor evolution in vivo using secreted luciferases.
- Author
-
Charles, J.P., Fuchs, J., Hefter, M., Vischedyk, J.B., Kleint, M., Vogiatzi, F., Nist, A., Timofeev, O., Wanzel, M., and Stiewe, T.
- Published
- 2016
- Full Text
- View/download PDF
38. 338 - Widespread epigenetic activation of ΔNp73 in small cell lung cancer causes vulnerability to Tip60-p400 inhibition.
- Author
-
Nist, A., Krampitz, A.M., Schlereth, K., Mernberger, M., Moßner, M.C., Muley, T., Dammann, R., and Stiewe, T.
- Published
- 2016
- Full Text
- View/download PDF
39. Molecular adaptation to neoadjuvant immunotherapy in triple-negative breast cancer.
- Author
-
Denkert C, Schneeweiss A, Rey J, Karn T, Hattesohl A, Weber KE, Rachakonda S, Braun M, Huober J, Jank P, Sinn HP, Zahm DM, Felder B, Hanusch C, Teply-Szymanski J, Marmé F, Fehm T, Thomalla J, Sinn BV, Stiewe T, Marczyk M, Blohmer JU, van Mackelenbergh M, Schem C, Staib P, Link T, Müller V, Stickeler E, Stover DG, Solbach C, Metzger-Filho O, Jackisch C, Geyer CE Jr, Fasching PA, Pusztai L, Nekljudova V, Untch M, and Loibl S
- Subjects
- Humans, Female, Gene Expression Regulation, Neoplastic drug effects, Antibodies, Monoclonal therapeutic use, Prognosis, Triple Negative Breast Neoplasms immunology, Triple Negative Breast Neoplasms therapy, Triple Negative Breast Neoplasms pathology, Triple Negative Breast Neoplasms drug therapy, Neoadjuvant Therapy methods, Immunotherapy methods, Tumor Microenvironment immunology, Tumor Microenvironment drug effects
- Abstract
Therapy-induced molecular adaptation of triple-negative breast cancer is crucial for immunotherapy response and resistance. We analyze tumor biopsies from three different time points in the randomized neoadjuvant GeparNuevo trial (NCT02685059), evaluating the combination of durvalumab with chemotherapy, for longitudinal alterations of gene expression. Durvalumab induces an activation of immune and stromal gene expression as well as a reduction of proliferation-related gene expression. Immune genes are positive prognostic factors irrespective of treatment, while proliferation genes are positive prognostic factors only in the durvalumab arm. We identify stromal-related gene expression as a contributor to immunotherapy resistance and poor therapy response. The results provide evidence from clinical trial cohorts suggesting a role for stromal reorganization in therapy resistance to immunotherapy and in the generation of an immune-suppressive microenvironment, which might be relevant for future therapy approaches targeting the tumor stroma parallel to immunotherapy, such as combinations of immunotherapy with anti-angiogenic therapy., Competing Interests: Declaration of interests C.D. reports grants from European Commission H2020, grants from German Cancer Aid Translational Oncology, grants from German Breast Group, and grants from BMBF to the institution during the conduct of the study; personal fees from Novartis, personal fees from Roche, personal fees from MSD Oncology, personal fees from Daiichi Sankyo, personal fees from AstraZeneca and Molecular Health, grants from Myriad, personal fees from Merck, and other funding from Sividon Diagnostics outside the submitted work; in addition, C.D. has a patent VMScope digital pathology software with royalties paid, a patent WO2020109570A1—cancer immunotherapy pending, and a patent WO2015114146A1 and WO2010076322A1—therapy response issued. J.R. declares to be a GBG Forschungs GmbH employee. GBG Forschungs GmbH received funding for research grants from AbbVie, Amgen, AstraZeneca, BMS, Daiichi Sankyo, Gilead, Molecular Health, Novartis, Pfizer, and Roche (paid to the institution). Funding was also received (non-financial/medical writing) from Daiichi Sankyo, Gilead, Novartis, Pfizer, Roche, and Seagen (paid to the institution). GBG Forschungs GmbH has royalties in VM Scope and patents pending: EP14153692.0, EP21152186.9, and EP15702464.7. T.K. reports a patent WO2020109570A1 pending. S.R. declares to be a GBG Forschungs GmbH employee. GBG Forschungs GmbH received funding for research grants from AbbVie, Amgen, AstraZeneca, BMS, Daiichi Sankyo, Gilead, Molecular Health, Novartis, Pfizer, and Roche (paid to the institution). J.H. reports research funding from Lilly; honoraria from Lilly, Novartis, Roche, Pfizer, AstraZeneca, Seagen, Gilead, and Daiichi; consulting and advisory relationships with Lilly, Novartis, Roche, Pfizer, AstraZeneca, Gilead, and Daiichi; travel expenses from Roche, Novartis, Daiichi, and Gilead. P.J. reports research funding and travel expenses from Gilead Sciences GmbH. C.H. reports an advisory role and speakers bureau role for AstraZeneca, Roche, Novartis, and Aristo Pharma. B.V.S. is an employee of BioNTech SE and reports a patent WO2020109570A1 pending. J.-U.B. reports consultation fees, honoraria, and reimbursement for attending symposia from AstraZeneca, Amgen, Daiichi Sankyo, Eisai, Gilead, Lilly, MSD, Novartis, Pfizer, Roche, and Seagen. M.v.M. reports personal fees, honoraria, or travel grants from Amgen, AstraZeneca, Daiichi Sankyo, Genomic Health, GSK, Lilly, Molecular Health, MSD, Mylan, Novartis, Pfizer, Pierre Fabre, Roche, and Seagen. C. Schem reports speaker activities for Roche, Pfizer, Novartis, Celgen, Novartis, Exact Sciences, MSD, AstraZeneca, Lilly, and Seagen, as well as advisory boards for Roche, Astra Zeneca, Pfizer, Novartis, MSD, Amgen, Exact Sciences, Stemline, Lilly, and Novartis. T.L. reports personal fees from Amgen, Roche, Teva, Clovis, Tesaro, MSD, Novartis, Pfizer, Lilly, GSK, Gilead, AstraZeneca, Daiichi Sankyo, Stemline, and Seagen outside of the submitted work. T.L. participates in advisory boards from Amgen, MSD, Tesaro, Roche, Pfizer, Lilly, Myriad, Esai, GSK, Gilead, Daiichi Sankyo, Roche, and AstraZeneca outside of the submitted work and T.L. received travel support from Pfizer, PharmaMar, MSD, Celgene, Roche, AstraZeneca, Gilead, Daiichi Sankyo, Stemline, and Clovis outside of the submitted work. P.S. reports grants, personal fees, and non-financial support from AbbVie, Amgen, AstraZeneca, Bristol Myers Squibb Company, MSD, Incyte, Janssen-Cilag, Novartis, Takeda, Pfizer, and Roche. V.M. received speaker honoraria from AstraZeneca, Daiichi Sankyo, Eisai, GSK, Pfizer, MSD, Medac, Novartis, Roche, Seagen, Onkowissen, high5 Oncology, Medscape, Gilead, and Pierre Fabre; consultancy honoraria from Roche, Pierre Fabre, Amgen, ClinSol, Novartis, MSD, Daiichi Sankyo, Eisai, Lilly, Sanofi, Seagen, Gilead, and Stemline; institutional research support from Novartis, Roche, Seagen, and Genentech; and travel grants from Roche, Pfizer, Daiichi Sankyo, and Gilead. L.P. has received consulting fees and honoraria for advisory board participation from Pfizer, AstraZeneca, Merck, Novartis, Bristol Myers Squibb, GlaxoSmithKline, Genentech/Roche, Personalis, Daiichi, Natera, and Exact Sciences and institutional research funding from Seagen, GlaxoSmithKline, AstraZeneca, Merck, Pfizer, and Bristol Myers Squibb. C.E.G. reports the following competing interests: Exact Sciences, Advisory Board, personal; AbbVie, Steering Committee member, institutional, co-chair of SC for BrighTNess; Daiichi Sankyo, member SC, institutional, co-chair of SC for DESTINY-Breast05; Genentech/Roche, SC member, institutional, co-chair of SC for lidERA; Genentech/Roche, coordinating PI, institutional, NSABP B-59/GeparDouze; and Genentech/Roche, SC member, institutional, co-chair of SC for KATHERINE. C.J. reports honoraria from AstraZeneca, Amgen, Daiichi Sankyo, Lilly, Roche, Pfizer, MSD Oncology, Pierre Fabre, Sanofi-Aventis, Seagen, Gilead, and Novartis and has a consulting or advisory role for Amgen, Lilly, Roche, Pfizer, Pierre Fabre, Novartis, MSD Oncology, Agendia, Seagen, Gilead, Lilly, Stemline, and Medac. V.N. declares to be a GBG Forschungs GmbH employee. GBG Forschungs GmbH received funding for research grants from AbbVie, AstraZeneca, BMS, Daiichi Sankyo, Gilead, Novartis, Pfizer, and Roche (paid to the institution). GBG Forschungs GmbH received other funding from Daiichi Sankyo, Gilead, Novartis, Pfizer, Roche, and Seagen (paid to the institution). GBG Forschungs GmbH has the following royalties/patents: EP14153692.0, EP21152186.9, EP15702464.7, EP19808852.8, and VM Scope GmbH. M.U. reports honoraria from AstraZeneca, Amgen, Daiichi Sankyo, Lilly, Roche, Pfizer, MSD Oncology, Pierre Fabre, Sanofi-Aventis, Myriad, Seagen, Gilead, and Novartis; has a consulting or advisory role for Amgen, Lilly, Roche, Pfizer, Pierre Fabre, Novartis, MSD Oncology, Agendia, Seagen, Gilead, Lily, Stemline, Genzyme, and Medac; and all honoraria and fees are paid to the employer/institution. S.L. reports grants and other funding from AbbVie; other funding from Amgen; grants and other funding from AstraZeneca; other funding from BMS; grants and other funding from Celgene; grants, non-financial support, and other funding from Daiichi Sankyo; other funding from EirGenix; other funding from Eisai Europe Ltd; other funding from GSK; grants, non-financial support, and other funding from Immunomedics/Gilead; other funding from Lilly; other funding from Merck; grants from Molecular Health; grants, non-financial support, and other funding from Novartis; grants, non-financial support, and other funding from Pfizer; other funding from Pierre Fabre; other funding from Relay Therapeutics; grants, non-financial support, and other funding from Roche; other funding from Sanofi; non-financial support and other funding from Seagen; and other funding from Olema Pharmaceuticals, outside the submitted work. In addition, S.L. has a patent EP14153692.0 pending, a patent EP21152186.9 pending, a patent EP15702464.7 issued, a patent EP19808852.8 pending, and a patent Digital Ki67 Evaluator with royalties paid., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
40. Arachidonic acid impairs natural killer cell functions by disrupting signaling pathways driven by activating receptors and reactive oxygen species.
- Author
-
Hammoud MK, Meena C, Dietze R, Hoffmann N, Szymanski W, Finkernagel F, Nist A, Stiewe T, Graumann J, von Strandmann EP, and Müller R
- Subjects
- Humans, Female, Cell Line, Tumor, Ovarian Neoplasms pathology, Ovarian Neoplasms metabolism, Ovarian Neoplasms genetics, STAT1 Transcription Factor metabolism, STAT1 Transcription Factor genetics, Cell Proliferation, Killer Cells, Natural immunology, Killer Cells, Natural metabolism, Reactive Oxygen Species metabolism, Signal Transduction, Arachidonic Acid metabolism
- Abstract
Background: High levels of the polyunsaturated fatty acid arachidonic acid (AA) within the ovarian carcinoma (OC) microenvironment correlate with reduced relapse-free survival. Furthermore, OC progression is tied to compromised immunosurveillance, partially attributed to the impairment of natural killer (NK) cells. However, potential connections between AA and NK cell dysfunction in OC have not been studied., Methods: We employed a combination of phosphoproteomics, transcriptional profiling and biological assays to investigate AA's impact on NK cell functions., Results: AA (i) disrupts interleukin-2/15-mediated expression of pro-inflammatory genes by inhibiting STAT1-dependent signaling, (ii) hampers signaling by cytotoxicity receptors through disruption of their surface expression, (iii) diminishes phosphorylation of NKG2D-induced protein kinases, including ERK1/2, LYN, MSK1/2 and STAT1, and (iv) alters reactive oxygen species production by transcriptionally upregulating detoxification. These modifications lead to a cessation of NK cell proliferation and a reduction in cytotoxicity., Conclusion: Our findings highlight significant AA-induced alterations in the signaling network that regulates NK cell activity. As low expression of several NK cell receptors correlates with shorter OC patient survival, these findings suggest a functional linkage between AA, NK cell dysfunction and OC progression., Competing Interests: Declarations Ethics approval and consent to participate Blood cells from healthy individuals were used with the informed consent of the donors and approved by the local ethics committee (205/10). Consent for publication All participants consented to submit the manuscript to the journal. Competing interests The authors declare no competing interests., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
41. TP53: the unluckiest of genes?
- Author
-
Joerger AC, Stiewe T, and Soussi T
- Abstract
The transcription factor p53 plays a key role in the cellular defense against cancer development. It is inactivated in virtually every tumor, and in every second tumor this inactivation is due to a mutation in the TP53 gene. In this perspective, we show that this diverse mutational spectrum is unique among all other cancer-associated proteins and discuss what drives the selection of TP53 mutations in cancer. We highlight that several factors conspire to make the p53 protein particularly vulnerable to inactivation by the mutations that constantly plague our genome. It appears that the TP53 gene has emerged as a victim of its own evolutionary past that shaped its structure and function towards a pluripotent tumor suppressor, but came with an increased structural fragility of its DNA-binding domain. TP53 loss of function - with associated dominant-negative effects - is the main mechanism that will impair TP53 tumor suppressive function, regardless of whether a neomorphic phenotype is associated with some of these variants., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
42. Distinct negative-sense RNA viruses induce a common set of transcripts encoding proteins forming an extensive network.
- Author
-
Hofmann N, Bartkuhn M, Becker S, Biedenkopf N, Böttcher-Friebertshäuser E, Brinkrolf K, Dietzel E, Fehling SK, Goesmann A, Heindl MR, Hoffmann S, Karl N, Maisner A, Mostafa A, Kornecki L, Müller-Kräuter H, Müller-Ruttloff C, Nist A, Pleschka S, Sauerhering L, Stiewe T, Strecker T, Wilhelm J, Wuerth JD, Ziebuhr J, Weber F, and Schmitz ML
- Subjects
- Humans, RNA Viruses genetics, Host-Pathogen Interactions genetics, Gene Expression Profiling, Gene Regulatory Networks, Cell Line, Tumor, RNA Virus Infections virology, Sequence Analysis, RNA, Signal Transduction, RNA, Viral genetics, RNA, Viral metabolism
- Abstract
The large group of negative-strand RNA viruses (NSVs) comprises many important pathogens. To identify conserved patterns in host responses, we systematically compared changes in the cellular RNA levels after infection of human hepatoma cells with nine different NSVs of different virulence degrees. RNA sequencing experiments indicated that the amount of viral RNA in host cells correlates with the number of differentially expressed host cell transcripts. Time-resolved differential gene expression analysis revealed a common set of 178 RNAs that are regulated by all NSVs analyzed. A newly developed open access web application allows downloads and visualizations of all gene expression comparisons for individual viruses over time or between several viruses. Most of the genes included in the core set of commonly differentially expressed genes (DEGs) encode proteins that serve as membrane receptors, signaling proteins and regulators of transcription. They mainly function in signal transduction and control immunity, metabolism, and cell survival. One hundred sixty-five of the DEGs encode host proteins from which 47 have already been linked to the regulation of viral infections in previous studies and 89 proteins form a complex interaction network that may function as a core hub to control NSV infections.IMPORTANCEThe infection of cells with negative-strand RNA viruses leads to the differential expression of many host cell RNAs. The differential spectrum of virus-regulated RNAs reflects a large variety of events including anti-viral responses, cell remodeling, and cell damage. Here, these virus-specific differences and similarities in the regulated RNAs were measured in a highly standardized model. A newly developed app allows interested scientists a wide range of comparisons and visualizations., Competing Interests: The authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
43. Haptoglobin buffers lipopolysaccharides to delay activation of NFκB.
- Author
-
Zein L, Grossmann J, Swoboda H, Borgel C, Wilke B, Awe S, Nist A, Stiewe T, Stehling O, Freibert SA, Adhikary T, and Chung HR
- Subjects
- Humans, Macrophages immunology, Macrophages metabolism, Animals, Toll-Like Receptor 4 metabolism, Mice, Protein Binding, Signal Transduction, RAW 264.7 Cells, Haptoglobins metabolism, Lipopolysaccharides, NF-kappa B metabolism
- Abstract
It has remained yet unclear which soluble factors regulate the anti-inflammatory macrophage phenotype observed in both homeostasis and tumourigenesis. We show here that haptoglobin, a major serum protein with elusive immunoregulatory properties, binds and buffers bacterial lipopolysaccharides to attenuate activation of NFκB in macrophages. Haptoglobin binds different lipopolysaccharides with low micromolar affinities. Given its abundance, haptoglobin constitutes a buffer for serum-borne lipopolysaccharides, shielding them to safeguard against aberrant inflammatory reactions by reducing the amount of free lipopolysaccharides available for binding to TLR4. Concordantly, NFκB activation by haptoglobin-associated lipopolysaccharides was markedly delayed relative to stimulation with pure lipopolysaccharide. Our findings warrant evaluation of therapeutic benefits of haptoglobin for inflammatory conditions and re-evaluation of purification strategies. Finally, they allow to elucidate mechanisms of enhanced immunosuppression by oncofetal haptoglobin., 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 © 2024 Zein, Grossmann, Swoboda, Borgel, Wilke, Awe, Nist, Stiewe, Stehling, Freibert, Adhikary and Chung.)
- Published
- 2024
- Full Text
- View/download PDF
44. DYRK1B blockade promotes tumoricidal macrophage activity in pancreatic cancer.
- Author
-
Brichkina A, Ems M, Suezov R, Singh R, Lutz V, Picard FSR, Nist A, Stiewe T, Graumann J, Daude M, Diederich WE, Finkernagel F, Chung HR, Bartsch DK, Roth K, Keber C, Denkert C, Huber M, Gress TM, and Lauth M
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Disease Models, Animal, Phagocytosis, Carcinoma, Pancreatic Ductal pathology, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal metabolism, Dyrk Kinases, Macrophages metabolism, Pancreatic Neoplasms pathology, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Protein Serine-Threonine Kinases metabolism, Protein Serine-Threonine Kinases genetics, Protein-Tyrosine Kinases antagonists & inhibitors, Protein-Tyrosine Kinases metabolism, Tumor Microenvironment
- Abstract
Objective: Highly malignant pancreatic ductal adenocarcinoma (PDAC) is characterised by an abundant immunosuppressive and fibrotic tumour microenvironment (TME). Future therapeutic attempts will therefore demand the targeting of tumours and stromal compartments in order to be effective. Here we investigate whether dual specificity and tyrosine phosphorylation-regulated kinase 1B (DYRK1B) fulfil these criteria and represent a promising anticancer target in PDAC., Design: We used transplantation and autochthonous mouse models of PDAC with either genetic Dyrk1b loss or pharmacological DYRK1B inhibition, respectively. Mechanistic interactions between tumour cells and macrophages were studied in direct or indirect co-culture experiments. Histological analyses used tissue microarrays from patients with PDAC. Additional methodological approaches included bulk mRNA sequencing (transcriptomics) and proteomics (secretomics)., Results: We found that DYRK1B is mainly expressed by pancreatic epithelial cancer cells and modulates the influx and activity of TME-associated macrophages through effects on the cancer cells themselves as well as through the tumour secretome. Mechanistically, genetic ablation or pharmacological inhibition of DYRK1B strongly attracts tumoricidal macrophages and, in addition, downregulates the phagocytosis checkpoint and 'don't eat me' signal CD24 on cancer cells, resulting in enhanced tumour cell phagocytosis. Consequently, tumour cells lacking DYRK1B hardly expand in transplantation experiments, despite their rapid growth in culture. Furthermore, combining a small-molecule DYRK1B-directed therapy with mammalian target of rapamycin inhibition and conventional chemotherapy stalls the growth of established tumours and results in a significant extension of life span in a highly aggressive autochthonous model of PDAC., Conclusion: In light of DYRK inhibitors currently entering clinical phase testing, our data thus provide a novel and clinically translatable approach targeting both the cancer cell compartment and its microenvironment., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2024
- Full Text
- View/download PDF
45. SAMD1 suppresses epithelial-mesenchymal transition pathways in pancreatic ductal adenocarcinoma.
- Author
-
Simon C, Brunke ID, Stielow B, Forné I, Steitz AM, Geller M, Rohner I, Weber LM, Fischer S, Jeude LM, Huber T, Nist A, Stiewe T, Huber M, Buchholz M, and Liefke R
- Subjects
- Animals, Humans, Cadherins metabolism, Cadherins genetics, Cell Line, Tumor, Cell Movement genetics, Intracellular Signaling Peptides and Proteins metabolism, Intracellular Signaling Peptides and Proteins genetics, Prognosis, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal pathology, Epithelial-Mesenchymal Transition genetics, F-Box Proteins metabolism, F-Box Proteins genetics, Gene Expression Regulation, Neoplastic, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms pathology, Receptors, LDL genetics, Receptors, LDL metabolism
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) poses a significant threat due to its tendency to evade early detection, frequent metastasis, and the subsequent challenges in devising effective treatments. Processes that govern epithelial-mesenchymal transition (EMT) in PDAC hold promise for advancing novel therapeutic strategies. SAMD1 (SAM domain-containing protein 1) is a CpG island-binding protein that plays a pivotal role in the repression of its target genes. Here, we revealed that SAMD1 acts as a repressor of genes associated with EMT. Upon deletion of SAMD1 in PDAC cells, we observed significantly increased migration rates. SAMD1 exerts its effects by binding to specific genomic targets, including CDH2, encoding N-cadherin, which emerged as a driver of enhanced migration upon SAMD1 knockout. Furthermore, we discovered the FBXO11-containing E3 ubiquitin ligase complex as an interactor and negative regulator of SAMD1, which inhibits SAMD1 chromatin-binding genome-wide. High FBXO11 expression in PDAC is associated with poor prognosis and increased expression of EMT-related genes, underlining an antagonistic relationship between SAMD1 and FBXO11. In summary, our findings provide insights into the regulation of EMT-related genes in PDAC, shedding light on the intricate role of SAMD1 and its interplay with FBXO11 in this cancer type., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2024 Simon et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2024
- Full Text
- View/download PDF
46. Intercellular pathways of cancer treatment-related cardiotoxicity and their therapeutic implications: the paradigm of radiotherapy.
- Author
-
Logotheti S, Pavlopoulou A, Rudsari HK, Galow AM, Kafalı Y, Kyrodimos E, Giotakis AI, Marquardt S, Velalopoulou A, Verginadis II, Koumenis C, Stiewe T, Zoidakis J, Balasingham I, David R, and Georgakilas AG
- Subjects
- Humans, Animals, Signal Transduction, Cardiovascular Diseases, Neoplasms radiotherapy, Neoplasms drug therapy, Cardiotoxicity etiology, Radiotherapy adverse effects
- Abstract
Advances in cancer therapeutics have improved patient survival rates. However, cancer survivors may suffer from adverse events either at the time of therapy or later in life. Cardiovascular diseases (CVD) represent a clinically important, but mechanistically understudied complication, which interfere with the continuation of best-possible care, induce life-threatening risks, and/or lead to long-term morbidity. These concerns are exacerbated by the fact that targeted therapies and immunotherapies are frequently combined with radiotherapy, which induces durable inflammatory and immunogenic responses, thereby providing a fertile ground for the development of CVDs. Stressed and dying irradiated cells produce 'danger' signals including, but not limited to, major histocompatibility complexes, cell-adhesion molecules, proinflammatory cytokines, and damage-associated molecular patterns. These factors activate intercellular signaling pathways which have potentially detrimental effects on the heart tissue homeostasis. Herein, we present the clinical crosstalk between cancer and heart diseases, describe how it is potentiated by cancer therapies, and highlight the multifactorial nature of the underlying mechanisms. We particularly focus on radiotherapy, as a case known to often induce cardiovascular complications even decades after treatment. We provide evidence that the secretome of irradiated tumors entails factors that exert systemic, remote effects on the cardiac tissue, potentially predisposing it to CVDs. We suggest how diverse disciplines can utilize pertinent state-of-the-art methods in feasible experimental workflows, to shed light on the molecular mechanisms of radiotherapy-related cardiotoxicity at the organismal level and untangle the desirable immunogenic properties of cancer therapies from their detrimental effects on heart tissue. Results of such highly collaborative efforts hold promise to be translated to next-generation regimens that maximize tumor control, minimize cardiovascular complications, and support quality of life in cancer survivors., Competing Interests: Declaration of competing interest The authors have no competing interests to declare., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
47. BAG6 restricts pancreatic cancer progression by suppressing the release of IL33-presenting extracellular vesicles and the activation of mast cells.
- Author
-
Alashkar Alhamwe B, Ponath V, Alhamdan F, Dörsam B, Landwehr C, Linder M, Pauck K, Miethe S, Garn H, Finkernagel F, Brichkina A, Lauth M, Tiwari DK, Buchholz M, Bachurski D, Elmshäuser S, Nist A, Stiewe T, Pogge von Strandmann L, Szymański W, Beutgen V, Graumann J, Teply-Szymanski J, Keber C, Denkert C, Jacob R, Preußer C, and Pogge von Strandmann E
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Cell Proliferation, Mice, Inbred C57BL, Carcinoma, Pancreatic Ductal pathology, Carcinoma, Pancreatic Ductal immunology, Carcinoma, Pancreatic Ductal genetics, Disease Progression, Extracellular Vesicles metabolism, Interleukin-33 metabolism, Interleukin-33 genetics, Mast Cells metabolism, Mast Cells immunology, Pancreatic Neoplasms pathology, Pancreatic Neoplasms immunology, Tumor Microenvironment
- Abstract
Recent studies reveal a critical role of tumor cell-released extracellular vesicles (EVs) in pancreatic cancer (PC) progression. However, driver genes that direct EV function, the EV-recipient cells, and their cellular response to EV uptake remain to be identified. Therefore, we studied the role of Bcl-2-associated-anthanogene 6 (BAG6), a regulator of EV biogenesis for cancer progression. We used a Cre recombinase/LoxP-based reporter system in combination with single-cell RNA sequencing to monitor in vivo EV uptake and tumor microenvironment (TME) changes in mouse models for pancreatic ductal adenocarcinoma (PDAC) in a Bag6 pro- or deficient background. In vivo data were validated using mouse and human organoids and patient samples. Our data demonstrated that Bag6-deficient subcutaneous and orthotopic PDAC tumors accelerated tumor growth dependent on EV release. Mechanistically, this was attributed to mast cell (MC) activation via EV-associated IL33. Activated MCs promoted tumor cell proliferation and altered the composition of the TME affecting fibroblast polarization and immune cell infiltration. Tumor cell proliferation and fibroblast polarization were mediated via the MC secretome containing high levels of PDGF and CD73. Patients with high BAG6 gene expression and high protein plasma level have a longer overall survival indicating clinical relevance. The current study revealed a so far unknown tumor-suppressing activity of BAG6 in PDAC. Bag6-deficiency allowed the release of EV-associated IL33 which modulate the TME via MC activation promoting aggressive tumor growth. MC depletion using imatinib diminished tumor growth providing a scientific rationale to consider imatinib for patients stratified with low BAG6 expression and high MC infiltration. EVs derived from BAG6-deficient pancreatic cancer cells induce MC activation via IL33/Il1rl1. The secretome of activated MCs induces tumor proliferation and changes in the TME, particularly shifting fibroblasts into an inflammatory cancer-associated fibroblast (iCAF) phenotype. Blocking EVs or depleting MCs restricts tumor growth., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
48. KRAS and TP53 co-mutation predicts benefit of immune checkpoint blockade in lung adenocarcinoma.
- Author
-
Budczies J, Romanovsky E, Kirchner M, Neumann O, Blasi M, Schnorbach J, Shah R, Bozorgmehr F, Savai R, Stiewe T, Peters S, Schirmacher P, Thomas M, Kazdal D, Christopoulos P, and Stenzinger A
- Subjects
- Humans, Female, Male, Aged, Middle Aged, Biomarkers, Tumor genetics, Immunotherapy methods, Prognosis, Aged, 80 and over, Adult, Cohort Studies, Tumor Suppressor Protein p53 genetics, Mutation, Proto-Oncogene Proteins p21(ras) genetics, Adenocarcinoma of Lung genetics, Adenocarcinoma of Lung drug therapy, Adenocarcinoma of Lung immunology, Adenocarcinoma of Lung pathology, Immune Checkpoint Inhibitors therapeutic use, Lung Neoplasms genetics, Lung Neoplasms drug therapy, Lung Neoplasms pathology, Lung Neoplasms immunology
- Abstract
Background: Predictive biomarkers in use for immunotherapy in advanced non-small cell lung cancer are of limited sensitivity and specificity. We analysed the potential of activating KRAS and pathogenic TP53 mutations to provide additional predictive information., Methods: The study cohort included 713 consecutive immunotherapy patients with advanced lung adenocarcinomas, negative for actionable genetic alterations. Additionally, two previously published immunotherapy and two surgical patient cohorts were analyzed. Therapy benefit was stratified by KRAS and TP53 mutations. Molecular characteristics underlying KRASmut/TP53mut tumours were revealed by the analysis of TCGA data., Results: An interaction between KRAS and TP53 mutations was observed in univariate and multivariate analyses of overall survival (Hazard ratio [HR] = 0.56, p = 0.0044 and HR = 0.53, p = 0.0021) resulting in a stronger benefit for KRASmut/TP53mut tumours (HR = 0.71, CI 0.55-0.92). This observation was confirmed in immunotherapy cohorts but not observed in surgical cohorts. Tumour mutational burden, proliferation, and PD-L1 mRNA were significantly higher in TP53-mutated tumours, regardless of KRAS status. Genome-wide expression analysis revealed 64 genes, including CX3CL1 (fractalkine), as specific transcriptomic characteristic of KRASmut/TP53mut tumours., Conclusions: KRAS/TP53 co-mutation predicts ICI benefit in univariate and multivariate survival analyses and is associated with unique molecular tumour features. Mutation testing of the two genes can be easily implemented using small NGS panels., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
49. IRF2BP2 counteracts the ATF7/JDP2 AP-1 heterodimer to prevent inflammatory overactivation in acute myeloid leukemia (AML) cells.
- Author
-
Fischer S, Weber LM, Stielow B, Frech M, Simon C, Geller M, Könnecke J, Finkernagel F, Forné I, Nist A, Bauer UM, Stiewe T, Neubauer A, and Liefke R
- Subjects
- Humans, Cell Line, Tumor, Activating Transcription Factors metabolism, Activating Transcription Factors genetics, Chromatin metabolism, Cell Proliferation, Repressor Proteins metabolism, Repressor Proteins genetics, HEK293 Cells, Gene Expression Regulation, Leukemic, Protein Multimerization, Transcription Factors metabolism, Transcription Factors genetics, DNA-Binding Proteins, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute pathology, Transcription Factor AP-1 metabolism, Transcription Factor AP-1 genetics, Inflammation genetics, Inflammation metabolism
- Abstract
Acute myeloid leukemia (AML) is a hematological malignancy characterized by abnormal proliferation and accumulation of immature myeloid cells in the bone marrow. Inflammation plays a crucial role in AML progression, but excessive activation of cell-intrinsic inflammatory pathways can also trigger cell death. IRF2BP2 is a chromatin regulator implicated in AML pathogenesis, although its precise role in this disease is not fully understood. In this study, we demonstrate that IRF2BP2 interacts with the AP-1 heterodimer ATF7/JDP2, which is involved in activating inflammatory pathways in AML cells. We show that IRF2BP2 is recruited by the ATF7/JDP2 dimer to chromatin and counteracts its gene-activating function. Loss of IRF2BP2 leads to overactivation of inflammatory pathways, resulting in strongly reduced proliferation. Our research indicates that a precise equilibrium between activating and repressive transcriptional mechanisms creates a pro-oncogenic inflammatory environment in AML cells. The ATF7/JDP2-IRF2BP2 regulatory axis is likely a key regulator of this process and may, therefore, represent a promising therapeutic vulnerability for AML. Thus, our study provides new insights into the molecular mechanisms underlying AML pathogenesis and identifies a potential therapeutic target for AML treatment., (© The Author(s) 2024. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2024
- Full Text
- View/download PDF
50. Transcriptomic response of prostate cancer cells to carbon ion and photon irradiation with focus on androgen receptor and TP53 signaling.
- Author
-
Hänze J, Mengen LM, Mernberger M, Tiwari DK, Plagge T, Nist A, Subtil FSB, Theiss U, Eberle F, Roth K, Lauth M, Hofmann R, Engenhart-Cabillic R, Stiewe T, and Hegele A
- Subjects
- Humans, Male, Carbon, Cell Line, Tumor, DNA Damage radiation effects, DNA Repair, Gene Expression Regulation, Neoplastic radiation effects, Gene Expression Regulation, Neoplastic drug effects, Heavy Ion Radiotherapy, Photons, Prostatic Neoplasms radiotherapy, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Receptors, Androgen metabolism, Receptors, Androgen genetics, Signal Transduction radiation effects, Transcriptome radiation effects, Tumor Suppressor Protein p53 metabolism
- Abstract
Background: Radiotherapy is essential in the treatment of prostate cancer. An alternative to conventional photon radiotherapy is the application of carbon ions, which provide a superior intratumoral dose distribution and less induced damage to adjacent healthy tissue. A common characteristic of prostate cancer cells is their dependence on androgens which is exploited therapeutically by androgen deprivation therapy in the advanced prostate cancer stage. Here, we aimed to analyze the transcriptomic response of prostate cancer cells to irradiation by photons in comparison to carbon ions, focusing on DNA damage, DNA repair and androgen receptor signaling., Methods: Prostate cancer cell lines LNCaP (functional TP53 and androgen receptor signaling) and DU145 (dysfunctional TP53 and androgen receptor signaling) were irradiated by photons or carbon ions and the subsequent DNA damage was assessed by immuno-cytofluorescence. Furthermore, the cells were treated with an androgen-receptor agonist. The effects of irradiation and androgen treatment on the gene regulation and the transcriptome were investigated by RT-qPCR and RNA sequencing, followed by bioinformatic analysis., Results: Following photon or carbon ion irradiation, both LNCaP and DU145 cells showed a dose-dependent amount of visible DNA damage that decreased over time, indicating occurring DNA repair. In terms of gene regulation, mRNAs involved in the TP53-dependent DNA damage response were significantly upregulated by photons and carbon ions in LNCaP but not in DU145 cells, which generally showed low levels of gene regulation after irradiation. Both LNCaP and DU145 cells responded to photons and carbon ions by downregulation of genes involved in DNA repair and cell cycle, partially resembling the transcriptome response to the applied androgen receptor agonist. Neither photons nor carbon ions significantly affected canonical androgen receptor-dependent gene regulation. Furthermore, certain genes that were specifically regulated by either photon or carbon ion irradiation were identified., Conclusion: Photon and carbon ion irradiation showed a significant congruence in terms of induced signaling pathways and transcriptomic responses. These responses were strongly impacted by the TP53 status. Nevertheless, irradiation mode-dependent distinct gene regulations with undefined implication for radiotherapy outcome were revealed. Androgen receptor signaling and irradiations shared regulation of certain genes with respect to DNA-repair and cell-cycle., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.