35 results on '"Venz, S"'
Search Results
2. Marine alkaloid Monanchocidin a overcomes drug resistance by induction of autophagy and lysosomal membrane permeabilization
- Author
-
Dyshlovoy, S A, Hauschild, J, Amann, K, Tabakmakher, K M, Venz, S, Walther, R, Guzii, A G, Makarieva, T N, Shubina, L K, Fedorov, S N, Stonik, V A, Bokemeyer, C, Balabanov, S, Honecker, F, von Amsberg, G, University of Zurich, and Dyshlovoy, S A
- Subjects
10032 Clinic for Oncology and Hematology ,610 Medicine & health ,2730 Oncology - Published
- 2015
3. A novel mouse model for inhibition of DOHH-mediated hypusine modification reveals a crucial function in embryonic development, proliferation and oncogenic transformation
- Author
-
Sievert, H, Pällmann, N, Miller, K K, Hermanns-Borgmeyer, I, Venz, S, Sendoel, A, Preukschas, M, Schweizer, M, Boettcher, S, Janiesch, P C, Streichert, T, Walther, R, Hengartner, M O, Manz, M G, Brümmendorf, T H, Bokemeyer, C, Braig, M, Hauber, J, Duncan, K E, Balabanov, S, Sievert, H, Pällmann, N, Miller, K K, Hermanns-Borgmeyer, I, Venz, S, Sendoel, A, Preukschas, M, Schweizer, M, Boettcher, S, Janiesch, P C, Streichert, T, Walther, R, Hengartner, M O, Manz, M G, Brümmendorf, T H, Bokemeyer, C, Braig, M, Hauber, J, Duncan, K E, and Balabanov, S
- Abstract
The central importance of translational control by post-translational modification has spurred major interest in regulatory pathways that control translation. One such pathway uniquely adds hypusine to eukaryotic initiation factor 5A (eIF5A), and thereby affects protein synthesis and, subsequently, cellular proliferation through an unknown mechanism. Using a novel conditional knockout mouse model and a Caenorhabditis elegans knockout model, we found an evolutionarily conserved role for the DOHH-mediated second step of hypusine synthesis in early embryonic development. At the cellular level, we observed reduced proliferation and induction of senescence in 3T3 Dohh-/- cells as well as reduced capability for malignant transformation. Furthermore, mass spectrometry showed that deletion of DOHH results in an unexpected complete loss of hypusine modification. Our results provide new biological insight into the physiological roles of the second step of the hypusination of eIF5A. Moreover, the conditional mouse model presented here provides a powerful tool for manipulating hypusine modification in a temporal and spatial manner, to analyse both how this unique modification normally functions in vivo as well as how it contributes to different pathological conditions.
- Published
- 2014
4. Expression of annexin AI in conventional renal cell carcinoma (CRCC) correlates with tumour stage, Fuhrman grade, amount of eosinophilic cells and clinical outcome
- Author
-
Zimmermann, U., Woenckhaus, C., Teller, S., Venz, S., Langheinrich, M., Protzel, C., Maile, S., Junker, H., and Giebel, J.
- Subjects
616.6 - Patología del sistema genitourinario ,Kidney ,Cancer - Abstract
There is increasing evidence that Annexin AI (ANX AI) expression is dysregulated in several carcinomas and tumour cell lines. In order to gain insight into the putative role of ANX AI in tumorigenesis, clinical outcome and metastatic potential of conventional renal cell carcinomas (CRCCs) we investigated the expression of ANX AI in CRCCs and metastases. Furthermore, it was elucidated whether ANX AI overexpression affects migratory potential in Caki-1 cells. ANX AI immunohistochemistry was performed on 33 samples of CRCCs and 10 metastases. ANX AI expression was assessed in 12 samples by 2-dimensional gelelectrophoresis (2-DE), subsequent mass spectrometry and RT-PCR. Immunohistochemical data were statistically correlated with pathological parameters, amount of eosinophilic cells and clinical outcome. Furthermore, a haptotactic migration assay was done on Caki-1 cells transfected with ANX AI. Immunostaining for ANX AI was found in 18 tumours and all metastases investigated. Intensity of immunohistochemical staining correlated to Fuhrman grade, amount of eosinophilic cells and clinical outcome. 2-DE and RT-PCR confirmed the presence of ANX AI in neoplastic tissue. Overexpression of ANX AI did not significantly influence cell migration. From these findings ANX AI expression seems to be related to Fuhrman grade, clinical outcome and metastatic potential of CRCCs. Thus ANX AI could serve as a prognostic marker for tumour progression.
- Published
- 2007
5. Combination of a proteomics approach and reengineering of meso scale network models for prediction of mode-of-action for tyrosine kinase inhibitors
- Author
-
Balabanov, S, Wilhelm, T, Venz, S, Keller, G, Scharf, C, Pospisil, H, Braig, M, Barett, C, Bokemeyer, C, Walther, R, Brümmendorf, T H, Schuppert, A, Balabanov, S, Wilhelm, T, Venz, S, Keller, G, Scharf, C, Pospisil, H, Braig, M, Barett, C, Bokemeyer, C, Walther, R, Brümmendorf, T H, and Schuppert, A
- Abstract
In drug discovery, the characterisation of the precise modes of action (MoA) and of unwanted off-target effects of novel molecularly targeted compounds is of highest relevance. Recent approaches for identification of MoA have employed various techniques for modeling of well defined signaling pathways including structural information, changes in phenotypic behavior of cells and gene expression patterns after drug treatment. However, efficient approaches focusing on proteome wide data for the identification of MoA including interference with mutations are underrepresented. As mutations are key drivers of drug resistance in molecularly targeted tumor therapies, efficient analysis and modeling of downstream effects of mutations on drug MoA is a key to efficient development of improved targeted anti-cancer drugs. Here we present a combination of a global proteome analysis, reengineering of network models and integration of apoptosis data used to infer the mode-of-action of various tyrosine kinase inhibitors (TKIs) in chronic myeloid leukemia (CML) cell lines expressing wild type as well as TKI resistance conferring mutants of BCR-ABL. The inferred network models provide a tool to predict the main MoA of drugs as well as to grouping of drugs with known similar kinase inhibitory activity patterns in comparison to drugs with an additional MoA. We believe that our direct network reconstruction approach, demonstrated on proteomics data, can provide a complementary method to the established network reconstruction approaches for the preclinical modeling of the MoA of various types of targeted drugs in cancer treatment. Hence it may contribute to the more precise prediction of clinically relevant on- and off-target effects of TKIs.
- Published
- 2013
6. Protein-protein-interaction network organization of the hypusine modification system
- Author
-
Sievert, H, Venz, S, Platas-Barradas, O, Dhople, V M, Schaletzky, M, Nagel, C H, Braig, M, Preukschas, M, Pällmann, N, Bokemeyer, C, Brümmendorf, T H, Pörtner, R, Walther, R, Duncan, K E, Hauber, J, Balabanov, S, Sievert, H, Venz, S, Platas-Barradas, O, Dhople, V M, Schaletzky, M, Nagel, C H, Braig, M, Preukschas, M, Pällmann, N, Bokemeyer, C, Brümmendorf, T H, Pörtner, R, Walther, R, Duncan, K E, Hauber, J, and Balabanov, S
- Abstract
Hypusine modification of eukaryotic initiation factor 5A (eIF-5A) represents a unique and highly specific post-translational modification with regulatory functions in cancer, diabetes, and infectious diseases. However, the specific cellular pathways that are influenced by the hypusine modification remain largely unknown. To globally characterize eIF-5A and hypusine-dependent pathways, we used an approach that combines large-scale bioreactor cell culture with tandem affinity purification and mass spectrometry: "bioreactor-TAP-MS/MS." By applying this approach systematically to all four components of the hypusine modification system (eIF-5A1, eIF-5A2, DHS, and DOHH), we identified 248 interacting proteins as components of the cellular hypusine network, with diverse functions including regulation of translation, mRNA processing, DNA replication, and cell cycle regulation. Network analysis of this data set enabled us to provide a comprehensive overview of the protein-protein interaction landscape of the hypusine modification system. In addition, we validated the interaction of eIF-5A with some of the newly identified associated proteins in more detail. Our analysis has revealed numerous novel interactions, and thus provides a valuable resource for understanding how this crucial homeostatic signaling pathway affects different cellular functions.
- Published
- 2012
7. Marine Alkaloid Monanchocidin: A Proteomic-Based Screening of Protein Targets in Cisplatin-Resistant Tumor Cells
- Author
-
Dyshlovoy, S.A., primary, Venz, S., additional, Guzii, A., additional, Makarieva, T., additional, Tabakmakher, K., additional, Stonik, V., additional, Balabanov, S., additional, Bokemeyer, C., additional, and Honecker, F., additional
- Published
- 2013
- Full Text
- View/download PDF
8. Poster session 3. Drug profiles - preclinical
- Author
-
Koronkiewicz, M., primary, Romiszewska, A., additional, Kazimierczuk, Z., additional, Chilmonczyk, Z., additional, Neto, M. D. S., additional, Ramos, S. P., additional, Curvello, R., additional, Bin, M., additional, Domingues, N. L. C., additional, Rinaldi, A. W., additional, de Souza, A. C. S., additional, Dyshlovoy, S. A., additional, Venz, S., additional, Guzii, A., additional, Makarieva, T., additional, Tabakmakher, K., additional, Stonik, V., additional, Balabanov, S., additional, Bokemeyer, C., additional, Honecker, F., additional, Flis, S., additional, Flis, K., additional, Statkiewicz, M., additional, Bin, M. E. L., additional, Shishido, S. M., additional, Dovat, S., additional, Song, C., additional, Gowda, C., additional, Petrovic-Dovat, L., additional, Payne, J., additional, Chen, L. T., additional, Tsai, H. J., additional, Kuo, S. H., additional, Cheng, A. L., additional, Chen, J., additional, Fu, L., additional, Kwong, D., additional, Guan, X., additional, Zalietok, S., additional, Samoylenko, O., additional, Zhuravel, O., additional, Gulua, L., additional, Orlovsky, O., additional, Chekhun, V., additional, Milinevska, V., additional, Karnaushenko, O., additional, Priya, S., additional, Reshma, R. S., additional, Rakesh, S. N., additional, Sreelatha, K. H., additional, Veena, S., additional, Nand, K., additional, Gupta, J. C., additional, Panda, A. K., additional, Jain, S. K., additional, Talwar, G. P., additional, Riva, P., additional, Oreal, P., additional, Lima, R. T., additional, Sousa, D., additional, Choosang, K., additional, Pakkong, P., additional, Palmeira, A., additional, Paiva, A. M., additional, Seca, H., additional, Cerqueira, F., additional, Pedro, M., additional, Pinto, M. M., additional, Sousa, E., additional, and Vasconcelos, M. H., additional
- Published
- 2013
- Full Text
- View/download PDF
9. Therapeutics
- Author
-
Azimi, A., primary, Kuznecovs, S., additional, Kuznecovs, J., additional, Blazejczyk, A., additional, Switalska, M., additional, Chlopicki, S., additional, Marcinek, A., additional, Gebicki, J., additional, Wietrzyk, J., additional, Egyhazi, S., additional, Azimi, A., additional, Ghasghgaei, S., additional, Frostvik Stolt, M., additional, Hertzman Johansson, C., additional, Hansson, J., additional, Delage, J. D., additional, Li, H., additional, Lu, H., additional, Cazin, L. H., additional, Vannier, J. P., additional, Drouet, L., additional, Dupuy, E., additional, Soria, J., additional, Varin, R., additional, Soria, C., additional, Castle, J., additional, Kreiter, S., additional, Diekmann, J., additional, Lower, M., additional, van der Roemer, N., additional, de Graaf, J., additional, Selmi, S., additional, Diken, M., additional, Boegel, S., additional, Paret, C., additional, Koslowski, M., additional, Kuhn, A. N., additional, Britten, C. M., additional, Huber, C., additional, Tureci, O., additional, Sahin, U., additional, Procopio, G., additional, Verzoni, E., additional, Testa, I., additional, de Braud, F., additional, Misale, S., additional, Yaeger, R., additional, Hobor, S., additional, Scala, E., additional, Janakiraman, M., additional, Liska, D., additional, Valtorta, E., additional, Schiavo, R., additional, Buscarino, M., additional, Siravergna, G., additional, Bencardino, K., additional, Cercek, A., additional, Chen, C., additional, Veronese, S., additional, Zanon, C., additional, Sartore-Bianchi, A., additional, Gambacorta, M., additional, Gallicchio, M., additional, Vakiani, E., additional, Boscaro, V., additional, Medico, E., additional, Weiser, M., additional, Siena, S., additional, di Nicolantonio, F., additional, Solit, D., additional, Bardelli, A., additional, Burbridge, M. F., additional, Dovat, S. P., additional, Song, C., additional, Payne, K. J., additional, Yang, L., additional, Cree, A., additional, Glaysher, M., additional, Bolton, L., additional, Johnson, P., additional, Atkey, N., additional, Torrance, C., additional, Bogush, T. A., additional, Dudko, E. A., additional, Shaturova, A. S., additional, Tikhomirov, M. V., additional, Bogush, E. A., additional, Polotsky, B. E., additional, Tjulandin, S. A., additional, Davydov, M. I., additional, Pernemalm, M., additional, Pawitan, Y., additional, Lazar, V., additional, Lundeberg, J., additional, Lehtio, J., additional, Rasul, A., additional, Ma, T., additional, Dyshlovoy, S. A., additional, Naeth, I., additional, Venz, S., additional, Fedorov, S. N., additional, Shubina, L. K., additional, Stonik, V. A., additional, Balabanov, S., additional, Honecker, F., additional, Kongpracha, P., additional, Tohtong, R., additional, Demidkina, V., additional, Kudryavtsev, V. A., additional, Kabakov, A. E., additional, Golan, T., additional, Atias, D., additional, Barshack, I., additional, Avivi, C., additional, Goldstein, R. S., additional, Berger, R., additional, Ben-Arieh, S., additional, Urban, D., additional, Maimon, N., additional, Leibowitz-Amit, R., additional, Keizman, D., additional, Biran, H., additional, Mishaeli, M., additional, Onn, A., additional, Gottfried, M., additional, Saraswati, S., additional, Agrawal, S. S., additional, Raval, P., additional, Patel, M., additional, Ganure, L., additional, Hanen, J. H., additional, Sonia, B. H. K., additional, Aya, M., additional, Zohra, H., additional, Touhami, M., additional, Cheng, X., additional, Shi, T. Y., additional, Yang, G., additional, Tu, X. Y., additional, Wu, X. H., additional, Wei, Q. Y., additional, Benboubker, H., additional, Zheng, B. Q., additional, Shi, Y. Q., additional, He, X. H., additional, Liang, L. H., additional, and Saied, G. M., additional
- Published
- 2012
- Full Text
- View/download PDF
10. P2.19 Aaptamine, Demethyloxyaaptamine, and Isoaaptamine: A Proteomic-Based Screening of Protein Targets in Cisplatin-Resistant Tumor Cells
- Author
-
Dyshlovoy, S.A., primary, Naeth, I., additional, Venz, S., additional, Fedorov, S.N., additional, Shubina, L.K., additional, Stonik, V.A., additional, Balabanov, S., additional, and Honecker, F., additional
- Published
- 2012
- Full Text
- View/download PDF
11. Removal of contrast media by different extracorporeal treatments.
- Author
-
Schindler, R, Stahl, C, Venz, S, Ludat, K, Krause, W, and Frei, U
- Abstract
Although the capability of extracorporeal treatments after administration of contrast media to prevent radiocontrast-induced nephropathy is controversial, haemodialysis is performed in many institutions after radiographic procedures. There are conflicting reports on the efficacy of different dialysers and treatment modalities to remove contrast media.
- Published
- 2001
- Full Text
- View/download PDF
12. DDI2 protease controls embryonic development and inflammation via TCF11/NRF1.
- Author
-
Nedomova M, Haberecht-Müller S, Möller S, Venz S, Prochazkova M, Prochazka J, Sedlak F, Chawengsaksophak K, Hammer E, Kasparek P, Adamek M, Sedlacek R, Konvalinka J, Krüger E, and Grantz Saskova K
- Abstract
DDI2 is an aspartic protease that cleaves polyubiquitinated substrates. Upon proteotoxic stress, DDI2 activates the transcription factor TCF11/NRF1 (NFE2L1), crucial for maintaining proteostasis in mammalian cells, enabling the expression of rescue factors, including proteasome subunits. Here, we describe the consequences of DDI2 ablation in vivo and in cells. DDI2 knock-out (KO) in mice caused embryonic lethality at E12.5 with severe developmental failure. Molecular characterization of embryos showed insufficient proteasome expression with proteotoxic stress, accumulation of high molecular weight ubiquitin conjugates and induction of the unfolded protein response (UPR) and cell death pathways. In DDI2 surrogate KO cells, proteotoxic stress activated the integrated stress response (ISR) and induced a type I interferon (IFN) signature and IFN-induced proliferative signaling, possibly ensuring survival. These results indicate an important role for DDI2 in the cell-tissue proteostasis network and in maintaining a balanced immune response., Competing Interests: The authors declare that they have no conflict of interest., (© 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
13. Anticancer Activity of the Marine Triterpene Glycoside Cucumarioside A 2 -2 in Human Prostate Cancer Cells.
- Author
-
Menchinskaya ES, Dyshlovoy SA, Venz S, Jacobsen C, Hauschild J, Rohlfing T, Silchenko AS, Avilov SA, Balabanov S, Bokemeyer C, Aminin DL, von Amsberg G, and Honecker F
- Subjects
- Male, Humans, Glycosides pharmacology, Prostate, Prostatic Neoplasms, Castration-Resistant, Triterpenes pharmacology
- Abstract
Despite recent advances in the treatment of metastatic castration-resistant prostate cancer (CRPC), treatment is inevitably hampered by the development of drug resistance. Thus, new drugs are urgently needed. We investigated the efficacy, toxicity, and mechanism of action of the marine triterpene glycoside cucumarioside A
2 -2 (CA2 -2) using an in vitro CRPC model. CA2 -2 induced a G2 /M-phase cell cycle arrest in human prostate cancer PC-3 cells and caspase-dependent apoptosis executed via an intrinsic pathway. Additionally, the drug inhibited the formation and growth of CRPC cell colonies at low micromolar concentrations. A global proteome analysis performed using the 2D-PAGE technique, followed by MALDI-MS and bioinformatical evaluation, revealed alterations in the proteins involved in cellular processes such as metastatic potential, invasion, and apoptosis. Among others, the regulation of keratin 81, CrkII, IL-1β, and cathepsin B could be identified by our proteomics approach. The effects were validated on the protein level by a 2D Western blotting analysis. Our results demonstrate the promising anticancer activity of CA2 -2 in a prostate cancer model and provide insights on the underlying mode of action.- Published
- 2023
- Full Text
- View/download PDF
14. Global Protein Profiling in Processed Immunohistochemistry Tissue Sections.
- Author
-
Venz S, von Bohlen Und Halbach V, Hentschker C, Junker H, Kuss AW, Sura T, Krüger E, Völker U, von Bohlen Und Halbach O, Jensen LR, and Hammer E
- Subjects
- Mice, Animals, Immunohistochemistry, Mice, Inbred C57BL, Proteins analysis, Tandem Mass Spectrometry, Paraffin Embedding, Tissue Fixation methods, Proteomics methods, Formaldehyde chemistry
- Abstract
Tissue sections, which are widely used in research and diagnostic laboratories and have already been examined by immunohistochemistry (IHC), may subsequently provide a resource for proteomic studies, even though only small amount of protein is available. Therefore, we established a workflow for tandem mass spectrometry-based protein profiling of IHC specimens and characterized defined brain area sections. We investigated the CA1 region of the hippocampus dissected from brain slices of adult C57BL/6J mice. The workflow contains detailed information on sample preparation from brain slices, including removal of antibodies and cover matrices, dissection of region(s) of interest, protein extraction and digestion, mass spectrometry measurement, and data analysis. The Gene Ontology (GO) knowledge base was used for further annotation. Literature searches and Gene Ontology annotation of the detected proteins verify the applicability of this method for global protein profiling using formalin-fixed and embedded material and previously used IHC slides.
- Published
- 2023
- Full Text
- View/download PDF
15. Immunoproteasomes control activation of innate immune signaling and microglial function.
- Author
-
Çetin G, Studencka-Turski M, Venz S, Schormann E, Junker H, Hammer E, Völker U, Ebstein F, and Krüger E
- Subjects
- Animals, Mice, Humans, Proteasome Endopeptidase Complex metabolism, Proteome metabolism, Proteomics, Phagocytosis, Protein Kinases metabolism, Interferons metabolism, Ubiquitins metabolism, Microglia, NF-kappa B metabolism
- Abstract
Microglia are the resident immune cells of the central nervous system (CNS) and play a major role in the regulation of brain homeostasis. To maintain their cellular protein homeostasis, microglia express standard proteasomes and immunoproteasomes (IP), a proteasome isoform that preserves protein homeostasis also in non-immune cells under challenging conditions. The impact of IP on microglia function in innate immunity of the CNS is however not well described. Here, we establish that IP impairment leads to proteotoxic stress and triggers the unfolded and integrated stress responses in mouse and human microglia models. Using proteomic analysis, we demonstrate that IP deficiency in microglia results in profound alterations of the ubiquitin-modified proteome among which proteins involved in the regulation of stress and immune responses. In line with this, molecular analysis revealed chronic activation of NF-κB signaling in IP-deficient microglia without further stimulus. In addition, we show that IP impairment alters microglial function based on markers for phagocytosis and motility. At the molecular level IP impairment activates interferon signaling promoted by the activation of the cytosolic stress response protein kinase R. The presented data highlight the importance of IP function for the proteostatic potential as well as for precision proteolysis to control stress and immune signaling in microglia function., 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 © 2022 Çetin, Studencka-Turski, Venz, Schormann, Junker, Hammer, Völker, Ebstein and Krüger.)
- Published
- 2022
- Full Text
- View/download PDF
16. Identification of the Regulatory Targets of miR-3687 and miR-4417 in Prostate Cancer Cells Using a Proteomics Approach.
- Author
-
Venz S, Junker H, Ultsch E, Hetke F, Krüger E, Burchardt M, Caetano-Pinto P, and Roennau C
- Subjects
- Androgens, Cell Line, Tumor, Gene Expression Regulation, Neoplastic, Humans, Male, Proteomics, Tandem Mass Spectrometry, MicroRNAs metabolism, Prostatic Neoplasms, Castration-Resistant pathology
- Abstract
MicroRNAs (miRNA) are ubiquitous non-coding RNAs that have a prominent role in cellular regulation. The expression of many miRNAs is often found deregulated in prostate cancer (PCa) and castration-resistant prostate cancer (CRPC). Although their expression can be associated with PCa and CRPC, their functions and regulatory activity in cancer development are poorly understood. In this study, we used different proteomics tools to analyze the activity of hsa-miR-3687-3p (miR-3687) and hsa-miR-4417-3p (miR-4417), two miRNAs upregulated in CRPC. PCa and CRPC cell lines were transfected with miR-3687 or miR-4417 to overexpress the miRNAs. Cell lysates were analyzed using 2D gel electrophoresis and proteins were subsequently identified using mass spectrometry (Maldi-MS/MS). A whole cell lysate, without 2D-gel separation, was analyzed by ESI-MS/MS. The expression of deregulated proteins found across both methods was further investigated using Western blotting. Gene ontology and cellular process network analysis determined that miR-3687 and miR-4417 are involved in diverse regulatory mechanisms that support the CRPC phenotype, including metabolism and inflammation. Moreover, both miRNAs are associated with extracellular vesicles, which point toward a secretory mechanism. The tumor protein D52 isoform 1 (TD52-IF1), which regulates neuroendocrine trans-differentiation, was found to be substantially deregulated in androgen-insensitive cells by both miR-3687 and miR-4417. These findings show that these miRNAs potentially support the CRPC by truncating the TD52-IF1 expression after the onset of androgen resistance.
- Published
- 2022
- Full Text
- View/download PDF
17. Deficiency in FTSJ1 Affects Neuronal Plasticity in the Hippocampal Formation of Mice.
- Author
-
von Bohlen Und Halbach V, Venz S, Nwakor S, Hentschker C, Hammer E, Junker H, Kuss AW, von Bohlen Und Halbach O, and Jensen LR
- Abstract
The role of the tRNA methyltransferase FTSJ1 in the brain is largely unknown. We analyzed whether FTSJ1-deficient mice (KO) displayed altered neuronal plasticity. We explored open field behavior (10 KO mice (aged 22-25 weeks)) and 11 age-matched control littermates (WT) and examined mean layer thickness (7 KO; 6 WT) and dendritic spines (5 KO; 5 WT) in the hippocampal area CA1 and the dentate gyrus. Furthermore, long-term potentiation (LTP) within area CA1 was investigated (5 KO; 5 WT), and mass spectrometry (MS) using CA1 tissue (2 each) was performed. Compared to controls, KO mice showed a significant reduction in the mean thickness of apical CA1 layers. Dendritic spine densities were also altered in KO mice. Stable LTP could be induced in the CA1 area of KO mice and remained stable at for at least 1 h, although at a lower level as compared to WTs, while MS data indicated differential abundance of several proteins, which play a role in neuronal plasticity. FTSJ1 has an impact on neuronal plasticity in the murine hippocampal area CA1 at the morphological and physiological levels, which, in conjunction with comparable changes in other cortical areas, might accumulate in disturbed learning and memory functions.
- Published
- 2022
- Full Text
- View/download PDF
18. Gelsolin Governs the Neuroendocrine Transdifferentiation of Prostate Cancer Cells and Suppresses the Apoptotic Machinery.
- Author
-
Oelrich F, Junker H, Stope MB, Erb HHH, Walther R, Venz S, and Zimmermann U
- Subjects
- Apoptosis drug effects, Cell Line, Tumor, Cell Transdifferentiation drug effects, Cell Transdifferentiation physiology, Gelsolin genetics, Gene Expression Regulation, Neoplastic, Humans, Interleukin-6 metabolism, Interleukin-6 pharmacology, Male, Neuroendocrine Cells pathology, Neuroendocrine Tumors genetics, Neuroendocrine Tumors mortality, Prostatic Neoplasms metabolism, Voltage-Dependent Anion Channel 1 metabolism, Gelsolin metabolism, Prostatic Neoplasms pathology
- Abstract
Background/aim: Interleukin 6 (IL6) is increased in patients with progressive prostate cancer and induces its transdifferentiation to neuroendocrine prostate cancer. Neuroendocrine prostate cancer has become one of the greatest challenges in treating castration-resistant disease and is linked to poor prognosis. It is necessary to understand better the cellular events associated with IL6-mediated neuroendocrine differentiation to prevent it and identify potential new therapeutic targets., Materials and Methods: In the present study, an IL6-inducible neuroendocrine differentiation model established specifically for this purpose was applied using LNCaP cells. Proteomics and western blot analyses were used to identify proteins involved in neuroendocrine differentiation. Subsequently, the role of gelsolin (GSN) in the neuroendocrine differentiation model was characterized (knock-down analyses, microscopic co-localization analyses, apoptosis assay) and GSN expression levels in patient material were investigated., Results: This study revealed that GSN is a crucial factor in the neuroendocrine differentiation process., Conclusion: It was shown that siRNA-mediated knock-down of GSN can inhibit neuroendocrine differentiation, making it a valid target for preventing IL6-mediated neuroendocrine differentiation., (Copyright © 2021 International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
19. Inspired by Sea Urchins: Warburg Effect Mediated Selectivity of Novel Synthetic Non-Glycoside 1,4-Naphthoquinone-6S-Glucose Conjugates in Prostate Cancer.
- Author
-
Dyshlovoy SA, Pelageev DN, Hauschild J, Sabutskii YE, Khmelevskaya EA, Krisp C, Kaune M, Venz S, Borisova KL, Busenbender T, Denisenko VA, Schlüter H, Bokemeyer C, Graefen M, Polonik SG, Anufriev VP, and Amsberg GV
- Subjects
- Animals, Antineoplastic Agents chemical synthesis, Antineoplastic Agents therapeutic use, Apoptosis drug effects, Cell Line, Tumor, Drug Screening Assays, Antitumor, Glucose chemical synthesis, Glucose pharmacology, Humans, Male, Membrane Potential, Mitochondrial drug effects, Mitochondrial Membranes drug effects, Naphthoquinones chemical synthesis, Naphthoquinones pharmacology, Naphthoquinones therapeutic use, Prostatic Neoplasms pathology, Antineoplastic Agents pharmacology, Pigments, Biological chemistry, Prostatic Neoplasms drug therapy, Sea Urchins chemistry, Warburg Effect, Oncologic drug effects
- Abstract
The phenomenon of high sugar consumption by tumor cells is known as Warburg effect. It results from a high glycolysis rate, used by tumors as preferred metabolic pathway even in aerobic conditions. Targeting the Warburg effect to specifically deliver sugar conjugated cytotoxic compounds into tumor cells is a promising approach to create new selective drugs. We designed, synthesized, and analyzed a library of novel 6-S-(1,4-naphthoquinone-2-yl)-d-glucose chimera molecules (SABs)-novel sugar conjugates of 1,4-naphthoquinone analogs of the sea urchin pigments spinochromes, which have previously shown anticancer properties. A sulfur linker (thioether bond) was used to prevent potential hydrolysis by human glycoside-unspecific enzymes. The synthesized compounds exhibited a Warburg effect mediated selectivity to human prostate cancer cells (including highly drug-resistant cell lines). Mitochondria were identified as a primary cellular target of SABs. The mechanism of action included mitochondria membrane permeabilization, followed by ROS upregulation and release of cytotoxic mitochondrial proteins (AIF and cytochrome C) to the cytoplasm, which led to the consequent caspase-9 and -3 activation, PARP cleavage, and apoptosis-like cell death. These results enable us to further clinically develop these compounds for effective Warburg effect targeting., Competing Interests: The authors declare no conflict of interest.
- Published
- 2020
- Full Text
- View/download PDF
20. Successful Targeting of the Warburg Effect in Prostate Cancer by Glucose-Conjugated 1,4-Naphthoquinones.
- Author
-
Dyshlovoy SA, Pelageev DN, Hauschild J, Borisova KL, Kaune M, Krisp C, Venz S, Sabutskii YE, Khmelevskaya EA, Busenbender T, Denisenko VA, Pokhilo ND, Atopkina LN, Graefen M, Schlüter H, Stonik VA, Bokemeyer C, Anufriev VP, and von Amsberg G
- Abstract
Treatment of castration-resistant prostate cancer (CRPC) remains challenging due to the development of drug resistance. The Warburg effect describes the ability of cancer cells to consume larger amounts of glucose compared to normal tissues. We identified derivatives of natural 1,4-naphthoquinones to be active in CRPC and further synthetically modified them via glucose conjugation to increase selectivity by Warburg effect targeting. Mechanisms of action were examined by quantitative proteomics followed by bioinformatical analysis and target validation. Four synthesized molecules revealed the highest selectivity towards human CRPC cells, which correlated with higher GLUT-1 activity and expression. The compounds were able to induce pro-apoptotic signs and to inhibit the pro-survival processes and mechanisms of drug resistance (i.e., AR-signaling and autophagy). Proteome analysis suggested a disruption of the mitochondria/oxidative phosphorylation, which was validated by further functional analysis: thus, mitochondria depolarization, elevated levels of cytotoxic ROS, an increase of Bax/Bcl-2 ratio as well as release of mitochondrial AIF and cytochrome C to cytoplasm were observed. In conclusion, glucose-conjugated 1,4-naphthoquinones show potent activity and selectivity in human CRPC exerted via mitochondrial targeting. The compounds can overcome drug resistance against current standard therapies and suppress pro-survival mechanisms. This unique combination of properties makes them new promising candidates for the treatment of CRPC.
- Published
- 2019
- Full Text
- View/download PDF
21. Nicotinamide N -Methyltransferase and Its Precursor Substrate Methionine Directly and Indirectly Control Malignant Metabolism During Progression of Renal Cell Carcinoma.
- Author
-
Holstein S, Venz S, Junker H, Walther R, Stope MB, and Zimmermann U
- Subjects
- Biomarkers, Tumor metabolism, Cell Line, Tumor, Cell Proliferation physiology, Disease Progression, HEK293 Cells, Humans, Interleukin-6 metabolism, Prognosis, Up-Regulation physiology, Carcinoma, Renal Cell metabolism, Kidney Neoplasms metabolism, Methionine metabolism, Nicotinamide N-Methyltransferase metabolism
- Abstract
Background/aim: Renal cell carcinoma (RCC) is one of the most common tumor diseases in adults, and new specific biomarkers are urgently needed to define diagnosis and prognosis of patients with RCC as well as monitor the outcome of therapeutic interventions. The enzyme nicotinamide N-methyltransferase (NNMT) is believed to represent such a marker molecule in RCC therapy., Materials and Methods: NNMT expression was examined by western blotting in samples from patients with RCC and in RCC cell lines. Effects of NNMT on cell growth and metabolism were assessed using the Hoechst 33342 reagent assay and Vita-Orange cell viability assay. Incubation experiments were performed to study the influence of methionine and interleukin-6 (IL6) on expression of NNMT., Results: In patient samples, NNMT was up-regulated depending on the stage of progression. Investigations in an RCC cell culture model showed that after modulation of NNMT expression, cellular metabolism, but not cell growth was affected. This regulatory function was also dependent on the presence of the NNMT precursor substrate methionine and IL6., Conclusion: The metabolism-regulatory activity of NNMT depends on the precursor substrate methionine and the presence of IL6. The function of methionine appears to be dependent on the stage of progression, since in individual RCC cell lines, opposing effects on metabolism were demonstrated. This, in turn, reflects the thoroughly complex situation in the clinic., (Copyright© 2019, International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
22. Inhibition of calpain-1 stabilizes TCF11/Nrf1 but does not affect its activation in response to proteasome inhibition.
- Author
-
Nowak K, Taubert RM, Haberecht S, Venz S, and Krüger E
- Subjects
- Endoplasmic Reticulum drug effects, Endoplasmic Reticulum genetics, Gene Expression Regulation drug effects, HEK293 Cells, Humans, Proteasome Endopeptidase Complex drug effects, Proteasome Inhibitors pharmacology, Protein Transport genetics, RNA, Small Interfering pharmacology, Ubiquitination genetics, Calpain genetics, NF-E2-Related Factor 1 genetics, Proteasome Endopeptidase Complex genetics
- Abstract
Protein degradation is essential to compensate for the damaging effects of proteotoxic stress. To ensure protein and redox homeostasis in response to proteasome inhibition, the cleavage and nuclear translocation of the endoplasmic reticulum (ER)-bound transcription factor TCF11/Nrf1 ( NFE2L1 ) is crucial for the activation of rescue factors including the synthesis of new proteasomal subunits. Even though TCF11/Nrf1 is an essential transcription factor, the exact mechanisms by which it is activated and stabilized are not fully understood. It was previously shown that the calcium-dependent protease calpain-1 interacts with TCF11/Nrf1 and the TCF11/Nrf1 cleavage site is a potential calpain target. Here, we tested the hypothesis that calpain-1 or -2 cleave TCF11/Nrf1. However, we did not find a role for calpain-1 or -2 in the activation of TCF11/Nrf1 after proteasome inhibition neither by using chemical inhibitors nor siRNA-mediated knockdown or overexpression of calpain subunits. Instead, we found that TCF11/Nrf1 is digested by calpain-1 in vitro and that calpain-1 inhibition slows down the degradation of membrane-bound TCF11/Nrf1 by the proteasome in cultured cells. Thus, we provide evidence that calpain-1 is involved in the degradation of TCF11/Nrf1. Furthermore, we confirmed DDI2 as an essential factor for TCF11/Nrf1 activation and demonstrate an undefined role of DDI2 and calpain-1 in TCF11/Nrf1 stability., (© 2018 The Author(s).)
- Published
- 2018
- Full Text
- View/download PDF
23. The Angiotensin-(1-7)/Mas Axis Improves Pancreatic β-Cell Function in Vitro and in Vivo.
- Author
-
Sahr A, Wolke C, Maczewsky J, Krippeit-Drews P, Tetzner A, Drews G, Venz S, Gürtler S, van den Brandt J, Berg S, Döring P, Dombrowski F, Walther T, and Lendeckel U
- Subjects
- Animals, Cyclic AMP metabolism, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Insulin genetics, Insulin Resistance physiology, Insulin Secretion, Insulin-Secreting Cells drug effects, Maf Transcription Factors, Large genetics, Maf Transcription Factors, Large metabolism, Mice, Mice, Knockout, Proto-Oncogene Mas, Proto-Oncogene Proteins genetics, Receptors, G-Protein-Coupled genetics, Trans-Activators genetics, Trans-Activators metabolism, Angiotensin I pharmacology, Insulin metabolism, Insulin-Secreting Cells metabolism, Peptide Fragments pharmacology, Proto-Oncogene Proteins metabolism, Receptors, G-Protein-Coupled metabolism, Signal Transduction drug effects
- Abstract
The angiotensin-converting enzyme 2/angiotensin (Ang)-(1-7)/Mas axis of the renin-angiotensin system often opposes the detrimental effects of the angiotensin-converting enzyme/Ang II/Ang II type 1 receptor axis and has been associated with beneficial effects on glucose homeostasis, whereas underlying mechanisms are mostly unknown. Here we investigate the effects of Ang-(1-7) and its receptor Mas on β-cell function. Isolated islets from Mas-deficient and wild-type mice were stimulated with Ang-(1-7) or its antagonists and effects on insulin secretion determined. Islets' cytoplasmic calcium and cAMP concentrations, mRNA amounts of Ins1, Ins2, Pdx1, and Mafa and effects of inhibitors of cAMP downstream signaling were determined. Ang-(1-7) was also applied to mice by osmotic pumps for 14 days and effects on glucose tolerance and insulin secretion were assessed. Ang-(1-7) increased insulin secretion from wild-type islets, whereas antagonists and genetic Mas deficiency led to reduced insulin secretion. The Mas-dependent effects of Ang-(1-7) on insulin secretion did not result from changes in insulin gene expression or changes in the excitation-secretion coupling but from increased intracellular cAMP involving exchange protein activated directly by cAMP. Administration of Ang-(1-7) in vivo had only marginal effects on glucose tolerance in wild-type mice but still resulted in improved insulin secretion from islets isolated of these mice. Interestingly, although less pronounced than in wild types, Ang-(1-7) still affected insulin secretion in Mas-deficient islets. The data indicate a significant function of Ang-(1-7) in the regulation of insulin secretion from mouse islets in vitro and in vivo, mainly, but not exclusively, by Mas-dependent signaling, modulating the accessory pathway of insulin secretion via increase in cAMP.
- Published
- 2016
- Full Text
- View/download PDF
24. Marine compound rhizochalinin shows high in vitro and in vivo efficacy in castration resistant prostate cancer.
- Author
-
Dyshlovoy SA, Otte K, Alsdorf WH, Hauschild J, Lange T, Venz S, Bauer CK, Bähring R, Amann K, Mandanchi R, Schumacher U, Schröder-Schwarz J, Makarieva TN, Guzii AG, Tabakmakher KM, Fedorov SN, Shubina LK, Kasheverov IE, Ehmke H, Steuber T, Stonik VA, Bokemeyer C, Honecker F, and von Amsberg G
- Subjects
- Animals, Apoptosis drug effects, Caspases physiology, Cell Line, Tumor, Docetaxel, Fatty Alcohols adverse effects, Fatty Alcohols pharmacology, Humans, Insulin-Like Growth Factor I analysis, Male, Mice, Potassium Channel Blockers pharmacology, Prostate-Specific Antigen analysis, Taxoids pharmacology, Fatty Alcohols therapeutic use, Prostatic Neoplasms, Castration-Resistant drug therapy
- Abstract
Development of drug resistance is an inevitable phenomenon in castration-resistant prostate cancer (CRPC) cells requiring novel therapeutic approaches. In this study, efficacy and toxicity of Rhizochalinin (Rhiz) - a novel sphingolipid-like marine compound - was evaluated in prostate cancer models, resistant to currently approved standard therapies. In vitro activity and mechanism of action of Rhiz were examined in the human prostate cancer cell lines PC-3, DU145, LNCaP, 22Rv1, and VCaP. Rhiz significantly reduced cell viability at low micromolar concentrations showing most pronounced effects in enzalutamide and abiraterone resistant AR-V7 positive cells. Caspase-dependent apoptosis, inhibition of pro-survival autophagy, downregulation of AR-V7, PSA and IGF-1 expression as well as inhibition of voltage-gated potassium channels were identified as mechanisms of action. Remarkably, Rhiz re-sensitized AR-V7 positive cells to enzalutamide and increased efficacy of taxanes.In vivo activity and toxicity were evaluated in PC-3 and 22Rv1 NOD SCID mouse xenograft models using i.p. administration. Rhiz significantly reduced growth of PC-3 and 22Rv1 tumor xenografts by 27.0% (p = 0.0156) and 46.8% (p = 0.047) compared with controls with an increased fraction of tumor cells showing apoptosis secondary to Rhiz exposure. In line with the in vitro data, Rhiz was most active in AR-V7 positive xenografts in vivo. In animals, no severe side effects were observed.In conclusion, Rhiz is a promising novel marine-derived compound characterized by a unique combination of anticancer properties. Its further clinical development is of high impact for patients suffering from drug resistant prostate cancer especially those harboring AR-V7 mediated resistance to enzalutamide and abiraterone.
- Published
- 2016
- Full Text
- View/download PDF
25. Cabazitaxel overcomes cisplatin resistance in germ cell tumour cells.
- Author
-
Gerwing M, Jacobsen C, Dyshlovoy S, Hauschild J, Rohlfing T, Oing C, Venz S, Oldenburg J, Oechsle K, Bokemeyer C, von Amsberg G, and Honecker F
- Subjects
- Apoptosis drug effects, Cell Cycle drug effects, Cell Line, Tumor, Humans, Male, Neoplasms, Germ Cell and Embryonal pathology, Paclitaxel pharmacology, Testicular Neoplasms pathology, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Cisplatin therapeutic use, Drug Resistance, Neoplasm drug effects, Neoplasms, Germ Cell and Embryonal drug therapy, Taxoids pharmacology, Testicular Neoplasms drug therapy
- Abstract
Background: Cisplatin-based chemotherapy is highly effective in metastasized germ cell tumours (GCT). However, 10-30 % of patients develop resistance to cisplatin, requiring salvage therapy. We investigated the in vitro activity of paclitaxel and the novel taxane cabazitaxel in cisplatin-sensitive and -resistant GCT cell lines., Methods: In vitro activity of paclitaxel and cabazitaxel was determined by proliferation assays, and mode of action of cabazitaxel was assessed by western blotting and two screening approaches, i.e. whole proteome analysis and a human apoptosis array., Results: Activity of paclitaxel and cabazitaxel was not affected by cisplatin resistance, suggesting that there is no cross-resistance between these agents in vitro. Cabazitaxel treatment showed a strong inhibitory effect on colony formation capacity. Cabazitaxel induced classical apoptosis in all cell lines, reflected by cleavage of PARP and caspase 3, without inducing specific changes in the cell cycle distribution. Using the proteomic and human apoptosis array screening approaches, differential regulation of several proteins, including members of the bcl-2 family, was found, giving first insights into the mode of action of cabazitaxel in GCT., Conclusion: Cabazitaxel shows promising in vitro activity in GCT cells, independent of levels of cisplatin resistance.
- Published
- 2016
- Full Text
- View/download PDF
26. Marine alkaloid Monanchocidin a overcomes drug resistance by induction of autophagy and lysosomal membrane permeabilization.
- Author
-
Dyshlovoy SA, Hauschild J, Amann K, Tabakmakher KM, Venz S, Walther R, Guzii AG, Makarieva TN, Shubina LK, Fedorov SN, Stonik VA, Bokemeyer C, Balabanov S, Honecker F, and von Amsberg G
- Subjects
- Apoptosis drug effects, Blotting, Western, Cell Cycle drug effects, Cell Line, Tumor, Cell Membrane Permeability drug effects, Flow Cytometry, Guanidine pharmacology, Humans, Intracellular Membranes drug effects, Mass Spectrometry, Microscopy, Electron, Transmission, Antineoplastic Agents pharmacology, Autophagy drug effects, Drug Resistance, Neoplasm drug effects, Guanidine analogs & derivatives, Lysosomes drug effects, Urogenital Neoplasms pathology
- Abstract
Monanchocidin A (MonA) is a novel alkaloid recently isolated from the marine sponge Monanchora pulchra. The compound reveals cytotoxic activity in genitourinary cancers including cisplatin-sensitive and -resistant germ cell tumor (GCT) cell lines, hormone-sensitive and castration-resistant prostate carcinoma cell lines and different bladder carcinoma cell lines. In contrast, non-malignant cells were significantly less sensitive. MonA is highly synergistic with cisplatin in GCT cells. Induction of autophagy at lower and lysosomal membrane permeabilization (LMP) at higher concentrations were identified as the dominating modes of action. Cytotoxicity and protein degradation could be inhibited by 3-methyladenine, an inhibitor of autophagy. LMP was confirmed by loss of acridine orange staining of lysosoms and by release of cathepsin B. In conclusion, MonA exerts cytotoxic activity by mechanisms different from "classical" apoptosis, and could be a promising new compound to overcome resistance to standard therapies in genitourinary malignancies.
- Published
- 2015
- Full Text
- View/download PDF
27. A novel mouse model for inhibition of DOHH-mediated hypusine modification reveals a crucial function in embryonic development, proliferation and oncogenic transformation.
- Author
-
Sievert H, Pällmann N, Miller KK, Hermans-Borgmeyer I, Venz S, Sendoel A, Preukschas M, Schweizer M, Boettcher S, Janiesch PC, Streichert T, Walther R, Hengartner MO, Manz MG, Brümmendorf TH, Bokemeyer C, Braig M, Hauber J, Duncan KE, and Balabanov S
- Subjects
- 3T3 Cells, Alleles, Animals, Caenorhabditis elegans, Cell Proliferation, Cellular Senescence, Disease Models, Animal, Embryo Loss metabolism, Embryo Loss pathology, Fibroblasts metabolism, Fibroblasts pathology, Gene Knockout Techniques, Hydroxylation, Lysine metabolism, Mice, Mixed Function Oxygenases metabolism, Oxidoreductases Acting on CH-NH Group Donors metabolism, Peptide Initiation Factors metabolism, Phenotype, Protein Biosynthesis, Proto-Oncogene Proteins c-myc metabolism, RNA-Binding Proteins metabolism, ras Proteins metabolism, Eukaryotic Translation Initiation Factor 5A, Cell Transformation, Neoplastic metabolism, Cell Transformation, Neoplastic pathology, Embryonic Development, Lysine analogs & derivatives, Mixed Function Oxygenases antagonists & inhibitors
- Abstract
The central importance of translational control by post-translational modification has spurred major interest in regulatory pathways that control translation. One such pathway uniquely adds hypusine to eukaryotic initiation factor 5A (eIF5A), and thereby affects protein synthesis and, subsequently, cellular proliferation through an unknown mechanism. Using a novel conditional knockout mouse model and a Caenorhabditis elegans knockout model, we found an evolutionarily conserved role for the DOHH-mediated second step of hypusine synthesis in early embryonic development. At the cellular level, we observed reduced proliferation and induction of senescence in 3T3 Dohh-/- cells as well as reduced capability for malignant transformation. Furthermore, mass spectrometry showed that deletion of DOHH results in an unexpected complete loss of hypusine modification. Our results provide new biological insight into the physiological roles of the second step of the hypusination of eIF5A. Moreover, the conditional mouse model presented here provides a powerful tool for manipulating hypusine modification in a temporal and spatial manner, to analyse both how this unique modification normally functions in vivo as well as how it contributes to different pathological conditions., (© 2014. Published by The Company of Biologists Ltd.)
- Published
- 2014
- Full Text
- View/download PDF
28. Identification of a dithiol-disulfide switch in collapsin response mediator protein 2 (CRMP2) that is toggled in a model of neuronal differentiation.
- Author
-
Gellert M, Venz S, Mitlöhner J, Cott C, Hanschmann EM, and Lillig CH
- Subjects
- Amino Acid Sequence, Cell Differentiation, Cell Line, Cysteine chemistry, Glutaredoxins genetics, Glutaredoxins metabolism, Humans, Intercellular Signaling Peptides and Proteins genetics, Molecular Sequence Data, Nerve Tissue Proteins genetics, Neurogenesis, Oxidation-Reduction, Protein Conformation, Protein Interaction Domains and Motifs, Protein Structure, Quaternary, Protein Subunits, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Sequence Homology, Amino Acid, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Intercellular Signaling Peptides and Proteins chemistry, Intercellular Signaling Peptides and Proteins metabolism, Models, Neurological, Nerve Tissue Proteins chemistry, Nerve Tissue Proteins metabolism, Neurons cytology, Neurons metabolism
- Abstract
Vertebrate-specific glutaredoxin 2 (Grx2) is expressed in at least two isoforms, mitochondrial Grx2a and cytosolic Grx2c. We have previously shown that cytosolic Grx2 is essential for embryonic development of the brain. In particular, we identified collapsin response mediator protein 2 (CRMP2/DPYSL2), a mediator of the semaphorin-plexin signaling pathway, as redox-regulated target of Grx2c and demonstrated that this regulation is required for normal axonal outgrowth. In this study, we demonstrate the molecular mechanism of this regulation, a specific and reversible intermolecular Cys-504-Cys-504 dithiol-disulfide switch in homotetrameric CRMP2. This switch determines two conformations of the quaternary CRMP2 complex that controls axonal outgrowth and thus neuronal development.
- Published
- 2013
- Full Text
- View/download PDF
29. Combination of a proteomics approach and reengineering of meso scale network models for prediction of mode-of-action for tyrosine kinase inhibitors.
- Author
-
Balabanov S, Wilhelm T, Venz S, Keller G, Scharf C, Pospisil H, Braig M, Barett C, Bokemeyer C, Walther R, Brümmendorf TH, and Schuppert A
- Subjects
- Animals, Apoptosis drug effects, Benzamides pharmacology, Benzamides therapeutic use, Blotting, Western, Cell Line, Tumor, Cluster Analysis, Drug Resistance, Neoplasm drug effects, Humans, Imatinib Mesylate, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Leukemia, Myelogenous, Chronic, BCR-ABL Positive pathology, Mice, Neoplasm Proteins metabolism, Piperazines pharmacology, Piperazines therapeutic use, Protein Kinase Inhibitors therapeutic use, Protein-Tyrosine Kinases metabolism, Pyrimidines pharmacology, Pyrimidines therapeutic use, Models, Biological, Protein Kinase Inhibitors pharmacology, Protein-Tyrosine Kinases antagonists & inhibitors, Proteomics methods, Signal Transduction drug effects
- Abstract
In drug discovery, the characterisation of the precise modes of action (MoA) and of unwanted off-target effects of novel molecularly targeted compounds is of highest relevance. Recent approaches for identification of MoA have employed various techniques for modeling of well defined signaling pathways including structural information, changes in phenotypic behavior of cells and gene expression patterns after drug treatment. However, efficient approaches focusing on proteome wide data for the identification of MoA including interference with mutations are underrepresented. As mutations are key drivers of drug resistance in molecularly targeted tumor therapies, efficient analysis and modeling of downstream effects of mutations on drug MoA is a key to efficient development of improved targeted anti-cancer drugs. Here we present a combination of a global proteome analysis, reengineering of network models and integration of apoptosis data used to infer the mode-of-action of various tyrosine kinase inhibitors (TKIs) in chronic myeloid leukemia (CML) cell lines expressing wild type as well as TKI resistance conferring mutants of BCR-ABL. The inferred network models provide a tool to predict the main MoA of drugs as well as to grouping of drugs with known similar kinase inhibitory activity patterns in comparison to drugs with an additional MoA. We believe that our direct network reconstruction approach, demonstrated on proteomics data, can provide a complementary method to the established network reconstruction approaches for the preclinical modeling of the MoA of various types of targeted drugs in cancer treatment. Hence it may contribute to the more precise prediction of clinically relevant on- and off-target effects of TKIs.
- Published
- 2013
- Full Text
- View/download PDF
30. Protein-protein-interaction network organization of the hypusine modification system.
- Author
-
Sievert H, Venz S, Platas-Barradas O, Dhople VM, Schaletzky M, Nagel CH, Braig M, Preukschas M, Pällmann N, Bokemeyer C, Brümmendorf TH, Pörtner R, Walther R, Duncan KE, Hauber J, and Balabanov S
- Subjects
- Animals, Computational Biology, DNA-Binding Proteins metabolism, Humans, Lysine metabolism, Mass Spectrometry, Mice, Mixed Function Oxygenases metabolism, Multivesicular Bodies metabolism, NIH 3T3 Cells, Nuclear Proteins metabolism, Nucleophosmin, Oxidoreductases Acting on CH-NH Group Donors metabolism, Peptide Fragments metabolism, Peptide Initiation Factors metabolism, Protein Transport, RNA-Binding Proteins metabolism, Recombinant Fusion Proteins metabolism, Reproducibility of Results, Ribosomal Proteins metabolism, Subcellular Fractions metabolism, Eukaryotic Translation Initiation Factor 5A, Lysine analogs & derivatives, Protein Interaction Maps, Protein Processing, Post-Translational
- Abstract
Hypusine modification of eukaryotic initiation factor 5A (eIF-5A) represents a unique and highly specific post-translational modification with regulatory functions in cancer, diabetes, and infectious diseases. However, the specific cellular pathways that are influenced by the hypusine modification remain largely unknown. To globally characterize eIF-5A and hypusine-dependent pathways, we used an approach that combines large-scale bioreactor cell culture with tandem affinity purification and mass spectrometry: "bioreactor-TAP-MS/MS." By applying this approach systematically to all four components of the hypusine modification system (eIF-5A1, eIF-5A2, DHS, and DOHH), we identified 248 interacting proteins as components of the cellular hypusine network, with diverse functions including regulation of translation, mRNA processing, DNA replication, and cell cycle regulation. Network analysis of this data set enabled us to provide a comprehensive overview of the protein-protein interaction landscape of the hypusine modification system. In addition, we validated the interaction of eIF-5A with some of the newly identified associated proteins in more detail. Our analysis has revealed numerous novel interactions, and thus provides a valuable resource for understanding how this crucial homeostatic signaling pathway affects different cellular functions.
- Published
- 2012
- Full Text
- View/download PDF
31. Identification of clinically relevant protein targets in prostate cancer with 2D-DIGE coupled mass spectrometry and systems biology network platform.
- Author
-
Ummanni R, Mundt F, Pospisil H, Venz S, Scharf C, Barett C, Fälth M, Köllermann J, Walther R, Schlomm T, Sauter G, Bokemeyer C, Sültmann H, Schuppert A, Brümmendorf TH, and Balabanov S
- Subjects
- Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Cluster Analysis, Humans, Male, Neoplasm Proteins genetics, Principal Component Analysis, Prostate cytology, Prostate metabolism, Prostate pathology, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology, Real-Time Polymerase Chain Reaction, Reproducibility of Results, Transcriptome, Neoplasm Proteins metabolism, Prostatic Neoplasms metabolism, Proteomics methods, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Systems Biology methods, Tandem Mass Spectrometry, Two-Dimensional Difference Gel Electrophoresis
- Abstract
Prostate cancer (PCa) is the most common type of cancer found in men and among the leading causes of cancer death in the western world. In the present study, we compared the individual protein expression patterns from histologically characterized PCa and the surrounding benign tissue obtained by manual micro dissection using highly sensitive two-dimensional differential gel electrophoresis (2D-DIGE) coupled with mass spectrometry. Proteomic data revealed 118 protein spots to be differentially expressed in cancer (n = 24) compared to benign (n = 21) prostate tissue. These spots were analysed by MALDI-TOF-MS/MS and 79 different proteins were identified. Using principal component analysis we could clearly separate tumor and normal tissue and two distinct tumor groups based on the protein expression pattern. By using a systems biology approach, we could map many of these proteins both into major pathways involved in PCa progression as well as into a group of potential diagnostic and/or prognostic markers. Due to complexity of the highly interconnected shortest pathway network, the functional sub networks revealed some of the potential candidate biomarker proteins for further validation. By using a systems biology approach, our study revealed novel proteins and molecular networks with altered expression in PCa. Further functional validation of individual proteins is ongoing and might provide new insights in PCa progression potentially leading to the design of novel diagnostic and therapeutic strategies.
- Published
- 2011
- Full Text
- View/download PDF
32. Stage-related alterations in renal cell carcinoma--comprehensive quantitative analysis by 2D-DIGE and protein network analysis.
- Author
-
Junker H, Venz S, Zimmermann U, Thiele A, Scharf C, and Walther R
- Subjects
- Adult, Aged, Aged, 80 and over, Blotting, Western, Cluster Analysis, Female, Humans, Immunohistochemistry, Kidney Neoplasms metabolism, Male, Mass Spectrometry, Middle Aged, Neoplasm Proteins chemistry, Neoplasm Proteins classification, Neoplasm Staging, Principal Component Analysis, Proteome metabolism, Reproducibility of Results, Carcinoma, Renal Cell metabolism, Carcinoma, Renal Cell pathology, Kidney Neoplasms pathology, Neoplasm Proteins metabolism, Proteomics, Signal Transduction, Two-Dimensional Difference Gel Electrophoresis methods
- Abstract
Renal cell carcinoma accounts for about 3% of adult malignancies and 85% of neoplasms arising from the kidney. To identify potential progression markers for kidney cancer we examined non-neoplastic and neoplastic kidney tissue from three groups of patients, which represent different tumor stages (pT1, pT2, pT3) by a fluorescence two-dimensional difference gel electrophoresis (2D-DIGE) approach combined with MALDI-ToF-MS/MS. Delta2D software package was used for gel image based quantification and statistical analysis. Thereby, a comprehensive Principal Component Analysis (PCA) could be performed and allowed a robust quality control of the experiment as well as a classification of the analyzed samples, which correlated with the predicted stages from the pathological examination. Additionally for selected candidate proteins we detected a correlation to the tumor grading as revealed by immunohistochemistry. On the 2D protein map 176 spots out of 989 were detected as at least 2-fold differentially expressed. These spots were analyzed by MALDI-ToF-MS/MS and 187 different proteins were identified. The functional clustering of the identified proteins revealed ten groups. Within these groups we found 86 enzymes, 63 proteins of unknown function, 14 transporter, 8 peptidases and 7 kinases. From the systems biology approach we could map many of these proteins in major pathways involved in remodelling of cytoskeleton, mitochondrial dysfunctions and changes in lipid metabolism. Due to complexity of the highly interconnected pathway network, further expression and functional validation of these proteins might provide new insights in kidney cancer progression to design novel diagnostic and therapeutic strategies.
- Published
- 2011
- Full Text
- View/download PDF
33. Altered expression of tumor protein D52 regulates apoptosis and migration of prostate cancer cells.
- Author
-
Ummanni R, Teller S, Junker H, Zimmermann U, Venz S, Scharf C, Giebel J, and Walther R
- Subjects
- Cell Line, Tumor, Cell Proliferation, Humans, Integrin alphaVbeta3, Male, Mitochondria physiology, Neoplasm Proteins physiology, Prostatic Neoplasms genetics, Proto-Oncogene Proteins c-akt, Signal Transduction, Apoptosis genetics, Cell Movement genetics, Gene Expression Regulation physiology, Neoplasm Proteins genetics, Prostatic Neoplasms pathology
- Abstract
Tumor protein D52 (TPD52) is a protein found to be overexpressed in prostate and breast cancer due to gene amplification. However, its physiological function remains under investigation. In the present study, we investigated the response of the LNCaP human prostate carcinoma cell line to deregulation of TPD52 expression. Proteomic analysis of prostate biopsies showed TPD52 overexpression at the protein level, whereas its transcriptional upregulation was demonstrated by real-time PCR. Transfection of LNCaP cells with a specific small hairpin RNA giving efficient knockdown of TPD52 resulted in significant cell death of the carcinoma LNCaP cells. As demonstrated by activation of caspases (caspase-3 and -9), and by the loss of mitochondrial membrane potential, cell death occurs due to apoptosis. The disruption of the mitochondrial membrane potential indicates that TPD52 acts upstream of the mitochondrial apoptotic reaction. To study the effect of TPD52 expression on cell proliferation, LNCaP cells were either transfected with enhanced green fluorescence protein-TPD52 or a specific small hairpin RNA. Enhanced green fluorescence protein-TPD52 overexpressing cells showed an increased proliferation rate, whereas TPD52-depleted cells showed the reverse effect. Additionally, we demonstrate that exogenous expression of TPD52 promotes cell migration via alphav beta3 integrin in prostate cancer cells through activation of the protein kinase B/Akt signaling pathway. From these results, we conclude that TPD52 plays an important role in various molecular events, particularly in the morphological diversification and dissemination of prostate carcinoma cells, and may be a promising target with respect to developing new therapeutic strategies to treat prostate cancer.
- Published
- 2008
- Full Text
- View/download PDF
34. Loss of dopamine-D2 receptor binding sites in Parkinsonian plus syndromes.
- Author
-
Hierholzer J, Cordes M, Venz S, Schelosky L, Harisch C, Richter W, Keske U, Hosten N, Mäurer J, Poewe W, and Felix R
- Subjects
- Adult, Aged, Benzamides, Binding Sites, Contrast Media, Corpus Striatum diagnostic imaging, Female, Humans, Iodine Radioisotopes, Male, Middle Aged, Multiple System Atrophy metabolism, Observer Variation, Olivopontocerebellar Atrophies metabolism, Parkinson Disease diagnostic imaging, Pyrrolidines, Reproducibility of Results, Supranuclear Palsy, Progressive metabolism, Syndrome, Time Factors, Tomography, Emission-Computed, Single-Photon, Corpus Striatum metabolism, Parkinson Disease metabolism, Receptors, Dopamine D2 metabolism
- Abstract
Unlabelled: This study analyzed temporal changes of striatal dopamine-D2 receptor binding during the course of different extrapyramidal movement disorders using 123I-iodobenzamide (IBZM) SPECT., Methods: Eighteen patients (9 with Parkinson's disease, 9 with parkinsonian plus syndrome) were followed for 11-53 mo. Dopamine-D2 receptor binding was assessed using 123I-IBZM SPECT at the beginning and at the end of the follow-up period. SPECT data were acquired 120 min postinjection of 3-5 mCi 123I-IBZM. A semiautomated algorithm was applied to the raw data for semiquantitative evaluation of regional cerebral receptor binding., Results: Intraobserver (r = 0.992) and interobserver (r = 0.930) variance was low for the semiautomated interpretation of the SPECT examination of the dopaminergic D2 receptor binding, reflecting a highly reproducible SPECT algorithm. Mean specific dopamine-D2 receptor binding was lower in patients with parkinsonian plus syndrome compared to patients with Parkinson's disease on the initial (p < 0.001) as well as the follow-up study (p < 0.001). In patients with Parkinson's disease, we observed an unaffected receptor binding compared to a reduced binding of radiotracer in patients with parkinsonian plus syndrome during the course of the disease (p < 0.001)., Conclusion: During the follow-up, patients with Parkinson's disease showed a constant dopamine-D2 receptor binding. In contrast, patients with parkinsonian plus syndrome revealed a decline of the binding of dopamine-D2 receptor. These findings are in agreement with histopathological data that demonstrated a preserved dopamine-D2 receptor status in patients with Parkinson's disease and a decline of the dopamine-D2 receptors in patients with parkinsonian plus syndrome. SPECT examinations using 123I-IBZM are useful for assessing dynamic changes of dopamine-D2 receptors in extrapyramidal movement disorders. Semiquantitative SPECT evaluations may provide valuable information for clinical management and prognosis of the patient with extrapyramidal movement disorders.
- Published
- 1998
35. Dopamine D2 receptor imaging with iodine-123-iodobenzamide SPECT in idiopathic rotational torticollis.
- Author
-
Hierholzer J, Cordes M, Schelosky L, Richter W, Keske U, Venz S, Semmler W, Poewe W, and Felix R
- Subjects
- Adult, Brain pathology, Cerebellum diagnostic imaging, Corpus Striatum diagnostic imaging, Female, Functional Laterality, Humans, Magnetic Resonance Imaging, Male, Middle Aged, Receptors, Dopamine D2 metabolism, Torticollis diagnostic imaging, Torticollis pathology, Benzamides metabolism, Cerebellum metabolism, Corpus Striatum metabolism, Iodine Radioisotopes, Pyrrolidines metabolism, Receptors, Dopamine D2 analysis, Tomography, Emission-Computed, Single-Photon, Torticollis metabolism
- Abstract
Unlabelled: The cause of idiopathic rotational torticollis (IRT) is not completely understood to date. However, basal ganglia are believed to be involved in the pathophysiology of IRT. To elucidate this disorder further, the value of iodobenzamide (IBZM) SPECT was studied for the evaluation of striatal dopamine D2 receptors in these patients., Methods: Striatal dopamine D2 receptor density was assessed in 10 patients with IRT using 123I-IBZM SPECT. The images were interpreted by a nuclear medicine physician initially to determine IBZM binding within the striatum and the cerebellum and, secondly, interstriatal IBZM binding. The results were correlated with the clinical parameters of the patients and compared with the results obtained from normal controls., Results: No difference was found in average, specific striatal IBZM binding (basal ganglia/cerebellum ratio) between patients and controls. However, interstriatal analysis of IBZM binding revealed a significantly higher binding in the striatum contralateral to the direction of the torticollis (p = 0.026, by chi-square test)., Conclusion: It was concluded that the striatal dopamine D2 receptor status is altered in patients with IRT.
- Published
- 1994
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.