48 results on '"Müller-Brüsselbach S"'
Search Results
2. Compensatory PI3-kinase/Akt/mTor activation regulates imatinib resistance development
- Author
-
Burchert, A, Wang, Y, Cai, D, von Bubnoff, N, Paschka, P, Müller-Brüsselbach, S, Ottmann, O G, Duyster, J, Hochhaus, A, and Neubauer, A
- Published
- 2005
- Full Text
- View/download PDF
3. Inverse PPARβ/δ agonists suppress oncogenic signaling to the ANGPTL4 gene and inhibit cancer cell invasion
- Author
-
Adhikary, T, Brandt, D T, Kaddatz, K, Stockert, J, Naruhn, S, Meissner, W, Finkernagel, F, Obert, J, Lieber, S, Scharfe, M, Jarek, M, Toth, P M, Scheer, F, Diederich, W E, Reinartz, S, Grosse, R, Müller-Brüsselbach, S, and Müller, R
- Published
- 2013
- Full Text
- View/download PDF
4. Diversität von Tumorzellsubpopulationen im Aszites von Ovarialkarzinompatienten und ihr Einfluss auf Chemoresistenz
- Author
-
Jansen, JM, primary, Broske, C, additional, Plaum, T, additional, Allmeroth, A, additional, Wagner, U, additional, Müller-Brüsselbach, S, additional, Müller, R, additional, and Reinartz, S, additional
- Published
- 2016
- Full Text
- View/download PDF
5. Etablierung eines humanen Ex-vivo-Primärtumormodells zur Untersuchung von Chemoresistenz-Mechanismen beim Ovarialkarzinom
- Author
-
Jansen, JM, primary, Bartholomäus, S, additional, Plaum, T, additional, Allmeroth, A, additional, Wagner, U, additional, Müller-Brüsselbach, S, additional, Müller, R, additional, and Reinartz, S, additional
- Published
- 2016
- Full Text
- View/download PDF
6. Der Zusammenhang zwischem dem Polarisationsphänotyp von Tumor-assoziierten Makrophagen (TAM), Zytokinspiegeln und dem progressionsfreien Überleben bei Ovarialkarzinompatientinnen
- Author
-
Jansen, JM, primary, Reinartz, S, additional, Müller, R, additional, Müller-Brüsselbach, S, additional, Schumann, T, additional, Finkernagel, F, additional, Wortmann, A, additional, Meissner, W, additional, Krause, M, additional, and Wagner, U, additional
- Published
- 2014
- Full Text
- View/download PDF
7. Inverse PPARβ/δ agonists suppress oncogenic signaling to the ANGPTL4 gene and inhibit cancer cell invasion
- Author
-
Adhikary, T, primary, Brandt, D T, additional, Kaddatz, K, additional, Stockert, J, additional, Naruhn, S, additional, Meissner, W, additional, Finkernagel, F, additional, Obert, J, additional, Lieber, S, additional, Scharfe, M, additional, Jarek, M, additional, Toth, P M, additional, Scheer, F, additional, Diederich, W E, additional, Reinartz, S, additional, Grosse, R, additional, Müller-Brüsselbach, S, additional, and Müller, R, additional
- Published
- 2012
- Full Text
- View/download PDF
8. Nerve Growth Factor overexpression in Clara cells suppresses metastasis and tumor growth in a mouse model of experimental lung metastasis
- Author
-
Seidler, K, primary, Sydykov, A, additional, Müller-Brüsselbach, S, additional, Müller, R, additional, Weißmann, N, additional, Renz, H, additional, and Nockher, A, additional
- Published
- 2012
- Full Text
- View/download PDF
9. Poster Abstracts
- Author
-
Littger, Ralf, primary, Alke, Alexandra, additional, Tewes, Bernhard, additional, Gropp, Felix, additional, Asai, T., additional, Watanabe, K., additional, Kuromi, K., additional, Kurohane, K., additional, Ogino, K., additional, Taki, T., additional, Tsukada, H., additional, Nakayama, J., additional, Oku, N., additional, Babai, I., additional, Matyas, G., additional, Baranji, L., additional, Milosevits, J., additional, Alving, C. R., additional, Bendas, G., additional, Rothe, U., additional, Scherphof, G. L., additional, Kamps, J. A. A. M., additional, Kessner, S., additional, Carafa, M., additional, Di Stefano, A., additional, Sozio, P., additional, Cacciatore, I., additional, Mosciatti, B., additional, Santucci, E., additional, Choice, E., additional, Harvie, P., additional, Galbraith, T., additional, Zunder, E., additional, Dutzar, B., additional, Anklesaria, P., additional, Paul, R., additional, Cocquyt, J., additional, De Cuyper, M., additional, Van der Meeren, P., additional, Cruz, M. E. M., additional, Gaspar, M. M., additional, Silva, M. T., additional, Dathe, M., additional, Nikolenko, H., additional, Wessolowski, A., additional, Schmieder, P., additional, Beyermann, M., additional, Bienert, M., additional, Santos, N. Dos, additional, Cox, K. A., additional, Allen, C., additional, Gallagher, R. C., additional, Ickenstein, L., additional, Mayer, L. D., additional, Bally, M. B., additional, Fischer, S., additional, Margalit, R., additional, Freisleben, H.-J., additional, Garidel, P., additional, Chen, H. C., additional, Moore, D., additional, Mendelsohn, R., additional, Keller, M., additional, Hildebrand, A., additional, Blume, A., additional, Girão da Cruz, M. T., additional, Simões, S., additional, Pedroso de Lima, M. C., additional, Graser, A., additional, Nahde, T., additional, Fahr, A., additional, Müller, R., additional, Müller-Brüsselbach, S., additional, Cudmore, S., additional, O'Mahony, D., additional, Hoving, S., additional, van Tiel, S. T., additional, Seynhaeve, A. L. B., additional, Ambagtsheer, G., additional, Eggermont, A. M. M., additional, ten Hagen, T. L. M., additional, Høyrup, P., additional, Jensen, S. S., additional, Jørgensen, K., additional, Iden, D., additional, Kuang, H., additional, Mullen, P., additional, Jacobs, C., additional, Roben, P., additional, Stevens, T., additional, Lollo, C., additional, Ishida, T., additional, Maeda, R., additional, Masuda, K., additional, Ichihara, M., additional, Kiwada, H., additional, Jung, K., additional, Reszka, R., additional, Kaiser, N., additional, Ohloff, I., additional, Linser-Haar, S., additional, Massing, U., additional, Schubert, R., additional, Kan, P., additional, Tsao, C. W., additional, Chen, W. K., additional, Wang, A. J., additional, Kimpfler, A., additional, Gerber, C., additional, Wieschollek, A., additional, Bruchelt, G., additional, Kobayashi, T., additional, Okada, Y., additional, Sone, S., additional, Harashima, H., additional, Maruyama, K., additional, Kondo, Masayo, additional, Lee, Chun Man, additional, Tanaka, Toshiyuki, additional, Su, Wei, additional, Kitagawa, Toru, additional, Ito, Toshinori, additional, Matsuda, Hikaru, additional, Murai, Toshiyuki, additional, Miyasaka, Masayuki, additional, Junji, Kimura, additional, Kondo, Masami, additional, Asai, Tomohiro, additional, Ogino, Koichi, additional, Taki, Takao, additional, Tsukada, Hideo, additional, Baba, Kazuhiko, additional, Oku, Naoto, additional, Koning, G. A., additional, Wauben, M. H. M., additional, Vestweber, D., additional, Everts, M., additional, Kok, R. J., additional, Schraa, A. J., additional, Molema, G., additional, Schiffelers, R. M., additional, Storm, G., additional, Kristl, J., additional, Šentjurc, M., additional, Abramović, Z., additional, Landry, S., additional, Perron, S., additional, Bestman-Smith, J., additional, Désormeaux, A., additional, Tremblay, M. J., additional, Bergeron, M. G., additional, Madeira, C., additional, Loura, L. M. S., additional, Fedorov, A., additional, Prieto, M., additional, Aires-Barros, M. R., additional, Marques, C. M., additional, Simões, S. I., additional, Cruz, M. E., additional, Cevc, G., additional, Martins, M. B., additional, Moreira, J. N., additional, Gaspar, R., additional, Allen, T. M., additional, Esposito, C., additional, Ortaggi, G., additional, Bianco, A., additional, Bonadies, F., additional, Malizia, D., additional, Napolitano, R., additional, Cametti, C., additional, Mossa, G., additional, Endert, Gerold, additional, Essler, Frank, additional, Lutz, Silke, additional, Panzner, Steffen, additional, Pastorino, F., additional, Brignole, C., additional, Pagnan, G., additional, Moase, E. H., additional, Ponzoni, M., additional, Pavelic, Z., additional, Škalko-Basnet, N., additional, Jalšenjak, I., additional, Penacho, N., additional, Pisano, C., additional, Bucci, F., additional, Serafini, S., additional, Martinelli, R., additional, Cupelli, A., additional, Marconi, A., additional, Ferrara, F. F., additional, Santaniello, M., additional, Critelli, L., additional, Tinti, O., additional, Luisi, P., additional, Carminati, P., additional, Galletti, B., additional, Sauer, I., additional, Schleef, M., additional, Voß, C., additional, Schmidt, T., additional, Flaschel, E., additional, König, S., additional, Wenger, T., additional, Dumond, J., additional, Bogetto, N., additional, Reboud-Ravaux, M., additional, Schramm, H. J., additional, Schramm, W., additional, Sheynis, T., additional, Rozner, S., additional, Kolusheva, S., additional, Satchell, D., additional, Jelnik, R., additional, Shigeta, Y., additional, Imanaka, H., additional, Ando, H., additional, Makino, T., additional, Baba, N., additional, Shimizu, K., additional, Takada, M., additional, Baba, K., additional, Namba, Y., additional, Simberg, Dmitri, additional, Danino, Dganit, additional, Talmon, Yeshayahu, additional, Minsky, Abraham, additional, Ferrari, Marilyn E., additional, Wheeler, Carl J., additional, Barenholz, Yechezkel, additional, Takada, Miki, additional, Shimizu, Kosuke, additional, Kuromi, Koici, additional, Takeuchi, Y., additional, North, J. R., additional, Nango, M., additional, Tewes, B., additional, Köchling, T., additional, Deissler, M., additional, Kühl, C., additional, Marx, U., additional, Strote, G., additional, Gropp, F., additional, Qualls, Marquita M., additional, Kim, Jong-Mok, additional, Thompson, David H., additional, Zhang, Zhi-Yi, additional, Shum, Pochi, additional, Collier, Joel H., additional, Hu, Bi-Huang, additional, Ruberti, Jeffrey W., additional, Messersmith, Phillip B., additional, Tsuruda, T., additional, Nakade, A., additional, Sadzuka, Y., additional, Hirota, S., additional, Sonobe, T., additional, Vorauer-Uhl, K., additional, Wagner, A., additional, Katinger, H., additional, Weeke-Klimp, A. H., additional, Bartsch, M., additional, Meijer, D. K. F., additional, Zeisig, R., additional, Walther, W., additional, Reß, A., additional, Fichtner, I., additional, Zschörnig, O., additional, Schiller, J., additional, Süß, M., additional, Bergmeier, C., additional, Arnold, K., additional, Nchinda, Godwin, additional, Überla, Klaus, additional, and Zschörnig, Olaf, additional
- Published
- 2003
- Full Text
- View/download PDF
10. Poster Abstracts
- Author
-
Littger, Ralf, Alke, Alexandra, Tewes, Bernhard, Gropp, Felix, Asai, T., Watanabe, K., Kuromi, K., Kurohane, K., Ogino, K., Taki, T., Tsukada, H., Nakayama, J., Oku, N., Babai, I., Matyas, G., Baranji, L., Milosevits, J., Alving, C. R., Bendas, G., Rothe, U., Scherphof, G. L., Kamps, J. A. A. M., Kessner, S., Rothe, U., Bendas, G., Carafa, M., Di Stefano, A., Sozio, P., Cacciatore, I., Mosciatti, B., Santucci, E., Choice, E., Harvie, P., Galbraith, T., Zunder, E., Dutzar, B., Anklesaria, P., Paul, R., Cocquyt, J., De Cuyper, M., Van der Meeren, P., Cruz, M. E. M., Gaspar, M. M., Silva, M. T., Dathe, M., Nikolenko, H., Wessolowski, A., Schmieder, P., Beyermann, M., Bienert, M., Santos, N. Dos, Cox, K. A., Allen, C., Gallagher, R. C., Ickenstein, L., Mayer, L. D., Bally, M. B., Fischer, S., Margalit, R., Freisleben, H.-J., Garidel, P., Chen, H. C., Moore, D., Mendelsohn, R., Garidel, P., Keller, M., Hildebrand, A., Blume, A., Girão da Cruz, M. T., Simões, S., Pedroso de Lima, M. C., Graser, A., Nahde, T., Fahr, A., Müller, R., Müller-Brüsselbach, S., Harvie, P., Dutzar, B., Choice, E., Cudmore, S., O'Mahony, D., Anklesaria, P., Paul, R., Hoving, S., van Tiel, S. T., Seynhaeve, A. L. B., Ambagtsheer, G., Eggermont, A. M. M., ten Hagen, T. L. M., Høyrup, P., Jensen, S. S., Jørgensen, K., Iden, D., Kuang, H., Mullen, P., Jacobs, C., Roben, P., Stevens, T., Lollo, C., Ishida, T., Maeda, R., Masuda, K., Ichihara, M., Kiwada, H., Jung, K., Reszka, R., Kaiser, N., Ohloff, I., Linser-Haar, S., Massing, U., Schubert, R., Kan, P., Tsao, C. W., Chen, W. K., Wang, A. J., Kimpfler, A., Gerber, C., Wieschollek, A., Bruchelt, G., Schubert, R., Kobayashi, T., Okada, Y., Ishida, T., Sone, S., Harashima, H., Maruyama, K., Kiwada, H., Kondo, Masayo, Lee, Chun Man, Tanaka, Toshiyuki, Su, Wei, Kitagawa, Toru, Ito, Toshinori, Matsuda, Hikaru, Murai, Toshiyuki, Miyasaka, Masayuki, Junji, Kimura, Kondo, Masami, Asai, Tomohiro, Ogino, Koichi, Taki, Takao, Tsukada, Hideo, Baba, Kazuhiko, Oku, Naoto, Koning, G. A., Wauben, M. H. M., ten Hagen, T. L. M., Vestweber, D., Everts, M., Kok, R. J., Schraa, A. J., Molema, G., Schiffelers, R. M., Storm, G., Kristl, J., Šentjurc, M., Abramovi, Z., Landry, S., Perron, S., Bestman-Smith, J., Désormeaux, A., Tremblay, M. J., Bergeron, M. G., Madeira, C., Loura, L. M. S., Fedorov, A., Prieto, M., Aires-Barros, M. R., Marques, C. M., Simões, S. I., Cruz, M. E., Cevc, G., Martins, M. B., Moreira, J. N., Gaspar, R., Allen, T. M., Esposito, C., Ortaggi, G., Bianco, A., Bonadies, F., Malizia, D., Napolitano, R., Cametti, C., Mossa, G., Endert, Gerold, Essler, Frank, Lutz, Silke, Panzner, Steffen, Pastorino, F., Brignole, C., Pagnan, G., Moase, E. H., Allen, T. M., Ponzoni, M., Pavelic, Z., Škalko-Basnet, N., Jalšenjak, I., Penacho, N., Simões, S., Pedroso de Lima, M. C., Pisano, C., Bucci, F., Serafini, S., Martinelli, R., Cupelli, A., Marconi, A., Ferrara, F. F., Santaniello, M., Critelli, L., Tinti, O., Luisi, P., Carminati, P., Santaniello, M., Bucci, F., Tinti, O., Pisano, C., Critelli, L., Galletti, B., Luisi, P., Carminati, P., Sauer, I., Nikolenko, H., Dathe, M., Schleef, M., Voß, C., Schmidt, T., Flaschel, E., König, S., Wenger, T., Dumond, J., Bogetto, N., Reboud-Ravaux, M., Schramm, H. J., Schramm, W., Sheynis, T., Rozner, S., Kolusheva, S., Satchell, D., Jelnik, R., Shigeta, Y., Imanaka, H., Ando, H., Makino, T., Kurohane, K., Oku, N., Baba, N., Shimizu, K., Asai, T., Takada, M., Baba, K., Namba, Y., Oku, N., Simberg, Dmitri, Danino, Dganit, Talmon, Yeshayahu, Minsky, Abraham, Ferrari, Marilyn E., Wheeler, Carl J., Barenholz, Yechezkel, Takada, Miki, Shimizu, Kosuke, Kuromi, Koici, Asai, Tomohiro, Baba, Kazuhiko, Oku, Naoto, Takeuchi, Y., Kurohane, K., North, J. R., Namba, Y., Nango, M., Oku, N., Tewes, B., Köchling, T., Deissler, M., Kühl, C., Marx, U., Strote, G., Gropp, F., Qualls, Marquita M., Kim, Jong-Mok, Thompson, David H., Zhang, Zhi-Yi, Shum, Pochi, Collier, Joel H., Hu, Bi-Huang, Ruberti, Jeffrey W., Messersmith, Phillip B., Thompson, David H., Tsuruda, T., Nakade, A., Sadzuka, Y., Hirota, S., Sonobe, T., Vorauer-Uhl, K., Wagner, A., Katinger, H., Wagner, A., Vorauer-Uhl, K., Katinger, H., Weeke-Klimp, A. H., Bartsch, M., Meijer, D. K. F., Scherphof, G. L., Kamps, J. A. A. M., Zeisig, R., Walther, W., Reß, A., Fichtner, I., Zschörnig, O., Schiller, J., Süß, M., Bergmeier, C., Arnold, K., Nchinda, Godwin, Überla, Klaus, and Zschörnig, Olaf
- Abstract
DOCSPER—A Synthetic Lipid Fit for In Vivo ApplicationDOCSPER [1,3-Dioleoyloxy-2-(N5-carbamoyl-spermine)-propane] is a cationic amphiphile consisting of a hydrophobic 1,3 dioleylglycerol moiety and threefold positively charged spermine head group (1). We optimised the 5-step-synthesis of the lipospermine and after up-scaling we have obtained sufficient amounts to initiate preclinical investigations. DOCSPER was tested for its ability to transfect eukaryotic cells in vitro. It has proven to possess high transfection efficiency in comparison to commercially available liposomal transfection agents. Furthermore, DOCSPER was extensively tested in several in vivo studies (23). These studies revealed a high transfection efficiency, whereas very low toxicity levels were detected. Thus, the results clearly indicate that the cationic lipid DOCSPER is a reliable, low-risk system for broad applications in gene therapy.Groth D. et al. Int J Pharm 1998; 162:143–157.Nikol S. et al. Int J Angiol 2000; 9:87–95.Armeanu S. et al. Mol Ther 2000; 1(4):366–375.
- Published
- 1982
- Full Text
- View/download PDF
11. Growth of transgenic RAF-induced lung adenomas is increased in mice with a disrupted PPARbeta/delta gene
- Author
-
Müller-Brüsselbach S, Ebrahimsade S, Jäkel J, Eckhardt J, Ur, Rapp, Jm, Peters, Moll R, and Rolf Müller
12. 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
13. TRAIL-dependent apoptosis of peritoneal mesothelial cells by NK cells promotes ovarian cancer invasion.
- Author
-
Steitz AM, Schröder C, Knuth I, Keber CU, Sommerfeld L, Finkernagel F, Jansen JM, Wagner U, Müller-Brüsselbach S, Worzfeld T, Huber M, Beutgen VM, Graumann J, Pogge von Strandmann E, Müller R, and Reinartz S
- Abstract
A crucial requirement for metastasis formation in ovarian high-grade serous carcinoma (HGSC) is the disruption of the protective peritoneal mesothelium. Using co-culture systems of primary human cells, we discovered that tumor-associated NK cells induce TRAIL-dependent apoptosis in mesothelial cells via death receptors DR4 and DR5 upon encounter with activated T cells. Upregulation of TRAIL expression in NK cells concomitant with enhanced cytotoxicity toward mesothelial cells was driven predominantly by T-cell-derived TNFα, as shown by affinity proteomics-based analysis of the T cell secretome in conjunction with functional studies. Consistent with these findings, we detected apoptotic mesothelial cells in the peritoneal fluid of HGSC patients. In contrast to mesothelial cells, HGSC cells express negligible levels of both DR4 and DR5 and are TRAIL resistant, indicating cell-type-selective killing by NK cells. Our data point to a cooperative action of T and NK in breaching the mesothelial barrier in HGSC patients., Competing Interests: The authors declare no competing interests., (© 2023 The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
14. The lysophosphatidic acid-regulated signal transduction network in ovarian cancer cells and its role in actomyosin dynamics, cell migration and entosis.
- Author
-
Ojasalu K, Lieber S, Sokol AM, Nist A, Stiewe T, Bullwinkel I, Finkernagel F, Reinartz S, Müller-Brüsselbach S, Grosse R, Graumann J, and Müller R
- Subjects
- Humans, Female, Entosis, Neoplasm Recurrence, Local, Signal Transduction, Cell Movement physiology, Actomyosin metabolism, Ovarian Neoplasms metabolism
- Abstract
Lysophosphatidic acid (LPA) species accumulate in the ascites of ovarian high-grade serous cancer (HGSC) and are associated with short relapse-free survival. LPA is known to support metastatic spread of cancer cells by activating a multitude of signaling pathways via G-protein-coupled receptors of the LPAR family. Systematic unbiased analyses of the LPA-regulated signal transduction network in ovarian cancer cells have, however, not been reported to date. Methods: LPA-induced signaling pathways were identified by phosphoproteomics of both patient-derived and OVCAR8 cells, RNA sequencing, measurements of intracellular Ca
2+ and cAMP as well as cell imaging. The function of LPARs and downstream signaling components in migration and entosis were analyzed by selective pharmacological inhibitors and RNA interference. Results: Phosphoproteomic analyses identified > 1100 LPA-regulated sites in > 800 proteins and revealed interconnected LPAR1, ROCK/RAC, PKC/D and ERK pathways to play a prominent role within a comprehensive signaling network. These pathways regulate essential processes, including transcriptional responses, actomyosin dynamics, cell migration and entosis. A critical component of this signaling network is MYPT1, a stimulatory subunit of protein phosphatase 1 (PP1), which in turn is a negative regulator of myosin light chain 2 (MLC2). LPA induces phosphorylation of MYPT1 through ROCK (T853) and PKC/ERK (S507), which is majorly driven by LPAR1. Inhibition of MYPT1, PKC or ERK impedes both LPA-induced cell migration and entosis, while interference with ROCK activity and MLC2 phosphorylation selectively blocks entosis, suggesting that MYPT1 figures in both ROCK/MLC2-dependent and -independent pathways. We finally show a novel pathway governed by LPAR2 and the RAC-GEF DOCK7 to be indispensable for the induction of entosis. Conclusion: We have identified a comprehensive LPA-induced signal transduction network controlling LPA-triggered cytoskeletal changes, cell migration and entosis in HGSC cells. Due to its pivotal role in this network, MYPT1 may represent a promising target for interfering with specific functions of PP1 essential for HGSC progression., Competing Interests: Competing Interests: The authors have declared that no competing interest exists., (© The author(s).)- Published
- 2023
- Full Text
- View/download PDF
15. Basal cell adhesion molecule promotes metastasis-associated processes in ovarian cancer.
- Author
-
Sivakumar S, Lieber S, Librizzi D, Keber C, Sommerfeld L, Finkernagel F, Roth K, Reinartz S, Bartsch JW, Graumann J, Müller-Brüsselbach S, and Müller R
- Subjects
- Animals, Female, Humans, Mice, Cell Adhesion physiology, Spheroids, Cellular, Cell Adhesion Molecules metabolism, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology
- Abstract
Background: Basal cell adhesion molecule (BCAM) is a laminin α5 (LAMA5) binding membrane-bound protein with a putative role in cancer. Besides full-length BCAM1, an isoform lacking most of the cytoplasmic domain (BCAM2), and a soluble form (sBCAM) of unknown function are known. In ovarian carcinoma (OC), all BCAM forms are abundant and associated with poor survival, yet BCAM's contribution to peritoneal metastatic spread remains enigmatic., Methods: Biochemical, omics-based and real-time cell assays were employed to identify the source of sBCAM and metastasis-related functions of different BCAM forms. OC cells, explanted omentum and a mouse model of peritoneal colonisation were used in loss- and gain-of-function experiments., Results: We identified ADAM10 as a major BCAM sheddase produced by OC cells and identified proteolytic cleavage sites proximal to the transmembrane domain. Recombinant soluble BCAM inhibited single-cell adhesion and migration identically to membrane-bound isoforms, confirming its biological activity in OC. Intriguingly, this seemingly anti-tumorigenic potential of BCAM contrasts with a novel pro-metastatic function discovered in the present study. Thus, all queried BCAM forms decreased the compactness of tumour cell spheroids by inhibiting LAMA5 - integrin β1 interactions, promoted spheroid dispersion in a three-dimensional collagen matrix, induced clearance of mesothelial cells at spheroid attachment sites in vitro and enhanced invasion of spheroids into omental tissue both ex vivo and in vivo., Conclusions: Membrane-bound BCAM as well as sBCAM shed by ADAM10 act as decoys rather than signalling receptors to modulate metastasis-related functions. While BCAM appears to have tumour-suppressive effects on single cells, it promotes the dispersion of OC cell spheroids by regulating LAMA5-integrin-β1-dependent compaction and thereby facilitating invasion of metastatic target sites. As peritoneal dissemination is majorly mediated by spheroids, these findings offer an explanation for the association of BCAM with a poor clinical outcome of OC, suggesting novel therapeutic options., (© 2023 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.)
- Published
- 2023
- Full Text
- View/download PDF
16. Prostacyclin Released by Cancer-Associated Fibroblasts Promotes Immunosuppressive and Pro-Metastatic Macrophage Polarization in the Ovarian Cancer Microenvironment.
- Author
-
Sommerfeld L, Knuth I, Finkernagel F, Pesek J, Nockher WA, Jansen JM, Wagner U, Nist A, Stiewe T, Müller-Brüsselbach S, Müller R, and Reinartz S
- Abstract
Metastasis of high-grade ovarian carcinoma (HGSC) is orchestrated by soluble mediators of the tumor microenvironment. Here, we have used transcriptomic profiling to identify lipid-mediated signaling pathways encompassing 41 ligand-synthesizing enzymes and 23 cognate receptors in tumor, immune and stroma cells from HGSC metastases and ascites. Due to its strong association with a poor clinical outcome, prostacyclin (PGI
2 ) synthase (PTGIS) is of particular interest in this signaling network. PTGIS is highly expressed by cancer-associated fibroblasts (CAF), concomitant with elevated PGI2 synthesis, whereas tumor-associated macrophages (TAM) exhibit the highest expression of its surface receptor (PTGIR). PTGIR activation by PGI2 agonists triggered cAMP accumulation and induced a mixed-polarization macrophage phenotype with altered inflammatory gene expression, including CXCL10 and IL12A repression, as well as reduced phagocytic capability. Co-culture experiments provided further evidence for the interaction of CAF with macrophages via PGI2 , as the effect of PGI2 agonists on phagocytosis was mitigated by cyclooxygenase inhibitors. Furthermore, conditioned medium from PGI2 -agonist-treated TAM promoted tumor adhesion to mesothelial cells and migration in a PTGIR-dependent manner, and PTGIR activation induced the expression of metastasis-associated and pro-angiogenic genes. Taken together, our study identifies a PGI2 /PTGIR-driven crosstalk between CAF, TAM and tumor cells, promoting immune suppression and a pro-metastatic environment.- Published
- 2022
- Full Text
- View/download PDF
17. Arachidonic acid, a clinically adverse mediator in the ovarian cancer microenvironment, impairs JAK-STAT signaling in macrophages by perturbing lipid raft structures.
- Author
-
Hammoud MK, Dietze R, Pesek J, Finkernagel F, Unger A, Bieringer T, Nist A, Stiewe T, Bhagwat AM, Nockher WA, Reinartz S, Müller-Brüsselbach S, Graumann J, and Müller R
- Subjects
- Arachidonic Acid metabolism, Humans, Macrophages metabolism, Membrane Microdomains metabolism, STAT1 Transcription Factor metabolism, Signal Transduction, Neoplasms metabolism, Tumor Microenvironment
- Abstract
Survival of ovarian carcinoma is associated with the abundance of immunosuppressed CD163
high CD206high tumor-associated macrophages (TAMs) and high levels of arachidonic acid (AA) in the tumor microenvironment. Here, we show that both associations are functionally linked. Transcriptional profiling revealed that high CD163 and CD206/MRC1 expression in TAMs is strongly associated with an inhibition of cytokine-triggered signaling, mirrored by an impaired transcriptional response to interferons and IL-6 in monocyte-derived macrophages by AA. This inhibition of pro-inflammatory signaling is caused by dysfunctions of the cognate receptors, indicated by the inhibition of JAK1, JAK2, STAT1, and STAT3 phosphorylation, and by the displacement of the interferon receptor IFNAR1, STAT1 and other immune-regulatory proteins from lipid rafts. AA exposure led to a dramatic accumulation of free AA in lipid rafts, which appears to be mechanistically crucial, as the inhibition of its incorporation into phospholipids did not affect the AA-mediated interference with STAT1 phosphorylation. Inhibition of interferon-triggered STAT1 phosphorylation by AA was reversed by water-soluble cholesterol, known to prevent the perturbation of lipid raft structure by AA. These findings suggest that the pharmacologic restoration of lipid raft functions in TAMs may contribute to the development new therapeutic approaches., (© 2022 The Authors. Molecular Oncology published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)- Published
- 2022
- Full Text
- View/download PDF
18. The multicellular signalling network of ovarian cancer metastases.
- Author
-
Sommerfeld L, Finkernagel F, Jansen JM, Wagner U, Nist A, Stiewe T, Müller-Brüsselbach S, Sokol AM, Graumann J, Reinartz S, and Müller R
- Subjects
- Cell Movement genetics, Cell Movement physiology, Female, Gene Regulatory Networks genetics, Humans, Neoplasm Metastasis immunology, Ovarian Neoplasms immunology, Signal Transduction genetics, Signal Transduction physiology, Gene Regulatory Networks physiology, Neoplasm Metastasis physiopathology, Ovarian Neoplasms physiopathology
- Abstract
Background: Transcoelomic spread is the major route of metastasis of ovarian high-grade serous carcinoma (HGSC) with the omentum as the major metastatic site. Its unique tumour microenvironment with its large populations of adipocytes, mesothelial cells and immune cells establishes an intercellular signaling network that is instrumental for metastatic growth yet poorly understood., Methods: Based on transcriptomic analysis of tumour cells, tumour-associated immune and stroma cells we defined intercellular signaling pathways for 284 cytokines and growth factors and their cognate receptors after bioinformatic adjustment for contaminating cell types. The significance of individual components of this network was validated by analysing clinical correlations and potentially pro-metastatic functions, including tumour cell migration, pro-inflammatory signal transduction and TAM expansion., Results: The data show an unexpected prominent role of host cells, and in particular of omental adipocytes, mesothelial cells and fibroblasts (CAF), in sustaining this signaling network. These cells, rather than tumour cells, are the major source of most cytokines and growth factors in the omental microenvironment (n = 176 vs. n = 13). Many of these factors target tumour cells, are linked to metastasis and are associated with a short survival. Likewise, tumour stroma cells play a major role in extracellular-matrix-triggered signaling. We have verified the functional significance of our observations for three exemplary instances. We show that the omental microenvironment (i) stimulates tumour cell migration and adhesion via WNT4 which is highly expressed by CAF; (ii) induces pro-tumourigenic TAM proliferation in conjunction with high CSF1 expression by omental stroma cells and (iii) triggers pro-inflammatory signaling, at least in part via a HSP70-NF-κB pathway., Conclusions: The intercellular signaling network of omental metastases is majorly dependent on factors secreted by immune and stroma cells to provide an environment that supports ovarian HGSC progression. Clinically relevant pathways within this network represent novel options for therapeutic intervention., (© 2021 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.)
- Published
- 2021
- Full Text
- View/download PDF
19. Phosphoproteomics identify arachidonic-acid-regulated signal transduction pathways modulating macrophage functions with implications for ovarian cancer.
- Author
-
Dietze R, Hammoud MK, Gómez-Serrano M, Unger A, Bieringer T, Finkernagel F, Sokol AM, Nist A, Stiewe T, Reinartz S, Ponath V, Preußer C, von Strandmann EP, Müller-Brüsselbach S, Graumann J, and Müller R
- Subjects
- Calcium metabolism, Extracellular Vesicles drug effects, Extracellular Vesicles metabolism, Female, Group II Phospholipases A2 metabolism, Humans, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local metabolism, Ovarian Neoplasms metabolism, Reactive Oxygen Species metabolism, Transcription, Genetic drug effects, Tumor Microenvironment drug effects, Arachidonic Acid pharmacology, Macrophages drug effects, Ovarian Neoplasms drug therapy, Phosphorylation drug effects, Signal Transduction drug effects
- Abstract
Arachidonic acid (AA) is a polyunsaturated fatty acid present at high concentrations in the ovarian cancer (OC) microenvironment and associated with a poor clinical outcome. In the present study, we have unraveled a potential link between AA and macrophage functions. Methods: AA-triggered signal transduction was studied in primary monocyte-derived macrophages (MDMs) by phosphoproteomics, transcriptional profiling, measurement of intracellular Ca
2+ accumulation and reactive oxygen species production in conjunction with bioinformatic analyses. Functional effects were investigated by actin filament staining, quantification of macropinocytosis and analysis of extracellular vesicle release. Results: We identified the ASK1 - p38δ/α (MAPK13/14) axis as a central constituent of signal transduction pathways triggered by non-metabolized AA. This pathway was induced by the Ca2+ -triggered activation of calmodulin kinase II, and to a minor extent by ROS generation in a subset of donors. Activated p38 in turn was linked to a transcriptional stress response associated with a poor relapse-free survival. Consistent with the phosphorylation of the p38 substrate HSP27 and the (de)phosphorylation of multiple regulators of Rho family GTPases, AA impaired actin filament organization and inhibited actin-driven macropinocytosis. AA also affected the phosphorylation of proteins regulating vesicle biogenesis, and consistently, AA enhanced the release of tetraspanin-containing exosome-like vesicles. Finally, we identified phospholipase A2 group 2A (PLA2G2A) as the clinically most relevant enzyme producing extracellular AA, providing further potentially theranostic options. Conclusion: Our results suggest that AA contributes to an unfavorable clinical outcome of OC by impacting the phenotype of tumor-associated macrophages. Besides critical AA-regulated signal transduction proteins identified in the present study, PLA2G2A might represent a potential prognostic tool and therapeutic target to interfere with OC progression., Competing Interests: Competing Interests: The authors have declared that no competing interest exists., (© The author(s).)- Published
- 2021
- Full Text
- View/download PDF
20. Upregulation of mesothelial genes in ovarian carcinoma cells is associated with an unfavorable clinical outcome and the promotion of cancer cell adhesion.
- Author
-
Ojasalu K, Brehm C, Hartung K, Nischak M, Finkernagel F, Rexin P, Nist A, Pavlakis E, Stiewe T, Jansen JM, Wagner U, Gattenlöhner S, Bräuninger A, Müller-Brüsselbach S, Reinartz S, and Müller R
- Subjects
- Apoptosis genetics, Ascites genetics, Ascites pathology, Biomarkers, Tumor metabolism, Cell Adhesion, Cell Line, Tumor, Cystadenocarcinoma, Serous genetics, Cystadenocarcinoma, Serous pathology, Disease-Free Survival, Female, Humans, Neoplasm Grading, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Peritoneal Neoplasms secondary, Polymorphism, Single Nucleotide genetics, Spheroids, Cellular pathology, Treatment Outcome, Epithelium pathology, Gene Expression Regulation, Neoplastic, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology, Up-Regulation genetics
- Abstract
A hallmark of ovarian high-grade serous carcinoma (HGSC) is its early and massive peritoneal dissemination via the peritoneal fluid. It is generally believed that tumor cells must breach the mesothelium of peritoneal organs to adhere to the underlying extracellular matrix (ECM) and initiate metastatic growth. However, the molecular mechanisms underlying these processes are only partially understood. Here, we have analyzed 52 matched samples of spheroids and solid tumor masses (suspected primary lesions and metastases) from 10 patients by targeted sequencing of 21 loci previously proposed as targets of HGSC driver mutations. This analysis revealed very similar patterns of genetic alterations in all samples. One exception was FAT3 with a strong enrichment of mutations in metastases compared with presumed primary lesions in two cases. FAT3 is a putative tumor suppressor gene that codes for an atypical cadherin, pointing a potential role in peritoneal dissemination in a subgroup of HGSC patients. By contrast, transcriptome data revealed clear and consistent differences between tumor cell spheroids from ascites and metastatic lesions, which were mirrored by the in vitro adherence of ascites-derived spheroids. The adhesion-induced transcriptional alterations in metastases and adherent cells resembled epithelial-mesenchymal transition, but surprisingly also included the upregulation of a specific subset of mesothelial genes, such as calretinin (CALB2) and podoplanin (PDPN). Consistent with this finding, calretinin staining was also observed in subsets of tumor cells in HGSC metastases, particularly at the invasive tumor edges. Intriguingly, a high expression of either CALB2 or PDPN was strongly associated with a poor clinical outcome. siRNA-mediated CALB2 silencing triggered the detachment of adherent HGSC cells in vitro and inhibited the adhesion of detached HGSC cells to collagen type I. Our data suggest that the acquisition of a mesenchymal-mesothelial phenotype contributes to cancer cell adhesion to the ECM of peritoneal organs and HGSC progression., (© 2020 The Authors. Published by FEBS Press and John Wiley & Sons Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
21. Cell type-selective pathways and clinical associations of lysophosphatidic acid biosynthesis and signaling in the ovarian cancer microenvironment.
- Author
-
Reinartz S, Lieber S, Pesek J, Brandt DT, Asafova A, Finkernagel F, Watzer B, Nockher WA, Nist A, Stiewe T, Jansen JM, Wagner U, Konzer A, Graumann J, Grosse R, Worzfeld T, Müller-Brüsselbach S, and Müller R
- Subjects
- Ascites metabolism, Cell Line, Tumor, Disease-Free Survival, Female, Gene Expression Regulation, Neoplastic, Humans, Macrophages metabolism, Macrophages pathology, Metabolome, Neoplasms, Cystic, Mucinous, and Serous pathology, Ovarian Neoplasms genetics, Receptors, Lysophosphatidic Acid metabolism, Treatment Outcome, Up-Regulation genetics, Lysophospholipids biosynthesis, Ovarian Neoplasms metabolism, Ovarian Neoplasms pathology, Signal Transduction, Tumor Microenvironment genetics
- Abstract
The peritoneal fluid of ovarian carcinoma patients promotes cancer cell invasion and metastatic spread with lysophosphatidic acid (LPA) as a potentially crucial mediator. However, the origin of LPA in ascites and the clinical relevance of individual LPA species have not been addressed. Here, we show that the levels of multiple acyl-LPA species are strongly elevated in ascites versus plasma and are associated with short relapse-free survival. Data derived from transcriptome and secretome analyses of primary ascite-derived cells indicate that (a) the major route of LPA synthesis is the consecutive action of a secretory phospholipase A
2 (PLA2 ) and autotaxin, (b) that the components of this pathway are coordinately upregulated in ascites, and (c) that CD163+CD206+ tumor-associated macrophages play an essential role as main producers of PLA2 G7 and autotaxin. The latter conclusion is consistent with mass spectrometry-based metabolomic analyses of conditioned medium from ascites cells, which showed that tumor-associated macrophages, but not tumor cells, are able to produce 20:4 acyl-LPA in lipid-free medium. Furthermore, our transcriptomic data revealed that LPA receptor (LPAR) genes are expressed in a clearly cell type-selective manner: While tumor cells express predominantly LPAR1-3, macrophages and T cells also express LPAR5 and LPAR6 at high levels, pointing to cell type-selective LPA signaling pathways. RNA profiling identified cytokines linked to cell motility and migration as the most conspicuous class of LPA-induced genes in macrophages, suggesting that LPA exerts protumorigenic properties at least in part via the tumor secretome., (© 2018 The Authors. Published by FEBS Press and John Wiley & Sons Ltd.)- Published
- 2019
- Full Text
- View/download PDF
22. Chromatin Binding of c -REL and p65 Is Not Limiting for Macrophage IL12B Transcription During Immediate Suppression by Ovarian Carcinoma Ascites.
- Author
-
Unger A, Finkernagel F, Hoffmann N, Neuhaus F, Joos B, Nist A, Stiewe T, Visekruna A, Wagner U, Reinartz S, Müller-Brüsselbach S, Müller R, and Adhikary T
- Abstract
Tumors frequently exploit homeostatic mechanisms that suppress expression of IL-12, a central mediator of inflammatory and anti-tumor responses. The p40 subunit of the IL-12 heterodimer, encoded by IL12B , is limiting for these functions. Ovarian carcinoma patients frequently produce ascites which exerts immunosuppression by means of soluble factors. The NFκB pathway is necessary for transcription of IL12B , which is not expressed in macrophages freshly isolated from ascites. This raises the possibility that ascites prevents IL12B expression by perturbing NFκB binding to chromatin. Here, we show that ascites-mediated suppression of IL12B induction by LPS plus IFNγ in primary human macrophages is rapid, and that suppression can be reversible after ascites withdrawal. Nuclear translocation of the NFκB transcription factors c -REL and p65 was strongly reduced by ascites. Surprisingly, however, their binding to the IL12B locus and to CXCL10 , a second NFκB target gene, was unaltered, and the induction of CXCL10 transcription was not suppressed by ascites. These findings indicate that, despite its reduced nuclear translocation, NFκB function is not generally impaired by ascites, suggesting that ascites-borne signals target additional pathways to suppress IL12B induction. Consistent with these data, IL-10, a clinically relevant constituent of ascites and negative regulator of NFκB translocation, only partially recapitulated IL12B suppression by ascites. Finally, restoration of a defective IL-12 response by appropriate culture conditions was observed only in macrophages from a subset of donors, which may have important implications for the understanding of patient-specific immune responses.
- Published
- 2018
- Full Text
- View/download PDF
23. Interferon signaling in ascites-associated macrophages is linked to a favorable clinical outcome in a subgroup of ovarian carcinoma patients.
- Author
-
Adhikary T, Wortmann A, Finkernagel F, Lieber S, Nist A, Stiewe T, Wagner U, Müller-Brüsselbach S, Reinartz S, and Müller R
- Subjects
- Biomarkers, Cluster Analysis, Cytokines, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Humans, Immunophenotyping, Macrophages pathology, Ovarian Neoplasms pathology, Prognosis, Reproducibility of Results, Transcriptome, Tumor Microenvironment, Ascites pathology, Interferons metabolism, Macrophages metabolism, Ovarian Neoplasms metabolism, Ovarian Neoplasms mortality, Signal Transduction
- Abstract
Background: Although tumor-associated macrophages (TAMs) are essential for cancer progression, connections between different clinical outcomes and transcriptional networks have not been reported. We have addressed this issue by analyzing global expression patterns of TAMs isolated from the ascites of ovarian cancer patients., Results: TAMs isolated from different ovarian cancer patients can be stratified by coexpression or principal component analysis into subgroups with specific biological features and associated with distinct clinical outcomes. A hallmark of subgroup A is a high expression of clinically unfavorable markers, including (i) high CD163 expression, a surface receptor characteristic of an anti-inflammatory activation state, (ii) increased PCOLCE2 expression, indicative of enhanced extracellular matrix organization, and (iii) elevated ascites levels of IL-6 and IL-10, linked to the aggressiveness of ovarian cancer and immune suppression. In contrast, subgroup B TAMs are characterized by the upregulation of genes linked to immune defense mechanisms and interferon (IFN) signaling. Intriguingly, analysis of published data for 1763 ovarian cancer patients revealed a strong association of this transcriptional signature with a longer overall survival. Consistent with these results, IFNγ was able to abrogate the suppressive effect of ovarian cancer ascites on the inducibility of IL12B expression and IL-12 secretion, a key determinant of a cytotoxic immune response., Conclusions: The survival of ovarian cancer patients is linked to the presence of TAMs with a transcriptional signature that is characterized by a low expression of protumorigenic and immunosuppressive markers and an upregulation of genes linked to interferon signaling. The observed IFNγ-mediated restoration of the inducibility of IL-12 in the presence of ascites provides a possible explanation for the association of an interferon signaling-associated signature with a favorable clinical outcome.
- Published
- 2017
- Full Text
- View/download PDF
24. The transcriptional signature of human ovarian carcinoma macrophages is associated with extracellular matrix reorganization.
- Author
-
Finkernagel F, Reinartz S, Lieber S, Adhikary T, Wortmann A, Hoffmann N, Bieringer T, Nist A, Stiewe T, Jansen JM, Wagner U, Müller-Brüsselbach S, and Müller R
- Subjects
- Female, Flow Cytometry, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Genome-Wide Association Study, Humans, Lymphocyte Activation immunology, Macrophages immunology, Macrophages pathology, Macrophages, Peritoneal immunology, Macrophages, Peritoneal metabolism, Macrophages, Peritoneal pathology, Ovarian Neoplasms immunology, Ovarian Neoplasms pathology, Phagocytosis, Tumor Microenvironment genetics, Tumor Microenvironment immunology, Extracellular Matrix metabolism, Macrophages metabolism, Ovarian Neoplasms genetics, Ovarian Neoplasms metabolism, Transcriptome
- Abstract
Macrophages occur as resident cells of fetal origin or as infiltrating blood monocyte-derived cells. Despite the critical role of tumor-associated macrophages (TAMs) in tumor progression, the contribution of these developmentally and functionally distinct macrophage subsets and their alteration by the tumor microenvironment are poorly understood. We have addressed this question by comparing TAMs from human ovarian carcinoma ascites, resident peritoneal macrophages (pMPHs) and monocyte-derived macrophages (MDMs). Our study revealed striking a similarity between TAMs and pMPHs, which was considerably greater that the resemblance of TAMs and MDMs, including their transcriptomes, their inflammation-related activation state, the presence of receptors mediating immune functions and the expression of tumor-promoting mediators. Consistent with these results, TAMs phagocytized bacteria, presented peptide antigens and activated cytotoxic T cells within their pathophysiological environment. These observations support the notion that tumor-promoting properties of TAMs may reflect, at least to some extent, normal features of resident macrophages rather than functions induced by the tumor microenvironment. In spite of these surprising similarities between TAMs and pMPHs, bioinformatic analyses identified a TAM-selective signature of 30 genes that are upregulated relative to both pMPHs and MDMs. The majority of these genes is linked to extracellular matrix (ECM) remodeling, supporting a role for TAMs in cancer cell invasion and ovarian cancer progression.
- Published
- 2016
- Full Text
- View/download PDF
25. A transcriptome-based global map of signaling pathways in the ovarian cancer microenvironment associated with clinical outcome.
- Author
-
Reinartz S, Finkernagel F, Adhikary T, Rohnalter V, Schumann T, Schober Y, Nockher WA, Nist A, Stiewe T, Jansen JM, Wagner U, Müller-Brüsselbach S, and Müller R
- Subjects
- Female, Gene Regulatory Networks, Humans, Lipid Metabolism genetics, Neoplasm Proteins biosynthesis, Neoplasm Proteins genetics, Ovarian Neoplasms pathology, STAT3 Transcription Factor biosynthesis, Signal Transduction, Transforming Growth Factor beta biosynthesis, Gene Expression Regulation, Neoplastic genetics, Ovarian Neoplasms genetics, Transcriptome genetics, Tumor Microenvironment genetics
- Abstract
Background: Soluble protein and lipid mediators play essential roles in the tumor environment, but their cellular origins, targets, and clinical relevance are only partially known. We have addressed this question for the most abundant cell types in human ovarian carcinoma ascites, namely tumor cells and tumor-associated macrophages., Results: Transcriptome-derived datasets were adjusted for errors caused by contaminating cell types by an algorithm using expression data derived from pure cell types as references. These data were utilized to construct a network of autocrine and paracrine signaling pathways comprising 358 common and 58 patient-specific signaling mediators and their receptors. RNA sequencing based predictions were confirmed for several proteins and lipid mediators. Published expression microarray results for 1018 patients were used to establish clinical correlations for a number of components with distinct cellular origins and target cells. Clear associations with early relapse were found for STAT3-inducing cytokines, specific components of WNT and fibroblast growth factor signaling, ephrin and semaphorin axon guidance molecules, and TGFβ/BMP-triggered pathways. An association with early relapse was also observed for secretory macrophage-derived phospholipase PLA2G7, its product arachidonic acid (AA) and signaling pathways controlled by the AA metabolites PGE2, PGI2, and LTB4. By contrast, the genes encoding norrin and its receptor frizzled 4, both selectively expressed by cancer cells and previously not linked to tumor suppression, show a striking association with a favorable clinical course., Conclusions: We have established a signaling network operating in the ovarian cancer microenvironment with previously unidentified pathways and have defined clinically relevant components within this network.
- Published
- 2016
- Full Text
- View/download PDF
26. Design and Synthesis of Highly Active Peroxisome Proliferator-Activated Receptor (PPAR) β/δ Inverse Agonists with Prolonged Cellular Activity.
- Author
-
Toth PM, Lieber S, Scheer FM, Schumann T, Schober Y, Nockher WA, Adhikary T, Müller-Brüsselbach S, Müller R, and Diederich WE
- Subjects
- Drug Design, Magnetic Resonance Spectroscopy, Mass Spectrometry, PPAR delta antagonists & inhibitors, PPAR-beta antagonists & inhibitors
- Abstract
Based on 3-(((4-(hexylamino)-2-methoxyphenyl)amino)sulfonyl)-2-thiophenecarboxylic acid methyl ester (ST247, compound 2), a recently described peroxisome proliferator-activated receptor (PPAR)β/δ-selective inverse agonist, we designed and synthesized a series of structurally related ligands. The structural modifications presented herein ultimately resulted in a series of ligands that display increased cellular activity relative to 2. Moreover, with methyl 3-(N-(2-(2-ethoxyethoxy)-4-(hexylamino)phenyl)sulfamoyl)thiophene-2-carboxylate (PT-S264, compound 9 u), biologically relevant plasma concentrations in mice were achieved. The compounds presented in this study will provide useful novel tools for future investigations addressing the role of PPARβ/δ in physiological and pathophysiological processes., (© 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2016
- Full Text
- View/download PDF
27. A multi-stage process including transient polyploidization and EMT precedes the emergence of chemoresistent ovarian carcinoma cells with a dedifferentiated and pro-inflammatory secretory phenotype.
- Author
-
Rohnalter V, Roth K, Finkernagel F, Adhikary T, Obert J, Dorzweiler K, Bensberg M, Müller-Brüsselbach S, and Müller R
- Subjects
- Antineoplastic Agents pharmacology, Cell Cycle Checkpoints drug effects, Cell Cycle Checkpoints genetics, Cell Dedifferentiation genetics, Cell Division drug effects, Cell Division genetics, Cell Line, Tumor, Cell Survival drug effects, Cell Survival genetics, Cellular Senescence drug effects, Cellular Senescence genetics, Drug Resistance, Neoplasm genetics, Epithelial-Mesenchymal Transition genetics, Female, Flow Cytometry, Humans, Microscopy, Fluorescence, Nuclear Proteins genetics, Nuclear Proteins metabolism, Ovarian Neoplasms genetics, Ovarian Neoplasms metabolism, Ovarian Neoplasms pathology, Time Factors, Time-Lapse Imaging, Carboplatin pharmacology, Cell Dedifferentiation drug effects, Drug Resistance, Neoplasm drug effects, Epithelial-Mesenchymal Transition drug effects, Inflammation Mediators metabolism, Polyploidy
- Abstract
DNA-damaging drugs induce a plethora of molecular and cellular alterations in tumor cells, but their interrelationship is largely obscure. Here, we show that carboplatin treatment of human ovarian carcinoma SKOV3 cells triggers an ordered sequence of events, which precedes the emergence of mitotic chemoresistant cells. The initial phase of cell death after initiation of carboplatin treatment is followed around day 14 by the emergence of a mixed cell population consisting of cycling, cell cycle-arrested and senescent cells. At this stage, giant cells make up >80% of the cell population, p21 (CDKN1A) in strongly induced, and cell numbers remain nearly static. Subsequently, cell death decreases, p21 expression drops to a low level and cell divisions increase, including regular mitoses of giant cells and depolyploidization by multi-daughter divisions. These events are accompanied by the upregulation of stemness markers and a pro-inflammatory secretory phenotype, peaking after approximately 14 days of treatment. At the same time the cells initiate epithelial to mesenchymal transition, which over the subsequent weeks continuously increases, concomitantly with the emergence of highly proliferative, migratory, dedifferentiated, pro-inflammatory and chemoresistant cells (SKOV3-R). These cells are anchorage-independent and grow in a 3D collagen matrix, while cells on day 14 do not survive under these conditions, indicating that SKOV3-R cells were generated thereafter by the multi-stage process described above. This process was essentially recapitulated with the ovarian carcinoma cell line IGROV-1. Our observations suggest that transitory cells characterized by polyploidy, features of stemness and a pro-inflammatory secretory phenotype contribute to the acquisition of chemoresistance.
- Published
- 2015
- Full Text
- View/download PDF
28. 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, Nockher WA, Toth PM, Diederich WE, Nist A, Stiewe T, Wagner U, Reinartz S, Müller-Brüsselbach S, and Müller R
- Subjects
- Animals, Case-Control Studies, Fatty Acids, Female, Humans, Ligands, Linoleic Acid blood, Macrophages metabolism, Mice, Neoplasm Recurrence, Local blood, Neoplasm Recurrence, Local genetics, PPAR delta blood, PPAR-beta blood, Linoleic Acid genetics, Macrophages pathology, Ovarian Neoplasms blood, Ovarian Neoplasms genetics, PPAR delta genetics, PPAR-beta genetics, Tumor Microenvironment genetics
- Abstract
The nuclear receptor peroxisome proliferator-activated receptor β/δ (PPARβ/δ) is a lipid ligand-inducible transcription factor associated with macrophage polarization. However, its function in tumor-associated macrophages (TAMs) has not been investigated to date. Here, we report the PPARβ/δ-regulated transcriptome and cistrome for TAMs from ovarian carcinoma patients. Comparison with monocyte-derived macrophages shows that the vast majority of direct PPARβ/δ target genes are upregulated in TAMs and largely refractory to synthetic agonists, but repressible by inverse agonists. Besides genes with metabolic functions, these include cell type-selective genes associated with immune regulation and tumor progression, e.g., LRP5, CD300A, MAP3K8 and ANGPTL4. This deregulation is not due to increased expression of PPARβ/δ or its enhanced recruitment to target genes. Instead, lipidomic analysis of malignancy-associated ascites revealed high concentrations of polyunsaturated fatty acids, in particular linoleic acid, acting as potent PPARβ/δ agonists in macrophages. These fatty acid ligands accumulate in lipid droplets in TAMs, thereby providing a reservoir of PPARβ/δ ligands. These observations suggest that the deregulation of PPARβ/δ target genes by ligands of the tumor microenvironment contributes to the pro-tumorigenic polarization of ovarian carcinoma TAMs. This conclusion is supported by the association of high ANGPTL4 expression with a shorter relapse-free survival in serous ovarian carcinoma.
- Published
- 2015
- Full Text
- View/download PDF
29. The transcriptional PPARβ/δ network in human macrophages defines a unique agonist-induced activation state.
- Author
-
Adhikary T, Wortmann A, Schumann T, Finkernagel F, Lieber S, Roth K, Toth PM, Diederich WE, Nist A, Stiewe T, Kleinesudeik L, Reinartz S, Müller-Brüsselbach S, and Müller R
- Subjects
- Cell Differentiation, Cell Line, Cells, Cultured, Gene Expression Regulation, Humans, Macrophages metabolism, Monocytes cytology, Monocytes metabolism, PPAR delta agonists, PPAR-beta agonists, Transcriptome, Gene Regulatory Networks, Macrophage Activation, Macrophages immunology, PPAR delta metabolism, PPAR-beta metabolism
- Abstract
Peroxisome proliferator-activated receptor β/δ (PPARβ/δ) is a lipid ligand-inducible transcription factor with established metabolic functions, whereas its anti-inflammatory function is poorly understood. To address this issue, we determined the global PPARβ/δ-regulated signaling network in human monocyte-derived macrophages. Besides cell type-independent, canonical target genes with metabolic and immune regulatory functions we identified a large number of inflammation-associated NFκB and STAT1 target genes that are repressed by agonists. Accordingly, PPARβ/δ agonists inhibited the expression of multiple pro-inflammatory mediators and induced an anti-inflammatory, IL-4-like morphological phenotype. Surprisingly, bioinformatic analyses also identified immune stimulatory effects. Consistent with this prediction, PPARβ/δ agonists enhanced macrophage survival under hypoxic stress and stimulated CD8(+) T cell activation, concomitantly with the repression of immune suppressive target genes and their encoded products CD274 (PD-1 ligand), CD32B (inhibitory Fcγ receptor IIB) and indoleamine 2,3-dioxygenase 1 (IDO-1), as well as a diminished release of the immune suppressive IDO-1 metabolite kynurenine. Comparison with published data revealed a significant overlap of the PPARβ/δ transcriptome with coexpression modules characteristic of both anti-inflammatory and pro-inflammatory cytokines. Our findings indicate that PPARβ/δ agonists induce a unique macrophage activation state with strong anti-inflammatory but also specific immune stimulatory components, pointing to a context-dependent function of PPARβ/δ in immune regulation., (© The Author(s) 2015. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2015
- Full Text
- View/download PDF
30. The inverse agonist DG172 triggers a PPARβ/δ-independent myeloid lineage shift and promotes GM-CSF/IL-4-induced dendritic cell differentiation.
- Author
-
Lieber S, Scheer F, Finkernagel F, Meissner W, Giehl G, Brendel C, Diederich WE, Müller-Brüsselbach S, and Müller R
- Subjects
- Acrylonitrile pharmacology, Animals, Bone Marrow Cells drug effects, Bone Marrow Cells metabolism, Cell Differentiation drug effects, Cell Differentiation physiology, Cell Lineage, Cells, Cultured, Dendritic Cells metabolism, Drug Inverse Agonism, Drug Synergism, Mice, Mice, Inbred C57BL, Mice, Knockout, PPAR gamma metabolism, PPAR-beta metabolism, Acrylonitrile analogs & derivatives, Dendritic Cells drug effects, Granulocyte-Macrophage Colony-Stimulating Factor pharmacology, Interleukin-4 pharmacology, PPAR gamma agonists, PPAR-beta agonists, Piperazines pharmacology
- Abstract
The stilbene derivative (Z)-2-(2-bromophenyl)-3-{[4-(1-methylpiperazine)amino]phenyl}acrylonitrile (DG172) was developed as a highly selective inhibitory peroxisome proliferator-activated receptor (PPAR)β/δ ligand. Here, we describe a novel PPARβ/δ-independent, yet highly specific, effect of DG172 on the differentiation of bone marrow cells (BMCs). DG172 strongly augmented granulocyte-macrophage-colony-stimulating factor (GM-CSF)-induced differentiation of primary BMCs from Ppard null mice into two specific populations, characterized as mature (CD11c(hi)MHCII(hi)) and immature (CD11c(hi)MHCII(lo)) dendritic cells (DCs). IL-4 synergized with DG172 to shift the differentiation from MHCII(lo) cells to mature DCs in vitro. The promotion of DC differentiation occurred at the expense of differentiation to granulocytic Gr1(+)Ly6B(+) cells. In agreement with these findings, transcriptome analyses showed a strong DG172-mediated repression of genes encoding neutrophilic markers in both differentiating wild-type and Ppard null cells, while macrophage/DC marker genes were up-regulated. DG172 also inhibited the expression of transcription factors driving granulocytic differentiation (Cebpe, Gfi1, and Klf5), and increased the levels of transcription factors promoting macrophage/DC differentiation (Irf4, Irf8, Spib, and Spic). DG172 exerted these effects only at an early stage of BMC differentiation induced by GM-CSF, did not affect macrophage-colony-stimulating factor-triggered differentiation to macrophages and had no detectable PPARβ/δ-independent effect on other cell types tested. Structure-function analyses demonstrated that the 4-methylpiperazine moiety in DG172 is required for its effect on DC differentiation, but is dispensable for PPARβ/δ binding. Based on these data we developed a new compound, (Z)-2-(4-chlorophenyl)-3-[4-(4-methylpiperazine-1-yl)phenyl]acrylonitrile (DG228), which enhances DC differentiation in the absence of significant PPARβ/δ binding., (Copyright © 2014 by The American Society for Pharmacology and Experimental Therapeutics.)
- Published
- 2015
- Full Text
- View/download PDF
31. Mixed-polarization phenotype of ascites-associated macrophages in human ovarian carcinoma: correlation of CD163 expression, cytokine levels and early relapse.
- Author
-
Reinartz S, Schumann T, Finkernagel F, Wortmann A, Jansen JM, Meissner W, Krause M, Schwörer AM, Wagner U, Müller-Brüsselbach S, and Müller R
- Subjects
- Antigens, CD biosynthesis, Antigens, CD immunology, Antigens, Differentiation, Myelomonocytic biosynthesis, Antigens, Differentiation, Myelomonocytic immunology, Cell Polarity, Cystadenocarcinoma, Serous metabolism, Cystadenocarcinoma, Serous pathology, Cytokines biosynthesis, Cytokines immunology, Disease-Free Survival, Enzyme-Linked Immunosorbent Assay, Female, Flow Cytometry, Humans, Kaplan-Meier Estimate, Macrophages metabolism, Macrophages pathology, Neoplasm Recurrence, Local immunology, Neoplasm Recurrence, Local metabolism, Neoplasm Recurrence, Local pathology, Oligonucleotide Array Sequence Analysis, Ovarian Neoplasms metabolism, Ovarian Neoplasms pathology, Phenotype, Receptors, Cell Surface biosynthesis, Receptors, Cell Surface immunology, Transcriptome, Ascites immunology, Cystadenocarcinoma, Serous immunology, Macrophages immunology, Ovarian Neoplasms immunology
- Abstract
Ovarian cancer is typically accompanied by the occurrence of malignant ascites containing large number of macrophages. It has been suggested that these tumor-associated macrophages (TAMs) are skewed to alternative polarization (M2) and thereby play an essential role in therapy resistance and metastatic spread. In our study, we have investigated the nature, regulation and clinical correlations of TAM polarization in serous ovarian cancer. Macrophage polarization markers on TAMs and ascites cytokine levels were analyzed for 30 patients and associated with relapse-free survival (RFS) in a prospective study with 20 evaluable patients. Surface expression of the M2 marker CD163 on TAMs was inversely associated with RFS (p < 0.01). However, global gene expression profiles determined for 17 of these patients revealed a mixed-polarization phenotype unrelated to the M1/M2 classification. CD163 surface expression also correlated with the ascites levels of IL-6 and IL-10 (p < 0.05), both cytokines induced CD163 expression, and their ascites levels showed a clear inverse association with RFS (p < 0.01). These findings define a subgroup of patients with high CD163 expression, high IL-6 and/or IL-10 levels and poor clinical outcome., (© 2013 The Authors. Published by Wiley Periodicals, Inc. on behalf of UICC.)
- Published
- 2014
- Full Text
- View/download PDF
32. Regulation of TAK1/TAB1-mediated IL-1β signaling by cytoplasmic PPARβ/δ.
- Author
-
Stockert J, Wolf A, Kaddatz K, Schnitzer E, Finkernagel F, Meissner W, Müller-Brüsselbach S, Kracht M, and Müller R
- Subjects
- Cytoplasm metabolism, Gene Expression Profiling, Gene Expression Regulation, HSP27 Heat-Shock Proteins metabolism, HeLa Cells, Humans, Interleukin-1beta pharmacology, MAP Kinase Kinase Kinases chemistry, Protein Binding, Protein Interaction Domains and Motifs, Transcription Factor RelA metabolism, Transcription, Genetic drug effects, Adaptor Proteins, Signal Transducing metabolism, Interleukin-1beta metabolism, MAP Kinase Kinase Kinases metabolism, PPAR delta metabolism, PPAR-beta metabolism, Signal Transduction
- Abstract
The peroxisome proliferator-activated receptor subtypes PPARα, PPARβ/δ, PPARγ are members of the steroid hormone receptor superfamily with well-established functions in transcriptional regulation. Here, we describe an unexpected cytoplasmic function of PPARβ/δ. Silencing of PPARβ/δ expression interferes with the expression of a large subset of interleukin-1β (IL-1β)-induced target genes in HeLa cells, which is preceded by an inhibition of the IL-1β-induced phosphorylation of TAK1 and its downstream effectors, including the NFκBα inhibitor IκBα (NFKBIA) and the NFκBα subunit p65 (RELA). PPARβ/δ enhances the interaction between TAK1 and the small heat-shock protein HSP27, a known positive modulator of TAK1-mediated IL-1β signaling. Consistent with these findings, PPARβ/δ physically interacts with both the endogenous cytoplasmic TAK1/TAB1 complex and HSP27, and PPARβ/δ overexpression increases the TAK1-induced transcriptional activity of NFκB. These observations suggest that PPARβ/δ plays a role in the assembly of a cytoplasmic multi-protein complex containing TAK1, TAB1, HSP27 and PPARβ/δ, and thereby participates in the NFκB response to IL-1β.
- Published
- 2013
- Full Text
- View/download PDF
33. (Z)-2-(2-bromophenyl)-3-{[4-(1-methyl-piperazine)amino]phenyl}acrylonitrile (DG172): an orally bioavailable PPARβ/δ-selective ligand with inverse agonistic properties.
- Author
-
Lieber S, Scheer F, Meissner W, Naruhn S, Adhikary T, Müller-Brüsselbach S, Diederich WE, and Müller R
- Subjects
- Acrylonitrile chemical synthesis, Acrylonitrile pharmacokinetics, Acrylonitrile pharmacology, Administration, Oral, Animals, Binding, Competitive, Biological Availability, Chromatin metabolism, Co-Repressor Proteins metabolism, Drug Inverse Agonism, Fluorescence Resonance Energy Transfer, High-Throughput Screening Assays, Humans, Ligands, Male, Mice, Myoblasts drug effects, Myoblasts metabolism, Piperazines pharmacokinetics, Piperazines pharmacology, Small Molecule Libraries, Stereoisomerism, Structure-Activity Relationship, Transcription, Genetic drug effects, Acrylonitrile analogs & derivatives, PPAR gamma antagonists & inhibitors, PPAR-beta antagonists & inhibitors, Piperazines chemical synthesis
- Abstract
The ligand-regulated nuclear receptor peroxisome proliferator-activated receptor β/δ (PPARβ/δ) is a potential pharmacological target due to its role in disease-related biological processes. We used TR-FRET-based competitive ligand binding and coregulator interaction assays to screen 2693 compounds of the Open Chemical Repository of the NCI/NIH Developmental Therapeutics Program for inhibitory PPARβ/δ ligands. One compound, (Z)-3-(4-dimethylamino-phenyl)-2-phenyl-acrylonitrile, was used for a systematic SAR study. This led to the design of derivative 37, (Z)-2-(2-bromophenyl)-3-{[4-(1-methyl-piperazine)amino]phenyl}acrylonitrile (DG172), a novel PPARβ/δ-selective ligand showing high binding affinity (IC(50) = 27 nM) and potent inverse agonistic properties. 37 selectively inhibited the agonist-induced activity of PPARβ/δ, enhanced transcriptional corepressor recruitment, and down-regulated transcription of the PPARβ/δ target gene Angptl4 in mouse myoblasts (IC(50) = 9.5 nM). Importantly, 37 was bioavailable after oral application to mice with peak plasma levels in the concentration range of its maximal inhibitory potency, suggesting that 37 will be an invaluable tool to elucidate the functions and therapeutic potential of PPARβ/δ.
- Published
- 2012
- Full Text
- View/download PDF
34. High-affinity peroxisome proliferator-activated receptor β/δ-specific ligands with pure antagonistic or inverse agonistic properties.
- Author
-
Naruhn S, Toth PM, Adhikary T, Kaddatz K, Pape V, Dörr S, Klebe G, Müller-Brüsselbach S, Diederich WE, and Müller R
- Subjects
- Animals, Base Sequence, Binding Sites, Cell Line, Chromatin Immunoprecipitation, DNA Primers, Fluorescence Resonance Energy Transfer, Humans, Ligands, Mice, PPAR delta antagonists & inhibitors, PPAR-beta antagonists & inhibitors, Real-Time Polymerase Chain Reaction, PPAR delta metabolism, PPAR-beta metabolism, Sulfonamides metabolism, Thiophenes metabolism
- Abstract
Peroxisome proliferator-activated receptor β/δ (PPARβ/δ) is a ligand-regulated nuclear receptor with essential functions in metabolism and inflammation. We have synthesized a new derivative [methyl 3-(N-(4-(hexylamino)-2-methoxyphenyl)sulfamoyl)thiophene-2-carboxylate (ST247) structurally related to the published PPARβ/δ inhibitory ligand methyl 3-(N-(2-methoxy-4-(phenylamino)phenyl)sulfamoyl)thiophene-2-carboxylate (GSK0660). ST247 has a higher affinity to PPARβ/δ than GSK0660, and at equimolar concentrations, it more efficiently 1) induces the interaction with corepressors both in vitro and in vivo, 2) inhibits the agonist-induced transcriptional activity of PPARβ/δ, and 3) down-regulates basal level expression of the peroxisome proliferator responsive element-driven PPARβ/δ target gene ANGPTL4. Methyl 3-(N-(4-(tert-butylamino)-2-methoxyphenyl)sulfamoyl)thiophene-2-carboxylate (PT-S58), another high-affinity derivative from our series, also efficiently inhibits agonist-induced transcriptional activation, but in contrast to ST247, it does not enhance the interaction of PPARβ/δ with corepressors. PT-S58 rather prevents corepressor recruitment triggered by the inverse agonist ST247. These findings classify ST247 as an inverse agonist, whereas PT-S58 is the first pure PPARβ/δ antagonist described to date. It is noteworthy that ST247 and PT-S58 are also effective on PPRE-independent functions of PPARβ/δ: in monocytic cells, both ligands modulate expression of the activation marker CCL2 in the opposite direction as an established PPARβ/δ agonist. The possibility to differentially modulate specific functions of PPARβ/δ makes these novel compounds invaluable tools to advance our understanding of PPARβ/δ biology.
- Published
- 2011
- Full Text
- View/download PDF
35. Genomewide analyses define different modes of transcriptional regulation by peroxisome proliferator-activated receptor-β/δ (PPARβ/δ).
- Author
-
Adhikary T, Kaddatz K, Finkernagel F, Schönbauer A, Meissner W, Scharfe M, Jarek M, Blöcker H, Müller-Brüsselbach S, and Müller R
- Subjects
- Cell Line, Gene Regulatory Networks, Genome-Wide Association Study, Humans, PPAR delta genetics, PPAR-beta genetics, Response Elements, Gene Expression Regulation, Genome, Human genetics, Peroxisome Proliferator-Activated Receptors genetics, Transcription, Genetic
- Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors with essential functions in lipid, glucose and energy homeostasis, cell differentiation, inflammation and metabolic disorders, and represent important drug targets. PPARs heterodimerize with retinoid X receptors (RXRs) and can form transcriptional activator or repressor complexes at specific DNA elements (PPREs). It is believed that the decision between repression and activation is generally governed by a ligand-mediated switch. We have performed genomewide analyses of agonist-treated and PPARβ/δ-depleted human myofibroblasts to test this hypothesis and to identify global principles of PPARβ/δ-mediated gene regulation. Chromatin immunoprecipitation sequencing (ChIP-Seq) of PPARβ/δ, H3K4me3 and RNA polymerase II enrichment sites combined with transcriptional profiling enabled the definition of 112 bona fide PPARβ/δ target genes showing either of three distinct types of transcriptional response: (I) ligand-independent repression by PPARβ/δ; (II) ligand-induced activation and/or derepression by PPARβ/δ; and (III) ligand-independent activation by PPARβ/δ. These data identify PPRE-mediated repression as a major mechanism of transcriptional regulation by PPARβ/δ, but, unexpectedly, also show that only a subset of repressed genes are activated by a ligand-mediated switch. Our results also suggest that the type of transcriptional response by a given target gene is connected to the structure of its associated PPRE(s) and the biological function of its encoded protein. These observations have important implications for understanding the regulatory PPAR network and PPARβ/δ ligand-based drugs.
- Published
- 2011
- Full Text
- View/download PDF
36. Reverse crosstalk of TGFβ and PPARβ/δ signaling identified by transcriptional profiling.
- Author
-
Stockert J, Adhikary T, Kaddatz K, Finkernagel F, Meissner W, Müller-Brüsselbach S, and Müller R
- Subjects
- Cell Differentiation, Cell Line, Co-Repressor Proteins metabolism, Fibroblasts cytology, Fibroblasts drug effects, Fibroblasts metabolism, Gene Expression Profiling, Humans, Response Elements, Thiazoles pharmacology, Gene Expression Regulation drug effects, PPAR delta agonists, PPAR-beta agonists, Signal Transduction, Transcription, Genetic drug effects, Transforming Growth Factor beta pharmacology
- Abstract
Previous work has provided strong evidence for a role of peroxisome proliferator-activated receptor β/δ (PPARβ/δ) and transforming growth factor-β (TGFβ) in inflammation and tumor stroma function, raising the possibility that both signaling pathways are interconnected. We have addressed this hypothesis by microarray analyses of human diploid fibroblasts induced to myofibroblastic differentiation, which revealed a substantial, mostly reverse crosstalk of both pathways and identified distinct classes of genes. A major class encompasses classical PPAR target genes, including ANGPTL4, CPT1A, ADRP and PDK4. These genes are repressed by TGFβ, which is counteracted by PPARβ/δ activation. This is mediated, at least in part, by the TGFβ-induced recruitment of the corepressor SMRT to PPAR response elements, and its release by PPARβ/δ ligands, indicating that TGFβ and PPARβ/δ signals are integrated by chromatin-associated complexes. A second class represents TGFβ-induced genes that are downregulated by PPARβ/δ agonists, exemplified by CD274 and IL6, which is consistent with the anti-inflammatory properties of PPARβ/δ ligands. Finally, cooperative regulation by both ligands was observed for a minor group of genes, including several regulators of cell proliferation. These observations indicate that PPARβ/δ is able to influence the expression of distinct sets of both TGFβ-repressed and TGFβ-activated genes in both directions.
- Published
- 2011
- Full Text
- View/download PDF
37. Transcriptional profiling identifies functional interactions of TGF β and PPAR β/δ signaling: synergistic induction of ANGPTL4 transcription.
- Author
-
Kaddatz K, Adhikary T, Finkernagel F, Meissner W, Müller-Brüsselbach S, and Müller R
- Subjects
- Angiopoietin-Like Protein 4, Animals, Cell Line, Chromatin Immunoprecipitation, Computational Biology, Electrophoretic Mobility Shift Assay, Enhancer Elements, Genetic genetics, Gene Expression Profiling, Humans, Mice, NIH 3T3 Cells, Oligonucleotide Array Sequence Analysis, PPAR delta agonists, PPAR-beta agonists, Protein Binding drug effects, Protein Binding genetics, RNA, Small Interfering, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction, Smad3 Protein genetics, Smad3 Protein metabolism, Thiazoles pharmacology, Transforming Growth Factor beta genetics, Angiopoietins genetics, PPAR delta metabolism, PPAR-beta metabolism, Transforming Growth Factor beta metabolism
- Abstract
Peroxisome proliferator-activated receptors (PPARs) not only play a key role in regulating metabolic pathways but also modulate inflammatory processes, pointing to a functional interaction between PPAR and cytokine signaling pathways. In this study, we show by genome-wide transcriptional profiling that PPARβ/δ and transforming growth factor-β (TGFβ) pathways functionally interact in human myofibroblasts and that a subset of these genes is cooperatively activated by TGFβ and PPARβ/δ. Using the angiopoietin-like 4 (ANGPTL4) gene as a model, we demonstrate that two enhancer regions cooperate to mediate the observed synergistic response. A TGFβ-responsive enhancer located ∼8 kb upstream of the transcriptional start site is regulated by a mechanism involving SMAD3, ETS1, RUNX, and AP-1 transcription factors that interact with multiple contiguous binding sites. A second enhancer (PPAR-E) consisting of three juxtaposed PPAR response elements is located in the third intron ∼3.5 kb downstream of the transcriptional start site. The PPAR-E is strongly activated by all three PPAR subtypes, with a novel type of PPAR response element motif playing a central role. Although the PPAR-E is not regulated by TGFβ, it interacts with SMAD3, ETS1, RUNX2, and AP-1 in vivo, providing a possible mechanistic explanation for the observed synergism.
- Published
- 2010
- Full Text
- View/download PDF
38. 15-hydroxyeicosatetraenoic acid is a preferential peroxisome proliferator-activated receptor beta/delta agonist.
- Author
-
Naruhn S, Meissner W, Adhikary T, Kaddatz K, Klein T, Watzer B, Müller-Brüsselbach S, and Müller R
- Subjects
- Amino Acid Sequence, Animals, CHO Cells, Cricetinae, Cricetulus, Fibroblasts chemistry, Fibroblasts metabolism, Humans, Hydroxyeicosatetraenoic Acids biosynthesis, Hydroxyeicosatetraenoic Acids metabolism, Mice, Mice, Knockout, Molecular Sequence Data, NIH 3T3 Cells, PPAR delta genetics, PPAR delta physiology, PPAR-beta genetics, PPAR-beta physiology, Trans-Activators biosynthesis, Trans-Activators genetics, Trans-Activators physiology, Hydroxyeicosatetraenoic Acids pharmacology, PPAR delta agonists, PPAR-beta agonists
- Abstract
Peroxisome proliferator-activated receptor (PPARs) modulate target gene expression in response to unsaturated fatty acid ligands, such as arachidonic acid (AA). Here, we report that the AA metabolite 15-hydroxyeicosatetraenoic acid (15-HETE) activates the ligand-dependent activation domain (AF2) of PPARbeta/delta in vivo, competes with synthetic agonists in a PPARbeta/delta ligand binding assay in vitro, and triggers the interaction of PPARbeta/delta with coactivator peptides. These agonistic effects were also seen with PPARalpha and PPARgamma, but to a significantly weaker extent. We further show that 15-HETE strongly induces the expression of the bona fide PPAR target gene Angptl4 in a PPARbeta/delta-dependent manner and, conversely, that inhibition of 15-HETE synthesis reduces PPARbeta/delta transcriptional activity. Consistent with its function as an agonistic ligand, 15-HETE triggers profound changes in chromatin-associated PPARbeta/delta complexes in vivo, including the recruitment of the coactivator cAMP response element-binding protein binding protein. Both 15R-HETE and 15S-HETE are similarly potent at inducing PPARbeta/delta coactivator binding and transcriptional activation, indicating that 15-HETE enantiomers generated by different pathways function as PPARbeta/delta agonists.
- Published
- 2010
- Full Text
- View/download PDF
39. The rotamase Pin1 is up-regulated, hypophosphorylated and required for cell cycle progression in head and neck squamous cell carcinomas.
- Author
-
Wiegand S, Dakic B, Rath AF, Makarova G, Sterz C, Meissner W, Bette M, Adamkiewicz J, Müller-Brüsselbach S, Müller R, Werner JA, and Mandic R
- Subjects
- Adult, Aged, Aged, 80 and over, Alkaline Phosphatase pharmacology, Apoptosis, Blotting, Western, Case-Control Studies, Cell Cycle physiology, Cell Proliferation, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Electrophoresis, Gel, Two-Dimensional, Enzyme Inhibitors pharmacology, Female, Humans, Male, Middle Aged, NIMA-Interacting Peptidylprolyl Isomerase, Naphthoquinones pharmacology, Peptidylprolyl Isomerase antagonists & inhibitors, Phosphorylation, RNA, Small Interfering pharmacology, Up-Regulation, Carcinoma, Squamous Cell metabolism, Head and Neck Neoplasms metabolism, Neoplasm Proteins metabolism, Peptidylprolyl Isomerase metabolism
- Abstract
The peptidyl-prolyl cis/trans isomerase Pin1 has been implicated in malignant transformation in multiple studies, however, little is known about its potential impact in head and neck cancer. This study evaluates the role of Pin1 in head and neck squamous cell carcinomas (HNSCCs). Pin1 expression and level of phosphorylation was evaluated by Western blot analysis and 2D-gel-electrophoresis. Pin1 was inhibited with juglone (5-hydroxy-1,4-naphthalenedione) or Pin1 specific siRNA and its influence on cell cycle checkpoint distribution was assessed by FACS analysis. Pin1 overexpression was found in HNSCC tissues and cell lines. 2D-gel-electrophoresis data pointed to Pin1 being hypophosphorylated in HNSCC cells which is consistent with overactivation of this rotamase. Inhibition of HNSCC cells with juglone or Pin1 siRNA induced the cell cycle inhibitor p21(WAF1/Cip1) with a concomitant reduction of cells in G2/M and an increased fraction of cells with fragmented DNA. Cell death did not correlate with significant levels of apoptosis in Pin1 depleted HNSCC cells. In summary, the data shows that Pin1 is overexpressed and hypophosphorylated in HNSCC, and that inhibition of Pin1 blocks cell cycle progression and triggers tumor cell death. Pin1 therefore could represent a new target for the development of improved HNSCC targeting drugs.
- Published
- 2009
- Full Text
- View/download PDF
40. Ligand-mediated regulation of peroxisome proliferator-activated receptor (PPAR) beta/delta: a comparative analysis of PPAR-selective agonists and all-trans retinoic acid.
- Author
-
Rieck M, Meissner W, Ries S, Müller-Brüsselbach S, and Müller R
- Subjects
- Animals, Fluorescence Resonance Energy Transfer, Gene Expression Regulation drug effects, Ligands, Mice, NIH 3T3 Cells, PPAR delta agonists, PPAR delta physiology, PPAR-beta agonists, PPAR-beta physiology, Transcription, Genetic physiology, PPAR delta metabolism, PPAR-beta metabolism, Tretinoin pharmacology
- Abstract
Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear hormone receptor superfamily that modulate target gene expression in response to natural fatty acid ligands and synthetic agonists. It is noteworthy that all trans-retinoic acid (atRA) has recently been reported to act as a ligand for PPARbeta/delta, to activate its transcriptional activity, and, in contrast to the "classic" function of atRA, to stimulate cell proliferation (Schug et al., 2007). Here, we report that in contrast to synthetic PPARbeta/delta agonists, atRA failed to induce the transcriptional activity of PPARbeta/delta using different types of reporter gene assays. Likewise, atRA did not affect the expression of the bona fide PPARbeta/delta target genes ADRP and ANGPTL4 but strongly increased expression of the retinoic acid target gene CYP26A under the identical experimental conditions. Consistent with these observations, atRA did not compete with established PPARbeta/delta agonists in a ligand binding assay, and atRA did not enable the interaction of PPARbeta/delta with a coactivator peptide in a time-resolved fluorescence resonance energy transfer assay in vitro. These results are in sharp contrast to the effect of established PPARbeta/delta agonists in both in vitro assays. Taken as a whole, these data strongly suggest that atRA does not function as a ligand of PPARbeta/delta in any of the experimental systems tested and that the previously reported atRA effects are more likely to reflect an uncharacterized and less direct mechanism.
- Published
- 2008
- Full Text
- View/download PDF
41. Regulation of Cell Proliferation and Differentiation by PPARbeta/delta.
- Author
-
Müller R, Rieck M, and Müller-Brüsselbach S
- Abstract
Peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) is a ligand-activated transcription factor with essential functions in the regulation of lipid catabolism, glucose homeostasis, and inflammation, which makes it a potentially relevant drug target for the treatment of major human diseases. In addition, there is strong evidence that PPARbeta/delta modulates oncogenic signaling pathways and tumor growth. Consistent with these observations, numerous reports have clearly documented a role for PPARbeta/delta in cell cycle control, differentiation, and apoptosis. However, the precise role of PPARbeta/delta in tumorigenesis and cell proliferation remains controversial. This review summarizes our current knowledge and proposes a model corroborating the discrepant data in this area of research.
- Published
- 2008
- Full Text
- View/download PDF
42. A Role for PPARbeta/delta in Tumor Stroma and Tumorigenesis.
- Author
-
Müller R, Kömhoff M, Peters JM, and Müller-Brüsselbach S
- Abstract
Peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) is a transcription factor that is activated by endogenous fatty acid ligands and by synthetic agonists. Its role in the regulation of skeletal muscle fatty acid catabolism, glucose homeostasis, and cellular differentiation has been established in multiple studies. On the contrary, a role for PPARbeta/delta in tumorigenesis is less clear because there are contradictory reports in the literature. However, the majority of these studies have not examined the role of PPARbeta/delta in the tumor stroma. Recent evidence suggests that stromal PPARbeta/delta regulates tumor endothelial cell proliferation and promotes differentiation leading to the properly orchestrated events required for tumor blood vessel formation. This review briefly summarizes the significance of these studies that may provide clues to help explain the reported discrepancies in the literature regarding the role of PPARbeta/delta in tumorigenesis.
- Published
- 2008
- Full Text
- View/download PDF
43. Expression level and agonist-binding affect the turnover, ubiquitination and complex formation of peroxisome proliferator activated receptor beta.
- Author
-
Rieck M, Wedeken L, Müller-Brüsselbach S, Meissner W, and Müller R
- Subjects
- Animals, Cell Line, Chromatography, Gel, Humans, Immunoprecipitation, Ligands, Mice, PPAR-beta agonists, Thiazoles pharmacology, PPAR-beta metabolism, Ubiquitin metabolism
- Abstract
Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear hormone receptor superfamily that modulate target gene expression in response to fatty acid ligands. Their regulation by post-translational modifications has been reported but is poorly understood. In the present study, we investigated whether ligand binding affects the turnover and ubiquitination of the PPARbeta subtype (also known as PPARdelta). Our data show that the ubiquitination and degradation of PPARbeta is not significantly influenced by the synthetic agonist GW501516 under conditions of moderate PPARbeta expression. By contrast, the overexpression of PPARbeta dramatically enhanced its degradation concomitant with its polyubiquitination and the formation of high molecular mass complexes containing multiple, presumably oligomerized PPARbeta molecules that lacked stoichiometical amounts of the obligatory PPARbeta dimerization partner, retinoid X receptor. The formation of these apparently aberrant complexes, as well as the ubiquitination and destabilization of PPARbeta, were strongly inhibited by GW501516. Our findings suggest that PPARbeta is subject to complex post-translational regulatory mechanisms that partly may serve to safeguard the cell against deregulated PPARbeta expression. Furthermore, our data have important implications regarding the widespread use of overexpression systems to evaluate the function and regulation of PPARs.
- Published
- 2007
- Full Text
- View/download PDF
44. Growth of transgenic RAF-induced lung adenomas is increased in mice with a disrupted PPARbeta/delta gene.
- Author
-
Müller-Brüsselbach S, Ebrahimsade S, Jäkel J, Eckhardt J, Rapp UR, Peters JM, Moll R, and Müller R
- Subjects
- Adenoma pathology, Animals, Genotype, Hypoglycemic Agents pharmacology, Lung Neoplasms metabolism, Lung Neoplasms pathology, Mice, Mice, Inbred C57BL, Mice, Transgenic, PPAR delta physiology, PPAR-beta physiology, Proto-Oncogene Proteins c-raf physiology, Rosiglitazone, Thiazolidinediones pharmacology, Transcription Factors metabolism, Transgenes, Adenoma genetics, Gene Expression Regulation, Neoplastic, Lung Neoplasms genetics, PPAR delta genetics, PPAR-beta genetics, Proto-Oncogene Proteins c-raf genetics
- Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors with essential functions in regulating lipid metabolism. Both the PPARbeta (also referred to as PPARdelta) and PPARgamma subtype have been reported to either attenuate or potentiate tumorigenesis in a number of different models of intestinal and skin carcinogenesis. In the present study, we have addressed the role of PPARbeta and PPARgamma in lung tumorigenesis in a transgenic mouse model of RAF-induced lung adenoma using two different strategies: i) crossing with PPARbeta null mice, and ii) chronic treatment with the PPARgamma agonist rosiglitazone. Histological examination revealed a significant enhancement of tumor growth in mice lacking one or both alleles of Pparb, but no significant effect in response to rosiglitazone. These observations indicate i) that RAF-induced lung tumorigenesis is attenuated in mice with a disrupted Pparb gene, and ii) that chronic PPARgamma activation does not affect lung adenoma growth. These results are relevant with respect to the clinical application of drugs modulating the activity of PPARbeta or PPARgamma.
- Published
- 2007
45. Deregulation of tumor angiogenesis and blockade of tumor growth in PPARbeta-deficient mice.
- Author
-
Müller-Brüsselbach S, Kömhoff M, Rieck M, Meissner W, Kaddatz K, Adamkiewicz J, Keil B, Klose KJ, Moll R, Burdick AD, Peters JM, and Müller R
- Subjects
- Animals, Cell Proliferation, Cyclin-Dependent Kinase Inhibitor p57 metabolism, Endothelium, Vascular pathology, Magnetic Resonance Imaging, Mice, Mice, Knockout, Neoplasms, Experimental pathology, Oligonucleotide Array Sequence Analysis, PPAR-beta genetics, PPAR-beta metabolism, Down-Regulation, Neoplasms, Experimental blood supply, Neovascularization, Pathologic, PPAR-beta physiology
- Abstract
The peroxisome proliferator-activated receptor-beta (PPARbeta) has been implicated in tumorigenesis, but its precise role remains unclear. Here, we show that the growth of syngeneic Pparb wild-type tumors is impaired in Pparb(-/-) mice, concomitant with a diminished blood flow and an abundance of hyperplastic microvascular structures. Matrigel plugs containing pro-angiogenic growth factors harbor increased numbers of morphologically immature, proliferating endothelial cells in Pparb(-/-) mice, and retroviral transduction of Pparb triggers microvessel maturation. We have identified the Cdkn1c gene encoding the cell cycle inhibitor p57(Kip2) as a PPARbeta target gene and a mediator of the PPARbeta-mediated inhibition of cell proliferation, which provides a possible mechanistic explanation for the observed tumor endothelial hyperplasia and deregulation of tumor angiogenesis in Pparb(-/-) mice. Our data point to an unexpected essential role for PPARbeta in constraining tumor endothelial cell proliferation to allow for the formation of functional tumor microvessels.
- Published
- 2007
- Full Text
- View/download PDF
46. Specific components of prostanoid-signaling pathways are present in non-small cell lung cancer cells.
- Author
-
Kreutzer M, Fauti T, Kaddatz K, Seifart C, Neubauer A, Schweer H, Kömhoff M, Müller-Brüsselbach S, and Müller R
- Subjects
- Animals, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung pathology, Cell Line, Tumor, Cyclooxygenase 2 metabolism, Dinoprostone metabolism, Epoprostenol metabolism, Gas Chromatography-Mass Spectrometry methods, Humans, Immunoblotting, Lung Neoplasms genetics, Lung Neoplasms metabolism, Lung Neoplasms pathology, Mass Spectrometry methods, Mice, NIH 3T3 Cells, RNA, Messenger genetics, RNA, Messenger metabolism, Receptors, Prostaglandin E metabolism, Receptors, Prostaglandin E, EP1 Subtype, Receptors, Prostaglandin E, EP2 Subtype, Receptors, Prostaglandin E, EP4 Subtype, Receptors, Thromboxane A2, Prostaglandin H2 genetics, Receptors, Thromboxane A2, Prostaglandin H2 metabolism, Reverse Transcriptase Polymerase Chain Reaction, Thromboxane A2 metabolism, Cyclooxygenase 2 genetics, Prostaglandins metabolism, Receptors, Prostaglandin E genetics, Signal Transduction
- Abstract
In the present study, we measured prostanoid synthesis and the expression of genes associated with prostanoid signaling in human non-small cell lung carcinoma (NSCLC) cell lines and in primary human tumors. Consistent with the proposed growth promoting role of PGE2, we found that NSCLC cell lines frequently co-expressed the genes encoding cyclooxygenase-2 and the prostaglandin E2 (PGE2) receptors EP1, 2 and 4 concomitant with the synthesis of PGE2. In contrast, NSCLC cells did not synthesize appreciable amounts of prostaglandin I2 (PGI2, prostacyclin), lacked PGI2 synthase (PGIS) and did not express the gene coding for the PGI2 receptor IP at detectable levels. In agreement with this finding, PGIS mRNA levels were dramatically diminished in primary human tumor samples as compared to matched normal lung tissue. Finally, thromboxane A2 (TxA2) was synthesized in NSCLC cell lines, but transcription of the gene coding for the TxA2 receptor TP was not observed in these cells. In marked contrast, lung fibroblasts synthesized all three prostanoids and their receptors at high levels. While the observed expression patterns were consistent with the existence of autocrine/paracrine PGE2 signaling loops in NSCLC cells, PGI2- and TxA2-mediated signals may play a role in tumor stroma cells.
- Published
- 2007
47. Proteomic profile of mouse fibroblasts with a targeted disruption of the peroxisome proliferator activated receptor-beta/delta gene.
- Author
-
Adamkiewicz J, Kaddatz K, Rieck M, Wilke B, Müller-Brüsselbach S, and Müller R
- Subjects
- Animals, Cells, Cultured, Chloride Channels genetics, Chloride Channels metabolism, Electrophoresis, Gel, Two-Dimensional, Endothelial Cells cytology, Endothelial Cells metabolism, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Molecular Sequence Data, PPAR delta metabolism, PPAR-beta metabolism, Retinol-Binding Proteins genetics, Retinol-Binding Proteins metabolism, Retinol-Binding Proteins, Cellular, Fibroblasts chemistry, Gene Targeting, PPAR delta genetics, PPAR-beta genetics, Proteome analysis
- Abstract
The peroxisome proliferator activated receptor-beta (PPARbeta) plays an essential role in lipid metabolism, immune modulation, differentiation and cell proliferation. There is also strong evidence for a function in oncogenesis and tumor vascularization, but the underlying molecular mechanisms remain elusive. In the present study, we have used fibroblasts derived from Pparb wild-type and null mice to determine by 2-DE and PMF analysis the contribution of PPARbeta to the protein profile of fibroblasts. Thirty-one differentially expressed proteins of different functional categories were identified. For at least two proteins a role in tumorigenesis and/or tumor vascularization has previously been reported: while the calcium intracellular channel-4 (CLIC4) was expressed at lower levels in Pparb null cells, expression of the cellular retinol binding protein 1 (CRBP1) was enhanced. Clic4 and Crbp1 gene expression patterns observed in different experimental settings in vitro and in vivo confirmed the proteomics data. Our findings indicate that the expression of a defined set of proteins is altered in fibroblasts and endothelial cells from Pparb null mice, that this is due to aberrant gene regulation, and that the altered expression of these proteins is consistent with the tumor vascularization phenotype of Pparb null mice.
- Published
- 2007
- Full Text
- View/download PDF
48. Induction of PPARbeta and prostacyclin (PGI2) synthesis by Raf signaling: failure of PGI2 to activate PPARbeta.
- Author
-
Fauti T, Müller-Brüsselbach S, Kreutzer M, Rieck M, Meissner W, Rapp U, Schweer H, Kömhoff M, and Müller R
- Subjects
- Animals, Autocrine Communication, CHO Cells, Cells, Cultured, Cricetinae, Cyclooxygenase 2 genetics, Cyclooxygenase 2 metabolism, Dinoprostone biosynthesis, Dinoprostone metabolism, Epoprostenol metabolism, Extracellular Signal-Regulated MAP Kinases metabolism, Intramolecular Oxidoreductases genetics, Intramolecular Oxidoreductases metabolism, Membrane Proteins genetics, Membrane Proteins metabolism, Mice, NIH 3T3 Cells, PPAR-beta genetics, Prostaglandin-E Synthases, Prostaglandins biosynthesis, Prostaglandins metabolism, Proto-Oncogene Proteins c-raf genetics, Proto-Oncogene Proteins c-raf pharmacology, Signal Transduction, Tamoxifen analogs & derivatives, Tamoxifen metabolism, Tamoxifen pharmacology, Time Factors, Transcriptional Activation, Transfection, Epoprostenol biosynthesis, PPAR-beta metabolism, Proto-Oncogene Proteins c-raf metabolism
- Abstract
A role for the nuclear receptor peroxisome proliferator-activated receptor-beta (PPARbeta) in oncogenesis has been suggested by a number of observations but its precise role remains elusive. Prostaglandin I2 (PGI2, prostacyclin), a major arachidonic acid (AA) derived cyclooxygenase (Cox) product, has been proposed as a PPARbeta agonist. Here, we show that the 4-hydroxytamoxifen (4-OHT) mediated activation of a C-Raf-estrogen receptor fusion protein leads to the induction of both the PPARbeta and Cox-2 genes, concomitant with a dramatic increase in PGI2 synthesis. Surprisingly, however, 4-OHT failed to activate PPARbeta transcriptional activity, indicating that PGI2 is insufficient for PPARbeta activation. In agreement with this conclusion, the overexpression of ectopic Cox-2 and PGI2 synthase (PGIS) resulted in massive PGI2 synthesis but did not activate the transcriptional activity of PPARbeta. Conversely, inhibition of PGIS blocked PGI2 synthesis but did not affect the AA mediated activation of PPARbeta. Our data obtained with four different cell types and different experimental strategies do not support the prevailing opinion that PGI2 plays a significant role in the regulation of PPARbeta.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.