Skip to main content
Top
Published in: Molecular Cancer 1/2014

Open Access 01-12-2014 | Research

A TrxR inhibiting gold(I) NHC complex induces apoptosis through ASK1-p38-MAPK signaling in pancreatic cancer cells

Authors: Xinlai Cheng, Palvo Holenya, Suzan Can, Hamed Alborzinia, Riccardo Rubbiani, Ingo Ott, Stefan Wölfl

Published in: Molecular Cancer | Issue 1/2014

Login to get access

Abstract

Background

Cancer cells in the advanced stage show aberrant antioxidant capacity to detoxify excessive ROS resulting in the compensation for intrinsic oxidative stress and therapeutic resistance. PDAC is one of the most lethal cancers and often associated with a high accumulation of ROS. Recent studies identified gold(I) NHC complexes as potent TrxR inhibitors suppressing cell growth in a wide spectrum of human malignant cell lines at the low micromolar concentration. However, the mechanism of action is not completely elucidated yet.

Methods

To understand the biological function of gold(I) NHC complexes in PDAC, we used a recently published gold(I) NHC complex, MC3, and evaluated its anti-proliferative effect in four PDAC cell lines, determined by MTT and SRB assays. In further detailed analysis, we analyzed cellular ROS levels using the ROS indicator DHE and mitochondrial membrane potential indicated by the dye JC-1 in Panc1. We also analyzed cell cycle arrest and apoptosis by FACS. To elucidate the role of specific cell signaling pathways in MC3-induced cell death, co-incubation with ROS scavengers, a p38-MAPK inhibitor and siRNA mediated depletion of ASK1 were performed, and results were analyzed by immunoblotting, ELISA-microarrays, qRT-PCR and immunoprecipitation.

Results

Our data demonstrate that MC3 efficiently suppressed cell growth, and induced cell cycle arrest and apoptosis in pancreatic cancer cells, in particular in the gemcitabine-resistant cancer cells Panc1 and ASPC1. Treatment with MC3 resulted in a substantial alteration of the cellular redox homeostasis leading to increased ROS levels and a decrease in the mitochondrial membrane potential. ROS scavengers suppressed ROS formation and rescued cells from damage. On the molecular level, MC3 blocked the interaction of Trx with ASK1 and subsequently activated p38-associated signaling. Furthermore, inhibition of this pathway by using ASK1 siRNA or a p38 inhibitor clearly attenuated the effect of MC3 on cell proliferation in Panc1 and ASPC1.

Conclusions

Our results confirm that MC3 is a TrxR inhibitor and show MC3 induced apoptosis in gemcitabine-resistant PDACs. MC3 mediated cell death could be blocked by using anti-oxidants, ASK1 siRNA or p38 inhibitor suggesting that the Trx-ASK1-p38 signal cascade played an important role in gold(I) NHC complexes-mediated cellular damage.
Appendix
Available only for authorised users
Literature
1.
go back to reference Rosenberg B, VanCamp L, Trosko JE, Mansour VH: Platinum compounds: a new class of potent antitumour agents. Nature. 1969, 222: 385-386. 10.1038/222385a0CrossRefPubMed Rosenberg B, VanCamp L, Trosko JE, Mansour VH: Platinum compounds: a new class of potent antitumour agents. Nature. 1969, 222: 385-386. 10.1038/222385a0CrossRefPubMed
2.
go back to reference Wong E, Giandomenico CM: Current status of platinum-based antitumor drugs. Chem Rev. 1999, 99: 2451-2466. 10.1021/cr980420vCrossRefPubMed Wong E, Giandomenico CM: Current status of platinum-based antitumor drugs. Chem Rev. 1999, 99: 2451-2466. 10.1021/cr980420vCrossRefPubMed
3.
go back to reference Galanski M, Jakupec MA, Keppler BK: Update of the preclinical situation of anticancer platinum complexes: novel design strategies and innovative analytical approaches. Curr Med Chem. 2005, 12: 2075-2094. 10.2174/0929867054637626CrossRefPubMed Galanski M, Jakupec MA, Keppler BK: Update of the preclinical situation of anticancer platinum complexes: novel design strategies and innovative analytical approaches. Curr Med Chem. 2005, 12: 2075-2094. 10.2174/0929867054637626CrossRefPubMed
4.
go back to reference Boulikas T, Vougiouka M: Cisplatin and platinum drugs at the molecular level. (Review). Oncol Rep. 2003, 10: 1663-1682.PubMed Boulikas T, Vougiouka M: Cisplatin and platinum drugs at the molecular level. (Review). Oncol Rep. 2003, 10: 1663-1682.PubMed
5.
go back to reference Pasini A, Zunino F: New Cisplatin Analogs - on the way to better antitumor agents. Angew Chem Int Ed Engl. 1987, 26: 615-624. 10.1002/anie.198706151.CrossRef Pasini A, Zunino F: New Cisplatin Analogs - on the way to better antitumor agents. Angew Chem Int Ed Engl. 1987, 26: 615-624. 10.1002/anie.198706151.CrossRef
6.
go back to reference Kasper C, Alborzinia H, Can S, Kitanovic I, Meyer A, Geldmacher Y, Oleszak M, Ott I, Wolfl S, Sheldrick WS: Synthesis and cellular impact of diene-ruthenium(II) complexes: a new class of organoruthenium anticancer agents. J Inorg Biochem. 2012, 106: 126-133. 10.1016/j.jinorgbio.2011.08.027CrossRefPubMed Kasper C, Alborzinia H, Can S, Kitanovic I, Meyer A, Geldmacher Y, Oleszak M, Ott I, Wolfl S, Sheldrick WS: Synthesis and cellular impact of diene-ruthenium(II) complexes: a new class of organoruthenium anticancer agents. J Inorg Biochem. 2012, 106: 126-133. 10.1016/j.jinorgbio.2011.08.027CrossRefPubMed
7.
go back to reference Alessio E, Mestroni G, Bergamo A, Sava G: Ruthenium antimetastatic agents. Curr Top Med Chem. 2004, 4: 1525-1535. 10.2174/1568026043387421CrossRefPubMed Alessio E, Mestroni G, Bergamo A, Sava G: Ruthenium antimetastatic agents. Curr Top Med Chem. 2004, 4: 1525-1535. 10.2174/1568026043387421CrossRefPubMed
8.
go back to reference Geldmacher Y, Kitanovic I, Alborzinia H, Bergerhoff K, Rubbiani R, Wefelmeier P, Prokop A, Gust R, Ott I, Wolfl S, Sheldrick WS: Cellular selectivity and biological impact of cytotoxic rhodium(III) and iridium(III) complexes containing methyl-substituted phenanthroline ligands. ChemMedChem. 2011, 6: 429-439. 10.1002/cmdc.201000517CrossRefPubMed Geldmacher Y, Kitanovic I, Alborzinia H, Bergerhoff K, Rubbiani R, Wefelmeier P, Prokop A, Gust R, Ott I, Wolfl S, Sheldrick WS: Cellular selectivity and biological impact of cytotoxic rhodium(III) and iridium(III) complexes containing methyl-substituted phenanthroline ligands. ChemMedChem. 2011, 6: 429-439. 10.1002/cmdc.201000517CrossRefPubMed
9.
go back to reference Rubbiani R, Can S, Kitanovic I, Alborzinia H, Stefanopoulou M, Kokoschka M, Monchgesang S, Sheldrick WS, Wolfl S, Ott I: Comparative in vitro evaluation of N-heterocyclic carbene gold(I) complexes of the benzimidazolylidene type. J Med Chem. 2011, 54: 8646-8657. 10.1021/jm201220nCrossRefPubMed Rubbiani R, Can S, Kitanovic I, Alborzinia H, Stefanopoulou M, Kokoschka M, Monchgesang S, Sheldrick WS, Wolfl S, Ott I: Comparative in vitro evaluation of N-heterocyclic carbene gold(I) complexes of the benzimidazolylidene type. J Med Chem. 2011, 54: 8646-8657. 10.1021/jm201220nCrossRefPubMed
10.
go back to reference Rubbiani R, Kitanovic I, Alborzinia H, Can S, Kitanovic A, Onambele LA, Stefanopoulou M, Geldmacher Y, Sheldrick WS, Wolber G, Prokop A, Wolfl S, Ott I: Benzimidazol-2-ylidene gold(I) complexes are thioredoxin reductase inhibitors with multiple antitumor properties. J Med Chem. 2010, 53: 8608-8618. 10.1021/jm100801eCrossRefPubMed Rubbiani R, Kitanovic I, Alborzinia H, Can S, Kitanovic A, Onambele LA, Stefanopoulou M, Geldmacher Y, Sheldrick WS, Wolber G, Prokop A, Wolfl S, Ott I: Benzimidazol-2-ylidene gold(I) complexes are thioredoxin reductase inhibitors with multiple antitumor properties. J Med Chem. 2010, 53: 8608-8618. 10.1021/jm100801eCrossRefPubMed
11.
go back to reference Fuertes MA, Alonso C, Perez JM: Biochemical modulation of Cisplatin mechanisms of action: enhancement of antitumor activity and circumvention of drug resistance. Chem Rev. 2003, 103: 645-662. 10.1021/cr020010dCrossRefPubMed Fuertes MA, Alonso C, Perez JM: Biochemical modulation of Cisplatin mechanisms of action: enhancement of antitumor activity and circumvention of drug resistance. Chem Rev. 2003, 103: 645-662. 10.1021/cr020010dCrossRefPubMed
12.
go back to reference Alborzinia H, Can S, Holenya P, Scholl C, Lederer E, Kitanovic I, Wolfl S: Real-time monitoring of cisplatin-induced cell death. PLoS One. 2011, 6: e19714- 10.1371/journal.pone.0019714PubMedCentralCrossRefPubMed Alborzinia H, Can S, Holenya P, Scholl C, Lederer E, Kitanovic I, Wolfl S: Real-time monitoring of cisplatin-induced cell death. PLoS One. 2011, 6: e19714- 10.1371/journal.pone.0019714PubMedCentralCrossRefPubMed
13.
go back to reference Alama A, Tasso B, Novelli F, Sparatore F: Organometallic compounds in oncology: implications of novel organotins as antitumor agents. Drug Discov Today. 2009, 14: 500-508. 10.1016/j.drudis.2009.02.002CrossRefPubMed Alama A, Tasso B, Novelli F, Sparatore F: Organometallic compounds in oncology: implications of novel organotins as antitumor agents. Drug Discov Today. 2009, 14: 500-508. 10.1016/j.drudis.2009.02.002CrossRefPubMed
14.
go back to reference Bedard K, Krause KH: The NOX family of ROS-generating NADPH oxidases: Physiology and pathophysiology. Physiol Rev. 2007, 87: 245-313. 10.1152/physrev.00044.2005CrossRefPubMed Bedard K, Krause KH: The NOX family of ROS-generating NADPH oxidases: Physiology and pathophysiology. Physiol Rev. 2007, 87: 245-313. 10.1152/physrev.00044.2005CrossRefPubMed
15.
go back to reference D’Autreaux B, Toledano MB: ROS as signalling molecules: mechanisms that generate specificity in ROS homeostasis. Nat Rev Mol Cell Biol. 2007, 8: 813-824. 10.1038/nrm2256CrossRefPubMed D’Autreaux B, Toledano MB: ROS as signalling molecules: mechanisms that generate specificity in ROS homeostasis. Nat Rev Mol Cell Biol. 2007, 8: 813-824. 10.1038/nrm2256CrossRefPubMed
16.
go back to reference Boonstra J, Post JA: Molecular events associated with reactive oxygen species and cell cycle progression in mammalian cells. Gene. 2004, 337: 1-13.CrossRefPubMed Boonstra J, Post JA: Molecular events associated with reactive oxygen species and cell cycle progression in mammalian cells. Gene. 2004, 337: 1-13.CrossRefPubMed
17.
go back to reference Schafer FQ, Buettner GR: Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple. Free Radic Biol Med. 2001, 30: 1191-1212. 10.1016/S0891-5849(01)00480-4CrossRefPubMed Schafer FQ, Buettner GR: Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple. Free Radic Biol Med. 2001, 30: 1191-1212. 10.1016/S0891-5849(01)00480-4CrossRefPubMed
18.
go back to reference Trachootham D, Alexandre J, Huang P: Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach?. Nat Rev Drug Discov. 2009, 8: 579-591. 10.1038/nrd2803CrossRefPubMed Trachootham D, Alexandre J, Huang P: Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach?. Nat Rev Drug Discov. 2009, 8: 579-591. 10.1038/nrd2803CrossRefPubMed
19.
go back to reference Ceccarelli J, Delfino L, Zappia E, Castellani P, Borghi M, Ferrini S, Tosetti F, Rubartelli A: The redox state of the lung cancer microenvironment depends on the levels of thioredoxin expressed by tumor cells and affects tumor progression and response to prooxidants. Int J Cancer. 2008, 123: 1770-1778. 10.1002/ijc.23709CrossRefPubMed Ceccarelli J, Delfino L, Zappia E, Castellani P, Borghi M, Ferrini S, Tosetti F, Rubartelli A: The redox state of the lung cancer microenvironment depends on the levels of thioredoxin expressed by tumor cells and affects tumor progression and response to prooxidants. Int J Cancer. 2008, 123: 1770-1778. 10.1002/ijc.23709CrossRefPubMed
20.
go back to reference Yip KW, Reed JC: Bcl-2 family proteins and cancer. Oncogene. 2008, 27: 6398-6406. 10.1038/onc.2008.307CrossRefPubMed Yip KW, Reed JC: Bcl-2 family proteins and cancer. Oncogene. 2008, 27: 6398-6406. 10.1038/onc.2008.307CrossRefPubMed
21.
go back to reference Kirkpatrick DL, Ehrmantraut G, Stettner S, Kunkel M, Powis G: Redox active disulfides: The thioredoxin system as a drug target. Oncol Res. 1997, 9: 351-356.PubMed Kirkpatrick DL, Ehrmantraut G, Stettner S, Kunkel M, Powis G: Redox active disulfides: The thioredoxin system as a drug target. Oncol Res. 1997, 9: 351-356.PubMed
22.
go back to reference Saitoh M, Nishitoh H, Fujii M, Takeda K, Tobiume K, Sawada Y, Kawabata M, Miyazono K, Ichijo H: Mammalian thioredoxin is a direct inhibitor of apoptosis signal-regulating kinase (ASK) 1. Embo J. 1998, 17: 2596-2606. 10.1093/emboj/17.9.2596PubMedCentralCrossRefPubMed Saitoh M, Nishitoh H, Fujii M, Takeda K, Tobiume K, Sawada Y, Kawabata M, Miyazono K, Ichijo H: Mammalian thioredoxin is a direct inhibitor of apoptosis signal-regulating kinase (ASK) 1. Embo J. 1998, 17: 2596-2606. 10.1093/emboj/17.9.2596PubMedCentralCrossRefPubMed
23.
go back to reference Fujino G, Noguchi T, Matsuzawa A, Yamauchi S, Saitoh M, Takeda K, Ichijo H: Thioredoxin and TRAF family proteins regulate reactive oxygen species-dependent activation of ASK1 through reciprocal modulation of the N-terminal homophilic interaction of ASK1. Mol Cell Biol. 2007, 27: 8152-8163. 10.1128/MCB.00227-07PubMedCentralCrossRefPubMed Fujino G, Noguchi T, Matsuzawa A, Yamauchi S, Saitoh M, Takeda K, Ichijo H: Thioredoxin and TRAF family proteins regulate reactive oxygen species-dependent activation of ASK1 through reciprocal modulation of the N-terminal homophilic interaction of ASK1. Mol Cell Biol. 2007, 27: 8152-8163. 10.1128/MCB.00227-07PubMedCentralCrossRefPubMed
24.
go back to reference Masutani H, Ueda S, Yodoi J: The thioredoxin system in retroviral infection and apoptosis. Cell Death Differ. 2005, 12 (Suppl 1): 991-998.CrossRefPubMed Masutani H, Ueda S, Yodoi J: The thioredoxin system in retroviral infection and apoptosis. Cell Death Differ. 2005, 12 (Suppl 1): 991-998.CrossRefPubMed
25.
go back to reference Huang P, Feng L, Oldham EA, Keating MJ, Plunkett W: Superoxide dismutase as a target for the selective killing of cancer cells. Nature. 2000, 407: 390-395. 10.1038/35030140CrossRefPubMed Huang P, Feng L, Oldham EA, Keating MJ, Plunkett W: Superoxide dismutase as a target for the selective killing of cancer cells. Nature. 2000, 407: 390-395. 10.1038/35030140CrossRefPubMed
26.
go back to reference Fang J, Sawa T, Akaike T, Greish K, Maeda H: Enhancement of chemotherapeutic response of tumor cells by a heme oxygenase inhibitor, pegylated zinc protoporphyrin. Int J Cancer. 2004, 109: 1-8. 10.1002/ijc.11644CrossRefPubMed Fang J, Sawa T, Akaike T, Greish K, Maeda H: Enhancement of chemotherapeutic response of tumor cells by a heme oxygenase inhibitor, pegylated zinc protoporphyrin. Int J Cancer. 2004, 109: 1-8. 10.1002/ijc.11644CrossRefPubMed
27.
go back to reference Welsh SJ, Williams RR, Birmingham A, Newman DJ, Kirkpatrick DL, Powis G: The thioredoxin redox inhibitors 1-methylpropyl 2-imidazolyl disulfide and pleurotin inhibit hypoxia-induced factor 1alpha and vascular endothelial growth factor formation. Mol Cancer Ther. 2003, 2: 235-243.PubMed Welsh SJ, Williams RR, Birmingham A, Newman DJ, Kirkpatrick DL, Powis G: The thioredoxin redox inhibitors 1-methylpropyl 2-imidazolyl disulfide and pleurotin inhibit hypoxia-induced factor 1alpha and vascular endothelial growth factor formation. Mol Cancer Ther. 2003, 2: 235-243.PubMed
28.
go back to reference Pelicano H, Carney D, Huang P: ROS stress in cancer cells and therapeutic implications. Drug Resist Updat. 2004, 7: 97-110.CrossRefPubMed Pelicano H, Carney D, Huang P: ROS stress in cancer cells and therapeutic implications. Drug Resist Updat. 2004, 7: 97-110.CrossRefPubMed
29.
go back to reference Kong Q, Lillehei KO: Antioxidant inhibitors for cancer therapy. Med Hypotheses. 1998, 51: 405-409. 10.1016/S0306-9877(98)90036-6CrossRefPubMed Kong Q, Lillehei KO: Antioxidant inhibitors for cancer therapy. Med Hypotheses. 1998, 51: 405-409. 10.1016/S0306-9877(98)90036-6CrossRefPubMed
30.
go back to reference Cabello CM, Bair WB, Wondrak GT: Experimental therapeutics: targeting the redox Achilles heel of cancer. Curr Opin Investig Drugs. 2007, 8: 1022-1037.PubMed Cabello CM, Bair WB, Wondrak GT: Experimental therapeutics: targeting the redox Achilles heel of cancer. Curr Opin Investig Drugs. 2007, 8: 1022-1037.PubMed
31.
go back to reference Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T, Thun MJ: Cancer statistics, 2008. CA Cancer J Clin. 2008, 58: 71-96. 10.3322/CA.2007.0010CrossRefPubMed Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T, Thun MJ: Cancer statistics, 2008. CA Cancer J Clin. 2008, 58: 71-96. 10.3322/CA.2007.0010CrossRefPubMed
32.
go back to reference Burris HA, Moore MJ, Andersen J, Green MR, Rothenberg ML, Madiano MR, Cripps MC, Portenoy RK, Storniolo AM, Tarassoff P, Nelson R, Dorr FA, Stephens CD, VanHoff DD: Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: A randomized trial. J Clin Oncol. 1997, 15: 2403-2413.PubMed Burris HA, Moore MJ, Andersen J, Green MR, Rothenberg ML, Madiano MR, Cripps MC, Portenoy RK, Storniolo AM, Tarassoff P, Nelson R, Dorr FA, Stephens CD, VanHoff DD: Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: A randomized trial. J Clin Oncol. 1997, 15: 2403-2413.PubMed
33.
go back to reference Bernas T, Dobrucki J: Mitochondrial and nonmitochondrial reduction of MTT: Interaction of MTT with TMRE, JC-1, and NAO mitochondrial fluorescent probes. Cytometry. 2002, 47: 236-242. 10.1002/cyto.10080CrossRefPubMed Bernas T, Dobrucki J: Mitochondrial and nonmitochondrial reduction of MTT: Interaction of MTT with TMRE, JC-1, and NAO mitochondrial fluorescent probes. Cytometry. 2002, 47: 236-242. 10.1002/cyto.10080CrossRefPubMed
34.
go back to reference Skehan P, Storeng R, Scudiero D, Monks A, Mcmahon J, Vistica D, Warren JT, Bokesch H, Kenney S, Boyd MR: New colorimetric cytotoxicity assay for anticancer-drug screening. J Natl Cancer Inst. 1990, 82: 1107-1112. 10.1093/jnci/82.13.1107CrossRefPubMed Skehan P, Storeng R, Scudiero D, Monks A, Mcmahon J, Vistica D, Warren JT, Bokesch H, Kenney S, Boyd MR: New colorimetric cytotoxicity assay for anticancer-drug screening. J Natl Cancer Inst. 1990, 82: 1107-1112. 10.1093/jnci/82.13.1107CrossRefPubMed
35.
go back to reference Hingorani SR, Wang LF, Multani AS, Combs C, Deramaudt TB, Hruban RH, Rustgi AK, Chang S, Tuveson DA: Trp53(R172H) and KraS(G12D) cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell. 2005, 7: 469-483. 10.1016/j.ccr.2005.04.023CrossRefPubMed Hingorani SR, Wang LF, Multani AS, Combs C, Deramaudt TB, Hruban RH, Rustgi AK, Chang S, Tuveson DA: Trp53(R172H) and KraS(G12D) cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell. 2005, 7: 469-483. 10.1016/j.ccr.2005.04.023CrossRefPubMed
36.
go back to reference Berrozpe G, Schaeffer J, Peinado MA, Real FX, Perucho M: Comparative-analysis of mutations in the P53 and K-Ras genes in pancreatic-cancer. Int J Cancer. 1994, 58: 185-191. 10.1002/ijc.2910580207CrossRefPubMed Berrozpe G, Schaeffer J, Peinado MA, Real FX, Perucho M: Comparative-analysis of mutations in the P53 and K-Ras genes in pancreatic-cancer. Int J Cancer. 1994, 58: 185-191. 10.1002/ijc.2910580207CrossRefPubMed
37.
go back to reference Kitanovic I, Can S, Alborzinia H, Kitanovic A, Pierroz V, Leonidova A, Pinto A, Spingler B, Ferrari S, Molteni R, Steffen A, Metzler-Nolte N, Wölfl S, Gasser G: A deadly organometallic luminescent probe: anticancer activity of a ReI bisquinoline complex. Chemistry. 2014, 20: 2496-2507. 10.1002/chem.201304012CrossRefPubMed Kitanovic I, Can S, Alborzinia H, Kitanovic A, Pierroz V, Leonidova A, Pinto A, Spingler B, Ferrari S, Molteni R, Steffen A, Metzler-Nolte N, Wölfl S, Gasser G: A deadly organometallic luminescent probe: anticancer activity of a ReI bisquinoline complex. Chemistry. 2014, 20: 2496-2507. 10.1002/chem.201304012CrossRefPubMed
38.
go back to reference Vermes I, Haanen C, Steffensnakken H, Reutelingsperger C: A novel assay for apoptosis - flow cytometric detection of phosphatidylserine expression on early apoptotic cells using fluorescein-labeled annexin-V. J Immunol Methods. 1995, 184: 39-51. 10.1016/0022-1759(95)00072-ICrossRefPubMed Vermes I, Haanen C, Steffensnakken H, Reutelingsperger C: A novel assay for apoptosis - flow cytometric detection of phosphatidylserine expression on early apoptotic cells using fluorescein-labeled annexin-V. J Immunol Methods. 1995, 184: 39-51. 10.1016/0022-1759(95)00072-ICrossRefPubMed
39.
go back to reference Modica-Napolitano JS, Renshaw PF: Ethanolamine and phosphoethanolamine inhibit mitochondrial function in vitro: implications for mitochondrial dysfunction hypothesis in depression and bipolar disorder. Biol Psychiatry. 2004, 55: 273-277. 10.1016/S0006-3223(03)00784-4CrossRefPubMed Modica-Napolitano JS, Renshaw PF: Ethanolamine and phosphoethanolamine inhibit mitochondrial function in vitro: implications for mitochondrial dysfunction hypothesis in depression and bipolar disorder. Biol Psychiatry. 2004, 55: 273-277. 10.1016/S0006-3223(03)00784-4CrossRefPubMed
40.
go back to reference Salvioli S, Ardizzoni A, Franceschi C, Cossarizza A: JC-1, but not DiOC(6)(3) or rhodamine 123, is a reliable fluorescent probe to assess Delta Psi changes in intact cells: Implications for studies on mitochondrial functionality during apoptosis. Febs Lett. 1997, 411: 77-82. 10.1016/S0014-5793(97)00669-8CrossRefPubMed Salvioli S, Ardizzoni A, Franceschi C, Cossarizza A: JC-1, but not DiOC(6)(3) or rhodamine 123, is a reliable fluorescent probe to assess Delta Psi changes in intact cells: Implications for studies on mitochondrial functionality during apoptosis. Febs Lett. 1997, 411: 77-82. 10.1016/S0014-5793(97)00669-8CrossRefPubMed
41.
go back to reference Holenya P, Kitanovic I, Heigwer F, Wolfl S: Microarray-based kinetic colorimetric detection for quantitative multiplex protein phosphorylation analysis. Proteomics. 2011, 11: 2129-2133. 10.1002/pmic.201000690CrossRefPubMed Holenya P, Kitanovic I, Heigwer F, Wolfl S: Microarray-based kinetic colorimetric detection for quantitative multiplex protein phosphorylation analysis. Proteomics. 2011, 11: 2129-2133. 10.1002/pmic.201000690CrossRefPubMed
42.
go back to reference Lee JC, Laydon JT, Mcdonnell PC, Gallagher TF, Kumar S, Green D, Mcnulty D, Blumenthal MJ, Heys JR, Landvatter SW, Strickler JE, Mclaughlin MM, Siemens IR, Fisher SM, Livi GP, White JR, Adams JL, Young PR: A Protein-Kinase Involved in the Regulation of Inflammatory Cytokine Biosynthesis. Nature. 1994, 372: 739-746. 10.1038/372739a0CrossRefPubMed Lee JC, Laydon JT, Mcdonnell PC, Gallagher TF, Kumar S, Green D, Mcnulty D, Blumenthal MJ, Heys JR, Landvatter SW, Strickler JE, Mclaughlin MM, Siemens IR, Fisher SM, Livi GP, White JR, Adams JL, Young PR: A Protein-Kinase Involved in the Regulation of Inflammatory Cytokine Biosynthesis. Nature. 1994, 372: 739-746. 10.1038/372739a0CrossRefPubMed
43.
go back to reference Raingeaud J, Gupta S, Rogers JS, Dickens M, Han JH, Ulevitch RJ, Davis RJ: Pro-inflammatory cytokines and environmental-stress cause P38 mitogen-activated protein-kinase activation by dual phosphorylation on tyrosine and threonine. J Biol Chem. 1995, 270: 7420-7426. 10.1074/jbc.270.13.7420CrossRefPubMed Raingeaud J, Gupta S, Rogers JS, Dickens M, Han JH, Ulevitch RJ, Davis RJ: Pro-inflammatory cytokines and environmental-stress cause P38 mitogen-activated protein-kinase activation by dual phosphorylation on tyrosine and threonine. J Biol Chem. 1995, 270: 7420-7426. 10.1074/jbc.270.13.7420CrossRefPubMed
44.
go back to reference Kim HS, Lee MS: Essential role of STAT1 in caspase-independent cell death of activated macrophages through the p38 mitogen-activated protein kinase/STAT1/reactive oxygen species pathway. Mol Cell Biol. 2005, 25: 6821-6833. 10.1128/MCB.25.15.6821-6833.2005PubMedCentralCrossRefPubMed Kim HS, Lee MS: Essential role of STAT1 in caspase-independent cell death of activated macrophages through the p38 mitogen-activated protein kinase/STAT1/reactive oxygen species pathway. Mol Cell Biol. 2005, 25: 6821-6833. 10.1128/MCB.25.15.6821-6833.2005PubMedCentralCrossRefPubMed
45.
go back to reference Cuadrado A, Lafarga V, Cheung PCF, Dolado I, Llanos S, Cohen P, Nebreda AR: A new p38 MAP kinase-regulated transcriptional coactivator that stimulates p53-dependent apoptosis. Embo J. 2007, 26: 2115-2126. 10.1038/sj.emboj.7601657PubMedCentralCrossRefPubMed Cuadrado A, Lafarga V, Cheung PCF, Dolado I, Llanos S, Cohen P, Nebreda AR: A new p38 MAP kinase-regulated transcriptional coactivator that stimulates p53-dependent apoptosis. Embo J. 2007, 26: 2115-2126. 10.1038/sj.emboj.7601657PubMedCentralCrossRefPubMed
46.
go back to reference Wellner U, Schubert J, Burk UC, Schmalhofer O, Zhu F, Sonntag A, Waldvogel B, Vannier C, Darling D, zur Hausen A, Brunton VG, Morton J, Sansom O, Schuler J, Stemmler MP, Herzberger C, Hopt U, Keck T, Brabletz S, Brabletz T: The EMT-activator ZEB1 promotes tumorigenicity by repressing stemness-inhibiting microRNAs. Nat Cell Biol. 2009, 11: 1487-U1236, 10.1038/ncb1998CrossRefPubMed Wellner U, Schubert J, Burk UC, Schmalhofer O, Zhu F, Sonntag A, Waldvogel B, Vannier C, Darling D, zur Hausen A, Brunton VG, Morton J, Sansom O, Schuler J, Stemmler MP, Herzberger C, Hopt U, Keck T, Brabletz S, Brabletz T: The EMT-activator ZEB1 promotes tumorigenicity by repressing stemness-inhibiting microRNAs. Nat Cell Biol. 2009, 11: 1487-U1236, 10.1038/ncb1998CrossRefPubMed
47.
go back to reference Wagner EF, Nebreda AR: Signal integration by JNK and p38 MAPK pathways in cancer development. Nat Rev Cancer. 2009, 9: 537-549. 10.1038/nrc2694CrossRefPubMed Wagner EF, Nebreda AR: Signal integration by JNK and p38 MAPK pathways in cancer development. Nat Rev Cancer. 2009, 9: 537-549. 10.1038/nrc2694CrossRefPubMed
48.
go back to reference Mateescu B, Batista L, Cardon M, Gruosso T, de Feraudy Y, Mariani O, Nicolas A, Meyniel JP, Cottu P, Sastre-Garau X, Mechta-Grigoriou F: miR-141 and miR-200a act on ovarian tumorigenesis by controlling oxidative stress response. Nat Med. 2011, 17: 1627-U1144, 10.1038/nm.2512CrossRefPubMed Mateescu B, Batista L, Cardon M, Gruosso T, de Feraudy Y, Mariani O, Nicolas A, Meyniel JP, Cottu P, Sastre-Garau X, Mechta-Grigoriou F: miR-141 and miR-200a act on ovarian tumorigenesis by controlling oxidative stress response. Nat Med. 2011, 17: 1627-U1144, 10.1038/nm.2512CrossRefPubMed
49.
go back to reference Dolado I, Swat A, Ajenjo N, De Vita G, Cuadrado A, Nebreda AR: p38alpha MAP kinase as a sensor of reactive oxygen species in tumorigenesis. Cancer Cell. 2007, 11: 191-205. 10.1016/j.ccr.2006.12.013CrossRefPubMed Dolado I, Swat A, Ajenjo N, De Vita G, Cuadrado A, Nebreda AR: p38alpha MAP kinase as a sensor of reactive oxygen species in tumorigenesis. Cancer Cell. 2007, 11: 191-205. 10.1016/j.ccr.2006.12.013CrossRefPubMed
50.
go back to reference Cheng X, Rasque P, Vatter S, Merz KH, Eisenbrand G: Synthesis and cytotoxicity of novel indirubin-5-carboxamides. Bioorg Med Chem. 2010, 18: 4509-4515. 10.1016/j.bmc.2010.04.066CrossRefPubMed Cheng X, Rasque P, Vatter S, Merz KH, Eisenbrand G: Synthesis and cytotoxicity of novel indirubin-5-carboxamides. Bioorg Med Chem. 2010, 18: 4509-4515. 10.1016/j.bmc.2010.04.066CrossRefPubMed
Metadata
Title
A TrxR inhibiting gold(I) NHC complex induces apoptosis through ASK1-p38-MAPK signaling in pancreatic cancer cells
Authors
Xinlai Cheng
Palvo Holenya
Suzan Can
Hamed Alborzinia
Riccardo Rubbiani
Ingo Ott
Stefan Wölfl
Publication date
01-12-2014
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2014
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/1476-4598-13-221

Other articles of this Issue 1/2014

Molecular Cancer 1/2014 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine