1
|
Omarova BA, Shults EE, Zhakipbekov KS, Abekova АО, Ishmuratova MY, Petrova TN, Kartbayeva EB. Biological effects and phytochemical study of the underground part of Iris scariosa Willd. ex Link extract: A new source of bioactive constituents. Fitoterapia 2024; 175:105920. [PMID: 38531480 DOI: 10.1016/j.fitote.2024.105920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 03/18/2024] [Accepted: 03/23/2024] [Indexed: 03/28/2024]
Abstract
The expected toxicity and resistance of chemotherapeutic agents necessitate and encourage for the use of natural chemotherapeutic sources of plant origin in the clinical stage of cancer therapy. Plants of the genus Iris (Iridaceae) used by local populations for the treatment of cancer, bacterial and viral infections. In this study, an ethanol extract of rhizomes of I. scariosa was prepared and tested for the cytotoxicity using the MTT assay. The extract exhibited the most potent cytotoxicity against the breast cancer cell line MCF7 (IC50 = 9.28 ± 0.49 μg/ml, selectively index ˃5), and induced apoptosis in MCF7 lines. Notably, the extract significantly inhibited the colony formation of MCF7 and HepG2 cancer cells at a concentration range from 10.6 to 85.0 μg/ml, including non-toxic concentrations for HepG2 cells. The ethanol extract was analyzed by HPLC, revealed the identification of 5 secondary metabolites (quercetin, rutin, myricetin, apigenin, artemisetin), the content of which was shown to reach around 15% of the extract. The petroleum ether (PE) part of the extract (yield 2.62%) was analyzed by GC-MS. The composition of tert-butyl methyl ether (TBME) part of the extract (yield 23.72%) was studied. Total of 15 individual compounds: two benzophenones, eight isoflavones, four flavones and a (2R)-flavanone were isolated. The pentamethoxyflavone artemisetin and flavanone pinocembrin were isolated for the first from Iris sp. The readily available isoflavones from the TBME part of extract (irilone, iriflogenin, irigenin and tectorigenin) may serve as new leads for the discovery of anticancer drugs.
Collapse
Affiliation(s)
- Botagoz A Omarova
- Asfendiyarov Kazakh National Medical University, Tole Bi St. 94, Almaty 050000, Republic of Kazakhstan
| | - Elvira E Shults
- Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentyev Ave. 9, 630090 Novosibirsk, Russia.
| | - Kairat S Zhakipbekov
- Asfendiyarov Kazakh National Medical University, Tole Bi St. 94, Almaty 050000, Republic of Kazakhstan.
| | - Аssima О Abekova
- JSC «Scientific Center for Anti-Infectious Drugs», al-Farabi Ave. 75A, 050060 Almaty, Republic of Kazakhstan
| | - Margarita Yu Ishmuratova
- NCJSC "Buketov Karaganda University", Universitetskaya Str., 28/3, 100028 Karaganda, Republic of Kazakhstan
| | - Tatyana N Petrova
- Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentyev Ave. 9, 630090 Novosibirsk, Russia
| | - Elmira B Kartbayeva
- Asfendiyarov Kazakh National Medical University, Tole Bi St. 94, Almaty 050000, Republic of Kazakhstan; Higher School of Medicine, Al-Farabi Kazakh National University, 71 al-Farabi Ave., Almaty 050040, Republic of Kazakhstan
| |
Collapse
|
2
|
Kim J, Jo YH, Jang M, Nguyen NNY, Yun HR, Ko SH, Shin Y, Lee JS, Kang I, Ha J, Choi TG, Kim SS. PAC-5 Gene Expression Signature for Predicting Prognosis of Patients with Pancreatic Adenocarcinoma. Cancers (Basel) 2019; 11:cancers11111749. [PMID: 31703415 PMCID: PMC6896100 DOI: 10.3390/cancers11111749] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/24/2019] [Accepted: 11/04/2019] [Indexed: 01/05/2023] Open
Abstract
Pancreatic adenocarcinoma (PAC) is one of the most aggressive malignancies. Intratumoural molecular heterogeneity impedes improvement of the overall survival rate. Current pathological staging system is not sufficient to accurately predict prognostic outcomes. Thus, accurate prognostic model for patient survival and treatment decision is demanded. Using differentially expressed gene analysis between normal pancreas and PAC tissues, the cancer-specific genes were identified. A prognostic gene expression model was computed by LASSO regression analysis. The PAC-5 signature (LAMA3, E2F7, IFI44, SLC12A2, and LRIG1) that had significant prognostic value in the overall dataset was established, independently of the pathological stage. We provided evidence that the PAC-5 signature further refined the selection of the PAC patients who might benefit from postoperative therapies. SLC12A2 and LRIG1 interacted with the proteins that were implicated in resistance of EGFR kinase inhibitor. DNA methylation was significantly involved in the gene regulations of the PAC-5 signature. The PAC-5 signature provides new possibilities for improving the personalised therapeutic strategies. We suggest that the PAC-5 genes might be potential drug targets for PAC.
Collapse
Affiliation(s)
- Jieun Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.K.); (N.N.Y.N.); (H.R.Y.); (Y.S.); (I.K.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (M.J.)
| | - Yong Hwa Jo
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (M.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Miran Jang
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (M.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Ngoc Ngo Yen Nguyen
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.K.); (N.N.Y.N.); (H.R.Y.); (Y.S.); (I.K.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (M.J.)
| | - Hyeong Rok Yun
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.K.); (N.N.Y.N.); (H.R.Y.); (Y.S.); (I.K.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (M.J.)
| | - Seok Hoon Ko
- Department of Emergency Medicine, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Yoonhwa Shin
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.K.); (N.N.Y.N.); (H.R.Y.); (Y.S.); (I.K.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (M.J.)
| | - Ju-Seog Lee
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Insug Kang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.K.); (N.N.Y.N.); (H.R.Y.); (Y.S.); (I.K.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (M.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Joohun Ha
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.K.); (N.N.Y.N.); (H.R.Y.); (Y.S.); (I.K.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (M.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Tae Gyu Choi
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (M.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (T.G.C.); (S.S.K.); Tel.: +82-961-028-7 (T.G.C.); +82-961-052-4 (S.S.K.)
| | - Sung Soo Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea; (J.K.); (N.N.Y.N.); (H.R.Y.); (Y.S.); (I.K.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea; (Y.H.J.); (M.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (T.G.C.); (S.S.K.); Tel.: +82-961-028-7 (T.G.C.); +82-961-052-4 (S.S.K.)
| |
Collapse
|
3
|
Oien DB, Garay T, Eckstein S, Chien J. Cisplatin and Pemetrexed Activate AXL and AXL Inhibitor BGB324 Enhances Mesothelioma Cell Death from Chemotherapy. Front Pharmacol 2018; 8:970. [PMID: 29375377 PMCID: PMC5768913 DOI: 10.3389/fphar.2017.00970] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 12/20/2017] [Indexed: 12/18/2022] Open
Abstract
Reactive oxygen species (ROS) can promote or inhibit tumorigenesis. In mesothelioma, asbestos exposure to serous membranes induces ROS through iron content and chronic inflammation, and ROS promote cell survival signaling in mesothelioma. Moreover, a current chemotherapy regimen for mesothelioma consisting of a platinum and antifolate agent combination also induce ROS. Mesothelioma is notoriously chemotherapy-resistant, and we propose that ROS induced by cisplatin and pemetrexed may promote cell survival signaling pathways, which ultimately may contribute to chemotherapy resistance. In The Cancer Genome Atlas datasets, we found AXL kinase expression is relatively high in mesothelioma compared to other cancer samples. We showed that ROS induce the phosphorylation of AXL, which was blocked by the selective inhibitor BGB324 in VMC40 and P31 mesothelioma cells. We also showed that cisplatin and pemetrexed induce the phosphorylation of AXL and Akt, which was also blocked by BGB324 as well as by N-acetylcysteine antioxidant. AXL knockdown in these cells enhances sensitivity to cisplatin and pemetrexed. Similarly, AXL inhibitor BGB324 also enhances sensitivity to cisplatin and pemetrexed. Finally, higher synergy was observed when cells were pretreated with BGB324 before adding chemotherapy. These results demonstrate cisplatin and pemetrexed induce ROS that activate AXL, and blocking AXL activation enhances the efficacy of cisplatin and pemetrexed. These results suggest AXL inhibition combined with the current chemotherapy regimen may represent an effective strategy to enhance the efficacy of chemotherapy in mesothelioma. This is the first study, to our knowledge, on chemotherapy-induced activation of AXL and cell survival pathways associated with ROS signaling.
Collapse
Affiliation(s)
- Derek B Oien
- Division of Molecular Medicine, Department of Internal Medicine, UNMHSC School of Medicine, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, United States
| | - Tamás Garay
- Second Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Sarah Eckstein
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Jeremy Chien
- Division of Molecular Medicine, Department of Internal Medicine, UNMHSC School of Medicine, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, United States
| |
Collapse
|
4
|
Geoghegan F, Buckland RJ, Rogers ET, Khalifa K, O'Connor EB, Rooney MF, Behnam-Motlagh P, Nilsson TK, Grankvist K, Porter RK. Bioenergetics of acquired cisplatin resistant H1299 non-small cell lung cancer and P31 mesothelioma cells. Oncotarget 2017; 8:94711-94725. [PMID: 29212260 PMCID: PMC5706906 DOI: 10.18632/oncotarget.21885] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 09/21/2017] [Indexed: 01/31/2023] Open
Abstract
Acquired cisplatin resistance is a common feature of tumours following cancer treatment with cisplatin and also of non-small cell lung cancer (H1299) and mesothelioma (P31) cell lines exposed to cisplatin. To elucidate the cellular basis of acquired cisplatin resistance, a comprehensive bioenergetic analysis was undertaken. We demonstrate that cellular oxygen consumption was significantly decreased in cisplatin resistant cells and that the reduction was primarily due to reduced mitochondrial activity as a result of reduced mitochondrial abundance. The differential mitochondrial abundance was supported by data showing reduced sirtuin 1 (SIRT1), peroxisome-proliferator activator receptor-γ co-activator 1-alpha (PGC1α), sirtuin 3 (SIRT3) and mitochondrial transcription factor A (TFAM) protein expression in resistant cells. Consistent with these data we observed increased reactive oxygen species (ROS) production and increased hypoxia inducible factor 1-alpha (HIF1α) stabilization in cisplatin resistant cells when compared to cisplatin sensitive controls. We also observed an increase in AMP kinase subunit α2 (AMPKα2) transcripts and protein expression in resistant H1299 cells. mRNA expression was also reduced for cisplatin resistant H1299 cells in these genes, however the pattern was not consistent in resistant P31 cells. There was very little change in DNA methylation of these genes, suggesting that the cells are not stably reprogrammed epigenetically. Taken together, our data demonstrate reduced oxidative metabolism, reduced mitochondrial abundance, potential for increased glycolytic flux and increased ROS production in acquired cisplatin resistant cells. This suggests that the metabolic changes are a result of reduced SIRT3 expression and increased HIF-1α stabilization.
Collapse
Affiliation(s)
- Fintan Geoghegan
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin 2, Ireland
| | - Robert J Buckland
- Dept of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, Sweden
| | - Eric T Rogers
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin 2, Ireland
| | - Karima Khalifa
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin 2, Ireland
| | - Emma B O'Connor
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin 2, Ireland
| | - Mary F Rooney
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin 2, Ireland
| | | | - Torbjörn K Nilsson
- Dept of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, Sweden
| | - Kjell Grankvist
- Dept of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, Sweden
| | - Richard K Porter
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
5
|
Hoda MA, Pirker C, Dong Y, Schelch K, Heffeter P, Kryeziu K, van Schoonhoven S, Klikovits T, Laszlo V, Rozsas A, Ozsvar J, Klepetko W, Döme B, Grusch M, Hegedüs B, Berger W. Trabectedin Is Active against Malignant Pleural Mesothelioma Cell and Xenograft Models and Synergizes with Chemotherapy and Bcl-2 Inhibition In Vitro. Mol Cancer Ther 2016; 15:2357-2369. [PMID: 27512118 DOI: 10.1158/1535-7163.mct-15-0846] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 07/26/2016] [Indexed: 11/16/2022]
Abstract
Malignant pleural mesothelioma (MPM) is characterized by widespread resistance to systemic therapy. Trabectedin is an antineoplastic agent targeting both the malignant cells and the tumor microenvironment that has been approved for the treatment of advanced soft tissue sarcoma and ovarian cancer. In this preclinical study, we evaluated the antineoplastic potential of trabectedin as a single agent and in drug combination approaches in human MPM. Therefore, we utilized an extended panel of MPM cell lines (n = 6) and primary cell cultures from surgical MPM specimens (n = 13), as well as nonmalignant pleural tissue samples (n = 2). Trabectedin exerted a dose-dependent cytotoxic effect in all MPM cell cultures in vitro when growing as adherent monolayers or nonadherent spheroids with IC50 values ≤ 2.6 nmol/L. Nonmalignant mesothelial cells were significantly less responsive. The strong antimesothelioma activity was based on cell-cycle perturbation and apoptosis induction. The activity of trabectedin against MPM cells was synergistically enhanced by coadministration of cisplatin, a drug routinely used for systemic MPM treatment. Comparison of gene expression signatures indicated an inverse correlation between trabectedin response and bcl-2 expression. Accordingly, bcl-2 inhibitors (Obatoclax, ABT-199) markedly synergized with trabectedin paralleled by deregulated expression of the bcl-2 family members bcl-2, bim, bax, Mcl-1, and bcl-xL as a consequence of trabectedin exposure. In addition, trabectedin exerted significant antitumor activity against an intraperitoneal MPM xenograft model. Together, these data suggest that trabectedin exerts strong activity in MPM and synergizes with chemotherapy and experimental bcl-2 inhibitors in vitro Thus, it represents a promising new therapeutic option for MPM. Mol Cancer Ther; 15(10); 2357-69. ©2016 AACR.
Collapse
Affiliation(s)
- Mir A Hoda
- Applied and Experimental Oncology, Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria. Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria
| | - Christine Pirker
- Applied and Experimental Oncology, Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Yawen Dong
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria
| | - Karin Schelch
- Applied and Experimental Oncology, Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria. Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria
| | - Petra Heffeter
- Applied and Experimental Oncology, Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Kushtrim Kryeziu
- Applied and Experimental Oncology, Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Sushilla van Schoonhoven
- Applied and Experimental Oncology, Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Thomas Klikovits
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria
| | - Viktoria Laszlo
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria
| | - Anita Rozsas
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria. National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Judit Ozsvar
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria
| | - Walter Klepetko
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria
| | - Balazs Döme
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria. National Koranyi Institute of Pulmonology, Budapest, Hungary. Department of Thoracic Surgery, National Institute of Oncology and Semmelweis University, Budapest, Hungary. Department of Biomedical Imaging and Image-guided Therapy, Division of Molecular and Gender Imaging, Medical University of Vienna, Vienna, Austria
| | - Michael Grusch
- Applied and Experimental Oncology, Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Balazs Hegedüs
- Translational Thoracic Oncology Laboratory, Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria. MTA-SE Molecular Oncology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Walter Berger
- Applied and Experimental Oncology, Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
6
|
Yang Y, Li H, Hou S, Hu B, Liu J, Wang J. Differences in gene expression profiles and carcinogenesis pathways involved in cisplatin resistance of four types of cancer. Oncol Rep 2013; 30:596-614. [PMID: 23733047 DOI: 10.3892/or.2013.2514] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Accepted: 03/04/2013] [Indexed: 11/06/2022] Open
Abstract
Cisplatin-based chemotherapy is the standard therapy used for the treatment of several types of cancer. However, its efficacy is largely limited by the acquired drug resistance. To date, little is known about the RNA expression changes in cisplatin-resistant cancers. Identification of the RNAs related to cisplatin resistance may provide specific insight into cancer therapy. In the present study, expression profiling of 7 cancer cell lines was performed using oligonucleotide microarray analysis data obtained from the GEO database. Bioinformatic analyses such as the Gene Ontology (GO) and KEGG pathway were used to identify genes and pathways specifically associated with cisplatin resistance. A signal transduction network was established to identify the core genes in regulating cancer cell cisplatin resistance. A number of genes were differentially expressed in 7 groups of cancer cell lines. They mainly participated in 85 GO terms and 11 pathways in common. All differential gene interactions in the Signal-Net were analyzed. CTNNB1, PLCG2 and SRC were the most significantly altered. With the use of bioinformatics, large amounts of data in microarrays were retrieved and analyzed by means of thorough experimental planning, scientific statistical analysis and collection of complete data on cancer cell cisplatin resistance. In the present study, a novel differential gene expression pattern was constructed and further study will provide new targets for the diagnosis and mechanisms of cancer cisplatin resistance.
Collapse
Affiliation(s)
- Yong Yang
- Beijing Key Laboratory of Respiratory and Pulmonary Circulation, Capital Medical University, Beijing 100069, PR China
| | | | | | | | | | | |
Collapse
|
7
|
Hermann G, Heffeter P, Falta T, Berger W, Hann S, Koellensperger G. In vitro studies on cisplatin focusing on kinetic aspects of intracellular chemistry by LC-ICP-MS. Metallomics 2013; 5:636-47. [DOI: 10.1039/c3mt20251h] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
8
|
Pichler V, Heffeter P, Valiahdi SM, Kowol CR, Egger A, Berger W, Jakupec MA, Galanski M, Keppler BK. Unsymmetric mono- and dinuclear platinum(IV) complexes featuring an ethylene glycol moiety: synthesis, characterization, and biological activity. J Med Chem 2012. [PMID: 23194425 DOI: 10.1021/jm301645g] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Eight novel mononuclear and two dinuclear platinum(IV) complexes were synthesized and characterized by elemental analysis, one- and two-dimensional NMR spectroscopy, mass spectrometry, and reversed-phase HPLC (log k(w)) and in one case by X-ray diffraction. Cytotoxicity of the compounds was studied in three human cancer cell lines (CH1, SW480, and A549) by means of the MTT assay, featuring IC(50) values to the low micromolar range. Furthermore a selected set of compounds was investigated in additional cancer cell lines (P31 and P31/cis, A2780 and A2780/cis, SW1573, 2R120, and 2R160) with regard to their resistance patterns, offering a distinctly different scheme compared to cisplatin. To gain further insights into the mode of action, drug uptake, DNA synthesis inhibition, cell cycle effects, and induction of apoptosis were determined for two characteristic substances.
Collapse
Affiliation(s)
- Verena Pichler
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Lamoral-Theys D, Wauthoz N, Heffeter P, Mathieu V, Jungwirth U, Lefranc F, Nève J, Dubois J, Dufrasne F, Amighi K, Berger W, Gailly P, Kiss R. Trivanillic polyphenols with anticancer cytostatic effects through the targeting of multiple kinases and intracellular Ca2+ release. J Cell Mol Med 2012; 16:1421-34. [PMID: 21810170 PMCID: PMC3823212 DOI: 10.1111/j.1582-4934.2011.01403.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Cancer cells exhibit de-regulation of multiple cellular signalling pathways and treatments of various types of cancers with polyphenols are promising. We recently reported the synthesis of a series of 33 novel divanillic and trivanillic polyphenols that displayed anticancer activity, at least in vitro, through inhibiting various kinases. This study revealed that minor chemical modifications of a trivanillate scaffold could convert cytotoxic compounds into cytostatic ones. Compound 13c, a tri-chloro derivative of trivanillic ester, displayed marked inhibitory activities against FGF-, VEGF-, EGF- and Src-related kinases, all of which are implicated not only in angiogenesis but also in the biological aggressiveness of various cancer types. The pan-anti-kinase activity of 13c occurs at less than one-tenth of its mean IC50in vitro growth inhibitory concentrations towards a panel of 12 cancer cell lines. Of the 26 kinases for which 13c inhibited their activity by >75%, eight (Yes, Fyn, FGF-R1, EGFR, Btk, Mink, Ret and Itk) are implicated in control of the actin cytoskeleton organization to varying degrees. Compound 13c accordingly impaired the typical organization of the actin cytoskeleton in human U373 glioblastoma cells. The pan-anti-kinase activity and actin cytoskeleton organization impairment provoked by 13c concomitantly occurs with calcium homeostasis impairment but without provoking MDR phenotype activation. All of these anticancer properties enabled 13c to confer therapeutic benefits in vivo in a mouse melanoma pseudometastatic lung model. These data argue in favour of further chemically modifying trivanillates to produce novel and potent anticancer drugs.
Collapse
Affiliation(s)
- Delphine Lamoral-Theys
- Laboratoire de Chimie BioAnalytique, Toxicologie et Chimie Physique Appliquée, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Temsirolimus inhibits malignant pleural mesothelioma growth in vitro and in vivo: synergism with chemotherapy. J Thorac Oncol 2011; 6:852-63. [PMID: 21358348 DOI: 10.1097/jto.0b013e31820e1a25] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Human malignant pleural mesothelioma (MPM) is an asbestos-related malignancy characterized by frequent resistance to chemotherapy and radiotherapy. Here, we investigated the feasibility of mammalian target of rapamycin (mTOR) inhibition by temsirolimus as an antimesothelioma strategy. METHODS Phosphorylation of mTOR (p-mTOR) was assessed by immunohistochemistry in MPM surgical specimens (n = 70). Activation of mTOR and impact of mTOR inhibition by temsirolimus was determined in MPM cell lines in vitro (n = 6) and in vivo as xenografts in severe combined immunodeficiency mice (n = 2) either as single agent or in combination with cisplatin. RESULTS Strong immunoreactivity for p-mTOR was predominantly detected in epitheloid and biphasic but not sarcomatoid MPM specimens while adjacent normal tissues remained widely unstained. Accordingly, all mesothelioma cell lines harbored activated mTOR, which was further confirmed by hyperphosphorylation of the downstream targets pS6K, S6, and 4EBP1. Temsirolimus potently blocked mTOR-mediated signals and exerted a cytostatic effect on mesothelioma cell lines in vitro cultured both as adherent monolayers and as nonadherent spheroids. Mesothelioma cells with intrinsic or acquired cisplatin resistance exhibited hypersensitivity against temsirolimus. Accordingly, cisplatin and temsirolimus exerted synergistic inhibition of the mTOR downstream signals and enhanced growth inhibition and/or apoptosis induction in mesothelioma cell lines. Finally, temsirolimus was highly active against MPM xenograft models in severe combined immunodeficiency mice both as a single agent and in combination with cisplatin. CONCLUSION The mTOR inhibitor temsirolimus is active against mesothelioma in vitro and in vivo and synergizes with chemotherapy. These data suggest mTOR inhibition as a promising novel therapeutic strategy against MPM.
Collapse
|
11
|
Tastesen HS, Holm JB, Møller J, Poulsen KA, Møller C, Stürup S, Hoffmann EK, Lambert IH. Pinpointing differences in cisplatin-induced apoptosis in adherent and non-adherent cancer cells. Cell Physiol Biochem 2011; 26:809-20. [PMID: 21220912 DOI: 10.1159/000323990] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2010] [Indexed: 11/19/2022] Open
Abstract
Platinum compounds are used in the treatment of cancer. We demonstrate that cisplatin-induced (10 μM) apoptosis (caspase-3 activity) is pronounced within 18 hours in non-adherent Ehrlich ascites tumour cells (EATC), whereas there is no increase in caspase-3 activity in the adherent Ehrlich Lettré ascites tumour cells (ELA). Loss of KCl and cell shrinkage are hallmarks in apoptosis and has been shown in EATC. However, we find no reduction in cell volume and only a minor loss of K(+) which is accompanied by net uptake of Na(+) following 18 hours cisplatin exposure in ELA. Glutathione and taurine have previously been demonstrated to protect cells from apoptosis. We find, however, that increase or decrease in the cellular content of glutathione and taurine has no effect on cisplatin-induced cell death in EATC and ELA. Nevertheless, knock-down of the taurine transporter TauT leads to a significant increase in apoptosis in ELA following cisplatin exposure. We find that cytosolic accumulation of cisplatin is similar in EATC and ELA. However, the nuclear accumulation and DNA-binding of cisplatin is significant lower in ELA compared to EATC. We suggest three putative reasons for the observed cisplatin insensitivity in the adherent tumor cells (ELA) compared to the non-adherent tumor cells (EATC): less nuclear cisplatin accumulation, increased TauT activity, and decreased anion and water loss.
Collapse
|
12
|
Resistance to caspase-8 and -9 fragments in a malignant pleural mesothelioma cell line with acquired cisplatin-resistance. Cell Death Dis 2010; 1:e78. [PMID: 21364680 PMCID: PMC3032340 DOI: 10.1038/cddis.2010.54] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Apoptotic cysteine–aspartate proteases (caspases) are essential for the progression and execution of apoptosis, and detection of caspase fragmentation or activity is often used as markers of apoptosis. Cisplatin (cis-diamminedichloroplatinum (II)) is a chemotherapeutic drug that is clinically used for the treatment of solid tumours. We compared a cisplatin-resistant pleural malignant mesothelioma cell line (P31res1.2) with its parental cell line (P31) regarding the consequences of in vitro acquired cisplatin-resistance on basal and cisplatin-induced (equitoxic and equiapoptotic cisplatin concentrations) caspase-3, -8 and -9 fragmentation and proteolytic activity. Acquisition of cisplatin-resistance resulted in basal fragmentation of caspase-8 and -9 without a concomitant increase in proteolytic activity, and there was an increased basal caspase-3/7 activity. Similarly, cisplatin-resistant non-small-cell lung cancer cells, H1299res, had increased caspase-3 and -9 content compared with the parental H1299 cells. In P31 cells, cisplatin exposure resulted in caspase-9-mediated caspase-3/7 activation, but in P31res1.2 cells the cisplatin-induced caspase-3/7 activation occurred before caspase-8 or -9 activation. We therefore concluded that in vitro acquisition of cisplatin-resistance rendered P31res1.2 cells resistant to caspase-8 and caspase-9 fragments and that cisplatin-induced, initiator-caspase independent caspase-3/7 activation was necessary to overcome this resistance. Finally, the results demonstrated that detection of cleaved caspase fragments alone might be insufficient as a marker of caspase activity and ensuing apoptosis induction.
Collapse
|
13
|
Heffeter P, Böck K, Atil B, Hoda MAR, Körner W, Bartel C, Jungwirth U, Keppler BK, Micksche M, Berger W, Koellensperger G. Intracellular protein binding patterns of the anticancer ruthenium drugs KP1019 and KP1339. J Biol Inorg Chem 2010; 15:737-48. [PMID: 20221888 PMCID: PMC3371400 DOI: 10.1007/s00775-010-0642-1] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2010] [Accepted: 02/06/2010] [Indexed: 11/26/2022]
Abstract
The ruthenium compound KP1019 has demonstrated promising anticancer activity in a pilot clinical trial. This study aims to evaluate the intracellular uptake/binding patterns of KP1019 and its sodium salt KP1339, which is currently in a phase I-IIa study. Although KP1339 tended to be moderately less cytotoxic than KP1019, IC(50) values in several cancer cell models revealed significant correlation of the cytotoxicity profiles, suggesting similar targets for the two drugs. Accordingly, both drugs activated apoptosis, indicated by caspase activation via comparable pathways. Drug uptake determined by inductively coupled plasma mass spectrometry (ICP-MS) was completed after 1 h, corresponding to full cytotoxicity as early as after 3 h of drug exposure. Surprisingly, the total cellular drug uptake did not correlate with cytotoxicity. However, distinct differences in intracellular distribution patterns suggested that the major targets for the two ruthenium drugs are cytosolic rather than nuclear. Consequently, drug-protein binding in cytosolic fractions of drug-treated cells was analyzed by native size-exclusion chromatography (SEC) coupled online with ICP-MS. Ruthenium-protein binding of KP1019- and KP1339-treated cells distinctly differed from the platinum binding pattern observed after cisplatin treatment. An adapted SEC-SEC-ICP-MS system identified large protein complexes/aggregates above 700 kDa as initial major binding partners in the cytosol, followed by ruthenium redistribution to the soluble protein weight fraction below 40 kDa. Taken together, our data indicate that KP1019 and KP1339 rapidly enter tumor cells, followed by binding to larger protein complexes/organelles. The different protein binding patterns as compared with those for cisplatin suggest specific protein targets and consequently a unique mode of action for the ruthenium drugs investigated.
Collapse
Affiliation(s)
- Petra Heffeter
- Department of Medicine I, Institute of Cancer Research, Medical University Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Katharina Böck
- Division of Analytical Chemistry, Department of Chemistry, University of Natural Resources and Applied Life Sciences, BOKU, Vienna, Austria
| | - Bihter Atil
- Department of Medicine I, Institute of Cancer Research, Medical University Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Mir Ali Reza Hoda
- Department of Medicine I, Institute of Cancer Research, Medical University Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Wilfried Körner
- Institute for Geological Sciences, University of Vienna, Vienna, Austria
| | - Caroline Bartel
- Institute of Inorganic Chemistry, University of Vienna, Vienna, Austria
| | - Ute Jungwirth
- Department of Medicine I, Institute of Cancer Research, Medical University Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | | | - Michael Micksche
- Department of Medicine I, Institute of Cancer Research, Medical University Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Walter Berger
- Department of Medicine I, Institute of Cancer Research, Medical University Vienna, Borschkegasse 8a, 1090 Vienna, Austria,
| | - Gunda Koellensperger
- Division of Analytical Chemistry, Department of Chemistry, University of Natural Resources and Applied Life Sciences, BOKU, Vienna, Austria
| |
Collapse
|