1
|
Zarkovic G, Ziegler P, Lee JH, Dresden B, Kumar A, Shuda M, Bäckerholm A, Shair KHY. Disruption of ATR Signaling by Epstein-Barr Virus Latent Membrane Protein 1 Sensitizes Nasopharyngeal Carcinoma Cells to Cisplatin. J Med Virol 2025; 97:e70407. [PMID: 40407079 PMCID: PMC12100692 DOI: 10.1002/jmv.70407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 05/06/2025] [Accepted: 05/08/2025] [Indexed: 05/26/2025]
Abstract
Nasopharyngeal carcinoma (NPC) occurs with high incidence in Southeast Asia where almost all tumors are associated with Epstein-Barr virus (EBV) infection. Cisplatin is used in combination chemotherapy. In this study, we determined that the EBV oncoprotein, latent membrane protein 1 (LMP1), perturbs DNA damage response (DDR) signaling, activation of cell cycle checkpoints, and sensitivity to cisplatin in NPC cells (HK1). Hypersensitivity was validated by LMP1 knockdown and CRISPR/Cas9 targeting in HK1-EBV cells with latent EBV infection. The conserved PxQxT motif (in CTAR1) and Y384 residue (in CTAR2) were required for the hypersensitivity. Inhibition of ATR (VE821 or AZD6738), but not ATM (KU55933 or AZD0156), phenocopied the G1 arrest and hypersensitivity. Attenuation of DDR signaling and hypersensitivity by LMP1 or ATR inhibition was also observed in the C17 NPC cell line with restored stable LMP1 expression. LMP1 expression in NPC tumors is highly variable. Publicly available RNA-sequencing data from microdissected NPC tumors showed that LMP1 expression in the primary tumors was the lowest in cisplatin-treated patients that experienced recurrence. These findings could have clinical significance in stratifying NPC patients such that tumors with limited or variable LMP1 expression might benefit from ATR inhibitor therapy.
Collapse
Affiliation(s)
- Gabriella Zarkovic
- University of Pittsburgh Medical Center (UPMC) Cancer Virology ProgramPittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Phillip Ziegler
- University of Pittsburgh Medical Center (UPMC) Cancer Virology ProgramPittsburghPennsylvaniaUSA
| | - Jennifer Hye‐Rim Lee
- University of Pittsburgh Medical Center (UPMC) Cancer Virology ProgramPittsburghPennsylvaniaUSA
| | - Brooke Dresden
- University of Pittsburgh Medical Center (UPMC) Cancer Virology ProgramPittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of PediatricsUPMC Children's Hospital of PittsburghPittsburghPennsylvaniaUSA
| | - Amit Kumar
- University of Pittsburgh Medical Center (UPMC) Cancer Virology ProgramPittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Masahiro Shuda
- University of Pittsburgh Medical Center (UPMC) Cancer Virology ProgramPittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Alan Bäckerholm
- University of Pittsburgh Medical Center (UPMC) Cancer Virology ProgramPittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Kathy Ho Yen Shair
- University of Pittsburgh Medical Center (UPMC) Cancer Virology ProgramPittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
2
|
Benedetti F, Mongodin EF, Badger JH, Munawwar A, Cellini A, Yuan W, Silvestri G, Kraus CN, Marini S, Rathinam CV, Salemi M, Tettelin H, Gallo RC, Zella D. Bacterial DnaK reduces the activity of anti-cancer drugs cisplatin and 5FU. J Transl Med 2024; 22:269. [PMID: 38475767 PMCID: PMC10935962 DOI: 10.1186/s12967-024-05078-x] [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: 02/16/2024] [Accepted: 03/07/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Chemotherapy is a primary treatment for cancer, but its efficacy is often limited by cancer-associated bacteria (CAB) that impair tumor suppressor functions. Our previous research found that Mycoplasma fermentans DnaK, a chaperone protein, impairs p53 activities, which are essential for most anti-cancer chemotherapeutic responses. METHODS To investigate the role of DnaK in chemotherapy, we treated cancer cell lines with M. fermentans DnaK and then with commonly used p53-dependent anti-cancer drugs (cisplatin and 5FU). We evaluated the cells' survival in the presence or absence of a DnaK-binding peptide (ARV-1502). We also validated our findings using primary tumor cells from a novel DnaK knock-in mouse model. To provide a broader context for the clinical significance of these findings, we investigated human primary cancer sequencing datasets from The Cancer Genome Atlas (TCGA). We identified F. nucleatum as a CAB carrying DnaK with an amino acid composition highly similar to M. fermentans DnaK. Therefore, we investigated the effect of F. nucleatum DnaK on the anti-cancer activity of cisplatin and 5FU. RESULTS Our results show that both M. fermentans and F. nucleatum DnaKs reduce the effectiveness of cisplatin and 5FU. However, the use of ARV-1502 effectively restored the drugs' anti-cancer efficacy. CONCLUSIONS Our findings offer a practical framework for designing and implementing novel personalized anti-cancer strategies by targeting specific bacterial DnaKs in patients with poor response to chemotherapy, underscoring the potential for microbiome-based personalized cancer therapies.
Collapse
Affiliation(s)
- Francesca Benedetti
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Emmanuel F Mongodin
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jonathan H Badger
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, DHHS, Bethesda, MD, USA
| | - Arshi Munawwar
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ashley Cellini
- Pathology Biorepository Shared Service, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD, 21201, USA
| | - Weirong Yuan
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Giovannino Silvestri
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Simone Marini
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
- Department of Epidemiology, University of Florida, Gainesville, FL, USA
| | - Chozha V Rathinam
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Marco Salemi
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
- Department of Pathology, University of Florida, Gainesville, FL, USA
| | - Hervé Tettelin
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Robert C Gallo
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA.
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Davide Zella
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA.
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
3
|
Yin Y, Peng J, Zheng X, Zhou J, Wang Y, Dai Y, Yin G, Tang Y. Extrinsic apoptosis and senescence involved in growth kinetics of seminoma to cisplatin. Clin Exp Pharmacol Physiol 2023; 50:140-148. [PMID: 36222180 DOI: 10.1111/1440-1681.13730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 09/15/2022] [Accepted: 10/07/2022] [Indexed: 01/05/2023]
Abstract
Seminoma is the most common type of testicular germ cell tumour and is highly sensitive to cisplatin therapy, which has not been fully elucidated. In this study, we comprehensively monitored dynamic changes of TCam-2 cells after cisplatin treatment. At an early stage, we found that both low and high concentrations of cisplatin induced the S-phase arrest in TCam-2 cells. By contrast, the G0G1 arrest was caused by cisplatin in teratoma NTERA-2 cells. Afterwards, high concentrations of cisplatin promoted the extrinsic apoptosis and high expressions of related genes (Fas/FasL-caspase-8/-3) in TCam-2 cells. However, when decreasing the cisplatin, the apoptotic cells were significantly reduced, and accompanied by cells showing senescence-like morphology, positive SA-β-gal staining and up-regulation of senescence-related genes (p53, p21 and p16). Furthermore, the cell cycle analysis revealed that most of the senescent TCam-2 cells were irreversibly arrested in the G2M phase. G2M arrest was also observed in NTERA-2 cells treated with a low concentration of cisplatin, while no senescence-related phenotype was discovered. In addition, we detected the γ-H2AX, a DNA damage marker protein, and reactive oxygen species (ROS) and found both of which were elevated with the increase of cisplatin in TCam-2 cells. In conclusion, the extrinsic apoptosis and senescence are involved in the growth kinetics of TCam-2 cells to cisplatin, which provide some implications for studies on cisplatin and seminoma.
Collapse
Affiliation(s)
- Yinghao Yin
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jingxuan Peng
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Xiaoping Zheng
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jun Zhou
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yong Wang
- Department of Urology Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Yingbo Dai
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Guangming Yin
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yuxin Tang
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China.,Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.,Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| |
Collapse
|
4
|
Cisplatin toxicity in the developing brain displays an absolute requirement for caspase-3. Exp Neurol 2022; 351:114010. [DOI: 10.1016/j.expneurol.2022.114010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 01/26/2022] [Accepted: 02/08/2022] [Indexed: 11/19/2022]
|
5
|
Shi YY, Meng XT, Xu YN, Tian XJ. Role of FOXO protein's abnormal activation through PI3K/AKT pathway in platinum resistance of ovarian cancer. J Obstet Gynaecol Res 2021; 47:1946-1957. [PMID: 33827148 DOI: 10.1111/jog.14753] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 02/04/2021] [Accepted: 03/05/2021] [Indexed: 01/12/2023]
Abstract
AIM Platinum-based chemotherapy is the standard treatment for ovarian cancer. However, tumor cells' resistance to platinum drugs often occurs. This paper provides a review of Forkhead box O (FOXO) protein's role in platinum resistance of ovarian cancer which hopefully may provide some further guidance for the treatment of platinum-resistant ovarian cancer. METHODS We reviewed a 128 published papers from authoritative and professional journals on FOXO and platinum-resistant ovarian cancer, and adopts qualitative analyses and interpretation based on the literature. RESULTS Ovarian cancer often has abnormal activation of cellular pathways, the most important of which is the PI3K/AKT pathway. FOXOs act as crucial downstream factor of the PI3K/Akt pathway and are negatively regulated by it. DNA damage response and apoptosis including the relationship between FOXOs and ATM-Chk2-p53 are essential for platinum resistance of ovarian cancer. Through gene expression analysis in platinum-resistant ovarian cancer cell model, it was found that FoxO-1 is decreased in platinum-resistant ovarian cancer, so studying the role of FOXO in the pathway on platinum-induced apoptosis may further guide the treatment of platinum-resistant ovarian cancer. CONCLUSIONS There are many drug resistance mechanisms in ovarian cancer, wherein the decrease in cancer cells apoptosis is one of the important causes. Constituted by a series of transcription factors evolving conservatively and mainly working in inhibiting cancer, FOXO proteins play various roles in cells' antitumor response. More and more evidence suggests that we need to re-understand the role that FOXOs have played in cancer development and treatment.
Collapse
Affiliation(s)
- Yun-Yue Shi
- Department of Obstetrics and gynecology, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiang-Tian Meng
- Department of Ophthalmology, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Ya-Nan Xu
- Department of Obstetrics and gynecology, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiu-Juan Tian
- Department of Obstetrics and gynecology, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
6
|
Aneuploidy increases resistance to chemotherapeutics by antagonizing cell division. Proc Natl Acad Sci U S A 2020; 117:30566-30576. [PMID: 33203674 DOI: 10.1073/pnas.2009506117] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aneuploidy, defined as whole chromosome gains and losses, is associated with poor patient prognosis in many cancer types. However, the condition causes cellular stress and cell cycle delays, foremost in G1 and S phase. Here, we investigate how aneuploidy causes both slow proliferation and poor disease outcome. We test the hypothesis that aneuploidy brings about resistance to chemotherapies because of a general feature of the aneuploid condition-G1 delays. We show that single chromosome gains lead to increased resistance to the frontline chemotherapeutics cisplatin and paclitaxel. Furthermore, G1 cell cycle delays are sufficient to increase chemotherapeutic resistance in euploid cells. Mechanistically, G1 delays increase drug resistance to cisplatin and paclitaxel by reducing their ability to damage DNA and microtubules, respectively. Finally, we show that our findings are clinically relevant. Aneuploidy correlates with slowed proliferation and drug resistance in the Cancer Cell Line Encyclopedia (CCLE) dataset. We conclude that a general and seemingly detrimental effect of aneuploidy, slowed proliferation, provides a selective benefit to cancer cells during chemotherapy treatment.
Collapse
|
7
|
Coverdale JC, Bridgewater HE, Song JI, Smith NA, Barry NPE, Bagley I, Sadler PJ, Romero-Canelón I. In Vivo Selectivity and Localization of Reactive Oxygen Species (ROS) Induction by Osmium Anticancer Complexes That Circumvent Platinum Resistance. J Med Chem 2018; 61:9246-9255. [PMID: 30230827 PMCID: PMC6204601 DOI: 10.1021/acs.jmedchem.8b00958] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Indexed: 12/21/2022]
Abstract
Platinum drugs are widely used for cancer treatment. Other precious metals are promising, but their clinical progress depends on achieving different mechanisms of action to overcome Pt-resistance. Here, we evaluate 13 organo-Os complexes: 16-electron sulfonyl-diamine catalysts [(η6-arene)Os( N, N')], and 18-electron phenylazopyridine complexes [(η6-arene)Os( N, N')Cl/I]+ (arene = p-cymene, biphenyl, or terphenyl). Their antiproliferative activity does not depend on p21 or p53 status, unlike cisplatin, and their selective potency toward cancer cells involves the generation of reactive oxygen species. Evidence of such a mechanism of action has been found both in vitro and in vivo. This work appears to provide the first study of osmium complexes in the zebrafish model, which has been shown to closely model toxicity in humans. A fluorescent osmium complex, derived from a lead compound, was employed to confirm internalization of the complex, visualize in vivo distribution, and confirm colocalization with reactive oxygen species generated in zebrafish.
Collapse
Affiliation(s)
| | | | - Ji-Inn Song
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
| | - Nichola A. Smith
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
| | - Nicolas P. E. Barry
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
- School
of Chemistry and Biosciences, University
of Bradford, Bradford BD7 1DP, U.K.
| | - Ian Bagley
- BSU
Research Technology Platform, University
of Warwick, Coventry CV4 7AL, U.K.
| | - Peter J. Sadler
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
| | - Isolda Romero-Canelón
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
- School
of Pharmacy, University of Birmingham, Birmingham B15 2TT, U.K.
| |
Collapse
|
8
|
Synergistic Antitumor Effect of Oligogalacturonides and Cisplatin on Human Lung Cancer A549 Cells. Int J Mol Sci 2018; 19:ijms19061769. [PMID: 29903991 PMCID: PMC6032352 DOI: 10.3390/ijms19061769] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 06/07/2018] [Accepted: 06/12/2018] [Indexed: 11/21/2022] Open
Abstract
Cisplatin (DPP), a clinically potent antineoplastic agent, is limited by its severe adverse effects. The aim of this study was to investigate the effect of oligogalacturonides (OGA) and DDP on human lung cancer A549 cells. The combined use of OGA and DDP had a synergistic effect on the growth inhibition of A549 cells, changed the cell cycle distribution, and enhanced apoptotic response, especially in sequential combination treatment group of DDP 12 h + OGA 12 h. Western blot analyses showed that the combination treatment of OGA and DDP upregulated Bax, p53, and Caspase-3 and downregulated Bcl-2 proteins. More importantly, DDP-induced toxicity was attenuated by OGA and DDP combination treatment in normal HEK293 cells. Our data suggests that the combined use of OGA from natural sources and DDP could be an important new adjuvant therapy for lung cancer as well as offer important insights for reducing kidney toxicity of DDP and delaying the development of DDP resistance.
Collapse
|
9
|
Bado I, Pham E, Soibam B, Nikolos F, Gustafsson JÅ, Thomas C. ERβ alters the chemosensitivity of luminal breast cancer cells by regulating p53 function. Oncotarget 2018; 9:22509-22522. [PMID: 29854295 PMCID: PMC5976481 DOI: 10.18632/oncotarget.25147] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 03/21/2018] [Indexed: 01/13/2023] Open
Abstract
Estrogen receptor α (ERα)-positive breast cancers tend to develop resistance to both endocrine therapy and chemotherapy. Despite recent progress in defining molecular pathways that confer endocrine resistance, the mechanisms that regulate chemotherapy response in luminal tumors remain largely elusive. Luminal tumors often express wild-type p53 that is a major determinant of the cellular DNA damage response. Similar to p53, the second ER subtype, ERβ, has been reported to inhibit breast tumorigenesis by acting alone or in collaboration with p53. However, a synergistic mechanism of action has not been described. Here, we suggest that ERβ relies on p53 to elicit its tumor repressive actions in ERα-positive breast cancer cells. Upregulation of ERβ and treatment with ERβ agonists potentiates the tumor suppressor function of p53 resulting in decreased survival. This effect requires molecular interaction between the two proteins that disrupts the inhibitory action of ERα on p53 leading to increased transcriptional activity of p53. In addition, we show that the same interaction alters the chemosensitivity of endocrine-resistant cells including their response to tamoxifen therapy. Our results suggest a collaboration of ERβ and p53 tumor suppressor activity in breast cancer cells that indicates the importance of ligand-regulated ERβ as a tool to target p53 activity and improve the clinical management of resistant disease.
Collapse
Affiliation(s)
- Igor Bado
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas, USA
| | - Eric Pham
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Benjamin Soibam
- Department of Computer Science and Engineering Technology, University of Houston-Downtown, Huston, Texas, USA
| | - Fotis Nikolos
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas, USA
| | - Jan-Åke Gustafsson
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas, USA
- Center for Innovative Medicine, Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Christoforos Thomas
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas, USA
| |
Collapse
|
10
|
Willis AJ, Indra R, Wohak LE, Sozeri O, Feser K, Mrizova I, Phillips DH, Stiborova M, Arlt VM. The impact of chemotherapeutic drugs on the CYP1A1-catalysed metabolism of the environmental carcinogen benzo[a]pyrene: Effects in human colorectal HCT116 TP53(+/+), TP53(+/-) and TP53(-/-) cells. Toxicology 2018; 398-399:1-12. [PMID: 29471073 PMCID: PMC6593262 DOI: 10.1016/j.tox.2018.02.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/29/2018] [Accepted: 02/16/2018] [Indexed: 02/02/2023]
Abstract
Polycyclic aromatic hydrocarbons such as benzo[a]pyrene (BaP) can induce cytochrome P450 1A1 (CYP1A1) via a p53-dependent mechanism. The effect of different p53-activating chemotherapeutic drugs on CYP1A1 expression, and the resultant effect on BaP metabolism, was investigated in a panel of isogenic human colorectal HCT116 cells with differing TP53 status. Cells that were TP53(+/+), TP53(+/-) or TP53(-/-) were treated for up to 48 h with 60 μM cisplatin, 50 μM etoposide or 5 μM ellipticine, each of which caused high p53 induction at moderate cytotoxicity (60-80% cell viability). We found that etoposide and ellipticine induced CYP1A1 in TP53(+/+) cells but not in TP53(-/-) cells, demonstrating that the mechanism of CYP1A1 induction is p53-dependent; cisplatin had no such effect. Co-incubation experiments with the drugs and 2.5 μM BaP showed that: (i) etoposide increased CYP1A1 expression in TP53(+/+) cells, and to a lesser extent in TP53(-/-) cells, compared to cells treated with BaP alone; (ii) ellipticine decreased CYP1A1 expression in TP53(+/+) cells in BaP co-incubations; and (iii) cisplatin did not affect BaP-mediated CYP1A1 expression. Further, whereas cisplatin and etoposide had virtually no influence on CYP1A1-catalysed BaP metabolism, ellipticine treatment strongly inhibited BaP bioactivation. Our results indicate that the underlying mechanisms whereby etoposide and ellipticine regulate CYP1A1 expression must be different and may not be linked to p53 activation alone. These results could be relevant for smokers, who are exposed to increased levels of BaP, when prescribing chemotherapeutic drugs. Beside gene-environment interactions, more considerations should be given to potential drug-environment interactions during chemotherapy.
Collapse
Affiliation(s)
- Alexandra J Willis
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Radek Indra
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40 Prague 2, Czech Republic
| | - Laura E Wohak
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Osman Sozeri
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Kerstin Feser
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Iveta Mrizova
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40 Prague 2, Czech Republic
| | - David H Phillips
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom; NIHR Health Protection Research Unit in Health Impact of Environmental Hazards at King's College London in partnership with Public Health England, London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Marie Stiborova
- Department of Biochemistry, Faculty of Science, Charles University, Albertov 2030, 128 40 Prague 2, Czech Republic
| | - Volker M Arlt
- Department of Analytical, Environmental and Forensic Sciences, MRC-PHE Centre for Environment and Health, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom; NIHR Health Protection Research Unit in Health Impact of Environmental Hazards at King's College London in partnership with Public Health England, London, 150 Stamford Street, London SE1 9NH, United Kingdom.
| |
Collapse
|
11
|
Ullah K, Rosendahl AH, Izzi V, Bergmann U, Pihlajaniemi T, Mäki JM, Myllyharju J. Hypoxia-inducible factor prolyl-4-hydroxylase-1 is a convergent point in the reciprocal negative regulation of NF-κB and p53 signaling pathways. Sci Rep 2017; 7:17220. [PMID: 29222481 PMCID: PMC5722952 DOI: 10.1038/s41598-017-17376-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 11/24/2017] [Indexed: 12/14/2022] Open
Abstract
Hypoxia-inducible factor 1α (HIF1α) induces the expression of several hundred genes in hypoxia aiming at restoration of oxygen homeostasis. HIF prolyl-4-hydroxylases (HIF-P4Hs) regulate the stability of HIF1α in an oxygen-dependent manner. Hypoxia is a common feature in inflammation and cancer and the HIF pathway is closely linked with the inflammatory NF-κB and tumor suppressor p53 pathways. Here we show that genetic inactivation or chemical inhibition of HIF-P4H-1 leads to downregulation of proinflammatory genes, while proapoptotic genes are upregulated. HIF-P4H-1 inactivation reduces the inflammatory response under LPS stimulus in vitro and in an acute skin inflammation model in vivo. Furthermore, HIF-P4H-1 inactivation increases p53 activity and stability and hydroxylation of proline 142 in p53 has an important role in this regulation. Altogether, our data suggest that HIF-P4H-1 inhibition may be a promising therapeutic candidate for inflammatory diseases and cancer, enhancing the reciprocal negative regulation of the NF-κB and p53 pathways.
Collapse
Affiliation(s)
- Karim Ullah
- Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, FIN-90014, Finland.,Biocenter Oulu, University of Oulu, Oulu, FIN-90014, Finland.,Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, FIN-90014, Finland
| | - Ann-Helen Rosendahl
- Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, FIN-90014, Finland.,Biocenter Oulu, University of Oulu, Oulu, FIN-90014, Finland.,Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, FIN-90014, Finland
| | - Valerio Izzi
- Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, FIN-90014, Finland.,Biocenter Oulu, University of Oulu, Oulu, FIN-90014, Finland.,Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, FIN-90014, Finland
| | - Ulrich Bergmann
- Biocenter Oulu, University of Oulu, Oulu, FIN-90014, Finland
| | - Taina Pihlajaniemi
- Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, FIN-90014, Finland.,Biocenter Oulu, University of Oulu, Oulu, FIN-90014, Finland.,Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, FIN-90014, Finland
| | - Joni M Mäki
- Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, FIN-90014, Finland.,Biocenter Oulu, University of Oulu, Oulu, FIN-90014, Finland.,Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, FIN-90014, Finland
| | - Johanna Myllyharju
- Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, FIN-90014, Finland. .,Biocenter Oulu, University of Oulu, Oulu, FIN-90014, Finland. .,Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, FIN-90014, Finland.
| |
Collapse
|
12
|
Spreckelmeyer S, Estrada-Ortiz N, Prins GGH, van der Zee M, Gammelgaard B, Stürup S, de Graaf IAM, Groothuis GMM, Casini A. On the toxicity and transport mechanisms of cisplatin in kidney tissues in comparison to a gold-based cytotoxic agent. Metallomics 2017; 9:1786-1795. [PMID: 29104982 DOI: 10.1039/c7mt00271h] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mechanisms of toxicity and cellular transport of anticancer metallodrugs, including platinum-based agents, have not yet been fully elucidated. Here, we studied the toxic effects and accumulation mechanisms of cisplatin in healthy rat kidneys ex vivo, using the Precision Cut Tissue Slices (PCTS) method. In addition, for the first time, we investigated the nephrotoxic effects of an experimental anticancer cyclometallated complex [Au(pyb-H)(PTA)Cl]PF6 (PTA = 1,3,5-triazaphosphaadamantane). The viability of the kidney slices after metallodrug treatment was evaluated by ATP content determination and histomorphology analysis. A concentration dependent decrease in viability of PCKS was observed after exposure to cisplatin or the Au(iii) complex, which correlated with the increase in slice content of Pt and Au, respectively. Metal accumulation in kidney slices was analysed by ICP-MS. The involvement of OCTs and MATE transporters in the accumulation of both metal compounds in kidneys was evaluated co-incubating the tissues with cimitedine, inhibitor of OCT and MATE. Studies of mRNA expression of the markers KIM-1, villin, p53 and Bax showed that cisplatin damages proximal tubules, whereas the Au(iii) complex preferentially affects the distal tubules. However, no effect of cimetidine on the toxicity or accumulation of cisplatin and the Au(iii) complex was observed. The effect of temperature on metallodrug accumulation in kidneys suggests the involvement of a carrier-mediated uptake process, other than OCT2, for cisplatin; while carrier-mediated excretion was suggested in the cases of the Au(iii) complex.
Collapse
Affiliation(s)
- Sarah Spreckelmeyer
- Dept. Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713AV Groningen, The Netherlands and Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| | - Natalia Estrada-Ortiz
- Dept. Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713AV Groningen, The Netherlands
| | - Gerian G H Prins
- Dept. Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713AV Groningen, The Netherlands
| | - Margot van der Zee
- Dept. Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713AV Groningen, The Netherlands
| | - Bente Gammelgaard
- Dept. of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Stefan Stürup
- Dept. of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Inge A M de Graaf
- Dept. Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713AV Groningen, The Netherlands
| | - Geny M M Groothuis
- Dept. Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713AV Groningen, The Netherlands
| | - Angela Casini
- Dept. Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713AV Groningen, The Netherlands and School of Chemistry, Cardiff University, Main Building, Park Place, CF103AT Cardiff, UK.
| |
Collapse
|
13
|
Estrada-Ortiz N, Guarra F, de Graaf IAM, Marchetti L, de Jager MH, Groothuis GMM, Gabbiani C, Casini A. Anticancer Gold N-Heterocyclic Carbene Complexes: A Comparative in vitro and ex vivo Study. ChemMedChem 2017; 12:1429-1435. [PMID: 28741878 DOI: 10.1002/cmdc.201700316] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/25/2017] [Indexed: 12/15/2022]
Abstract
A series of organometallic AuI N-heterocyclic carbene (NHC) complexes was synthesized and characterized for anticancer activity in four human cancer cell lines. The compounds' toxicity in healthy tissue was determined using precision-cut kidney slices (PCKS) as a tool to determine the potential selectivity of the gold complexes ex vivo. All evaluated compounds presented cytotoxic activity toward the cancer cells in the nano- or low micromolar range. The mixed AuI NHC complex, (tert-butylethynyl)-1,3-bis-(2,6-diisopropylphenyl)imidazol-2-ylidene gold(I), bearing an alkynyl moiety as ancillary ligand, showed high cytotoxicity in cancer cells in vitro, while being barely toxic in healthy rat kidney tissues. The obtained results open new perspectives toward the design of mixed NHC-alkynyl gold complexes for cancer therapy.
Collapse
Affiliation(s)
- Natalia Estrada-Ortiz
- Department of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713, AV, Groningen, The Netherlands
| | - Federica Guarra
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Moruzzi, 3, 56124, Pisa, Italy
| | - Inge A M de Graaf
- Department of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713, AV, Groningen, The Netherlands
| | - Lorella Marchetti
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Moruzzi, 3, 56124, Pisa, Italy
| | - Marina H de Jager
- Department of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713, AV, Groningen, The Netherlands
| | - Geny M M Groothuis
- Department of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713, AV, Groningen, The Netherlands
| | - Chiara Gabbiani
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Moruzzi, 3, 56124, Pisa, Italy
| | - Angela Casini
- Department of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713, AV, Groningen, The Netherlands.,School of Chemistry, Cardiff University, Main Building, Park Place, CF103AT, Cardiff, UK
| |
Collapse
|
14
|
Skwarska A, Ramachandran S, Dobrynin G, Leszczynska KB, Hammond EM. The imidazoacridinone C-1311 induces p53-dependent senescence or p53-independent apoptosis and sensitizes cancer cells to radiation. Oncotarget 2017; 8:31187-31198. [PMID: 28415717 PMCID: PMC5458200 DOI: 10.18632/oncotarget.16102] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 03/01/2017] [Indexed: 01/01/2023] Open
Abstract
C-1311 is a small molecule, which has shown promise in a number of pre-clinical and clinical studies. However, the biological response to C-1311 exposure is complicated and has been reported to involve a number of cell fates. Here, we investigated the molecular signaling which determines the response to C-1311 in both cancer and non-cancer cell lines. For the first time we demonstrate that the tumor suppressor, p53 plays a key role in cell fate determination after C-1311 treatment. In the presence of wild-type p53, cells exposed to C-1311 entered senescence. In contrast, cells lines without functional p53 underwent mitotic catastrophe and apoptosis. C-1311 also induced autophagy in a non-p53-dependent manner. Cells in hypoxic conditions also responded to C-1311 in a p53-dependent manner, suggesting that our observations are physiologically relevant. Most importantly, we show that C-1311 can be effectively combined with radiation to improve the radiosensitivity of a panel of cancer cell lines. Together, our data suggest that C-1311 warrants further clinical testing in combination with radiotherapy for the treatment of solid tumors.
Collapse
Affiliation(s)
- Anna Skwarska
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, The University of Oxford, Oxford, UK
- Department of Pharmaceutical Technology and Biochemistry, Chemical Faculty, Gdańsk University of Technology, Gdańsk, Poland
| | - Shaliny Ramachandran
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, The University of Oxford, Oxford, UK
| | - Grzegorz Dobrynin
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, The University of Oxford, Oxford, UK
| | - Katarzyna B. Leszczynska
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, The University of Oxford, Oxford, UK
| | - Ester M. Hammond
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, The University of Oxford, Oxford, UK
| |
Collapse
|
15
|
Jo DW, Kim YK, Yun PY. The influence of p53 mutation status on the anti-cancer effect of cisplatin in oral squamous cell carcinoma cell lines. J Korean Assoc Oral Maxillofac Surg 2016; 42:337-344. [PMID: 28053903 PMCID: PMC5206238 DOI: 10.5125/jkaoms.2016.42.6.337] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 10/05/2016] [Accepted: 10/07/2016] [Indexed: 01/08/2023] Open
Abstract
Objectives The purpose of this study was to evaluate the anti-cancer activity of cisplatin by studying its effects on cell viability and identifying the mechanisms underlying the induction of cell cycle arrest and apoptosis on oral squamous cell carcinoma (OSCC) cell lines with varying p53 mutation status. Materials and Methods Three OSCC cell lines, YD-8 (p53 point mutation), YD-9 (p53 wild type), and YD-38 (p53 deletion) were used. To determine the cytotoxic effect of cisplatin, MTS assay was performed. The cell cycle alteration and apoptosis were analyzed using flow cytometry. Western blot analysis was used to detect the expression of cell cycle alteration- or apoptosis-related proteins as well as p53. Results Cisplatin showed a time- and dose-dependent anti-proliferative effect in all cell lines. Cisplatin induced G2/M cell accumulation in the three cell lines after treatment with 0.5 and 1.0 µg/mL of cisplatin for 48 hours. The proportion of annexin V-FITC-stained cells increased following treatment with cisplatin. The apoptotic proportion was lower in the YD-38 cell line than in the YD-9 or YD-8 cell lines. Also, immunoblotting analysis indicated that p53 and p21 were detected only in YD-8 and YD-9 cell lines after cisplatin treatment. Conclusion In this study, cisplatin showed anti-cancer effects via G2/M phase arrest and apoptosis, with some difference among OSCC cell lines. The mutation status of p53 might have influenced the difference observed among cell lines. Further studies on p53 mutation status are needed to understand the biological behavior and characteristics of OSCCs and to establish appropriate treatment.
Collapse
Affiliation(s)
- Deuk-Won Jo
- Department of Prosthodontics, Section of Dentistry, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Young-Kyun Kim
- Department of Oral and Maxillofacial Surgery, Section of Dentistry, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Pil-Young Yun
- Department of Oral and Maxillofacial Surgery, Section of Dentistry, Seoul National University Bundang Hospital, Seongnam, Korea
| |
Collapse
|
16
|
Flores ER. Commentary on “Apoptosis, p53, and Tumor Cell Sensitivity to Anticancer Agents”. Cancer Res 2016; 76:6763-6764. [DOI: 10.1158/0008-5472.can-16-2997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 11/02/2016] [Indexed: 11/16/2022]
|
17
|
Guanidine Alkaloids from the Marine Sponge Monanchora pulchra Show Cytotoxic Properties and Prevent EGF-Induced Neoplastic Transformation in Vitro. Mar Drugs 2016; 14:md14070133. [PMID: 27428983 PMCID: PMC4962023 DOI: 10.3390/md14070133] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/05/2016] [Accepted: 07/08/2016] [Indexed: 12/13/2022] Open
Abstract
Guanidine alkaloids from sponges Monanchora spp. represent diverse bioactive compounds, however, the mechanisms underlying bioactivity are very poorly understood. Here, we report results of studies on cytotoxic action, the ability to inhibit EGF-induced neoplastic transformation, and the effects on MAPK/AP-1 signaling of eight rare guanidine alkaloids, recently isolated from the marine sponge Monanchora pulchra, namely: monanchocidin A (1), monanchocidin B (2), monanchomycalin C (3), ptilomycalin A (4), monanchomycalin B (5), normonanchocidin D (6), urupocidin A (7), and pulchranin A (8). All of the compounds induced cell cycle arrest (apart from 8) and programmed death of cancer cells. Ptilomycalin A-like compounds 1–6 activated JNK1/2 and ERK1/2, following AP-1 activation and caused p53-independent programmed cell death. Compound 7 induced p53-independent cell death without activation of AP-1 or caspase-3/7, and the observed JNK1/2 activation did not contribute to the cytotoxic effect of the compound. Alkaloid 8 induced JNK1/2 (but not ERK1/2) activation leading to p53-independent cell death and strong suppression of AP-1 activity. Alkaloids 1–4, 7, and 8 were able to inhibit the EGF-induced neoplastic transformation of JB6 P+ Cl41 cells. Our results suggest that investigated guanidine marine alkaloids hold potential to eliminate human cancer cells and prevent cancer cell formation and spreading.
Collapse
|
18
|
Bose RN, Moghaddas S, Belkacemi L, Tripathi S, Adams NR, Majmudar P, McCall K, Dezvareh H, Nislow C. Absence of Activation of DNA Repair Genes and Excellent Efficacy of Phosphaplatins against Human Ovarian Cancers: Implications To Treat Resistant Cancers. J Med Chem 2015; 58:8387-401. [PMID: 26455832 DOI: 10.1021/acs.jmedchem.5b00732] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Phosphaplatins, platinum(II) and platinum(IV) complexes coordinated to a pyrophosphate moiety, exhibit excellent antitumor activities against a variety of cancers. To determine whether phosphaplatins trigger resistance to treatment by engaging DNA damage repair genes, a yeast genome-wide fitness assay was used. Treatment of yeast cells with pyrodach-2 (D2) or pyrodach-4 (D4) revealed no particular sensitivity to nucleotide excision repair, homologous recombination repair, or postreplication repair when compared with platin control compounds. Also, TNF receptor superfamily member 6 (FAS) protein was overexpressed in phosphaplatin-treated ovarian tumor cells, and platinum colocalized with FAS protein in lipid rafts. An overactivation of sphingomyelinase (ASMase) was noted in the treated cells, indicating participation of an extrinsic apoptotic mechanism due to increased ceramide release. Our results indicate that DNA is not the target of phosphaplatins and accordingly, that phosphaplatins might not cause resistance to treatment. Activation of ASMase and FAS along with the colocalization of platinum with FAS in lipid rafts support an extrinsic apoptotic signaling mechanism that is mediated by phosphaplatins.
Collapse
Affiliation(s)
- Rathindra N Bose
- Departments of Biology and Biochemistry, University of Houston , Houston, Texas 77204, United States
| | - Shadi Moghaddas
- Departments of Biology and Biochemistry, University of Houston , Houston, Texas 77204, United States
| | - Louiza Belkacemi
- Departments of Biology and Biochemistry, University of Houston , Houston, Texas 77204, United States
| | - Swarnendu Tripathi
- Departments of Biology and Biochemistry, University of Houston , Houston, Texas 77204, United States
| | - Nyssa R Adams
- Department of Specialty Medicine, Ohio University Heritage College of Osteopathic Medicine, Ohio University , Athens, Ohio 45701, United States
| | - Pooja Majmudar
- Department of Specialty Medicine, Ohio University Heritage College of Osteopathic Medicine, Ohio University , Athens, Ohio 45701, United States
| | - Kelly McCall
- Department of Specialty Medicine, Ohio University Heritage College of Osteopathic Medicine, Ohio University , Athens, Ohio 45701, United States
| | - Homa Dezvareh
- Departments of Biology and Biochemistry, University of Houston , Houston, Texas 77204, United States
| | - Corey Nislow
- Department of Pharmaceutical Sciences, University of British Columbia , Vancouver, British Columbia V6T 1Z3, Canada
| |
Collapse
|
19
|
Markowska A, Kasprzak B, Jaszczyńska-Nowinka K, Lubin J, Markowska J. Noble metals in oncology. Contemp Oncol (Pozn) 2015; 19:271-5. [PMID: 26557773 PMCID: PMC4631304 DOI: 10.5114/wo.2015.54386] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 03/05/2015] [Accepted: 07/20/2015] [Indexed: 11/17/2022] Open
Abstract
Worldwide research groups are searching for anticancer compounds, many of them are organometalic complexes having platinum group metals as their active centers. Most commonly used cytostatics from this group are cisplatin, carboplatin and oxaliplatin. Cisplatin was used fot the first time in 1978, from this time many platinum derivatives were created. In this review we present biological properties and probable future clinical use of platinum, gold, silver, iridium and ruthenium derivatives. Gold derivative Auranofin has been studied extensively. Action of silver nanoparticles on different cell lines was analysed. Iridium isotopes are commonly used in brachyterapy. Ruthenium compound new anti-tumour metastasis inhibitor (NAMI-A) is used in managing lung cancer metastases. Electroporation of another ruthenium based compound KP1339 was also studied. Most of described complexes have antiproliferative and proapoptotic properties. Further studies need to be made. Nevertheless noble metal based chemotherapheutics and compounds seem to be an interesting direction of research.
Collapse
Affiliation(s)
- Anna Markowska
- Perinatology and Gynecology Department, Poznan University of Medical Sciences, Poznan, Poland
| | | | | | - Jolanta Lubin
- University Hospital of Lord's Transfiguration, Poznan, Poland
| | - Janina Markowska
- Division of Gynecology, Department of Oncology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
20
|
Choi YM, Kim HK, Shim W, Anwar MA, Kwon JW, Kwon HK, Kim HJ, Jeong H, Kim HM, Hwang D, Kim HS, Choi S. Mechanism of Cisplatin-Induced Cytotoxicity Is Correlated to Impaired Metabolism Due to Mitochondrial ROS Generation. PLoS One 2015; 10:e0135083. [PMID: 26247588 PMCID: PMC4527592 DOI: 10.1371/journal.pone.0135083] [Citation(s) in RCA: 203] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 07/16/2015] [Indexed: 12/18/2022] Open
Abstract
The chemotherapeutic use of cisplatin is limited by its severe side effects. In this study, by conducting different omics data analyses, we demonstrated that cisplatin induces cell death in a proximal tubular cell line by suppressing glycolysis- and tricarboxylic acid (TCA)/mitochondria-related genes. Furthermore, analysis of the urine from cisplatin-treated rats revealed the lower expression levels of enzymes involved in glycolysis, TCA cycle, and genes related to mitochondrial stability and confirmed the cisplatin-related metabolic abnormalities. Additionally, an increase in the level of p53, which directly inhibits glycolysis, has been observed. Inhibition of p53 restored glycolysis and significantly reduced the rate of cell death at 24 h and 48 h due to p53 inhibition. The foremost reason of cisplatin-related cytotoxicity has been correlated to the generation of mitochondrial reactive oxygen species (ROS) that influence multiple pathways. Abnormalities in these pathways resulted in the collapse of mitochondrial energy production, which in turn sensitized the cells to death. The quenching of ROS led to the amelioration of the affected pathways. Considering these observations, it can be concluded that there is a significant correlation between cisplatin and metabolic dysfunctions involving mROS as the major player.
Collapse
Affiliation(s)
- Yong-Min Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, 443–749, Korea
| | - Han-Kyul Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 443–749, Korea
| | - Wooyoung Shim
- Department of Molecular Science and Technology, Ajou University, Suwon, 443–749, Korea
| | - Muhammad Ayaz Anwar
- Department of Molecular Science and Technology, Ajou University, Suwon, 443–749, Korea
| | - Ji-Woong Kwon
- Department of Molecular Science and Technology, Ajou University, Suwon, 443–749, Korea
| | - Hyuk-Kwon Kwon
- Department of Molecular Science and Technology, Ajou University, Suwon, 443–749, Korea
| | - Hyung Joong Kim
- Division of Energy Systems Research, Ajou University, Suwon, 443–749, Korea
| | - Hyobin Jeong
- School of Interdisciplinary Bioscience and Bioengineering, POSTECH, Pohang, 790–784, Korea
| | - Hwan Myung Kim
- Division of Energy Systems Research, Ajou University, Suwon, 443–749, Korea
| | - Daehee Hwang
- School of Interdisciplinary Bioscience and Bioengineering, POSTECH, Pohang, 790–784, Korea
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, 440–746, Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, 443–749, Korea
| |
Collapse
|
21
|
Karasawa T, Steyger PS. An integrated view of cisplatin-induced nephrotoxicity and ototoxicity. Toxicol Lett 2015; 237:219-27. [PMID: 26101797 DOI: 10.1016/j.toxlet.2015.06.012] [Citation(s) in RCA: 357] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 06/06/2015] [Accepted: 06/14/2015] [Indexed: 12/20/2022]
Abstract
Cisplatin is one of the most widely-used drugs to treat cancers. However, its nephrotoxic and ototoxic side-effects remain major clinical limitations. Recent studies have improved our understanding of the molecular mechanisms of cisplatin-induced nephrotoxicity and ototoxicity. While cisplatin binding to DNA is the major cytotoxic mechanism in proliferating (cancer) cells, nephrotoxicity and ototoxicity appear to result from toxic levels of reactive oxygen species and protein dysregulation within various cellular compartments. In this review, we discuss molecular mechanisms of cisplatin-induced nephrotoxicity and ototoxicity. We also discuss potential clinical strategies to prevent nephrotoxicity and ototoxicity and their current limitations.
Collapse
Affiliation(s)
- Takatoshi Karasawa
- Oregon Hearing Research Center, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, United States
| | - Peter S Steyger
- Oregon Hearing Research Center, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, United States.
| |
Collapse
|
22
|
Chen JJ, Liu SX, Chen MZ, Zhao ZY. Has‑miR‑125a and 125b are induced by treatment with cisplatin in nasopharyngeal carcinoma and inhibit apoptosis in a p53‑dependent manner by targeting p53 mRNA. Mol Med Rep 2015; 12:3569-3574. [PMID: 26017674 DOI: 10.3892/mmr.2015.3863] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 04/22/2015] [Indexed: 02/05/2023] Open
Abstract
MicroRNA (miRNA) is a class of non‑coding RNA, which targets mRNAs of interest and suppresses its expression by degradation or translational inhibition. miRNA (miR)‑125a and miR‑125b were previously demonstrated to translationally and transcriptionally inhibit the expression of p53. The observed downregulation of the protein level of p53 in cisplatin‑treated patients with nasopharyngeal carcinoma (NPC) indicates the association between cisplatin resistance, miR‑125a and miR‑125b. In the present study, through the detection of the expression levels of miR‑125a and miR‑125b, a significant upregulation of these miRs was demonstrated in cisplatin‑treated patients with NPC. As a consequence, the protein expression level of p53 decreased notably. To confirm the induction of miR‑125a and miR‑125b by treatment with cisplatin, a cisplatin‑resistant TW03 cell model (TW03/DDP) was constructed. As expected, in the TW03/DDP cells, the expression levels of miR‑125a and miR‑125b were upregulated, and this caused downregulation of p53. Ectopic expression of these miRNAs in the TW03 cell model sensitized TW03 to cisplatin by decreasing the protein expression levels of p53, whereas ectopic expression in the antisense oligos of these microRNAs demonstrated the opposite effect. In addition, the present demonstrated that the cisplatin‑induced expression of miR‑125a and miR‑125b inhibited cisplatin‑induced apoptosis in the TW03 cells by decreasing the protein expression levels of p53. Taken together, the present study revealed for the first time, to the best of our knowledge, that induction of the expression of miR‑125a and miR‑125b by treatment with cisplatin resulted in resistance to the cisplatin drug in the NPC cells.
Collapse
Affiliation(s)
- Jian Jun Chen
- Department of Medical, The First People's Hospital of Neijiang, Neijiang, Sichuan 641000, P.R. China
| | - Shi Xi Liu
- Department of E.N.T, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Mian Zhi Chen
- Department of E.N.T, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Zi Yi Zhao
- Central Laboratory, Teaching Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610000, P.R. China
| |
Collapse
|
23
|
Aljada A, Saleh AM, Al Suwaidan S. Modulation of insulin/IGFs pathways by sirtuin-7 inhibition in drug-induced chemoreistance. Diagn Pathol 2014; 9:94. [PMID: 24885964 PMCID: PMC4229859 DOI: 10.1186/1746-1596-9-94] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Accepted: 04/15/2014] [Indexed: 01/08/2023] Open
Abstract
Background Insulin and insulin-like growth factors (IGFs) are key regulators of metabolism and growth. Recent evidences suggest a key role of these pathways in non-classical tissues and the metabolic pathways by which these hormones exert their effects in neoplasia is unclear. Aims To study insulin/IGFs pathways in drug sensitive and resistant cancer cells representing breast cancer (MCF-7), osteosarcoma (SaOS-2), and ovarian cancer (A2780) and to examine the effect of Sirtuin-7 (Sirt7) inhibition on insulin/IGFs pathways in MCF-7 cell line. Methods Drug resistant cells were generated by continuous incubation of parental cell lines with stepwise increases in Doxorubicin or Cisplatin over a period of 3 to 6 months. MCF-7 cells were transfected with cloned hairpin siRNA template for Sirt7 using the Amaxa GmbH transfection system. mRNA expression of Sirt7, INSR, IRS-1, IRS-2, IRS-4, IGF-1, IGF-2, MDR-1, MRP-1, BCRP was measured by qPCR and Sirt7 by standard Western blotting. FITC-insulin uptake was imaged with Leica Confocal Microscope. Results Insulin receptor (INSR), insulin receptor substrate-1 (IRS-1) were inhibited in drug-induced resistance, whereas IRS-2 was significantly induced in all the chemoresistant cells tested when compared to their parental counterparts. IGF-1 and IGF-2 were also upregulated in all the drug resistant cells tested. Sirt7 was significantly reduced in all chemoresistant cells tested. Knockdown of Sirt7 expression in human breast MCF-7 cell line by siRNA induced premature senescence-like phenotype and multi-drug resistance, suggesting that this gene may play an active role in regulating cancer cell response to stress. Suppression of Sirt7 selectively inhibited INSR and IRS-1, whereas it had minimal effect on that of IRS-2. Sirt7 suppression in MCF-7 also inhibited insulin uptake. Additionally, Sirt7 inhibition upregulated IGF-1, IGF-2 and IGFR expression. Conclusion Our data demonstrate that stress-induced Sirt7 inhibition significantly increases stress resistance and modulates insulin/IGF-1 signaling pathways. More importantly, this study links Sir2 family proteins to insulin/IGF signaling in drug-induced stress resistance in neoplasia. Virtual Slides The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/1135426681234493
Collapse
Affiliation(s)
- Ahmad Aljada
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, P, O, Box 22490, Riyadh 11426, Kingdom of Saudi Arabia.
| | | | | |
Collapse
|
24
|
Menon VR, Peterson EJ, Valerie K, Farrell NP, Povirk LF. Ligand modulation of a dinuclear platinum compound leads to mechanistic differences in cell cycle progression and arrest. Biochem Pharmacol 2013; 86:1708-20. [PMID: 24161784 DOI: 10.1016/j.bcp.2013.10.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 10/07/2013] [Accepted: 10/14/2013] [Indexed: 11/25/2022]
Abstract
Despite similar structures and DNA binding profiles, two recently synthesized dinuclear platinum compounds are shown to elicit highly divergent effects on cell cycle progression. In colorectal HCT116 cells, BBR3610 shows a classical G2/M arrest with initial accumulation in S phase, but the derivative compound BBR3610-DACH, formed by introduction of the 1,2-diaminocyclohexane (DACH) as carrier ligand, results in severe G1/S as well as G2/M phase arrest, with nearly complete S phase depletion. The origin of this unique effect was studied. Cellular interstrand crosslinking as assayed by comet analysis was similar for both compounds, confirming previous in vitro results obtained on plasmid DNA. Immunoblotting revealed a stabilization of p53 and concomitant transient increases in p21 and p27 proteins after treatment with BBR3610-DACH. Cell viability assays and cytometric analysis of p53 and p21 null cells indicated that BBR3610-DACH-induced cell cycle arrest was p21-dependent and partially p53-dependent. However, an increase in the levels of cyclin E was observed with steady state levels of CDK2 and Cdc25A, suggesting that the G1 block occurs downstream of CDK/cyclin complex formation. The G2/M block was corroborated with decreased levels of cyclin A and cyclin B1. Surprisingly, BBR3610-DACH-induced G1 block was independent of ATM and ATR. Finally, both compounds induced apoptosis, with BBR3610-DACH showing a robust PARP-1 cleavage that was not associated with caspase-3/7 cleavage. In summary, BBR3610-DACH is a DNA binding platinum agent with unique inhibitory effects on cell cycle progression that could be further developed as a chemotherapeutic agent complementary to cisplatin and oxaliplatin.
Collapse
Affiliation(s)
- Vijay R Menon
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA 23298-0613, United States; Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298-0613, United States
| | | | | | | | | |
Collapse
|
25
|
Sancho-Martínez SM, Prieto-García L, Prieto M, López-Novoa JM, López-Hernández FJ. Subcellular targets of cisplatin cytotoxicity: An integrated view. Pharmacol Ther 2012; 136:35-55. [DOI: 10.1016/j.pharmthera.2012.07.003] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 06/28/2012] [Indexed: 12/29/2022]
|
26
|
Song W, Wang J, Yang Y, Jing N, Zhang X, Chen L, Wu J. Rewiring drug-activated p53-regulatory network from suppressing to promoting tumorigenesis. J Mol Cell Biol 2012; 4:197-206. [DOI: 10.1093/jmcb/mjs029] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
27
|
Michnov O, Solomayer E, Fehm T, Stubenrauch F, Iftner T. Knock down of p53 or its ubiquitin ligase E6AP does not affect the sensitivity of human papillomavirus-positive cervical cancer cells to cisplatin. Am J Cancer Res 2012; 2:309-321. [PMID: 22679561 PMCID: PMC3365809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Accepted: 04/20/2012] [Indexed: 06/01/2023] Open
Abstract
The persistent infection with high risk human papillomaviruses (hrHPV) is a necessary risk factor for the development of cervical cancer, which is the second most frequent cancer in women worldwide. Cisplatin-based radiotherapy represents the current treatment regimen. However, the results for advanced and recurrent disease are far from optimal. Since almost all cervical cancers contain wild type (wt) p53, which is degraded by the complex of hrHPV E6 and the ubiquitin ligase E6AP, we addressed if the reconstitution of p53 via silencing of E6AP sensitizes cervical cancer cells towards cisplatin treatment. For this we established and characterized two novel cervical cancer cell lines that contain integrated HPV16 genomes. Long-term established HeLa and SiHa cells and the novel cervical cancer cell lines at low passage numbers were treated with different concentrations of cisplatin. Cell viability was measured by the WST-1 assay. In addition, single cisplatin treatment was combined with the silencing of E6AP or p53. The comparison to HeLa and SiHa cells revealed a higher sensitivity of the novel cell lines to cisplatin treatment, which caused p53 accumulation and transcriptional induction of p21. Silencing of E6AP further increased p53 protein levels, but had no effect on cell viability when combined with cisplatin treatment. Interestingly, silencing of p53 had also no effect. We therefore conclude that reactivation of p53 via silencing of E6AP does not increase the sensitivity of cervical cancer cells towards cisplatin treatment.
Collapse
Affiliation(s)
- Olga Michnov
- Institute for Medical Virology and Epidemiology of Viral Diseases, Division of Experimental Virology, University Hospital TuebingenElfriede-Aulhorn-Str. 6, 72076 Tuebingen, Germany
| | - Erich Solomayer
- Department of Obstetrics and Gynecology, University of SaarlandKirrbergerstr. 100, 66424, Homburg, Saar, Germany
| | - Tanja Fehm
- Department of Obstetrics & Gynecology, University Hospital of TuebingenCalwer Str. 7, 72076 Tuebingen, Germany
| | - Frank Stubenrauch
- Institute for Medical Virology and Epidemiology of Viral Diseases, Division of Experimental Virology, University Hospital TuebingenElfriede-Aulhorn-Str. 6, 72076 Tuebingen, Germany
| | - Thomas Iftner
- Institute for Medical Virology and Epidemiology of Viral Diseases, Division of Experimental Virology, University Hospital TuebingenElfriede-Aulhorn-Str. 6, 72076 Tuebingen, Germany
| |
Collapse
|
28
|
Hostetter AA, Osborn MF, DeRose VJ. RNA-Pt adducts following cisplatin treatment of Saccharomyces cerevisiae. ACS Chem Biol 2012; 7:218-25. [PMID: 22004017 DOI: 10.1021/cb200279p] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The numerous regulatory roles of cellular RNAs suggest novel potential drug targets, but establishing intracellular drug-RNA interactions is challenging. Cisplatin (cis-diamminedichloridoplatinum(II)) is a leading anticancer drug that forms exchange-inert complexes with nucleic acids, allowing its distribution on cellular RNAs to be followed ex vivo. Although Pt adduct formation on DNA is well-known, a complete characterization of cellular RNA-Pt adducts has not been performed. In this study, the action of cisplatin on S. cerevisiae in minimal media was established with growth curves, clonogenic assays, and tests for apoptotic markers. Despite high toxicity, cisplatin-induced apoptosis in S. cerevisiae was not observed under these conditions. In-cell Pt concentrations and Pt accumulation on poly(A)-mRNA, rRNA, total RNA, and DNA quantified via ICP-MS indicate ∼4- to 20-fold more Pt accumulation in total cellular RNA than in DNA. Interestingly, similar Pt accumulation is observed on rRNA and total RNA, corresponding to one Pt per (14,600 ± 1,500) and (5760 ± 580) nucleotides on total RNA following 100 and 200 μM cisplatin treatments, respectively. Specific Pt adducts mapped by primer extension analysis of a solvent-accessible 18S rRNA helix occur at terminal and internal loop regions and appear as soon as 1 h post-treatment. Pt per nucleotide accumulation on poly(A)-mRNA is 4- to 6-fold lower than on rRNA but could have consequences for low copy-number or highly regulated transcripts. Taken together, these data demonstrate significant accumulation of Pt adducts on cellular RNA species following in cellulo cisplatin treatment. These and other small molecule-RNA interactions could disrupt processes regulated by RNA.
Collapse
Affiliation(s)
- Alethia A. Hostetter
- Department of Chemistry, University of Oregon, Eugene, Oregon 97403, United States
| | - Maire F. Osborn
- Department of Chemistry, University of Oregon, Eugene, Oregon 97403, United States
| | - Victoria J. DeRose
- Department of Chemistry, University of Oregon, Eugene, Oregon 97403, United States
| |
Collapse
|
29
|
Seifrtová M, Havelek R, Ćmielová J, Jiroutová A, Soukup T, Brůčková L, Mokrý J, English D, Řezáčová M. The response of human ectomesenchymal dental pulp stem cells to cisplatin treatment. Int Endod J 2011; 45:401-12. [DOI: 10.1111/j.1365-2591.2011.01990.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
30
|
Lovejoy KS, Serova M, Bieche I, Emami S, D'Incalci M, Broggini M, Erba E, Gespach C, Cvitkovic E, Faivre S, Raymond E, Lippard SJ. Spectrum of cellular responses to pyriplatin, a monofunctional cationic antineoplastic platinum(II) compound, in human cancer cells. Mol Cancer Ther 2011; 10:1709-19. [PMID: 21750216 PMCID: PMC3170455 DOI: 10.1158/1535-7163.mct-11-0250] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Pyriplatin, cis-diammine(pyridine)chloroplatinum(II), a platinum-based antitumor drug candidate, is a cationic compound with anticancer properties in mice and is a substrate for organic cation transporters that facilitate oxaliplatin uptake. Unlike cisplatin and oxaliplatin, which form DNA cross-links, pyriplatin binds DNA in a monofunctional manner. The antiproliferative effects of pyriplatin, alone and in combination with known anticancer drugs (paclitaxel, gemcitabine, SN38, cisplatin, and 5-fluorouracil), were evaluated in a panel of epithelial cancer cell lines, with direct comparison to cisplatin and oxaliplatin. The effects of pyriplatin on gene expression and platinum-DNA adduct formation were also investigated. Pyriplatin exhibited cytotoxic effects against human cell lines after 24 hours (IC(50) = 171-443 μmol/L), with maximum cytotoxicity in HOP-62 non-small cell lung cancer cells after 72 hours (IC(50) = 24 μmol/L). Pyriplatin caused a G(2)-M cell cycle block similar to that induced by cisplatin and oxaliplatin. Induction of apoptotsis and DNA damage response was supported by Annexin-V analysis and detection of phosphorylated Chk2 and H2AX. Treatment with pyriplatin increased CDKN1/p21 and decreased ERCC1 mRNA expression. On a platinum-per-nucleotide basis, pyriplatin-DNA adducts are less cytotoxic than those of cisplatin and oxaliplatin. The mRNA levels of genes implicated in drug transport and DNA damage repair, including GSTP1 and MSH2, correlate with pyriplatin cellular activity in the panel of cell lines. Synergy occurred for combinations of pyriplatin with paclitaxel. Because its spectrum of activity differs significantly from those of cisplatin or oxaliplatin, pyriplatin is a lead compound for developing novel drug candidates with cytotoxicity profiles unlike those of drugs currently in use.
Collapse
Affiliation(s)
- Katherine S. Lovejoy
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139 USA
| | - Maria Serova
- RayLab-U728 and Department of Medical Oncology, Beaujon University Hospital, Clichy, France
| | - Ivan Bieche
- Laboratory of Molecular Genetics, Beaujon University Hospital, Clichy, France
| | - Shahin Emami
- INSERM U482, Saint-Antoine Hospital, Paris, France
| | | | | | | | | | - Esteban Cvitkovic
- RayLab-U728 and Department of Medical Oncology, Beaujon University Hospital, Clichy, France
| | - Sandrine Faivre
- RayLab-U728 and Department of Medical Oncology, Beaujon University Hospital, Clichy, France
| | - Eric Raymond
- RayLab-U728 and Department of Medical Oncology, Beaujon University Hospital, Clichy, France
| | - Stephen J. Lippard
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139 USA
| |
Collapse
|
31
|
He G, Kuang J, Khokhar AR, Siddik ZH. The impact of S- and G2-checkpoint response on the fidelity of G1-arrest by cisplatin and its comparison to a non-cross-resistant platinum(IV) analog. Gynecol Oncol 2011; 122:402-9. [PMID: 21592546 PMCID: PMC3601787 DOI: 10.1016/j.ygyno.2011.04.034] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Revised: 04/18/2011] [Accepted: 04/23/2011] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Cisplatin is a DNA-damaging antitumor agent that is highly effective in treating ovarian cancer. It activates the p53/p21 pathway for its cytotoxic mode of action, but it does not induce p21-dependent cell cycle arrest in G1. Therefore, we investigated this paradox, and used the model analog DAP as a positive control for p21-dependent G1-arrest. METHODS Studies were conducted in p53-proficient ovarian A2780 tumor cells to examine Cdk activity, cell cycle distribution and DNA damage signaling after cisplatin or DAP in combination with the mitotic inhibitor nocodazole. RESULTS Cisplatin consistently induced transient S-phase arrest by inhibiting Cdk2/cyclin A complex in S-phase at 12 h and then a durable G2/M-arrest by inhibiting Cdc2/cyclin B complex at 12-18 h. These inhibitions were associated with Chk1 and Chk2 activation and resultant increase in inhibitory tyrosine phosphorylation of Cdk2 and Cdc2. Cisplatin also potently inhibited G1-phase Cdk4/cyclin D1 and Cdk2/cyclin E activities at ~18 h. In agreement, exposure of cisplatin-treated A2780, HCT-116(p53-/-) and HCT-116(p21-/-) tumor cells to nocodazole revealed limited G1-arrest that was dependent on p53 and p21. In contrast, the durable G1-arrest by DAP, which failed to activate Chk1 and Chk2, was unaffected by nocodazole. CONCLUSIONS Cisplatin induced G1-arrest, but at an attenuated level. This was primarily due to orchestration of Cdk inhibition in S-phase first, then in G2, and finally in G1 that effectively blocked cells in G2 and prevented cells from progressing and arresting in G1. These studies demonstrate that cisplatin unequivocally activates G1-checkpoint response, but the fidelity of G1-arrest is compromised by Chk1/2 activation and checkpoint response in S- and G2/M-phase.
Collapse
Affiliation(s)
- Guangan He
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jian Kuang
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Abdul R. Khokhar
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zahid H. Siddik
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
32
|
Disruption of the MDM2-p53 interaction strongly potentiates p53-dependent apoptosis in cisplatin-resistant human testicular carcinoma cells via the Fas/FasL pathway. Cell Death Dis 2011; 2:e148. [PMID: 21509038 PMCID: PMC3122064 DOI: 10.1038/cddis.2011.33] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Wild-type p53 has a major role in the response and execution of apoptosis after chemotherapy in many cancers. Although high levels of wild-type p53 and hardly any TP53 mutations are found in testicular cancer (TC), chemotherapy resistance is still observed in a significant subgroup of TC patients. In the present study, we demonstrate that p53 resides in a complex with MDM2 at higher cisplatin concentrations in cisplatin-resistant human TC cells compared with cisplatin-sensitive TC cells. Inhibition of the MDM2–p53 interaction using either Nutlin-3 or MDM2 RNA interference resulted in hyperactivation of the p53 pathway and a strong induction of apoptosis in cisplatin-sensitive and -resistant TC cells. Suppression of wild-type p53 induced resistance to Nutlin-3 in TC cells, demonstrating the key role of p53 for Nutlin-3 sensitivity. More specifically, our results indicate that p53-dependent induction of Fas membrane expression (∼threefold) and enhanced Fas/FasL interactions at the cell surface are important mechanisms of Nutlin-3-induced apoptosis in TC cells. Importantly, an analogous Fas-dependent mechanism of apoptosis upon Nutlin-3 treatment is executed in wild-type p53 expressing Hodgkin lymphoma and acute myeloid leukaemia cell lines. Finally, we demonstrate that Nutlin-3 strongly augmented cisplatin-induced apoptosis and cell kill via the Fas death receptor pathway. This effect is most pronounced in cisplatin-resistant TC cells.
Collapse
|
33
|
Tripathi R, Samadder T, Gupta S, Surolia A, Shaha C. Anticancer activity of a combination of cisplatin and fisetin in embryonal carcinoma cells and xenograft tumors. Mol Cancer Ther 2011; 10:255-68. [PMID: 21216935 DOI: 10.1158/1535-7163.mct-10-0606] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Use of chemotherapeutic drug cisplatin is limited because of its toxicity. Therefore, efforts continue for the discovery of novel combination therapies with cisplatin to reduce its effective treatment dose. This study evaluates the potential of fisetin, a flavonoid, to increase cisplatin cytotoxicity in human embryonal carcinoma NT2/D1 cells. Addition of fisetin to cisplatin enhanced cisplatin cytotoxicity in vitro at four times lower dose than that required by cisplatin monotherapy for similar cytotoxic effects. Cisplatin, fisetin monotherapy, and addition of fisetin to cisplatin in a combination increased FasL expression. Cisplatin and fisetin as single agents activated caspases-8 and -3 and caspases-9 and -7, respectively, whereas combination treatment activated all 4 caspases. Increases in p53 and p21 and decreases in cyclin B1 and survivin occurred, all effects being more exaggerated with the combination. Fisetin, with or without cisplatin, increased expression of proapoptotic protein Bak and induced its mitochondrial oligomerization. Bid truncation and mitochondrial translocation of Bid and p53 was induced by fisetin in the presence or absence of cisplatin. Downregulation of p53 by short hairpin RNA during drug treatment decreased p21 levels but caused survivin increase, thus reducing cell death. Upstream to p53, inhibition of p38 phosphorylation reduced p53 phosphorylation and cell death. In a NT2/D1 mouse xenograft model, combination therapy was most effective in reducing tumor size. In summary, findings of this study suggest that addition of fisetin to cisplatin activates both the mitochondrial and the cell death receptor pathway and could be a promising regimen for the elimination of embryonal carcinoma cells.
Collapse
Affiliation(s)
- Rakshamani Tripathi
- Cell Death and Differentiation Research Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | | | | | | | | |
Collapse
|
34
|
Puliyappadamba VT, Cheriyan VT, Thulasidasan AKT, Bava SV, Vinod BS, Prabhu PR, Varghese R, Bevin A, Venugopal S, Anto RJ. Nicotine-induced survival signaling in lung cancer cells is dependent on their p53 status while its down-regulation by curcumin is independent. Mol Cancer 2010; 9:220. [PMID: 20727180 PMCID: PMC2936340 DOI: 10.1186/1476-4598-9-220] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Accepted: 08/20/2010] [Indexed: 12/23/2022] Open
Abstract
Background Lung cancer is the most lethal cancer and almost 90% of lung cancer is due to cigarette smoking. Even though nicotine, one of the major ingredients of cigarette smoke and the causative agent for addiction, is not a carcinogen by itself, several investigators have shown that nicotine can induce cell proliferation and angiogenesis. We observed that the proliferative index of nicotine is different in the lung cancer cell lines H1299 (p53-/-) and A549 (p53+/+) which indicates that the mode of up-regulation of survival signals by nicotine might be different in cells with and without p53. Results While low concentrations of nicotine induced activation of NF-κB, Akt, Bcl2, MAPKs, AP1 and IAPs in H1299, it failed to induce NF-κB in A549, and compared to H1299, almost 100 times higher concentration of nicotine was required to induce all other survival signals in A549. Transfection of WT-p53 and DN-p53 in H1299 and A549 respectively, reversed the mode of activation of survival signals. Curcumin down-regulated all the survival signals induced by nicotine in both the cells, irrespective of their p53 status. The hypothesis was confirmed when lower concentrations of nicotine induced NF-κB in two more lung cancer cells, Hop-92 and NCI-H522 with mutant p53 status. Silencing of p53 in A549 using siRNA made the cells susceptible to nicotine-induced NF-κB nuclear translocation as in A549 DN-p53 cells. Conclusions The present study reveals a detrimental role of nicotine especially in lung cancer patients with impaired p53 status and identifies curcumin as a potential chemopreventive.
Collapse
Affiliation(s)
- Vineshkumar T Puliyappadamba
- Integrated Cancer Research Program, Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Di Pasqua AJ, Hong C, Wu MY, McCracken E, Wang X, Mi L, Chung FL. Sensitization of non-small cell lung cancer cells to cisplatin by naturally occurring isothiocyanates. Chem Res Toxicol 2010; 23:1307-9. [PMID: 20707406 PMCID: PMC2924773 DOI: 10.1021/tx100187f] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We show that naturally occurring isothiocyanates (ITCs) sensitize human non-small cell lung cancer cells to cisplatin. Moreover, the structure of the ITC side chain moiety is important for sensitization. In NCI-H596 cells, 20 microM benzyl isothiocyanate (BITC) and phenethyl isothiocyanate (PEITC) enhance the efficacy of various concentrations of cisplatin, but sulforaphane (SFN) does not. Reducing the concentration of BITC and PEITC to 10 microM still allows for the sensitization of cells to cisplatin. Neither cellular platinum accumulation nor DNA platination account for this increased cytotoxicity. BITC and PEITC deplete beta-tubulin, but SFN does not; this correlates with and may be important for sensitization.
Collapse
Affiliation(s)
- Anthony J. Di Pasqua
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3800 Reservoir Road, LL 128A, Box 571465, Washington D. C. 20057
| | - Charles Hong
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3800 Reservoir Road, LL 128A, Box 571465, Washington D. C. 20057
| | - Mona Y Wu
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3800 Reservoir Road, LL 128A, Box 571465, Washington D. C. 20057
| | - Erin McCracken
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3800 Reservoir Road, LL 128A, Box 571465, Washington D. C. 20057
| | - Xiantao Wang
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3800 Reservoir Road, LL 128A, Box 571465, Washington D. C. 20057
| | - Lixin Mi
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3800 Reservoir Road, LL 128A, Box 571465, Washington D. C. 20057
| | - Fung-Lung Chung
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, 3800 Reservoir Road, LL 128A, Box 571465, Washington D. C. 20057
| |
Collapse
|
36
|
Basu A, Krishnamurthy S. Cellular responses to Cisplatin-induced DNA damage. J Nucleic Acids 2010; 2010:201367. [PMID: 20811617 PMCID: PMC2929606 DOI: 10.4061/2010/201367] [Citation(s) in RCA: 335] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Accepted: 06/28/2010] [Indexed: 12/29/2022] Open
Abstract
Cisplatin is one of the most effective anticancer agents widely used in the treatment of solid tumors. It is generally considered as a cytotoxic drug which kills cancer cells by damaging DNA and inhibiting DNA synthesis. How cells respond to cisplatin-induced DNA damage plays a critical role in deciding cisplatin sensitivity. Cisplatin-induced DNA damage activates various signaling pathways to prevent or promote cell death. This paper summarizes our current understandings regarding the mechanisms by which cisplatin induces cell death and the bases of cisplatin resistance. We have discussed various steps, including the entry of cisplatin inside cells, DNA repair, drug detoxification, DNA damage response, and regulation of cisplatin-induced apoptosis by protein kinases. An understanding of how various signaling pathways regulate cisplatin-induced cell death should aid in the development of more effective therapeutic strategies for the treatment of cancer.
Collapse
Affiliation(s)
- Alakananda Basu
- Department of Molecular Biology & Immunology, University of North Texas Health Science Center and Institute for Cancer Research, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, USA
| | - Soumya Krishnamurthy
- Department of Molecular Biology & Immunology, University of North Texas Health Science Center and Institute for Cancer Research, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, USA
| |
Collapse
|
37
|
Ahmad S. Platinum-DNA interactions and subsequent cellular processes controlling sensitivity to anticancer platinum complexes. Chem Biodivers 2010; 7:543-66. [PMID: 20232326 DOI: 10.1002/cbdv.200800340] [Citation(s) in RCA: 157] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Platinum-based compounds are widely used as chemotherapeutics for the treatment of a variety of cancers. The anticancer activity of cisplatin and other platinum drugs is believed to arise from their interaction with DNA. Several cellular pathways are activated in response to this interaction, which include recognition by high-mobility group and repair proteins, translesion synthesis by polymerases, and induction of apoptosis. The apoptotic process is regulated by activation of caspases, p53 gene, and several proapoptotic and antiapoptotic proteins. Such cellular processing eventually leads to an inhibition of the replication or transcription machinery of the cell. Deactivation of platinum drugs by thiols, increased nucleotide excision repair of Pt-DNA adducts, decreased mismatch repair, and defective apoptosis result in resistance to platinum therapy. The differences in cytotoxicity of various platinum complexes are attributed to the differential recognition of their adducts by cellular proteins. Cisplatin and oxaliplatin both produce mainly 1,2-GG intrastrand cross-links as major adducts, but oxaliplatin is found to be more active particularly against cisplatin-resistant tumor cells. Mismatch repair and replicative bypass appear to be the processes most likely involved in differentiating the molecular responses to these two agents. This review describes the formation of Pt-DNA adducts, their interaction with cellular components, and biological effects of this interaction.
Collapse
Affiliation(s)
- Saeed Ahmad
- Department of Chemistry, University of Engineering and Technology, Lahore 54890, Pakistan.
| |
Collapse
|
38
|
Chandra S. Quantitative imaging of chemical composition in single cells by secondary ion mass spectrometry: cisplatin affects calcium stores in renal epithelial cells. Methods Mol Biol 2010; 656:113-30. [PMID: 20680587 PMCID: PMC2974903 DOI: 10.1007/978-1-60761-746-4_6] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
A detailed protocol for quantitative single cell mass spectrometry imaging (MSI) analysis is described in this chapter with examples of the treatment of cells with anticancer drug, cisplatin. Cisplatin, cis-diamminedichloridoplatinum ii (CDDP), is widely used for the treatment of many malignancies, including testicular, ovarian, bladder, cervical, head and neck, and small cell and non-small cell lung cancers. The possibility of renal injury by cisplatin treatment is a major dose-limiting factor in this cancer therapy. At present, the mechanisms of cisplatin-induced renal cytotoxicity are poorly understood. In this work, secondary ion mass spectrometry (SIMS) was used for investigating cisplatin-induced alterations in intracellular chemical composition in a well-established model (LLC-PK(1) cell line) for studying renal injury. The cells were cryogenically prepared by the sandwich freeze-fracture method for subcellular imaging analysis of chemical composition (total concentrations of K(+), Na(+), and Ca(2+)) in individual cells. The single cell analysis of these diffusible ions necessitates the use of reliable cryogenic sample preparations for SIMS. The sandwich freeze-fracture method offers a simple approach for cryogenically preserving diffusible ions and molecules inside the cells for SIMS analysis. A CAMECA IMS-3f SIMS ion microscope instrument capable of producing chemical images of single cells with 500-nm spatial resolution was used in the study. In cisplatin-treated cells, SIMS imaging showed the presence of detectable amount of platinum at mass 195, as (195)Pt(+) secondary ions in individual cells. SIMS observations also revealed that individual cells differed in their response to cisplatin. While the chemical composition of some cells was unaffected by cisplatin, others showed a reduction in cytoplasmic calcium stores that was not associated with changes in their intracellular K or Na concentrations. Another population of cells displayed an increase in cytoplasmic calcium concentration that was associated with higher levels of intracellular Na and a reduction in K concentration of the same cells. Since the loss of intracellular K and the gain of Na and Ca are typical symptoms of cell injury, it is plausible that the initial response of the cell to cisplatin treatment is the reduction in cytoplasmic calcium pool in stores. If, somehow, the calcium stores are compromised with cisplatin, then maintenance of free Ca(2+) homeostasis would become uncontrollable in the cell. These observations open new avenues of research for understanding of the mode of action of cisplatin in cell injury. This study also demonstrates the need and vast potential of single cell imaging mass spectrometry techniques in cell biology and medicine.
Collapse
Affiliation(s)
- Subhash Chandra
- Cornell SIMS Laboratory, Department of Biomedical Engineering, Snee Hall, Cornell University, Ithaca, NY 14853, U.S.A. ()
| |
Collapse
|
39
|
Kutuk O, Arisan ED, Tezil T, Shoshan MC, Basaga H. Cisplatin overcomes Bcl-2-mediated resistance to apoptosis via preferential engagement of Bak: critical role of Noxa-mediated lipid peroxidation. Carcinogenesis 2009; 30:1517-27. [PMID: 19578044 DOI: 10.1093/carcin/bgp165] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Increased expression of antiapoptotic Bcl-2 proteins confers therapeutic resistance in various cancer types. Targeting Bcl-2 proteins by small molecules or activating alternative pathways to bypass Bcl-2-mediated protection to promote apoptosis are two approaches to overcoming therapeutic resistance. Here, we show that cisplatin triggers a Bak-dependent pathway to induce apoptosis in Bcl-2-overexpressing MCF-7 cells. p53-mediated induction of Noxa expression, generation of lipid peroxidation end products and induction of Noxa-Mcl-1 interaction are necessary for this pathway to function. Although Puma is also induced by cisplatin treatment, it is not required for apoptosis. Similarly, reactive oxygen species production by cisplatin did not have any effect on cisplatin-induced apoptosis in MCF-7 Bcl-2 cells. Furthermore, p53 promotes cisplatin-induced apoptosis by directly binding and counteracting Bcl-x(L) antiapoptotic function. In conclusion, our findings suggest a novel mode of action for cisplatin to overcome Bcl-2-mediated protection against apoptosis, which requires preferential activation of Bak and p53-mediated upregulation of Noxa protein levels and lipid peroxidation.
Collapse
Affiliation(s)
- Ozgur Kutuk
- Biological Sciences and Bioengineering Program, Sabanci University, 34956 Tuzla, Istanbul, Turkey
| | | | | | | | | |
Collapse
|
40
|
Roy S, Hagen K, Maheswari P, Lutz M, Spek A, Reedijk J, van Wezel G. Phenanthroline Derivatives with Improved Selectivity as DNA-Targeting Anticancer or Antimicrobial Drugs. ChemMedChem 2008; 3:1427-34. [DOI: 10.1002/cmdc.200800097] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
41
|
Lau ATY, Wang Y, Chiu JF. Reactive oxygen species: current knowledge and applications in cancer research and therapeutic. J Cell Biochem 2008; 104:657-67. [PMID: 18172854 DOI: 10.1002/jcb.21655] [Citation(s) in RCA: 215] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Reactive oxygen species (ROS) are natural products inevitably generated along cellular metabolism. Due to their highly reactive nature, which can damage DNA, proteins and lipids, cells utilize antioxidative or defense systems to balance these toxic products to keep the cells in a state of redox homeostasis. However, under the situation of imbalance in redox status, depending on the magnitude of ROS encountered, high levels of ROS can induce apoptosis, whereas chronic low levels of ROS promote vascular diseases such as arteriosclerosis. Although ROS seem to be catastrophic to life, accumulating evidence points to the beneficial roles of ROS by virtue of the ability as chemotherapeutic agents to cure human diseases. Many anti-cancer drugs have been developed in this way which can generate ROS and cause oxidative stress-induced apoptosis in cancer cells. The effects of ROS are paradoxical because they can act as both disease culprits and chemotherapeutic agents. In this review, the current knowledge of ROS and the potential applications of ROS in cancer therapeutic will be discussed.
Collapse
Affiliation(s)
- Andy T Y Lau
- Department of Anatomy, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China
| | | | | |
Collapse
|
42
|
Hoffmann TK, Sonkoly E, Hauser U, van Lierop A, Whiteside TL, Klussmann JP, Hafner D, Schuler P, Friebe-Hoffmann U, Scheckenbach K, Erjala K, Grénman R, Schipper J, Bier H, Balz V. Alterations in the p53 pathway and their association with radio- and chemosensitivity in head and neck squamous cell carcinoma. Oral Oncol 2008; 44:1100-9. [PMID: 18487078 DOI: 10.1016/j.oraloncology.2008.02.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2008] [Revised: 02/14/2008] [Accepted: 02/15/2008] [Indexed: 11/30/2022]
Abstract
Chemotherapy and/or radiotherapy are established measures in treatment protocols of head and neck squamous cell carcinoma (HNSCC). However, we still lack reliable predictive markers for the response to radio- and chemotherapy. The p53 pathway is involved in stress response and thus might influence chemo-/radiosensitivity. Using 29 HNSCC cell lines previously characterized for p53 mutations, we simultaneously analyzed several key players in the p53 pathway by RT-PCR, transcript sequencing and immunohistochemistry, and investigated their association with chemosensitivity and radiosensitivity. Cell lines with p53 mutations were slightly more sensitive to cisplatin than those with wild-type p53. The type of mutation did not influence radio- or chemosensitivity. p14(ARF), an activator of p53, was lost or mutated in all cell lines. Three cell lines showed overexpression of HDM-2, a major negative regulator of p53; however, HDM-2 levels did not correlate with radio- or chemosensitivity. HPV-16 oncoproteins were detected in one highly chemoresistant cell line. Our findings suggest that molecular events resulting in the inactivation of the p53 pathway occur in all HNSCC cell lines. However, single alterations in the p53 pathway are not reliable predictors for the response to radio- or chemotherapy in HNSCC.
Collapse
Affiliation(s)
- Thomas K Hoffmann
- Department of Otorhinolaryngology, Heinrich-Heine-University Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Keter FK, Kanyanda S, Lyantagaye SSL, Darkwa J, Rees DJG, Meyer M. In vitro evaluation of dichloro-bis(pyrazole)palladium(II) and dichloro-bis(pyrazole)platinum(II) complexes as anticancer agents. Cancer Chemother Pharmacol 2008; 63:127-38. [PMID: 18350297 DOI: 10.1007/s00280-008-0721-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2007] [Accepted: 02/24/2008] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Cisplatin (cis-diamminedichloroplatinum) was first identified for its anti-bacterial activity, and was later also shown to be an efficient anticancer agent. However, the therapeutic use of this anticancer drug is somewhat limited by its toxic side effects, which include nephrotoxicity, nausea, and vomiting. Furthermore the development of drug-resistant tumours is commonly observed following therapy with cisplatin. Hence there is a need for improved platinum derived drugs to overcome these limitations. AIMS Apoptosis contributes significantly to the cytotoxic effects of anticancer agents such as cisplatin; therefore in this study the potential anticancer properties of a series of pyrazole palladium(II) and platinum(II) complexes, [(3,5-R(2)pz)(2)PdCl(2)] [R = H (1), R = Me (2)] and [(3,5-R(2)pz)(2)PtCl(2)] [R = H (3), R = Me (4)], were evaluated by assessment of their pro-apoptotic activity. METHODS The induction of apoptosis was measured in CHO cells by the detection of phosphatidylserine (PS) exposure using the annexin V and APOPercentage assays; DNA fragmentation using the Terminal deoxynucleotide transferase dUTP Nick End Labelling (TUNEL) assay; and the detection of activated caspase-3. RESULTS The platinum complexes were shown to be considerably more active than the palladium complexes, with complex 3 demonstrating the highest level of cytotoxic and pro-apoptotic activity. The LD(50) values for complex 3 and cisplatin were 20 and 70 microM, respectively, demonstrating that the cytotoxic activity for complex 3 was three times higher than for cisplatin. Various human cancer cell lines, including CaSki, HeLa, as well as the p53 mutant Jurkat T cell line were also shown to be susceptible to complex 3. CONCLUSIONS Collectively, this in vitro study provides insights into action of palladium and platinum complexes and demonstrates the potential use of these compounds, and in particular complex 3, in the development of new anticancer agents.
Collapse
Affiliation(s)
- Frankline K Keter
- Department of Chemistry, University of Johannesburg, P.O. Box 524, Auckland Park, 2006, South Africa
| | | | | | | | | | | |
Collapse
|
44
|
Lo Nigro C, Arnolfo E, Taricco E, Fruttero A, Russi EG, Lucio F, Ribero S, Comino A, Merlano M, Ungari S. The cisplatin-irradiation combination suggests that apoptosis is not a major determinant of clonogenic death. Anticancer Drugs 2007; 18:659-67. [PMID: 17762394 DOI: 10.1097/cad.0b013e328087388f] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
It is commonly believed that tumor cells treated with anticancer agents, chemotherapy and/or radiation, die by apoptosis and that tumors which do not undergo apoptosis are resistant to treatment. In this study, we investigated the molecular basis underlying cisplatin cytotoxicity in the murine teratocarcinoma F9 cell line to see whether irradiation enhances cisplatin-induced cytotoxicity. We compared the apoptosis induced by chemo and/or radiotherapy with other cellular effects such as cell survival, clonogenic capability, cell cycle perturbation, expression of p53 and p53-related mRNAs, and necrosis. When combined with radiation, a clear additive cytotoxic effect of cisplatin was demonstrated. We found that both cisplatin and radiation induced cell death, but the level of induced apoptosis was low and there was no correlation with the results of the clonogenic assays: we noted a difference between cytotoxic effects in the clonogenic assay and the extent of apoptosis by fluorescence-activated cell sorter analysis, suggesting that cell killing reflected not only apoptosis but also cell cycle arrest, and that apoptosis, cell kinetics and clonogenicity suppression were independent processes.
Collapse
Affiliation(s)
- Cristiana Lo Nigro
- Molecular Biology Unit, Azienda Ospedaliera S. Croce e Carle, Cuneo, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Duale N, Lindeman B, Komada M, Olsen AK, Andreassen A, Soderlund EJ, Brunborg G. Molecular portrait of cisplatin induced response in human testis cancer cell lines based on gene expression profiles. Mol Cancer 2007; 6:53. [PMID: 17711579 PMCID: PMC1988831 DOI: 10.1186/1476-4598-6-53] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2007] [Accepted: 08/21/2007] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Testicular germ cell tumors (TGCTs) respond well to cisplatin-based chemotherapy and show a low incidence of acquired resistance compared to most somatic tumors. The reasons for these specific characteristics are not known in detail but seem to be multifactorial. We have studied gene expression profiles of testicular and colon cancer derived cell lines treated with cisplatin. The main goal of this study was to identify novel gene expression profiles with their functional categories and the biochemical pathways that are associated with TGCT cells' response to cisplatin. RESULTS Genes that were differentially expressed between the TGCT cell lines vs the (somatic) HCT116 cell line, after cisplatin treatment, were identified using the significance analysis of microarrays (SAM) method. The response of TGCT cells was strikingly different from that of HCT116, and we identified 1794 genes that were differentially expressed. Functional classification of these genes showed that they participate in a variety of different and widely distributed functional categories and biochemical pathways. Database mining showed significant association of genes (n = 41) induced by cisplatin in our study, and genes previously reported to by expressed in differentiated TGCT cells. We identified 37 p53-responsive genes that were altered after cisplatin exposure. We also identified 40 target genes for two microRNAs, hsa-mir-372 and 373 that may interfere with p53 signaling in TGCTs. The tumor suppressor genes NEO1 and LATS2, and the estrogen receptor gene ESR1, all have binding sites for p53 and hsa-mir-372/373. NEO1 and LATS2 were down-regulated in TGCT cells following cisplatin exposure, while ESR1 was up-regulated in TGCT cells. Cisplatin-induced genes associated with terminal growth arrest through senescence were identified, indicating associations which were not previously described for TGCT cells. CONCLUSION By linking our gene expression data to publicly available databases and literature, we provide a global pattern of cisplatin induced cellular response that is specific for testicular cancer cell lines. We have identified cisplatin-responsive functional classes and pathways, such as the angiogenesis, Wnt, integrin, and cadherin signaling pathways. The identification of differentially expressed genes in this study may contribute to a better understanding of the unusual sensitivity of TGCT to some DNA-damaging agents.
Collapse
Affiliation(s)
- Nur Duale
- Department of Chemical Toxicology, Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway
| | - Birgitte Lindeman
- Department of Chemical Toxicology, Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway
| | - Mitsuko Komada
- Department of Chemical Toxicology, Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway
| | - Ann-Karin Olsen
- Department of Chemical Toxicology, Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway
| | - Ashild Andreassen
- Department of Chemical Toxicology, Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway
| | - Erik J Soderlund
- Department of Chemical Toxicology, Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway
| | - Gunnar Brunborg
- Department of Chemical Toxicology, Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
46
|
Affiliation(s)
- Yongwon Jung
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139-4307, USA
| | | |
Collapse
|
47
|
Sarma MS, Mazumder S, Ghosh D, Roy A, Duthie A, Tiekink ERT. Synthesis, spectroscopic characterization and biocidal activity of some diorganotin(IV) complexes of salicylaldehydethiosemicarbazones and related ligands. Molecular and supramolecular structures of [R2Sn(OArCHNNCSNH2)], where R = Me, Ph and Ar = C6H4, C6H3(5-Br) and C6H3(5-Cl), and of [Me2Sn{OC6H3(5-Br)CHNNCSNH2}]·OH2. Appl Organomet Chem 2007. [DOI: 10.1002/aoc.1301] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
48
|
Bodo J, Hunakova L, Kvasnicka P, Jakubikova J, Duraj J, Kasparkova J, Sedlak J. Sensitisation for cisplatin-induced apoptosis by isothiocyanate E-4IB leads to signalling pathways alterations. Br J Cancer 2006; 95:1348-53. [PMID: 17060935 PMCID: PMC2360594 DOI: 10.1038/sj.bjc.6603434] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
A new synthetic isothiocyanate (ITC) derivative, ethyl 4-isothiocyanatobutanoate (E-4IB), appeared to be an effective modulator of cellular proliferation and potent inducer of apoptosis. In cooperation with cisplatin, this compound exerted synergistic effects in human ovarian carcinoma A2780 cells. In the present study we investigated in more detail E4IB-sensitisation for cisplatin-induced apoptosis. Sequential administration of both cytostatic agents led to increased intracellular platinum accumulation, glutathione level depletion and mitochondrial membrane potential dissipation. These events were accompanied with poly (ADP-ribosyl) polymerase cleavage, stimulation of caspase-3 activity, upregulation of p53, FasL and Gadd45alpha, cyclin B1 downregulation and an increase in mitogen-activated protein kinases JNK, ERK and p38 phosphorylation as well as PI3K level alterations. The presented results might have implications for developing new strategies aimed at therapeutic benefit of natural or synthetic ITCs in cooperation with various anticancer drugs.
Collapse
Affiliation(s)
- J Bodo
- Laboratory of Tumour Immunology, Cancer Research Institute, Slovak Academy of Sciences, Vlarska 7, Bratislava 833 91, Slovakia
| | - L Hunakova
- Laboratory of Tumour Immunology, Cancer Research Institute, Slovak Academy of Sciences, Vlarska 7, Bratislava 833 91, Slovakia
| | - P Kvasnicka
- Department of Nuclear Physics and Biophysics, Faculty of Mathematics, Physics and Informatics, Comenius University, Mlynska dolina F1, Bratislava 842 15, Slovakia
| | - J Jakubikova
- Laboratory of Tumour Immunology, Cancer Research Institute, Slovak Academy of Sciences, Vlarska 7, Bratislava 833 91, Slovakia
| | - J Duraj
- Laboratory of Tumour Immunology, Cancer Research Institute, Slovak Academy of Sciences, Vlarska 7, Bratislava 833 91, Slovakia
| | - J Kasparkova
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Kralovopolska 135, Brno CZ-61265, Czech Republic
| | - J Sedlak
- Laboratory of Tumour Immunology, Cancer Research Institute, Slovak Academy of Sciences, Vlarska 7, Bratislava 833 91, Slovakia
- E-mail:
| |
Collapse
|
49
|
Giuliano CJ, Freemantle SJ, Spinella MJ. Testicular Germ Cell Tumors: A Paradigm for the Successful Treatment of Solid Tumor Stem Cells. CURRENT CANCER THERAPY REVIEWS 2006; 2:255-270. [PMID: 24482633 PMCID: PMC3904303 DOI: 10.2174/157339406777934681] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Treatment of testicular germ cell tumors (TGCTs) has been a success primarily due to the exquisite responsiveness of this solid tumor to cisplatin-based therapy. Despite the promise of cure for the majority of TGCT patients, the effectiveness of therapy for some patients is limited by toxicity and the problem of resistance. There is compelling rationale to further understand the biology of TGCTs in order to better treat other solid tumors and to address the shortcomings of present TGCT therapies. TGCTs contain undifferentiated pluripotent stem cells, known as embryonal carcinoma, that share many properties with human embryonic stem cells. The importance of cancer stem cells in the initiation, progression and treatment of solid tumors is beginning to emerge. We discuss TGCTs in the context of solid tumor curability and targeted cancer stem cell therapy.
Collapse
Affiliation(s)
- Caryl J. Giuliano
- Department of Pharmacology and Toxicology, Dartmouth Medical School, and the Norris Cotton Cancer Center, Dartmouth Hitchcock-Medical Center, Hanover, NH 03755, USA
| | - Sarah J. Freemantle
- Department of Pharmacology and Toxicology, Dartmouth Medical School, and the Norris Cotton Cancer Center, Dartmouth Hitchcock-Medical Center, Hanover, NH 03755, USA
| | - Michael J. Spinella
- Department of Pharmacology and Toxicology, Dartmouth Medical School, and the Norris Cotton Cancer Center, Dartmouth Hitchcock-Medical Center, Hanover, NH 03755, USA
| |
Collapse
|
50
|
Widlak P, Pietrowska M, Lanuszewska J. The role of chromatin proteins in DNA damage recognition and repair. Histochem Cell Biol 2006; 125:119-26. [PMID: 16163486 DOI: 10.1007/s00418-005-0053-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The structure of chromatin is the major factor determining the rate and efficiency of DNA repair. Chromatin remodeling events such as rearrangement of nucleosomes and higher order chromatin structures are indispensable features of repair processes. During the last decade numerous chromatin proteins have been identified that preferentially bind to different types of DNA damage. The HMGB proteins, which preferentially interact with DNA intrastrand crosslinks induced by cisplatin, are the archetypal example of such proteins. Several hypothetical models have been proposed describing the role of such damage-binding chromatin proteins. The damage shielding model postulates that binding of chromatin proteins to damaged DNA might disturb damage recognition by repair factors and impair its removal. Alternatively, the damage-recognition/signaling model proposes that the binding of specific chromatin proteins to damaged DNA could serve as a hallmark to be recognized by repair proteins. Additionally, the binding of specific chromatin proteins to damaged DNA could induce chromatin remodeling at the damage site and indirectly affect its repair. This paper aims to critically review current experimental data in relation to such possible roles of chromatin proteins.
Collapse
Affiliation(s)
- Piotr Widlak
- Department of Experimental and Clinical Radiobiology, Maria Sklodowska-Curie Cancer Center and Institute of Oncology, Wybrzeze AK 15, 44-100, Gliwice, Poland.
| | | | | |
Collapse
|