1
|
Bork T, Hernando-Erhard C, Liang W, Tian Z, Yamahara K, Huber TB. Cisplatin Nephrotoxicity Is Critically Mediated by the Availability of BECLIN1. Int J Mol Sci 2024; 25:2560. [PMID: 38473806 DOI: 10.3390/ijms25052560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/10/2024] [Accepted: 02/13/2024] [Indexed: 03/14/2024] Open
Abstract
Cisplatin nephrotoxicity is a critical limitation of solid cancer treatment. Until now, the complex interplay of various pathophysiological mechanisms leading to proximal tubular cell apoptosis after cisplatin exposure has not been fully understood. In our study, we assessed the role of the autophagy-related protein BECLIN1 (ATG6) in cisplatin-induced acute renal injury (AKI)-a candidate protein involved in autophagy and with putative impact on apoptosis by harboring a B-cell lymphoma 2 (BCL2) interaction site of unknown significance. By using mice with heterozygous deletion of Becn1, we demonstrate that reduced intracellular content of BECLIN1 does not impact renal function or autophagy within 12 months. However, these mice were significantly sensitized towards cisplatin-induced AKI, and by using Becn1+/-;Sglt2-Cre;Tomato/EGFP mice with subsequent primary cell analysis, we confirmed that nephrotoxicity depends on proximal tubular BECLIN1 content. Mechanistically, BECLIN1 did not impact autophagy or primarily the apoptotic pathway. In fact, a lack of BECLIN1 sensitized mice towards cisplatin-induced ER stress. Accordingly, the ER stress inhibitor tauroursodeoxycholic acid (TUDCA) blunted cisplatin-induced cell death in Becn1 heterozygosity. In conclusion, our data first highlight a novel role of BECLIN1 in protecting against cellular ER stress independent from autophagy. These novel findings open new therapeutic avenues to intervene in this important intracellular stress response pathway with a promising impact on future AKI management.
Collapse
Affiliation(s)
- Tillmann Bork
- Department of Medicine IV, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Camila Hernando-Erhard
- Department of Medicine IV, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Wei Liang
- Department of Medicine IV, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430064, China
| | - Zhejia Tian
- Department of Nephrology and Hypertension, Hannover Medical School, 30625 Hannover, Germany
| | - Kosuke Yamahara
- Department of Medicine IV, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Department of Medicine, Shiga University of Medical Science, Tsukinowa-cho, Otsu 520-2192, Shiga, Japan
| | - Tobias B Huber
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
2
|
Yang GM, Xu L, Wang RM, Tao X, Zheng ZW, Chang S, Ma D, Zhao C, Dong Y, Wu S, Guo J, Wu ZY. Structures of the human Wilson disease copper transporter ATP7B. Cell Rep 2023; 42:112417. [PMID: 37074913 DOI: 10.1016/j.celrep.2023.112417] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 02/22/2023] [Accepted: 04/05/2023] [Indexed: 04/20/2023] Open
Abstract
The P-type ATPase ATP7B exports cytosolic copper and plays an essential role in the regulation of cellular copper homeostasis. Mutants of ATP7B cause Wilson disease (WD), an autosomal recessive disorder of copper metabolism. Here, we present cryoelectron microscopy (cryo-EM) structures of human ATP7B in the E1 state in the apo, the putative copper-bound, and the putative cisplatin-bound forms. In ATP7B, the N-terminal sixth metal-binding domain (MBD6) binds at the cytosolic copper entry site of the transmembrane domain (TMD), facilitating the delivery of copper from the MBD6 to the TMD. The sulfur-containing residues in the TMD of ATP7B mark the copper transport pathway. By comparing structures of the E1 state human ATP7B and E2-Pi state frog ATP7B, we propose the ATP-driving copper transport model of ATP7B. These structures not only advance our understanding of the mechanisms of ATP7B-mediated copper export but can also guide the development of therapeutics for the treatment of WD.
Collapse
Affiliation(s)
- Guo-Min Yang
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Lingyi Xu
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Rou-Min Wang
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Xin Tao
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China
| | - Zi-Wei Zheng
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Shenghai Chang
- Department of Biophysics, and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; Center of Cryo Electron Microscopy, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Demin Ma
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Cheng Zhao
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Yi Dong
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Shan Wu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, Hubei 430062, China.
| | - Jiangtao Guo
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; State Key Laboratory of Plant Physiology and Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Department of Cardiology, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310058, China; Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, Zhejiang 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China.
| | - Zhi-Ying Wu
- Department of Medical Genetics and Center for Rare Diseases, and Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, Zhejiang 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China.
| |
Collapse
|
3
|
Su Y, Zhang X, Li S, Xie W, Guo J. Emerging roles of the copper-CTR1 axis in tumorigenesis. Mol Cancer Res 2022; 20:1339-1353. [PMID: 35604085 DOI: 10.1158/1541-7786.mcr-22-0056] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/09/2022] [Accepted: 05/17/2022] [Indexed: 11/16/2022]
Abstract
Physiological roles of copper in metabolic homeostasis have been well established, however, whether and how copper is dysregulated in tumors and contributes to tumorigenesis are not recapitulated. Here, we comprehensively summarize the potential origins of copper accumulation in diseases especially in cancers by dysregulating copper transporter 1 (CTR1) or ATPase copper transporting alpha/beta (ATP7A/B) and further demonstrate the underlying mechanism of copper contributing to tumorigenesis. Specifically, in addition to modulating reactive oxygen species (ROS), angiogenesis, immune response, and metabolic homeostasis, copper recently has drawn more attention by directly binding to oncoproteins such as MEK, ULK, Memo, and PDK1 to activate distinct oncogenic signals and account for tumorigenesis. In the end, we disclose the emerging applications of copper in cancer diagnosis and highlight the promising strategies to target the copper-CTR1 axis for cancer therapies.
Collapse
Affiliation(s)
- Yaqing Su
- First Affiliated Hospital of Sun Yat-sen University, guangzhou, guangdong, China
| | - Xiaomei Zhang
- First Affiliated Hospital of Sun Yat-sen University, China
| | - Shaoqiang Li
- The First Affiliatd Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Xie
- First Affiliated Hospital of Sun Yat-sen University, China
| | - Jianping Guo
- First Affiliated Hospital of Sun Yat-sen University, guangzhou, guangdong, China
| |
Collapse
|
4
|
Ovarian Cancer: Treatment and Resistance to Pharmacotherapy. REPRODUCTIVE MEDICINE 2022. [DOI: 10.3390/reprodmed3020011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Despite advances in surgical techniques and chemotherapy, ovarian cancer is still a leading cause of death among gynecological cancers. In addition to the late detection of the disease, the main reason for poor prognosis is resistance to pharmacotherapy, mostly platinum compounds. About a third of patients do not respond to primary platinum-based chemotherapy treatment, and over time, eventually, 80% of other patients develop chemoresistance, which makes the recurrence of disease incurable. In this review, we describe a difficult clinical hurdle faced in ovarian cancer therapy as a result of platinum resistance, as well as resistance to newer targeted therapy with PARP inhibitors and bevacizumab. We, furthermore, give attention also to the role of the tumor microenvironment as it is less well understood than the tumor cell-intrinsic mechanism. Because a central goal in ovarian cancer research is the development of novel strategies to overcome chemoresistance, treatment for cancer is moving toward personalized therapy.
Collapse
|
5
|
Interference between copper transport systems and platinum drugs. Semin Cancer Biol 2021; 76:173-188. [PMID: 34058339 DOI: 10.1016/j.semcancer.2021.05.023] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/17/2021] [Indexed: 01/06/2023]
Abstract
Cisplatin, or cis-diamminedichloridoplatinum(II) cis-[PtCl2(NH3)2], is a platinum-based anticancer drug largely used for the treatment of various types of cancers, including testicular, ovarian and colorectal carcinomas, sarcomas, and lymphomas. Together with other platinum-based drugs, cisplatin triggers malignant cell death by binding to nuclear DNA, which appears to be the ultimate target. In addition to passive diffusion across the cell membrane, other transport systems, including endocytosis and some active or facilitated transport mechanisms, are currently proposed to play a pivotal role in the uptake of platinum-based drugs. In this review, an updated view of the current literature regarding the intracellular transport and processing of cisplatin will be presented, with special emphasis on the plasma membrane copper permease CTR1, the Cu-transporting ATPases, ATP7A and ATP7B, located in the trans-Golgi network, and the soluble copper chaperone ATOX1. Their role in eliciting cisplatin efficacy and their exploitation as pharmacological targets will be addressed.
Collapse
|
6
|
Mittal D, Biswas L, Verma AK. Redox resetting of cisplatin-resistant ovarian cancer cells by cisplatin-encapsulated nanostructured lipid carriers. Nanomedicine (Lond) 2021; 16:979-995. [PMID: 33970681 DOI: 10.2217/nnm-2020-0400] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aim: To sensitize cisplatin (Cis)-resistant ovarian cancer cells toward Cis using Cis-loaded nanostructured lipid carriers (CisNLCs). Materials & methods: CisNLCs were synthesized and characterized using dynamic light scattering, Fourier transform IR and x-ray diffraction (XRD). Sensitivity of PA-1 and CaOV3 cells to Cis and its biotoxicity were assessed. Further, expression of the Cis-resistance markers GSTPi and ATP7B, and apoptotic markers Bax, Bcl2 and Cas9 were quantified by real-time PCR. Results: The size of synthesized CisNLCs was approximately 179.3 ± 2.32 nm and surface charge was -33.9 ± 1.47 mV. IC50 was 210 μg/ml in PA-1 and 500 μg/ml in CaOV3. CisNLCs modulated reactive oxygen species levels in CaOV3 cells. Reduced GSTPi and decreased Cis efflux via ATP7B sequestration caused Cis to accumulate in cytoplasm, thereby augmenting apoptosis in cells. Conclusion: CisNLCs sensitize CaOV3 by redox resetting, indicating their immense therapeutic potential.
Collapse
Affiliation(s)
- Disha Mittal
- Department of Zoology, Nanobiotech Lab, Kirori Mal College, University of Delhi, Delhi, 110007, India
| | - Largee Biswas
- Department of Zoology, Nanobiotech Lab, Kirori Mal College, University of Delhi, Delhi, 110007, India
| | - Anita Kamra Verma
- Department of Zoology, Nanobiotech Lab, Kirori Mal College, University of Delhi, Delhi, 110007, India
| |
Collapse
|
7
|
Lukanović D, Herzog M, Kobal B, Černe K. The contribution of copper efflux transporters ATP7A and ATP7B to chemoresistance and personalized medicine in ovarian cancer. Biomed Pharmacother 2020; 129:110401. [DOI: 10.1016/j.biopha.2020.110401] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/23/2020] [Accepted: 06/13/2020] [Indexed: 02/08/2023] Open
|
8
|
Curnock R, Cullen PJ. Mammalian copper homeostasis requires retromer-dependent recycling of the high-affinity copper transporter 1. J Cell Sci 2020; 133:133/16/jcs249201. [PMID: 32843536 PMCID: PMC7473646 DOI: 10.1242/jcs.249201] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/20/2020] [Indexed: 12/13/2022] Open
Abstract
The concentration of essential micronutrients, such as copper (used here to describe both Cu+ and Cu2+), within the cell is tightly regulated to avoid their adverse deficiency and toxicity effects. Retromer-mediated sorting and recycling of nutrient transporters within the endo-lysosomal network is an essential process in regulating nutrient balance. Cellular copper homeostasis is regulated primarily by two transporters: the copper influx transporter copper transporter 1 (CTR1; also known as SLC31A1), which controls the uptake of copper, and the copper-extruding ATPase ATP7A, a recognised retromer cargo. Here, we show that in response to fluctuating extracellular copper, retromer controls the delivery of CTR1 to the cell surface. Following copper exposure, CTR1 is endocytosed to prevent excessive copper uptake. We reveal that internalised CTR1 localises on retromer-positive endosomes and, in response to decreased extracellular copper, retromer controls the recycling of CTR1 back to the cell surface to maintain copper homeostasis. In addition to copper, CTR1 plays a central role in the trafficking of platinum. The efficacy of platinum-based cancer drugs has been correlated with CTR1 expression. Consistent with this, we demonstrate that retromer-deficient cells show reduced sensitivity to the platinum-based drug cisplatin. Summary: CTR1 (SLC31A1) is the only known mammalian importer of copper. We show that CTR1 is a retromer complex cargo protein, and that retromer is required for cellular sensitivity to extracellular copper.
Collapse
Affiliation(s)
- Rachel Curnock
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol BS8 1TD, UK
| | - Peter J Cullen
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
9
|
Davis CN, Winters A, Milic I, Devitt A, Cookson A, Brophy PM, Morphew RM. Evidence of sequestration of triclabendazole and associated metabolites by extracellular vesicles of Fasciola hepatica. Sci Rep 2020; 10:13445. [PMID: 32778698 PMCID: PMC7418001 DOI: 10.1038/s41598-020-69970-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/21/2020] [Indexed: 12/21/2022] Open
Abstract
Fascioliasis is a neglected zoonotic disease that infects humans and ruminant species worldwide. In the absence of vaccines, control of fascioliasis is primarily via anthelminthic treatment with triclabendazole (TCBZ). Parasitic flatworms, including Fasciola hepatica, are active secretors of extracellular vesicles (EVs), but research has not been undertaken investigating EV anthelmintic sequestration. Adult F. hepatica were cultured in lethal and sub-lethal doses of TCBZ and its active metabolites, in order to collect EVs and evaluate their morphological characteristics, production and anthelmintic metabolite content. Transmission electron microscopy demonstrated that F. hepatica exposed to TCBZ and its metabolites produced EVs of similar morphology, compared to non-TCBZ exposed controls, even though TCBZ dose and/or TCBZ metabolite led to measurable structural changes in the treated F. hepatica tegument. qNano particle analysis revealed that F. hepatica exposed to TCBZ and its metabolites produced at least five times greater EV concentrations than non-TCBZ controls. A combined mass spectrometry and qNano particle analysis confirmed the presence of TCBZ and the TCBZ–sulphoxide metabolite in anthelmintic exposed EVs, but limited TCBZ sulphone was detectable. This data suggests that EVs released from adult F. hepatica have a biological role in the sequestration of TCBZ and additional toxic xenobiotic metabolites.
Collapse
Affiliation(s)
- Chelsea N Davis
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, UK.
| | - Ana Winters
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, UK
| | - Ivana Milic
- School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Andrew Devitt
- School of Life and Health Sciences, Aston University, Birmingham, UK
| | - Alan Cookson
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, UK
| | - Peter M Brophy
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, UK
| | - Russell M Morphew
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, UK
| |
Collapse
|
10
|
Synthetic Lethality Screening Identifies FDA-Approved Drugs that Overcome ATP7B-Mediated Tolerance of Tumor Cells to Cisplatin. Cancers (Basel) 2020; 12:cancers12030608. [PMID: 32155756 PMCID: PMC7139527 DOI: 10.3390/cancers12030608] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 02/27/2020] [Accepted: 03/01/2020] [Indexed: 12/26/2022] Open
Abstract
Tumor resistance to chemotherapy represents an important challenge in modern oncology. Although platinum (Pt)-based drugs have demonstrated excellent therapeutic potential, their effectiveness in a wide range of tumors is limited by the development of resistance mechanisms. One of these mechanisms includes increased cisplatin sequestration/efflux by the copper-transporting ATPase, ATP7B. However, targeting ATP7B to reduce Pt tolerance in tumors could represent a serious risk because suppression of ATP7B might compromise copper homeostasis, as happens in Wilson disease. To circumvent ATP7B-mediated Pt tolerance we employed a high-throughput screen (HTS) of an FDA/EMA-approved drug library to detect safe therapeutic molecules that promote cisplatin toxicity in the IGROV-CP20 ovarian carcinoma cells, whose resistance significantly relies on ATP7B. Using a synthetic lethality approach, we identified and validated three hits (Tranilast, Telmisartan, and Amphotericin B) that reduced cisplatin resistance. All three drugs induced Pt-mediated DNA damage and inhibited either expression or trafficking of ATP7B in a tumor-specific manner. Global transcriptome analyses showed that Tranilast and Amphotericin B affect expression of genes operating in several pathways that confer tolerance to cisplatin. In the case of Tranilast, these comprised key Pt-transporting proteins, including ATOX1, whose suppression affected ability of ATP7B to traffic in response to cisplatin. In summary, our findings reveal Tranilast, Telmisartan, and Amphotericin B as effective drugs that selectively promote cisplatin toxicity in Pt-resistant ovarian cancer cells and underscore the efficiency of HTS strategy for identification of biosafe compounds, which might be rapidly repurposed to overcome resistance of tumors to Pt-based chemotherapy.
Collapse
|
11
|
Tang FK, Zhu J, Kong FKW, Ng M, Bian Q, Yam VWW, Tse AKW, Tse YC, Leung KCF. A BODIPY-based fluorescent sensor for the detection of Pt2+ and Pt drugs. Chem Commun (Camb) 2020; 56:2695-2698. [DOI: 10.1039/d0cc00027b] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
A novel BODIPY-based fluorescent sensor PS was designed for imaging Pt2+, cisplatin and nedaplatin in aqueous medium and biological environments, providing great potential for studying the Pt-drug metabolism and the development of new platinum drugs.
Collapse
Affiliation(s)
- Fung-Kit Tang
- Department of Chemistry, State Key Laboratory of Environmental and Biological Analysis
- Hong Kong Baptist University
- Kowloon Tong
- P. R. China
| | - Jiaqian Zhu
- Centre for Cancer and Inflammation Research
- School of Chinese Medicine
- Hong Kong Baptist University
- P. R. China
| | | | - Maggie Ng
- Department of Chemistry
- The University of Hong Kong
- P. R. China
| | - Qingyuan Bian
- Department of Chemistry, State Key Laboratory of Environmental and Biological Analysis
- Hong Kong Baptist University
- Kowloon Tong
- P. R. China
| | | | - Anfernee Kai-Wing Tse
- Programme of Food Science and Technology, Division of Science and Technology
- Beijing Normal University-Hong Kong Baptist University United International College
- Zhuhai 519087
- P. R. China
| | - Yu-Chung Tse
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research
- Department of Biology
- Southern University of Science and Technology (SUSTech)
- Shenzhen
- P. R. China
| | - Ken Cham-Fai Leung
- Department of Chemistry, State Key Laboratory of Environmental and Biological Analysis
- Hong Kong Baptist University
- Kowloon Tong
- P. R. China
| |
Collapse
|
12
|
Petruzzelli R, Polishchuk RS. Activity and Trafficking of Copper-Transporting ATPases in Tumor Development and Defense against Platinum-Based Drugs. Cells 2019; 8:E1080. [PMID: 31540259 PMCID: PMC6769697 DOI: 10.3390/cells8091080] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/10/2019] [Accepted: 09/12/2019] [Indexed: 02/06/2023] Open
Abstract
Membrane trafficking pathways emanating from the Golgi regulate a wide range of cellular processes. One of these is the maintenance of copper (Cu) homeostasis operated by the Golgi-localized Cu-transporting ATPases ATP7A and ATP7B. At the Golgi, these proteins supply Cu to newly synthesized enzymes which use this metal as a cofactor to catalyze a number of vitally important biochemical reactions. However, in response to elevated Cu, the Golgi exports ATP7A/B to post-Golgi sites where they promote sequestration and efflux of excess Cu to limit its potential toxicity. Growing tumors actively consume Cu and employ ATP7A/B to regulate the availability of this metal for oncogenic enzymes such as LOX and LOX-like proteins, which confer higher invasiveness to malignant cells. Furthermore, ATP7A/B activity and trafficking allow tumor cells to detoxify platinum (Pt)-based drugs (like cisplatin), which are used for the chemotherapy of different solid tumors. Despite these noted activities of ATP7A/B that favor oncogenic processes, the mechanisms that regulate the expression and trafficking of Cu ATPases in malignant cells are far from being completely understood. This review summarizes current data on the role of ATP7A/B in the regulation of Cu and Pt metabolism in malignant cells and outlines questions and challenges that should be addressed to understand how ATP7A and ATP7B trafficking mechanisms might be targeted to counteract tumor development.
Collapse
Affiliation(s)
- Raffaella Petruzzelli
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli (NA), Italy.
| | - Roman S Polishchuk
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli (NA), Italy.
| |
Collapse
|
13
|
Han Y, Wang T, Liu H, Zhang S, Zhang H, Li M, Sun Q, Li Z. The release and detection of copper ions from ultrasmall theranostic Cu 2-xSe nanoparticles. NANOSCALE 2019; 11:11819-11829. [PMID: 31184674 DOI: 10.1039/c9nr02884f] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Nanoscale copper chalcogenides have been widely used in nanomedicine, however, their pharmacokinetics, degradation, and biological effects of released copper ions are usually overlooked, which are crucial for their future clinical translation. Herein, we report the in vitro and in vivo release of copper ions from polyvinylpyrrolidone (PVP) functionalized ultrasmall copper selenide (Cu2-xSe) theranostic nanoparticles. We synthesized a Cu2+-specific fluorescent probe (NCM), which can quickly and specifically react with copper ions to exhibit very strong near infrared fluorescence. The in vitro study shows that copper ions can be slowly released from Cu2-xSe nanoparticles in aqueous solution with the progress of their oxidation. The release of copper ions from Cu2-xSe nanoparticles in RAW 264.7 murine macrophages is very fast, evidenced by the gradual increase of fluorescence intensity and the diffusion of fluorescence from cytoplasm into nuclei. We also demonstrate the distribution, degradation, and the metabolism of ultrasmall Cu2-xSe nanoparticles by the in vivo fluorescence imaging, the blood routine test, blood biochemistry and histology analysis, and the characterization of copper transport and binding proteins. The results show that ultrasmall Cu2-xSe nanoparticles were mainly eliminated through feces and urine from the body within 72 h after intravenous injection, and the released copper ions did not cause severe toxicity. Our research highlights the great potential of copper chalcogenide nanoparticles in nanomedicine.
Collapse
Affiliation(s)
- Yaobao Han
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Ding D, Jiang H, Zhang J, Xu X, Qi W, Shi H, Yin S, Salvi R. Cisplatin-induced vestibular hair cell lesion-less damage at high doses. J Otol 2018; 13:115-121. [PMID: 30671086 PMCID: PMC6335437 DOI: 10.1016/j.joto.2018.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/10/2018] [Accepted: 08/20/2018] [Indexed: 12/04/2022] Open
Abstract
Cisplatin, a widely used anticancer drug, damages hair cells in cochlear organotypic cultures at low doses, but paradoxically causes little damage at high doses resulting in a U-shaped dose-response function. To determine if the cisplatin dose-response function for vestibular hair cells follows a similar pattern, we treated vestibular organotypic cultures with doses of cisplatin ranging from 10 to 1000 μM. Vestibular hair cell lesions progressively increased as the dose of cisplatin increased with maximum damage occurring around 50–100 μM, but the lesions progressively decreased at higher doses resulting in little hair cell loss at 1000 μM. The U-shaped dose-response function for cisplatin-treated vestibular hair cells in culture appears to be regulated by copper transporters, Ctr1, ATP7A and ATP7B, that dose-dependently regulate the uptake, sequestration and extrusion of cisplatin.
Collapse
Affiliation(s)
- Dalian Ding
- Center for Hearing and Deafness, University at Buffalo, USA.,Department of Otolaryngology Head and Neck Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Haiyan Jiang
- Center for Hearing and Deafness, University at Buffalo, USA
| | - Jianhui Zhang
- Department of Otolaryngology Head and Neck Surgery, The Third People's Hospital of Chengdu, China
| | | | - Weidong Qi
- Department of Otolaryngology Head and Neck Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Haibo Shi
- Department of Otorhinolaryngology, Affiliated Sixth People's Hospital of Shanghai Jiaotong University, Shanghai, China
| | - Shankai Yin
- Department of Otorhinolaryngology, Affiliated Sixth People's Hospital of Shanghai Jiaotong University, Shanghai, China
| | - Richard Salvi
- Center for Hearing and Deafness, University at Buffalo, USA.,Department of Audiology and Speech-Language Pathology, Asia University, China
| |
Collapse
|
15
|
Li YQ, Yin JY, Liu ZQ, Li XP. Copper efflux transporters ATP7A and ATP7B: Novel biomarkers for platinum drug resistance and targets for therapy. IUBMB Life 2018; 70:183-191. [PMID: 29394468 DOI: 10.1002/iub.1722] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 01/12/2018] [Indexed: 12/22/2022]
Abstract
Platinum-based chemotherapy agents are widely used in the treatment of various solid malignancies. However, their efficacy is limited by drug resistance. Recent studies suggest that copper efflux transporters, which are encoded by ATP7A and ATP7B, play an important role in platinum drug resistance. Over-expressions of ATP7A and ATP7B are observed in multiple cancers. Moreover, their expressions are associated with cancer prognosis and treatment outcomes of platinum-based chemotherapy. In our review, we highlight the roles of ATP7A/7B in platinum drug resistance and cancer progression. We also discuss the possible mechanisms of platinum drug resistance mediated by ATP7A/7B and provide novel strategies for overcoming resistance. This review may be helpful for understanding the roles of ATP7A and ATP7B in platinum drug resistance. © 2018 IUBMB Life, 70(3):183-191, 2018.
Collapse
Affiliation(s)
- Yue-Qin Li
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, People's Republic of China
| | - Ji-Ye Yin
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, People's Republic of China
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, People's Republic of China
| | - Xiang-Ping Li
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, People's Republic of China
| |
Collapse
|
16
|
Guttmann S, Chandhok G, Groba SR, Niemietz C, Sauer V, Gomes A, Ciarimboli G, Karst U, Zibert A, Schmidt HH. Organic cation transporter 3 mediates cisplatin and copper cross-resistance in hepatoma cells. Oncotarget 2017; 9:743-754. [PMID: 29416650 PMCID: PMC5787505 DOI: 10.18632/oncotarget.23142] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 11/15/2017] [Indexed: 12/12/2022] Open
Abstract
Platinum-based drugs are first-line compounds in the treatment of many solid cancers. Major obstacles are tumors that become resistant and toxic side effects, both largely due to the expression of transporters that mediate the cellular processing of platinum. In this study, we addressed the establishment of cisplatin resistance in the absence of copper transporter ATP7B that has been previously found to be overexpressed in various resistant cells. Cisplatin sensitivity, induction of apoptosis, drug accumulation, and transporter gene expression were determined in hepatoma cell lines. Knockout or overexpression of copper transporter ATP7B did not affect cisplatin sensitivity. Cisplatin resistant cells showed a stably reduced cisplatin accumulation and a downregulation of organic cation transporter 3 (OCT3). In contrast, OCT3 overexpression could reverse resistance. Reduced MT1 expression was detected in the resistant cell line, however transient and highly dependent on the presence of cisplatin. Cross-resistance to copper was also associated with OCT3 downregulation. Our results suggest that a decreased level of OCT3 expression results in resistance to cisplatin and copper. OCT3 may represent a novel target for improved prognosis and anticancer therapy, including HCC.
Collapse
Affiliation(s)
- Sarah Guttmann
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany
| | - Gursimran Chandhok
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany.,Present address: Monash Biomedicine Discovery Institute, and Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Sara Reinartz Groba
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany
| | - Christoph Niemietz
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany
| | - Vanessa Sauer
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany
| | - Amanda Gomes
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany.,Present address: Wilson Disease Clinic, Kokilaben Dhirubhai Ambani Hospital and Medical Research Institute, Mumbai, India
| | - Giuliano Ciarimboli
- Universitätsklinikum Münster, Medizinische Klinik D, Experimentelle Nephrologie, Münster, Germany
| | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany
| | - Andree Zibert
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany
| | - Hartmut H Schmidt
- Medizinische Klinik B für Gastroenterologie und Hepatologie, Universitätsklinikum Münster, Münster, Germany
| |
Collapse
|
17
|
Moinuddin FM, Shinsato Y, Komatsu M, Mitsuo R, Minami K, Yamamoto M, Kawahara K, Hirano H, Arita K, Furukawa T. ATP7B expression confers multidrug resistance through drug sequestration. Oncotarget 2017; 7:22779-90. [PMID: 26988911 PMCID: PMC5008400 DOI: 10.18632/oncotarget.8059] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 02/23/2016] [Indexed: 11/30/2022] Open
Abstract
We previously reported that ATP7B is involved in cisplatin resistance and ATP7A confers multidrug resistance (MDR) in cancer cells. In this study, we show that ATP7B expressing cells also are resistant to doxorubicin, SN-38, etoposide, and paclitaxel as well as cisplatin. In ATP7B expressing cells, doxorubicin relocated from the nuclei to the late-endosome at 4 hours after doxorubicin exposure. EGFP-ATP7B mainly colocalized with doxorubicin. ATP7B has six metal binding sites (MBSs) in the N-terminal cytoplasmic region. To investigate the role of the MBSs of ATP7B in doxorubicin resistance, we used three mutant ATP7B (Cu0, Cu6 and M6C/S) expressing cells. Cu0 has no MBSs, Cu6 has only the sixth MBS and M6C/S carries CXXC to SXXS mutation in the sixth MBS. Cu6 expressing cells were less resistance to the anticancer agents than wild type ATP7B expressing cells, and had doxorubicin sequestration in the late-endosome. Cu0- and M6C/S-expressing cells were sensitive to doxorubicin. In these cells, doxorubicin did not relocalize to the late-endosome. EGFP-M6C/S mainly localized to the trans-Golgi network (TGN) even in the presence of copper. Thus the cysteine residues in the sixth MBS of ATP7B are essential for MDR phenotype. Finally, we found that ammonium chloride and tamoxifen suppressed late endosomal sequestration of doxorubicin, thereby attenuating drug resistance. These results suggest that the sequestration depends on the acidity of the vesicles partly. We here demonstrate that ATP7B confers MDR by facilitating nuclear drug efflux and late endosomal drug sequestration.
Collapse
Affiliation(s)
- F M Moinuddin
- Department of Neurosurgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan.,Department of Molecular Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Yoshinari Shinsato
- Department of Molecular Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan.,Center for the Research of Advanced Diagnosis and Therapy of Cancer, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Masaharu Komatsu
- Division of Food and Chemical Biology, Faculty of Fisheries, Kagoshima University, 4-50-20, Shimoarata, Kagoshima 890-0056, Japan
| | - Ryoichi Mitsuo
- Department of Molecular Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Kentaro Minami
- Department of Molecular Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan.,Center for the Research of Advanced Diagnosis and Therapy of Cancer, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Masatatsu Yamamoto
- Department of Molecular Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan.,Center for the Research of Advanced Diagnosis and Therapy of Cancer, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Kohich Kawahara
- Department of Molecular Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan.,Center for the Research of Advanced Diagnosis and Therapy of Cancer, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Hirofumi Hirano
- Department of Neurosurgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Kazunori Arita
- Department of Neurosurgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Tatsuhiko Furukawa
- Department of Molecular Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan.,Center for the Research of Advanced Diagnosis and Therapy of Cancer, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| |
Collapse
|
18
|
A fluorescent oxaliplatin derivative for investigation of oxaliplatin resistance using imaging techniques. J Biol Inorg Chem 2017; 22:1295-1304. [DOI: 10.1007/s00775-017-1502-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 10/13/2017] [Indexed: 01/01/2023]
|
19
|
Zhu S, Shanbhag V, Wang Y, Lee J, Petris M. A Role for The ATP7A Copper Transporter in Tumorigenesis and Cisplatin Resistance. J Cancer 2017; 8:1952-1958. [PMID: 28819394 PMCID: PMC5559955 DOI: 10.7150/jca.19029] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/25/2017] [Indexed: 01/24/2023] Open
Abstract
The ATP7A protein is a ubiquitously expressed copper-translocating P-type ATPase that controls cytoplasmic copper concentrations by mediating cellular copper egress. In vitro studies have previously demonstrated that ATP7A abundance in various tumor cell lines is correlated with increased resistance to cisplatin, a widely-used chemotherapy agent. However, to date no studies have examined a role for ATP7A in tumor growth or cisplatin sensitivity in vivo. In this study, we deleted ATP7A in H-RAS transformed tumorigenic mouse embryonic fibroblasts (MEFRAS7A-). Interestingly, loss of ATP7A was found to markedly suppress tumorigenesis in MEFRAS7A- cells relative to wild type parental cells. This was associated with hyperaccumulation of copper and sensitivity to reactive oxygen species and hypoxia. Tumor grafts lacking ATP7A were markedly more sensitive to cisplatin chemotherapy compared to ATP7A-expressing control tumors. These findings identify ATP7A at the nexus between tumorigenesis and cisplatin resistance pathways, underscoring its potential as a therapeutic target for regulating both tumor growth and the efficacy of cisplatin treatment.
Collapse
Affiliation(s)
- Sha Zhu
- Department of Biochemistry, University of Missouri, Columbia, MO, 65211.,The Christopher S. Bond Life Science Center, University of Missouri, Columbia, MO, 65211
| | - Vinit Shanbhag
- Department of Biochemistry, University of Missouri, Columbia, MO, 65211.,The Christopher S. Bond Life Science Center, University of Missouri, Columbia, MO, 65211
| | - Yanfang Wang
- Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, 65211.,The Christopher S. Bond Life Science Center, University of Missouri, Columbia, MO, 65211
| | - Jaekwon Lee
- Redox Biology Center, Department of Biochemistry, University of Nebraska, Lincoln, Nebraska, 68588
| | - Michael Petris
- Department of Biochemistry, University of Missouri, Columbia, MO, 65211.,Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, 65211.,The Christopher S. Bond Life Science Center, University of Missouri, Columbia, MO, 65211
| |
Collapse
|
20
|
Legin AA, Theiner S, Schintlmeister A, Reipert S, Heffeter P, Jakupec MA, Mayr J, Varbanov HP, Kowol CR, Galanski M, Berger W, Wagner M, Keppler BK. Multi-scale imaging of anticancer platinum(iv) compounds in murine tumor and kidney. Chem Sci 2016; 7:3052-3061. [PMID: 29997796 PMCID: PMC6004953 DOI: 10.1039/c5sc04383b] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 12/22/2015] [Indexed: 01/31/2023] Open
Abstract
Nano-scale secondary ion mass spectrometry (NanoSIMS) enables trace element and isotope analyses with high spatial resolution. This unique capability has recently been exploited in several studies analyzing the subcellular distribution of Au and Pt anticancer compounds. However, these studies were restricted to cell culture systems. To explore the applicability to the in vivo setting, we developed a combined imaging approach consisting of laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS), NanoSIMS and transmission electron microscopy (TEM) suitable for multi-scale detection of the platinum distribution in tissues. Applying this approach to kidney and tumor samples upon administration of selected platinum(iv) anticancer prodrugs revealed uneven platinum distributions on both the organ and subcellular scales. Spatial platinum accumulation patterns were quantitatively assessed by LA-ICP-MS in histologically heterogeneous organs (e.g., higher platinum accumulation in kidney cortex than in medulla) and used to select regions of interest for subcellular-scale imaging with NanoSIMS. These analyses revealed cytoplasmic sulfur-rich organelles accumulating platinum in both kidney and malignant cells. Those in the tumor were subsequently identified as organelles of lysosomal origin, demonstrating the potential of the combinatorial approach for investigating therapeutically relevant drug concentrations on a submicrometer scale.
Collapse
Affiliation(s)
- A A Legin
- Institute of Inorganic Chemistry , Research Platform "Translational Cancer Therapy Research," and Research Network "Chemistry meets Microbiology" , University of Vienna , Währinger Straße 42 , A-1090 Vienna , Austria . ; Tel: +43-1-4277-52600
| | - S Theiner
- Institute of Inorganic Chemistry , Research Platform "Translational Cancer Therapy Research," and Research Network "Chemistry meets Microbiology" , University of Vienna , Währinger Straße 42 , A-1090 Vienna , Austria . ; Tel: +43-1-4277-52600
| | - A Schintlmeister
- Department of Microbiology and Ecosystem Science , Research Network "Chemistry meets Microbiology", and Large-Instrument Facility for Advanced Isotope Research , University of Vienna , A-1090 Vienna , Austria
| | - S Reipert
- Core Facility of Cell Imaging and Ultrastructure Research , University of Vienna , A-1090 Vienna , Austria
| | - P Heffeter
- Institute of Cancer Research , Comprehensive Cancer Center and Research Platform "Translational Cancer Therapy Research" , Medical University of Vienna , A-1090 Vienna , Austria
| | - M A Jakupec
- Institute of Inorganic Chemistry , Research Platform "Translational Cancer Therapy Research," and Research Network "Chemistry meets Microbiology" , University of Vienna , Währinger Straße 42 , A-1090 Vienna , Austria . ; Tel: +43-1-4277-52600
| | - J Mayr
- Institute of Inorganic Chemistry , Research Platform "Translational Cancer Therapy Research," and Research Network "Chemistry meets Microbiology" , University of Vienna , Währinger Straße 42 , A-1090 Vienna , Austria . ; Tel: +43-1-4277-52600
| | - H P Varbanov
- Institute of Inorganic Chemistry , Research Platform "Translational Cancer Therapy Research," and Research Network "Chemistry meets Microbiology" , University of Vienna , Währinger Straße 42 , A-1090 Vienna , Austria . ; Tel: +43-1-4277-52600
| | - C R Kowol
- Institute of Inorganic Chemistry , Research Platform "Translational Cancer Therapy Research," and Research Network "Chemistry meets Microbiology" , University of Vienna , Währinger Straße 42 , A-1090 Vienna , Austria . ; Tel: +43-1-4277-52600
| | - M Galanski
- Institute of Inorganic Chemistry , Research Platform "Translational Cancer Therapy Research," and Research Network "Chemistry meets Microbiology" , University of Vienna , Währinger Straße 42 , A-1090 Vienna , Austria . ; Tel: +43-1-4277-52600
| | - W Berger
- Institute of Cancer Research , Comprehensive Cancer Center and Research Platform "Translational Cancer Therapy Research" , Medical University of Vienna , A-1090 Vienna , Austria
| | - M Wagner
- Department of Microbiology and Ecosystem Science , Research Network "Chemistry meets Microbiology", and Large-Instrument Facility for Advanced Isotope Research , University of Vienna , A-1090 Vienna , Austria
| | - B K Keppler
- Institute of Inorganic Chemistry , Research Platform "Translational Cancer Therapy Research," and Research Network "Chemistry meets Microbiology" , University of Vienna , Währinger Straße 42 , A-1090 Vienna , Austria . ; Tel: +43-1-4277-52600
| |
Collapse
|
21
|
Sun T, Li Z, Xie Z, Jing X. Amphiphilic Cyanine-Platinum Conjugates as Fluorescent Nanodrugs. Chem Asian J 2015; 11:221-5. [DOI: 10.1002/asia.201501163] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 11/08/2015] [Indexed: 01/08/2023]
Affiliation(s)
- Tingting Sun
- State Key Laboratory of Polymer Physics and Chemistry; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 China
- University of Chinese Academy of Sciences; Beijing 100049 China
| | - Zhensheng Li
- State Key Laboratory of Polymer Physics and Chemistry; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 China
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 China
| | - Xiabin Jing
- State Key Laboratory of Polymer Physics and Chemistry; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun 130022 China
| |
Collapse
|
22
|
Jagodinsky JC, Sulima A, Cao Y, Poprawski JE, Blackman BN, Lloyd JR, Swenson RE, Gottesman MM, Hall MD. Evaluation of fluorophore-tethered platinum complexes to monitor the fate of cisplatin analogs. J Biol Inorg Chem 2015; 20:1081-95. [PMID: 26323351 DOI: 10.1007/s00775-015-1290-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 08/01/2015] [Indexed: 12/28/2022]
Abstract
The platinum drugs cisplatin, carboplatin, and oxaliplatin are highly utilized in the clinic and as a consequence have been extensively studied in the laboratory setting, sometimes by generating fluorophore-tagged analogs. Here, we synthesized two Pt(II) complexes containing ethane-1,2-diamine ligands linked to a BODIPY fluorophore, and compared their biological activity with previously reported Pt(II) complexes conjugated to carboxyfluorescein and carboxyfluorescein diacetate. The cytotoxicity and DNA damage capacity of Pt-fluorophore complexes was compared to cisplatin, and the Pt-BODIPY complexes were found to be more cytotoxic with reduced cytotoxicity in cisplatin-resistant cells. Microscopy revealed a predominately cytosolic localization, with nuclear distribution at higher concentrations. Spheroids grown from parent and resistant cells revealed penetration of Pt-BODIPY into spheroids, and retention of the cisplatin-resistant spheroid phenotype. While most activity profiles were retained for the Pt-BODIPY complexes, accumulation in resistant cells was only slightly affected, suggesting that some aspects of Pt-fluorophore cellular pharmacology deviate from cisplatin.
Collapse
Affiliation(s)
- Justin C Jagodinsky
- Laboratory of Cell Biology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, 37 Convent Drive, Rm. 2108, Bethesda, MD, 20892, USA
| | - Agnieszka Sulima
- Imaging Probe Development Center, National Institutes of Health, Rockville, MD, USA
| | - Yiqi Cao
- Laboratory of Cell Biology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, 37 Convent Drive, Rm. 2108, Bethesda, MD, 20892, USA
| | - Joanna E Poprawski
- Laboratory of Cell Biology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, 37 Convent Drive, Rm. 2108, Bethesda, MD, 20892, USA
| | - Burchelle N Blackman
- Imaging Probe Development Center, National Institutes of Health, Rockville, MD, USA
| | - John R Lloyd
- Advanced Mass Spectrometry Facility, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Rolf E Swenson
- Imaging Probe Development Center, National Institutes of Health, Rockville, MD, USA
| | - Michael M Gottesman
- Laboratory of Cell Biology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, 37 Convent Drive, Rm. 2108, Bethesda, MD, 20892, USA.
| | - Matthew D Hall
- Laboratory of Cell Biology, National Cancer Institute, Center for Cancer Research, National Institutes of Health, 37 Convent Drive, Rm. 2108, Bethesda, MD, 20892, USA
| |
Collapse
|
23
|
Harrach S, Ciarimboli G. Role of transporters in the distribution of platinum-based drugs. Front Pharmacol 2015; 6:85. [PMID: 25964760 PMCID: PMC4408848 DOI: 10.3389/fphar.2015.00085] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 04/02/2015] [Indexed: 12/21/2022] Open
Abstract
Platinum derivatives used as chemotherapeutic drugs such as cisplatin and oxaliplatin have a potent antitumor activity. However, severe side effects such as nephro-, oto-, and neurotoxicity are associated with their use. Effects and side effects of platinum-based drugs are in part caused by their transporter-mediated uptake in target and non target cells. In this mini review, the transport systems involved in cellular handling of platinum derivatives are illustrated, focusing on transporters for cisplatin. The copper transporter 1 seems to be of particular importance for cisplatin uptake in tumor cells, while the organic cation transporter (OCT) 2, due to its specific organ distribution, may play a major role in the development of undesired cisplatin side effects. In polarized cells, e.g., in renal proximal tubule cells, apically expressed transporters, such as multidrug and toxin extrusion protein 1, mediate secretion of cisplatin and in this way contribute to the control of its toxic effects. Specific inhibition of cisplatin uptake transporters such as the OCTs may be an attractive therapeutic option to reduce its toxicity, without impairing its antitumor efficacy.
Collapse
Affiliation(s)
- Saliha Harrach
- Experimental Nephrology, Medical Clinic D, University of Münster, University Hospital MünsterMünster, Germany
| | - Giuliano Ciarimboli
- Experimental Nephrology, Medical Clinic D, University of Münster, University Hospital MünsterMünster, Germany
- Interdisciplinary Center for Clinical Research (IZKF), University of Münster, University Hospital MünsterMünster, Germany
| |
Collapse
|
24
|
Sun T, Guan X, Zheng M, Jing X, Xie Z. Mitochondria-Localized Fluorescent BODIPY-Platinum Conjugate. ACS Med Chem Lett 2015; 6:430-3. [PMID: 25941554 DOI: 10.1021/acsmedchemlett.5b00041] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/11/2015] [Indexed: 12/14/2022] Open
Abstract
A convenient synthesis of a BODIPY (1,3,5,7-tetramethyl-8-(4-pyridyl)-4,4'-difluoroboradiazaindacene) labeled platinum compound (BODIPY-Pt) was developed by direct conjugation of cisplatin with the pyridine group of BODIPY. The membrane permeability and selective uptake of BODIPY-Pt in the mitochondria was studied using confocal laser scanning microscopy (CLSM). The fluorescent BODIPY-Pt conjugate showed high cellular proliferation inhibition against human cervical carcinoma (HeLa) and human breast cancer (MCF-7) cells, with half maximal inhibitory concentrations (IC50) of 27.37 and 12.14 μM, respectively. This work highlights the potential of using BODIPY labeled Pt compounds to realize the visualization of Pt distribution in living cells.
Collapse
Affiliation(s)
- Tingting Sun
- State Key Laboratory
of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- The University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Xingang Guan
- State Key Laboratory
of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Life Science Research Center, Beihua University, Jilin 132013, P. R. China
| | - Min Zheng
- State Key Laboratory of Luminescence and Applications, Changchun
Institute of Optics, Fine Mechanics, and Physics, Chinese Academy of Sciences, 3888 East Nanhu Road, Changchun, Jilin 130033, P. R. China
| | - Xiabin Jing
- State Key Laboratory
of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Zhigang Xie
- State Key Laboratory
of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| |
Collapse
|
25
|
Peña B, Barhoumi R, Burghardt RC, Turro C, Dunbar KR. Confocal fluorescence microscopy studies of a fluorophore-labeled dirhodium compound: visualizing metal-metal bonded molecules in lung cancer (A549) cells. J Am Chem Soc 2014; 136:7861-4. [PMID: 24854400 PMCID: PMC4063186 DOI: 10.1021/ja503774m] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Indexed: 02/08/2023]
Abstract
The new dirhodium compound [Rh2(μ-O2CCH3)2(η(1)-O2CCH3)(phenbodipy)(H2O)3][O2CCH3] (1), which incorporates a bodipy fluorescent tag, was prepared and studied by confocal fluorescence microscopy in human lung adenocarcinoma (A549) cells. It was determined that 1 localizes mainly in lysosomes and mitochondria with no apparent nuclear localization in the 1-100 μM range. These results support the conclusion that cellular organelles rather than the nucleus can be targeted by modification of the ligands bound to the Rh2(4+) core. This is the first study of a fluorophore-labeled metal-metal bonded compound, work that opens up new venues for the study of intracellular distribution of dinuclear transition metal anticancer complexes.
Collapse
Affiliation(s)
- Bruno Peña
- Department
of Chemistry, Texas A&M University, College Station, Texas 77842, United States
| | - Rola Barhoumi
- Department
of Veterinary Integrative Biosciences, Texas
A&M University, College Station, Texas 77843, United States
| | - Robert C. Burghardt
- Department
of Veterinary Integrative Biosciences, Texas
A&M University, College Station, Texas 77843, United States
| | - Claudia Turro
- Department
of Chemistry and Biochemistry, The Ohio
State University, Columbus, Ohio 43210, United States
| | - Kim R. Dunbar
- Department
of Chemistry, Texas A&M University, College Station, Texas 77842, United States
| |
Collapse
|
26
|
Li XP, Yin JY, Wang Y, He H, Li X, Gong WJ, Chen J, Qian CY, Zheng Y, Li F, Yin T, Gong ZC, Zhou BT, Zhang Y, Xiao L, Zhou HH, Liu ZQ. The ATP7B genetic polymorphisms predict clinical outcome to platinum-based chemotherapy in lung cancer patients. Tumour Biol 2014; 35:8259-65. [PMID: 24852429 DOI: 10.1007/s13277-014-2072-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 05/06/2014] [Indexed: 01/04/2023] Open
Abstract
This study aims to investigate the influence of ATP7B genetic polymorphism to platinum-based chemotherapy in Chinese Han lung cancer patients. A total of 338 Chinese Han lung cancer patients were enrolled in this study. All patients underwent at least two cycles of platinum-based chemotherapy. Four tag SNPs of ATP7B (rs1061472, rs9535826, rs7999812, and rs9535828) were selected to evaluate their impacts to platinum-based chemotherapy in these patients. ATP7B rs9535828 and rs9535826 were found to be associated with platinum resistance in Chinese Han lung cancer patients. Patients with A allele in ATP7B rs9535828 presented an increased susceptibility to platinum drugs (OR 1.96, 95 % CI 1.17-3.30, p < 0.01). Patients with G allele in ATP7B rs9535826 had the highest susceptibility to platinum drugs (OR 2.05, 95 % CI 1.19-3.52, p < 0.01). Our findings suggest that ATP7B genetic polymorphisms could affect the therapeutic efficacy of platinum-based chemotherapy, and ATP7B gene might be considered as predictive markers for the efficacy evaluation of platinum-based chemotherapy in Chinese Han lung cancer patients.
Collapse
Affiliation(s)
- Xiang-Ping Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 410008, Changsha, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Galliani A, Losacco M, Lasorsa A, Natile G, Arnesano F. Cisplatin handover between copper transporters: the effect of reducing agents. J Biol Inorg Chem 2014; 19:705-14. [DOI: 10.1007/s00775-014-1138-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 04/09/2014] [Indexed: 12/26/2022]
|
28
|
Miller MA, Askevold B, Yang KS, Kohler RH, Weissleder R. Platinum compounds for high-resolution in vivo cancer imaging. ChemMedChem 2014; 9:1131-5. [PMID: 24504646 DOI: 10.1002/cmdc.201300502] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Indexed: 12/15/2022]
Abstract
Platinum(II) compounds, principally cisplatin and carboplatin, are commonly used front-line cancer therapeutics. Despite their widespread use and continued interest in the development of new derivatives, including nanoformulations with improved properties, it has been difficult to visualize platinum compounds in live subjects, in real time, and with subcellular resolution. Here, we present four novel cisplatin- and carboplatin-derived fluorescent imaging compounds for quantitative intravital cancer imaging. We conjugated 4,4-difluoro-5,7-dimethyl-4-bora-3a,4a-daiza-s-indacene (BODIPY) to Pt(II) complexes to generate derivatives with robust in vivo fluorescence and retained DNA-damaging and cytotoxic properties. We successfully applied these compounds to image pharmacokinetics and tumor uptake in a xenograft cancer mouse model. By using a genetic reporter of single-cell DNA damage for in vivo imaging, Pt drug accumulation and resultant DNA damage could be monitored in individual tumor cells, at subcellular resolution, and in real time in a live animal model of cancer. These derivatives represent promising imaging tools that will be useful in understanding further the distribution and interactions of platinum within tumors.
Collapse
Affiliation(s)
- Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 (USA)
| | | | | | | | | |
Collapse
|
29
|
Przybyłkowski A, Gromadzka G, Wawer A, Grygorowicz T, Cybulska A, Członkowska A. Intestinal expression of metal transporters in Wilson's disease. Biometals 2013; 26:925-34. [PMID: 23963605 PMCID: PMC3825560 DOI: 10.1007/s10534-013-9668-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 08/09/2013] [Indexed: 11/30/2022]
Abstract
In Wilson’s disease (WND), biallelic ATP7B gene mutation is responsible for pathological copper accumulation in the liver, brain and other organs. It has been proposed that copper transporter 1 (CTR1) and the divalent metal transporter 1 (DMT1) translocate copper across the human intestinal epithelium, while Cu-ATPases: ATP7A and ATP7B serve as copper efflux pumps. In this study, we investigated the expression of CTR1, DMT1 and ATP7A in the intestines of both WND patients and healthy controls to examine whether any adaptive mechanisms to systemic copper overload function in the enterocytes. Duodenal biopsy samples were taken from 108 patients with Wilson’s disease and from 90 controls. CTR1, DMT1, ATP7A and ATP7B expression was assessed by polymerase chain reaction and Western blot. Duodenal CTR1 mRNA and protein expression was decreased in WND patients in comparison to control subjects, while ATP7A mRNA and protein production was increased. The variable expression of copper transporters may serve as a defense mechanism against systemic copper overload resulting from functional impairment of ATP7B.
Collapse
Affiliation(s)
- Adam Przybyłkowski
- Department of Clinical and Experimental Pharmacology, Medical University of Warsaw, ul. Krakowskie Przedmieście 26/28, 00-927, Warsaw, Poland,
| | | | | | | | | | | |
Collapse
|
30
|
Palm-Espling ME, Andersson CD, Björn E, Linusson A, Wittung-Stafshede P. Determinants for simultaneous binding of copper and platinum to human chaperone Atox1: hitchhiking not hijacking. PLoS One 2013; 8:e70473. [PMID: 23936210 PMCID: PMC3728025 DOI: 10.1371/journal.pone.0070473] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 06/18/2013] [Indexed: 11/19/2022] Open
Abstract
Cisplatin (CisPt) is an anticancer agent that has been used for decades to treat a variety of cancers. CisPt treatment causes many side effects due to interactions with proteins that detoxify the drug before reaching the DNA. One key player in CisPt resistance is the cellular copper-transport system involving the uptake protein Ctr1, the cytoplasmic chaperone Atox1 and the secretory path ATP7A/B proteins. CisPt has been shown to bind to ATP7B, resulting in vesicle sequestering of the drug. In addition, we and others showed that the apo-form of Atox1 could interact with CisPt in vitro and in vivo. Since the function of Atox1 is to transport copper (Cu) ions, it is important to assess how CisPt binding depends on Cu-loading of Atox1. Surprisingly, we recently found that CisPt interacted with Cu-loaded Atox1 in vitro at a position near the Cu site such that unique spectroscopic features appeared. Here, we identify the binding site for CisPt in the Cu-loaded form of Atox1 using strategic variants and a combination of spectroscopic and chromatographic methods. We directly prove that both metals can bind simultaneously and that the unique spectroscopic signals originate from an Atox1 monomer species. Both Cys in the Cu-site (Cys12, Cys15) are needed to form the di-metal complex, but not Cys41. Removing Met10 in the conserved metal-binding motif makes the loop more floppy and, despite metal binding, there are no metal-metal electronic transitions. In silico geometry minimizations provide an energetically favorable model of a tentative ternary Cu-Pt-Atox1 complex. Finally, we demonstrate that Atox1 can deliver CisPt to the fourth metal binding domain 4 of ATP7B (WD4), indicative of a possible drug detoxification mechanism.
Collapse
Affiliation(s)
| | | | - Erik Björn
- Department of Chemistry, Umeå University, Umeå, Sweden
| | - Anna Linusson
- Department of Chemistry, Umeå University, Umeå, Sweden
| | | |
Collapse
|
31
|
Moreno-Smith M, Halder J, Meltzer PS, Gonda TA, Mangala LS, Rupaimoole R, Lu C, Nagaraja AS, Gharpure KM, Kang Y, Rodriguez-Aguayo C, Vivas-Mejia PE, Zand B, Schmandt R, Wang H, Langley RR, Jennings NB, Ivan C, Coffin JE, Armaiz GN, Bottsford-Miller J, Kim SB, Halleck MS, Hendrix MJ, Bornman W, Bar-Eli M, Lee JS, Siddik ZH, Lopez-Berestein G, Sood AK. ATP11B mediates platinum resistance in ovarian cancer. J Clin Invest 2013; 123:2119-30. [PMID: 23585472 PMCID: PMC3635722 DOI: 10.1172/jci65425] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 02/14/2013] [Indexed: 11/17/2022] Open
Abstract
Platinum compounds display clinical activity against a wide variety of solid tumors; however, resistance to these agents is a major limitation in cancer therapy. Reduced platinum uptake and increased platinum export are examples of resistance mechanisms that limit the extent of DNA damage. Here, we report the discovery and characterization of the role of ATP11B, a P-type ATPase membrane protein, in cisplatin resistance. We found that ATP11B expression was correlated with higher tumor grade in human ovarian cancer samples and with cisplatin resistance in human ovarian cancer cell lines. ATP11B gene silencing restored the sensitivity of ovarian cancer cell lines to cisplatin in vitro. Combined therapy of cisplatin and ATP11B-targeted siRNA significantly decreased cancer growth in mice bearing ovarian tumors derived from cisplatin-sensitive and -resistant cells. In vitro mechanistic studies on cellular platinum content and cisplatin efflux kinetics indicated that ATP11B enhances the export of cisplatin from cells. The colocalization of ATP11B with fluorescent cisplatin and with vesicular trafficking proteins, such as syntaxin-6 (STX6) and vesicular-associated membrane protein 4 (VAMP4), strongly suggests that ATP11B contributes to secretory vesicular transport of cisplatin from Golgi to plasma membrane. In conclusion, inhibition of ATP11B expression could serve as a therapeutic strategy to overcome cisplatin resistance.
Collapse
Affiliation(s)
- Myrthala Moreno-Smith
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - J.B. Halder
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Paul S. Meltzer
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Tamas A. Gonda
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Lingegowda S. Mangala
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Rajesha Rupaimoole
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Chunhua Lu
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Archana S. Nagaraja
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Kshipra M. Gharpure
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yu Kang
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Cristian Rodriguez-Aguayo
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Pablo E. Vivas-Mejia
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Behrouz Zand
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Rosemarie Schmandt
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Hua Wang
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Robert R. Langley
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Nicholas B. Jennings
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Cristina Ivan
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jeremy E. Coffin
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Guillermo N. Armaiz
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Justin Bottsford-Miller
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Sang Bae Kim
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Margaret S. Halleck
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Mary J.C. Hendrix
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - William Bornman
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Menashe Bar-Eli
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ju-Seog Lee
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Zahid H. Siddik
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Gabriel Lopez-Berestein
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Anil K. Sood
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Center for Cancer Research, Genetics Branch, National Cancer Institute, Bethesda, Maryland, USA.
Department of Medicine, Columbia University, New York, New York, USA.
Center for RNA Interference and Non-coding RNA and
Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Cancer Center, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico.
Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.
Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, USA.
Children’s Memorial Research Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
32
|
Martens-de Kemp SR, Dalm SU, Wijnolts FMJ, Brink A, Honeywell RJ, Peters GJ, Braakhuis BJM, Brakenhoff RH. DNA-bound platinum is the major determinant of cisplatin sensitivity in head and neck squamous carcinoma cells. PLoS One 2013; 8:e61555. [PMID: 23613873 PMCID: PMC3629194 DOI: 10.1371/journal.pone.0061555] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 03/11/2013] [Indexed: 01/21/2023] Open
Abstract
PURPOSE The combination of systemic cisplatin with local and regional radiotherapy as primary treatment of head and neck squamous cell carcinoma (HNSCC) leads to cure in approximately half of the patients. The addition of cisplatin has significant effects on outcome, but despite extensive research the mechanism underlying cisplatin response is still not well understood. METHODS We examined 19 HNSCC cell lines with variable cisplatin sensitivity. We determined the TP53 mutational status of each cell line and investigated the expression levels of 11 potentially relevant genes by quantitative real-time PCR. In addition, we measured cisplatin accumulation and retention, as well as the level of platinum-DNA adducts. RESULTS We found that the IC50 value was significantly correlated with the platinum-DNA adduct levels that accumulated during four hours of cisplatin incubation (p = 0.002). We could not find a significant correlation between cisplatin sensitivity and any of the other parameters tested, including the expression levels of established cisplatin influx and efflux transporters. Furthermore, adduct accumulation did not correlate with mRNA expression of the investigated influx pumps (CTR1 and OCT3) nor with that of the examined DNA repair genes (ATR, ATM, BRCA1, BRCA2 and ERCC1). CONCLUSION Our findings suggest that the cisplatin-DNA adduct level is the most important determinant of cisplatin sensitivity in HNSCC cells. Imaging with radio-labeled cisplatin might have major associations with outcome.
Collapse
MESH Headings
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/pathology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cisplatin/metabolism
- Cisplatin/pharmacology
- Cisplatin/therapeutic use
- DNA Adducts/metabolism
- DNA Adducts/pharmacology
- DNA, Neoplasm/metabolism
- Drug Screening Assays, Antitumor
- Gene Expression Regulation, Neoplastic/drug effects
- Genes, Neoplasm/genetics
- Head and Neck Neoplasms/drug therapy
- Head and Neck Neoplasms/genetics
- Head and Neck Neoplasms/pathology
- Humans
- Inhibitory Concentration 50
- Mutation/genetics
- Platinum/pharmacology
- Platinum/therapeutic use
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Squamous Cell Carcinoma of Head and Neck
- Statistics, Nonparametric
- Tumor Suppressor Protein p53/genetics
Collapse
Affiliation(s)
- Sanne R Martens-de Kemp
- Department of Otolaryngology/Head-Neck Surgery, VU University Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Ciarimboli G. Membrane transporters as mediators of Cisplatin effects and side effects. SCIENTIFICA 2012; 2012:473829. [PMID: 24278698 PMCID: PMC3820462 DOI: 10.6064/2012/473829] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 10/23/2012] [Indexed: 06/02/2023]
Abstract
Transporters are important mediators of specific cellular uptake and thus, not only for effects, but also for side effects, metabolism, and excretion of many drugs such as cisplatin. Cisplatin is a potent cytostatic drug, whose use is limited by its severe acute and chronic nephro-, oto-, and peripheral neurotoxicity. For this reason, other platinum derivatives, such as carboplatin and oxaliplatin, with less toxicity but still with antitumoral action have been developed. Several transporters, which are expressed on the cell membranes, have been associated with cisplatin transport across the plasma membrane and across the cell: the copper transporter 1 (Ctr1), the copper transporter 2 (Ctr2), the P-type copper-transporting ATPases ATP7A and ATP7B, the organic cation transporter 2 (OCT2), and the multidrug extrusion transporter 1 (MATE1). Some of these transporters are also able to accept other platinum derivatives as substrate. Since membrane transporters display a specific tissue distribution, they can be important molecules that mediate the entry of platinum derivatives in target and also nontarget cells possibly mediating specific effects and side effects of the chemotherapeutic drug. This paper summarizes the literature on toxicities of cisplatin compared to that of carboplatin and oxaliplatin and the interaction of these platinum derivatives with membrane transporters.
Collapse
Affiliation(s)
- Giuliano Ciarimboli
- Experimentelle Nephrologie, Medizinische Klinik D, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude A14, 48149 Münster, Germany
| |
Collapse
|
34
|
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]
|
35
|
Kalayda GV, Wagner CH, Jaehde U. Relevance of copper transporter 1 for cisplatin resistance in human ovarian carcinoma cells. J Inorg Biochem 2012; 116:1-10. [PMID: 23010323 DOI: 10.1016/j.jinorgbio.2012.07.010] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 07/09/2012] [Accepted: 07/09/2012] [Indexed: 11/27/2022]
Abstract
Defects in intracellular accumulation of the antitumour drug cisplatin are a commonly observed feature in the cells selected for cisplatin resistance. Copper transporter 1 (CTR1) has been suggested to play an important role in drug uptake and resistance. Here, we describe a detailed investigation of the involvement of CTR1 in cisplatin uptake and its relevance for cisplatin resistance using a well characterised sensitive/cisplatin-resistant cell line pair: A2780 human ovarian carcinoma cell line and its cisplatin-resistant variant A2780cis. A2780cis cells showed decreased cisplatin accumulation and lower CTR1 expression compared to A2780 cells. Co-incubation with copper sulphate affected neither cisplatin accumulation (determined by flameless atomic absorption spectrometry) nor its cytotoxicity (determined using an MTT-assay, MTT=3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide). In both cell lines, CTR1 was localised near the nucleus as found using confocal fluorescence microscopy. The steady-state localisation of the protein in perinuclear region appears to involve its continuous endocytosis from cell surface. In contrast to copper, cisplatin exposure had no influence on the sub cellular localisation of CTR1. Co-localisation between CTR1 and a fluorescent cisplatin analogue labelled with carboxyfluorescein-diacetate could be observed in vesicular structures when continuous retrieval of the protein from cell membrane was inhibited. Our results strongly suggest that CTR1 mediates cisplatin uptake in the cell lines studied. Upon its transport across the plasma membrane by CTR1 the platinum drug is likely to be internalised along with the protein. Our findings imply that reduced CTR1 expression accounts for decreased cisplatin accumulation and represents one of the determinants of cisplatin resistance in A2780cis cell line.
Collapse
Affiliation(s)
- Ganna V Kalayda
- Pharma Center Bonn, Institute of Pharmacy, Department of Clinical Pharmacy, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany.
| | | | | |
Collapse
|
36
|
Safaei R, Adams PL, Maktabi MH, Mathews RA, Howell SB. The CXXC motifs in the metal binding domains are required for ATP7B to mediate resistance to cisplatin. J Inorg Biochem 2012; 110:8-17. [PMID: 22459168 DOI: 10.1016/j.jinorgbio.2012.02.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Revised: 02/01/2012] [Accepted: 02/20/2012] [Indexed: 01/01/2023]
Abstract
The copper (Cu) exporter ATP7B mediates resistance to cisplatin (cDDP) but details of the mechanism are unknown. We explored the role of the CXXC motifs in the metal binding domains (MBDs) of ATP7B by investigating binding of cDDP to the sixth metal binding domain (MBD6) or a variant in which the CXXC motif was converted to SXXS. Platinum measurement showed that cDDP bound to wild type MBD6 but not to the SXXS variant. Wild type ATP7B rendered ovarian 2008 cells resistant to cDDP. In 2008 and in HEK293T cells, wild type ATP7B trafficked from TGN to peripheral locations in response to Cu or cDDP. A variant in which the CXXC motifs in all 6 MBDs were converted to SXXS localized correctly to the TGN but failed to traffic when exposed to either Cu or cDDP. Deletion of either the first 5 MBDs or all 6 MBDs resulted in failure to localize to the TGN. Neither the SXXS variant nor the deletion variant was able to mediate resistance to cDDP. We conclude that cDDP binds to the CXXC motifs of ATP7B and that this interaction is essential to the trafficking of ATP7B and to its ability to mediate resistance to cDDP.
Collapse
Affiliation(s)
- Roohangiz Safaei
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093-0819, USA.
| | | | | | | | | |
Collapse
|
37
|
Affiliation(s)
- Francisco Zaera
- Department of Chemistry, University of California, Riverside, California 92521, United States
| |
Collapse
|
38
|
Jing-chun H, Da-lian D, Dong-zhen Y, Hai-yan J, Shan-kai Y, Salvi R. Modulation of copper transporters in protection against cisplatin-induced cochlear hair cell damage. J Otol 2011. [DOI: 10.1016/s1672-2930(11)50022-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
39
|
Abstract
The modern era of evidence-based ototoxicity emerged in the 1940s following the discovery of aminoglycosides and their ototoxic side effects. New classes of ototoxins have been identified in subsequent decades, notably loop diuretics, antineoplastic drugs, and metal chelators. Ototoxic drugs are frequently nephrotoxic, as both organs regulate fluid and ion composition. The mechanisms of ototoxicity are as diverse as the pharmacological properties of each ototoxin, though the generation of toxic levels of reactive oxygen species appears to be a common denominator. As mechanisms of cytotoxicity for each ototoxin continue to be elucidated, a new frontier in ototoxicity is emerging: How do ototoxins cross the blood-labyrinth barrier that tightly regulates the composition of the inner ear fluids? Increased knowledge of the mechanisms by which systemic ototoxins are trafficked across the blood-labyrinth barrier into the inner ear is critical to developing new pharmacotherapeutic agents that target the blood-labyrinth barrier to prevent trafficking of ototoxic drugs and their cytotoxic sequelae.
Collapse
Affiliation(s)
- Peter S Steyger
- Oregon Hearing Research Center, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
40
|
Human copper transporters: mechanism, role in human diseases and therapeutic potential. Future Med Chem 2011; 1:1125-42. [PMID: 20454597 DOI: 10.4155/fmc.09.84] [Citation(s) in RCA: 190] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Normal copper homeostasis is essential for human growth and development. Copper deficiency, caused by genetic mutations, inadequate diet or surgical interventions, may lead to cardiac hypertrophy, poor neuronal myelination, blood vessel abnormalities and impaired immune response. Copper overload is associated with morphological and metabolic changes in tissues and, if untreated, eventual death. Recent reports also indicate that changes in the expression of copper transporters alter the sensitivity of cancer cells to major chemotherapeutic drugs, such as cisplatin, although the mechanism behind this important phenomenon remains unclear. This review summarizes current information on the molecular characteristics of copper transporters CTR1, CTR2, ATP7A and ATP7B, their roles in mammalian copper homeostasis and the physiological consequences of their inactivation. The mechanisms through which copper transporters may influence cell sensitivity to cisplatin are discussed. Regulation of human copper homeostasis has significant therapeutic potential and requires the detailed understanding of copper transport mechanisms.
Collapse
|
41
|
Shapira A, Livney YD, Broxterman HJ, Assaraf YG. Nanomedicine for targeted cancer therapy: towards the overcoming of drug resistance. Drug Resist Updat 2011; 14:150-63. [PMID: 21330184 DOI: 10.1016/j.drup.2011.01.003] [Citation(s) in RCA: 319] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2010] [Revised: 01/12/2011] [Accepted: 01/14/2011] [Indexed: 12/11/2022]
Abstract
Anticancer drug resistance almost invariably emerges and poses major obstacles towards curative therapy of various human malignancies. In the current review we will distinguish between mechanisms of chemoresistance that are predominantly mediated by ATP-driven multidrug resistance (MDR) efflux transporters, typically of the ATP-binding cassette (ABC) superfamily, and those that are independent of such drug efflux pumps. In recent years, multiple nanoparticle (NP)-based therapeutic systems have been developed that were rationally designed to overcome drug resistance by neutralizing, evading or exploiting various drug efflux pumps and other resistance mechanisms. NPs are being exploited for selective drug delivery to tumor cells, to cancer stem/tumor initiating cells and/or to the supportive cancer cell microenvironment, i.e. stroma or tumor vasculature. Some of these NPs are currently undergoing preclinical in vivo studies as well as advanced stages of clinical evaluation with promising results. Nanovehicles harboring a payload of therapeutic drug combinations for the selective targeting and elimination of tumor cells as well as the simultaneous overcoming of mechanisms of drug resistance are a subject of intense research efforts, some of which are expected to enter clinical trials in the near future. In the present review we highlight novel approaches to selectively target cancer cells and overcome drug resistance phenomena, through the use of various nanometric drug delivery systems. In the near future, it is anticipated that innovative theragnostic nanovehicles will be developed which will harbor four major components: (1) a selective targeting moiety, (2) a diagnostic imaging aid for the localization of the malignant tumor and its micro- or macrometastases, (3) a cytotoxic, small molecule drug(s) or novel therapeutic biological(s), and (4) a chemosensitizing agent aimed at neutralizing a resistance mechanism, or exploiting a molecular "Achilles hill" of drug resistant cells. We propose to name these envisioned four element-containing nanovehicle platform, "quadrugnostic" nanomedicine. This targeted strategy holds promise in paving the way for the introduction of highly effective nanoscopic vehicles for cancer therapeutics while overcoming drug resistance.
Collapse
Affiliation(s)
- Alina Shapira
- Russell Berrie Nanotechnology Institute, Technion, Haifa, Israel
| | | | | | | |
Collapse
|
42
|
Chapman EG, Hostetter AA, Osborn MF, Miller AL, DeRose VJ. Binding of kinetically inert metal ions to RNA: the case of platinum(II). Met Ions Life Sci 2011; 9:347-77. [PMID: 22010278 PMCID: PMC4080900 DOI: 10.1039/9781849732512-00347] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
In this chapter several aspects of Pt(II) are highlighted that focus on the properties of Pt(II)-RNA adducts and the possibility that they influence RNA-based processes in cells. Cellular distribution of Pt(II) complexes results in significant platination of RNA, and localization studies find Pt(II) in the nucleus, nucleolus, and a distribution of other sites in cells. Treatment with Pt(II) compounds disrupts RNA-based processes including enzymatic processing, splicing, and translation, and this disruption may be indicative of structural changes to RNA or RNA-protein complexes. Several RNA-Pt(II) adducts have been characterized in vitro by biochemical and other methods. Evidence for Pt(II) binding in non-helical regions and for Pt(II) cross-linking of internal loops has been found. Although platinated sites have been identified, there currently exists very little in the way of detailed structural characterization of RNA-Pt(II) adducts. Some insight into the details of Pt(II) coordination to RNA, especially RNA helices, can be gained from DNA model systems. Many RNA structures, however, contain complex tertiary folds and common, purine-rich structural elements that present suitable Pt(II) nucleophiles in unique arrangements which may hold the potential for novel types of platinum-RNA adducts. Future research aimed at structural characterization of platinum-RNA adducts may provide further insights into platinum-nucleic acid binding motifs, and perhaps provide a rationale for the observed inhibition by Pt(II) complexes of splicing, translation, and enzymatic processing.
Collapse
Affiliation(s)
- Erich G. Chapman
- Department of Chemistry University of Oregon Eugene OR 97403 USA
| | | | - Maire F. Osborn
- Department of Chemistry University of Oregon Eugene OR 97403 USA
| | - Amanda L. Miller
- Department of Chemistry University of Oregon Eugene OR 97403 USA
| | | |
Collapse
|
43
|
Roles of COMM-domain-containing 1 in stability and recruitment of the copper-transporting ATPase in a mouse hepatoma cell line. Biochem J 2010; 429:53-61. [PMID: 20433422 DOI: 10.1042/bj20100223] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A novel function of COMMD1 {COMM [copper metabolism MURR1 (mouse U2af1-rs1 region 1)]-domain-containing 1}, a protein relevant to canine copper toxicosis, was examined in the mouse hepatoma cell line Hepa 1-6 with multi-disciplinary techniques consisting of molecular and cellular biological techniques, speciation and elemental imaging. To clarify the function of COMMD1, COMMD1-knockdown was accomplished by introducing siRNA (small interfering RNA) into the cells. Although COMMD1-knockdown did not affect copper incorporation, it inhibited copper excretion, resulting in copper accumulation, which predominantly existed in the form bound to MT (metallothionein). It is known that the liver copper transporter Atp7b (ATP-dependent copper transporter 7beta), localizes on the trans-Golgi network membrane under basal copper conditions and translocates to cytoplasmic vesicles to excrete copper when its concentration exceeds a certain threshold, with the vesicles dispersing in the periphery of the cell. COMMD1-knockdown reduced the expression of Atp7b, and abolished the relocation of Atp7b back from the periphery to the trans-Golgi network membrane when the copper concentration was reduced by treatment with a Cu(I) chelator. The same phenomena were observed during COMMD1-knockdown when another Atp7b substrate, cis-diamminedichloroplatinum, and its sequestrator, glutathione ethylester, were applied. These results suggest that COMMD1 maintains the amount of Atp7b and facilitates recruitment of Atp7b from cytoplasmic vesicles to the trans-Golgi network membrane, i.e. COMMD1 is required to shuttle Atp7b when the intracellular copper level returns below the threshold.
Collapse
|
44
|
Inoue Y, Matsumoto H, Yamada S, Kawai K, Suemizu H, Gika M, Takanami I, Nakamura M, Iwazaki M. ATP7B expression is associated with in vitro sensitivity to cisplatin in non-small cell lung cancer. Oncol Lett 2010; 1:279-282. [PMID: 22966294 DOI: 10.3892/ol_00000049] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Accepted: 08/31/2009] [Indexed: 11/05/2022] Open
Abstract
Copper-transporting P-type adenosine triphosphatase (ATP7B) is reportedly associated with platinum drug resistance in various solid tumors. However, the impact of ATP7B on platinum drug resistance in non-small cell lung cancer (NSCLC) remains unclear. We investigated in vitro cisplatin (CDDP) sensitivity using the collagen gel-droplet embedded culture drug sensitivity test. The ATP7B mRNA expression level in each specimen was also examined using real-time polymerase chain reaction. The relationship between ATP7B expression and in vitro CDDP sensitivity was then evaluated. The ATP7B mRNA expression levels in CDDP-resistant tumors were significantly higher than those in the CDDP-sensitive group (p=0.015; Mann-Whitney U test). Our results suggested that ATP7B expression is a promising chemoresistance marker for cisplatin.
Collapse
Affiliation(s)
- Yoshimasa Inoue
- Department of General Thoracic Surgery, Tokai University Hachioji Hospital, Tokyo 192-0032
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Howell SB, Safaei R, Larson CA, Sailor MJ. Copper transporters and the cellular pharmacology of the platinum-containing cancer drugs. Mol Pharmacol 2010; 77:887-94. [PMID: 20159940 DOI: 10.1124/mol.109.063172] [Citation(s) in RCA: 260] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Multiple lines of evidence indicate that the platinum-containing cancer drugs enter cells, are distributed to various subcellular compartments, and are exported from cells via transporters that evolved to manage copper homeostasis. The cytotoxicity of the platinum drugs is directly related to how much drug enters the cell, and almost all cells that have acquired resistance to the platinum drugs exhibit reduced drug accumulation. The major copper influx transporter, copper transporter 1 (CTR1), has now been shown to control the tumor cell accumulation and cytotoxic effect of cisplatin, carboplatin, and oxaliplatin. There is a good correlation between change in CTR1 expression and acquired cisplatin resistance among ovarian cancer cell lines, and genetic knockout of CTR1 renders cells resistant to cisplatin in vivo. The expression of CTR1 is regulated at the transcriptional level by copper via Sp1 and at the post-translational level by the proteosome. Copper and cisplatin both trigger the down-regulation of CTR1 via a process that involves ubiquitination and proteosomal degradation and requires the copper chaperone antioxidant protein 1 (ATOX1). The cisplatin-induced degradation of CTR1 can be blocked with the proteosome inhibitor bortezomib, and this increases the cellular uptake and the cytotoxicity of cisplatin in a synergistic manner. Copper and platinum(II) have similar sulfur binding characteristics, and the presence of stacked rings of methionines and cysteines in the CTR1 trimer suggest a mechanism by which CTR1 selectively transports copper and the platinum-containing drugs via sequential transchelation reactions similar to the manner in which copper is passed from ATOX1 to the copper efflux transporters.
Collapse
Affiliation(s)
- Stephen B Howell
- Department of Medicine, Moores UCSD Cancer Center, 3855 Health Sciences Drive, La Jolla, CA 92093, USA.
| | | | | | | |
Collapse
|
46
|
Shahzad MMK, Lopez-Berestein G, Sood AK. Novel strategies for reversing platinum resistance. Drug Resist Updat 2009; 12:148-52. [PMID: 19805003 DOI: 10.1016/j.drup.2009.09.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Revised: 09/08/2009] [Accepted: 09/09/2009] [Indexed: 12/19/2022]
Abstract
Platinum-based drugs continue to be the mainstay of therapy for ovarian cancer. Along with adverse effects, chemoresistance (intrinsic or acquired) has become a major limitation in the management of recurrent disease. Even though much is known about the effects of platinum drugs on cancer cells, the mechanisms underlying resistance are poorly understood. In this review, we summarize the current data on chemoresistance and discuss novel strategies to reverse resistance to platinum-based drugs. The most important targets highlighted here include Aurora kinases, PARP, ATP7B, and ERCC1. Furthermore, we discuss the implications of these novel approaches for ovarian cancer treatment.
Collapse
Affiliation(s)
- Mian M K Shahzad
- Department of Gynecologic Oncology, U.T.M.D. Anderson Cancer Center, 1155 Herman Pressler, Unit 1362, Houston, TX 77030, USA
| | | | | |
Collapse
|
47
|
Jandial DD, Messer K, Farshchi-Heydari S, Pu M, Howell SB. Tumor platinum concentration following intraperitoneal administration of cisplatin versus carboplatin in an ovarian cancer model. Gynecol Oncol 2009; 115:362-6. [PMID: 19775736 DOI: 10.1016/j.ygyno.2009.08.028] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Revised: 08/28/2009] [Accepted: 08/31/2009] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Current intraperitoneal (IP) regimens for the treatment of ovarian cancer rely on cisplatin (DDP) whereas intravenous regimens rely on carboplatin (CBDCA). A major question in the field is whether CBDCA can replace DDP for IP treatment. We compared the uptake of IP administered DDP and CBDCA into human ovarian carcinoma nodules of various sizes growing on the peritoneal surface of nu/nu mice. METHODS Human 2008 cells expressing GFP were inoculated IP in nu/nu mice. When small tumor nodules became visible by external imaging, a maximum tolerated dose of DDP, or either an equimolar or equitoxic dose of CBDCA, was injected IP. Platinum (Pt) concentration in tumor nodules was measured by inductively coupled plasma mass spectrometry. RESULTS A total of 749 tumors harvested from 33 mice were analyzed for Pt concentration. DDP produced a 3.4-fold higher level of Pt in tumor nodules when compared to an equimolar dose of CBDCA (p=0.02). However, when DDP and CBDCA were injected at doses that were equitoxic to the mice, tumor Pt levels were equivalent (p=0.63). Although Pt concentrations of equal-sized nodules were highly variable, tumor Pt content (ng Pt/mg tumor) decreased with increasing nodule size following IP DDP, an effect not seen with IP administration of equitoxic doses of CBDCA (p<0.001). CONCLUSIONS These results suggest that IP CBDCA has comparable or better drug penetration when compared to DDP given at equitoxic doses, and thus provide support for replacing DDP with CBDCA in the IP treatment of patients with ovarian cancer.
Collapse
Affiliation(s)
- Danielle D Jandial
- Moores Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0819, USA.
| | | | | | | | | |
Collapse
|
48
|
Affiliation(s)
- Alice V. Klein
- School of Chemistry, The University of Sydney, NSW 2006, Australia
| | | |
Collapse
|
49
|
Mangala LS, Zuzel V, Schmandt R, Leshane ES, Halder JB, Armaiz-Pena GN, Spannuth WA, Tanaka T, Shahzad MMK, Lin YG, Nick AM, Danes CG, Lee JW, Jennings NB, Vivas-Mejia PE, Wolf JK, Coleman RL, Siddik ZH, Lopez-Berestein G, Lutsenko S, Sood AK. Therapeutic Targeting of ATP7B in Ovarian Carcinoma. Clin Cancer Res 2009; 15:3770-80. [PMID: 19470734 DOI: 10.1158/1078-0432.ccr-08-2306] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE Resistance to platinum chemotherapy remains a significant problem in ovarian carcinoma. Here, we examined the biological mechanisms and therapeutic potential of targeting a critical platinum resistance gene, ATP7B, using both in vitro and in vivo models. EXPERIMENTAL DESIGN Expression of ATP7A and ATP7B was examined in ovarian cancer cell lines by real-time reverse transcription-PCR and Western blot analysis. ATP7A and ATP7B gene silencing was achieved with targeted small interfering RNA (siRNA) and its effects on cell viability and DNA adduct formation were examined. For in vivo therapy experiments, siRNA was incorporated into the neutral nanoliposome 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine (DOPC). RESULTS ATP7A and ATP7B genes were expressed at higher levels in platinum-resistant cells compared with sensitive cells; however, only differences in ATP7B reached statistical significance. ATP7A gene silencing had no significant effect on the sensitivity of resistant cells to cisplatin, but ATP7B silencing resulted in 2.5-fold reduction of cisplatin IC(50) levels and increased DNA adduct formation in cisplatin-resistant cells (A2780-CP20 and RMG2). Cisplatin was found to bind to the NH(2)-terminal copper-binding domain of ATP7B, which might be a contributing factor to cisplatin resistance. For in vivo therapy experiments, ATP7B siRNA was incorporated into DOPC and was highly effective in reducing tumor growth in combination with cisplatin (70-88% reduction in both models compared with controls). This reduction in tumor growth was accompanied by reduced proliferation, increased tumor cell apoptosis, and reduced angiogenesis. CONCLUSION These data provide a new understanding of cisplatin resistance in cancer cells and may have implications for therapeutic reversal of drug resistance.
Collapse
Affiliation(s)
- Lingegowda S Mangala
- Departments of Gynecologic Oncology, Experimental Therapeutics, and Cancer Biology, The University of Texas M. D. Anderson Cancer Center, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Martinez-Balibrea E, Martínez-Cardús A, Musulén E, Ginés A, Manzano JL, Aranda E, Plasencia C, Neamati N, Abad A. Increased levels of copper efflux transporter ATP7B are associated with poor outcome in colorectal cancer patients receiving oxaliplatin-based chemotherapy. Int J Cancer 2009; 124:2905-10. [PMID: 19296535 DOI: 10.1002/ijc.24273] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recently, the copper efflux transporters ATP7B and ATP7A have been implicated in the transport of and resistance to platinum drugs in breast and ovarian cancers. Because of the extensive use of oxaliplatin in colorectal cancer (CRC), we examined the expression of both transporters in tumors from CRC patients treated with oxaliplatin/5FU and sought to determine whether their expression can predict clinical outcome in these patients. ATP7B and ATP7A levels were determined by quantitative real-time PCR in 50 primary tumors of previously untreated patients with advanced colorectal adenocarcinoma who were subsequently treated with oxaliplatin/5FU. Additionally, ATP7B protein expression was assessed by immunohistochemical staining using a tissue microarray. Patients with the lowest mRNA expression levels of ATP7B had a significantly longer time to progression (TTP) (p = 0.0009) than patients with the highest levels (12.14 months vs. 6.43 months) who also had an increased risk of progression (HR = 3.56; 95% CI, 1.6-7.9; p = 0.002). Furthermore, patients with low levels of both protein and mRNA of ATP7B derived the maximum benefit from oxaliplatin/5FU with the longest TTP as compared with patients with high levels of ATP7B protein and mRNA (14.64 months vs. 4.63 months, respectively, p = 0.01) and showed a nonsignificant trend toward a lower response rate (37.5% and 75%, respectively). In conclusion, ATP7B mRNA and protein expression in colorectal tumors is associated with clinical outcome to oxaliplatin/5FU. Prospective studies are required to evaluate the role of this marker in tailoring chemotherapy.
Collapse
Affiliation(s)
- Eva Martinez-Balibrea
- Medical Oncology Service, Hospital Universitari Germans Trias i Pujol, Institut Català d'Oncologia, Badalona, Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|