151
|
Oh GS, Kim HJ, Shen A, Lee SB, Khadka D, Pandit A, So HS. Cisplatin-induced Kidney Dysfunction and Perspectives on Improving Treatment Strategies. Electrolyte Blood Press 2014; 12:55-65. [PMID: 25606044 PMCID: PMC4297704 DOI: 10.5049/ebp.2014.12.2.55] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 12/05/2014] [Indexed: 01/14/2023] Open
Abstract
Cisplatin is one of the most widely used and highly effective drug for the treatment of various solid tumors; however, it has dose-dependent side effects on the kidney, cochlear, and nerves. Nephrotoxicity is the most well-known and clinically important toxicity. Numerous studies have demonstrated that several mechanisms, including oxidative stress, DNA damage, and inflammatory responses, are closely associated with cisplatin-induced nephrotoxicity. Even though the establishment of cisplatin-induced nephrotoxicity can be alleviated by diuretics and pre-hydration of patients, the prevalence of cisplatin nephrotoxicity is still high, occurring in approximately one-third of patients who have undergone cisplatin therapy. Therefore it is imperative to develop treatments that will ameliorate cisplatin-nephrotoxicity. In this review, we discuss the mechanisms of cisplatin-induced renal toxicity and the new strategies for protecting the kidneys from the toxic effects without lowering the tumoricidal activity.
Collapse
Affiliation(s)
- Gi-Su Oh
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, Korea
| | - Hyung-Jin Kim
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, Korea
| | - AiHua Shen
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, Korea
| | - Su Bin Lee
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, Korea
| | - Dipendra Khadka
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, Korea
| | - Arpana Pandit
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, Korea
| | - Hong-Seob So
- Center for Metabolic Function Regulation, Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk, Korea
| |
Collapse
|
152
|
Abstract
Pharmaceutical agents provide diagnostic and therapeutic utility that are central to patient care. However, all agents also carry adverse drug effect profiles. While most of these are clinically insignificant, some drugs may cause unacceptable toxicity that impacts negatively on patient morbidity and mortality. Recognizing adverse effects is important for administering appropriate drug doses, instituting preventive strategies, and withdrawing the offending agent due to toxicity. In the present article, we will review those drugs that are associated with impaired renal function. By focusing on pharmaceutical agents that are currently in clinical practice, we will provide an overview of nephrotoxic drugs that a treating physician is most likely to encounter. In doing so, we will summarize risk factors for nephrotoxicity, describe clinical manifestations, and address preventive and treatment strategies.
Collapse
Affiliation(s)
| | - Anushree C Shirali
- Section of Nephrology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
153
|
Fatima S, Al-Mohaimeed N, Arjumand S, Banu N, Al-Jameil N, Al-Shaikh Y. Effect of pre- and post-combined multidoses of epigallocatechin gallate and coenzyme Q10 on cisplatin-induced oxidative stress in rat kidney. J Biochem Mol Toxicol 2014; 29:91-7. [PMID: 25382014 DOI: 10.1002/jbt.21671] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 08/27/2014] [Accepted: 09/08/2014] [Indexed: 12/29/2022]
Abstract
The nephroprotective effect of coenzyme Q10 and epigallocatechin gallate was investigated in rats with acute renal injury induced by a single nephrotoxic dose of cisplatin. Two days prior to cisplatin administration, epigallocatechin gallate and coenzyme Q10 alone and in four different combinations were given for 6 days. The treatment with antioxidants significantly protected the cisplatin-induced increase in the levels of blood urea nitrogen and serum creatinine. Both the antioxidants alone or in different combinations significantly compensated the increased malondialdehyde and reduced glutathione levels. Moreover, the decrease in the activities of superoxide dismutase, catalase, and glutathione peroxidase and the concentration of selenium, zinc, and copper ions were significantly attenuated in renal tissue. In conclusion, epigallocatechin gallate and coenzyme Q10 are equally effective against cisplatin-induced nephrotoxicity, whereas the intervention by combining these two antioxidants was found to be highly effective at low doses in attenuating oxidative stress in rat kidney.
Collapse
Affiliation(s)
- Sabiha Fatima
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi, Arabia.
| | | | | | | | | | | |
Collapse
|
154
|
Wilmes A, Bielow C, Ranninger C, Bellwon P, Aschauer L, Limonciel A, Chassaigne H, Kristl T, Aiche S, Huber CG, Guillou C, Hewitt P, Leonard MO, Dekant W, Bois F, Jennings P. Mechanism of cisplatin proximal tubule toxicity revealed by integrating transcriptomics, proteomics, metabolomics and biokinetics. Toxicol In Vitro 2014; 30:117-27. [PMID: 25450742 DOI: 10.1016/j.tiv.2014.10.006] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 09/18/2014] [Accepted: 10/02/2014] [Indexed: 11/19/2022]
Abstract
Cisplatin is one of the most widely used chemotherapeutic agents for the treatment of solid tumours. The major dose-limiting factor is nephrotoxicity, in particular in the proximal tubule. Here, we use an integrated omics approach, including transcriptomics, proteomics and metabolomics coupled to biokinetics to identify cell stress response pathways induced by cisplatin. The human renal proximal tubular cell line RPTEC/TERT1 was treated with sub-cytotoxic concentrations of cisplatin (0.5 and 2 μM) in a daily repeat dose treating regime for up to 14 days. Biokinetic analysis showed that cisplatin was taken up from the basolateral compartment, transported to the apical compartment, and accumulated in cells over time. This is in line with basolateral uptake of cisplatin via organic cation transporter 2 and bioactivation via gamma-glutamyl transpeptidase located on the apical side of proximal tubular cells. Cisplatin affected several pathways including, p53 signalling, Nrf2 mediated oxidative stress response, mitochondrial processes, mTOR and AMPK signalling. In addition, we identified novel pathways changed by cisplatin, including eIF2 signalling, actin nucleation via the ARP/WASP complex and regulation of cell polarization. In conclusion, using an integrated omic approach together with biokinetics we have identified both novel and established mechanisms of cisplatin toxicity.
Collapse
Affiliation(s)
- Anja Wilmes
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck 6020, Austria.
| | - Chris Bielow
- Institute of Computer Science, Department of Mathematics and Computer Science, Freie Universität Berlin, Berlin 14195, Germany
| | - Christina Ranninger
- Department of Molecular Biology, Division of Chemistry and Bioanalytics, University of Salzburg, Salzburg 5020, Austria
| | - Patricia Bellwon
- Department of Toxicology, University of Würzburg, Würzburg 97078, Germany
| | - Lydia Aschauer
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Alice Limonciel
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Hubert Chassaigne
- European Commission, Joint Research Centre (JRC), Institute for Health and Consumer Protection, Chemical Assessment and Testing Unit, Via Enrico Fermi 2749, I-21027 Ispra, Italy
| | - Theresa Kristl
- Department of Molecular Biology, Division of Chemistry and Bioanalytics, University of Salzburg, Salzburg 5020, Austria
| | - Stephan Aiche
- Institute of Computer Science, Department of Mathematics and Computer Science, Freie Universität Berlin, Berlin 14195, Germany
| | - Christian G Huber
- Department of Toxicology, University of Würzburg, Würzburg 97078, Germany
| | - Claude Guillou
- European Commission, Joint Research Centre (JRC), Institute for Health and Consumer Protection, Chemical Assessment and Testing Unit, Via Enrico Fermi 2749, I-21027 Ispra, Italy
| | - Philipp Hewitt
- Merck KGaA, Merck Serono, Nonclinical Safety, Darmstadt 64293, Germany
| | - Martin O Leonard
- Centre for Radiation, Chemical and Environmental Hazard, Public Health England, Chilton, Didcot OX11 0RQ, UK
| | - Wolfgang Dekant
- Department of Toxicology, University of Würzburg, Würzburg 97078, Germany
| | - Frederic Bois
- Université de Technologie de Compiègne, Compiègne Cedex 60205, France
| | - Paul Jennings
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck 6020, Austria
| |
Collapse
|
155
|
Spreckelmeyer S, Orvig C, Casini A. Cellular transport mechanisms of cytotoxic metallodrugs: an overview beyond cisplatin. Molecules 2014; 19:15584-610. [PMID: 25268716 PMCID: PMC6271550 DOI: 10.3390/molecules191015584] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 09/17/2014] [Accepted: 09/22/2014] [Indexed: 12/21/2022] Open
Abstract
The field of medicinal inorganic chemistry has grown consistently during the past 50 years; however, metal-containing coordination compounds represent only a minor proportion of drugs currently on the market, indicating that research in this area has not yet been thoroughly realized. Although platinum-based drugs as cancer chemotherapeutic agents have been widely studied, exact knowledge of the mechanisms governing their accumulation in cells is still lacking. However, evidence suggests active uptake and efflux mechanisms are involved; this may be involved also in other experimental metal coordination and organometallic compounds with promising antitumor activities in vitro and in vivo, such as ruthenium and gold compounds. Such knowledge would be necessary to elucidate the balance between activity and toxicity profiles of metal compounds. In this review, we present an overview of the information available on the cellular accumulation of Pt compounds from in vitro, in vivo and clinical studies, as well as a summary of reports on the possible accumulation mechanisms for different families of experimental anticancer metal complexes (e.g., Ru Au and Ir). Finally, we discuss the need for rationalization of the investigational approaches available to study metallodrug cellular transport.
Collapse
Affiliation(s)
- Sarah Spreckelmeyer
- Dept. Pharmacokinetics, Toxicology and Targeting, Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen 9713 AV, The Netherlands
| | - Chris Orvig
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T1Z1, Canada
| | - Angela Casini
- Dept. Pharmacokinetics, Toxicology and Targeting, Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen 9713 AV, The Netherlands.
| |
Collapse
|
156
|
Bajwa A, Rosin DL, Chroscicki P, Lee S, Dondeti K, Ye H, Kinsey GR, Stevens BK, Jobin K, Kenwood BM, Hoehn KL, Lynch KR, Okusa MD. Sphingosine 1-phosphate receptor-1 enhances mitochondrial function and reduces cisplatin-induced tubule injury. J Am Soc Nephrol 2014; 26:908-25. [PMID: 25145931 DOI: 10.1681/asn.2013121351] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Sphingosine 1-phosphate (S1P), the natural sphingolipid ligand for a family of five G protein- coupled receptors (S1P1-S1P5Rs), regulates cell survival and lymphocyte circulation. We have shown that the pan-S1PR agonist, FTY720, attenuates kidney ischemia-reperfusion injury by directly activating S1P1 on proximal tubule (PT) cells, independent of the canonical lymphopenic effects of S1P1 activation on B and T cells. FTY720 also reduces cisplatin-induced AKI. Therefore, in this study, we used conditional PT-S1P1-null (PepckCreS1pr1(fl/fl)) and control (PepckCreS1pr1(w/wt)) mice to determine whether the protective effect of FTY720 in AKI is mediated by PT-S1P1. Cisplatin induced more renal injury in PT-S1P1-null mice than in controls. Although FTY720 produced lymphopenia in both control and PT-S1P1-null mice, it reduced injury only in control mice. Furthermore, the increase in proinflammatory cytokine (CXCL1, MCP-1, TNF-α, and IL-6) expression and infiltration of neutrophils and macrophages induced by cisplatin treatment was attenuated by FTY720 in control mice but not in PT-S1P1-null mice. Similarly, S1P1 deletion rendered cultured PT cells more susceptible to cisplatin-induced injury, whereas S1P1 overexpression protected PT cells from injury and preserved mitochondrial function. We conclude that S1P1 may have an important role in stabilizing mitochondrial function and that FTY720 administration represents a novel strategy in the prevention of cisplatin-induced AKI.
Collapse
Affiliation(s)
- Amandeep Bajwa
- Departments of Medicine and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Diane L Rosin
- Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia Pharmacology, and
| | - Piotr Chroscicki
- Departments of Medicine and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Sangju Lee
- Departments of Medicine and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Krishna Dondeti
- Departments of Medicine and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Hong Ye
- Departments of Medicine and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Gilbert R Kinsey
- Departments of Medicine and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Brian K Stevens
- Departments of Medicine and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Katarzyna Jobin
- Departments of Medicine and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | | | | | | | - Mark D Okusa
- Departments of Medicine and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
157
|
Pathophysiology of cisplatin-induced acute kidney injury. BIOMED RESEARCH INTERNATIONAL 2014; 2014:967826. [PMID: 25165721 PMCID: PMC4140112 DOI: 10.1155/2014/967826] [Citation(s) in RCA: 489] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 07/18/2014] [Accepted: 07/19/2014] [Indexed: 02/06/2023]
Abstract
Cisplatin and other platinum derivatives are the most widely used chemotherapeutic agents to treat solid tumors including ovarian, head and neck, and testicular germ cell tumors. A known complication of cisplatin administration is acute kidney injury (AKI). The nephrotoxic effect of cisplatin is cumulative and dose-dependent and often necessitates dose reduction or withdrawal. Recurrent episodes of AKI may result in chronic kidney disease. The pathophysiology of cisplatin-induced AKI involves proximal tubular injury, oxidative stress, inflammation, and vascular injury in the kidney. There is predominantly acute tubular necrosis and also apoptosis in the proximal tubules. There is activation of multiple proinflammatory cytokines and infiltration of inflammatory cells in the kidney. Inhibition of the proinflammatory cytokines TNF-α or IL-33 or depletion of CD4+ T cells or mast cells protects against cisplatin-induced AKI. Cisplatin also causes endothelial cell injury. An understanding of the pathogenesis of cisplatin-induced AKI is important for the development of adjunctive therapies to prevent AKI, to lessen the need for dose decrease or drug withdrawal, and to lessen patient morbidity and mortality.
Collapse
|
158
|
Solanki MH, Chatterjee PK, Gupta M, Xue X, Plagov A, Metz MH, Mintz R, Singhal PC, Metz CN. Magnesium protects against cisplatin-induced acute kidney injury by regulating platinum accumulation. Am J Physiol Renal Physiol 2014; 307:F369-84. [PMID: 24944268 DOI: 10.1152/ajprenal.00127.2014] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Despite its success as a potent antineoplastic agent, ∼25% of patients receiving cisplatin experience acute kidney injury (AKI) and must discontinue therapy. Impaired magnesium homeostasis has been linked to cisplatin-mediated AKI, and because magnesium deficiency is widespread, we examined the effect of magnesium deficiency and replacement on cisplatin-induced AKI in physiologically relevant older female mice. Magnesium deficiency significantly increased cisplatin-associated weight loss and markers of renal damage (plasma blood urea nitrogen and creatinine), histological changes, inflammation, and renal cell apoptosis and modulated signaling pathways (e.g., ERK1/2, p53, and STAT3). Conversely, these damaging effects were reversed by magnesium. Magnesium deficiency alone significantly induced basal and cisplatin-mediated oxidative stress, whereas magnesium replacement attenuated these effects. Similar results were observed using cisplatin-treated LLC-PK1 renal epithelial cells exposed to various magnesium concentrations. Magnesium deficiency significantly amplified renal platinum accumulation, whereas magnesium replacement blocked the augmented platinum accumulation after magnesium deficiency. Increased renal platinum accumulation during magnesium deficiency was accompanied by reduced renal efflux transporter expression, which was reversed by magnesium replacement. These findings demonstrate the role of magnesium in regulating cisplatin-induced AKI by enhancing oxidative stress and thus promoting cisplatin-mediated damage. Additional in vitro experiments using ovarian, breast, and lung cancer cell lines showed that magnesium supplementation did not compromise cisplatin's chemotherapeutic efficacy. Finally, because no consistently successful therapy to prevent or treat cisplatin-mediated AKI is available for humans, these results support developing more conservative magnesium replacement guidelines for reducing cisplatin-induced AKI in cancer patients at risk for magnesium deficiency.
Collapse
Affiliation(s)
- Malvika H Solanki
- Elmezzi Graduate School of Molecular Medicine, Manhasset, New York; The Center for Immunology and Inflammation, Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, New York; and
| | - Prodyot K Chatterjee
- The Center for Immunology and Inflammation, Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, New York; and
| | - Madhu Gupta
- Elmezzi Graduate School of Molecular Medicine, Manhasset, New York; The Center for Immunology and Inflammation, Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, New York; and
| | - Xiangying Xue
- The Center for Immunology and Inflammation, Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, New York; and
| | - Andrei Plagov
- The Center for Immunology and Inflammation, Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, New York; and
| | - Margot H Metz
- The Center for Immunology and Inflammation, Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, New York; and
| | - Rachel Mintz
- The Center for Immunology and Inflammation, Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, New York; and
| | - Pravin C Singhal
- The Center for Immunology and Inflammation, Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, New York; and Hofstra North Shore-LIJ School of Medicine, Manhasset, New York
| | - Christine N Metz
- Elmezzi Graduate School of Molecular Medicine, Manhasset, New York; The Center for Immunology and Inflammation, Feinstein Institute for Medical Research, North Shore-LIJ Health System, Manhasset, New York; and Hofstra North Shore-LIJ School of Medicine, Manhasset, New York
| |
Collapse
|
159
|
Fung KL, Tepede AK, Pluchino KM, Pouliot LM, Pixley JN, Hall MD, Gottesman MM. Uptake of compounds that selectively kill multidrug-resistant cells: the copper transporter SLC31A1 (CTR1) increases cellular accumulation of the thiosemicarbazone NSC73306. Mol Pharm 2014; 11:2692-702. [PMID: 24800945 PMCID: PMC4137994 DOI: 10.1021/mp500114e] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Acquired drug resistance in cancer continues to be a challenge in cancer therapy, in part due to overexpression of the drug efflux transporter P-glycoprotein (P-gp, MDR1, ABCB1). NSC73306 is a thiosemicarbazone compound that displays greater toxicity against cells expressing functional P-gp than against other cells. Here, we investigate the cellular uptake of NSC73306, and examine its interaction with P-gp and copper transporter 1 (CTR1, SLC31A1). Overexpression of P-gp sensitizes LLC-PK1 cells to NSC73306. Cisplatin (IC50 = 77 μM), cyclosporin A (IC50 = 500 μM), and verapamil (IC50 = 700 μM) inhibited cellular accumulation of [(3)H]NSC73306. Cellular hypertoxicity of NSC73306 to P-gp-expressing cells was inhibited by cisplatin in a dose-dependent manner. Cells transiently expressing the cisplatin uptake transporter CTR1 (SLC31A1) showed increased [(3)H]NSC73306 accumulation. In contrast, CTR1 knockdown decreased [(3)H]NSC73306 accumulation. The presence of NSC73306 reduced CTR1 levels, similar to the negative feedback of CTR1 levels by copper or cisplatin. Surprisingly, although cisplatin is a substrate of CTR1, we found that CTR1 protein was overexpressed in high-level cisplatin-resistant KB-CP20 and BEL7404-CP20 cell lines. We confirmed that the CTR1 protein was functional, as uptake of NSC73306 was increased in KB-CP20 cells compared to their drug-sensitive parental cells, and downregulation of CTR1 in KB-CP20 cells reduced [(3)H]NSC73306 accumulation. These results suggest that NSC73306 is a transport substrate of CTR1.
Collapse
Affiliation(s)
- King Leung Fung
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, Maryland 20892, United States
| | | | | | | | | | | | | |
Collapse
|
160
|
Sprowl JA, Lancaster CS, Pabla N, Hermann E, Kosloske AM, Gibson AA, Li L, Zeeh D, Schlatter E, Janke LJ, Ciarimboli G, Sparreboom A. Cisplatin-induced renal injury is independently mediated by OCT2 and p53. Clin Cancer Res 2014; 20:4026-35. [PMID: 24916697 DOI: 10.1158/1078-0432.ccr-14-0319] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
PURPOSE Tubular secretion of cisplatin is abolished in mice deficient for the organic cation transporters Oct1 and Oct2 (Oct1/2(-/-)mice), and these animals are protected from severe cisplatin-induced kidney damage. Since tubular necrosis is not completely absent in Oct1/2(-/-)mice, we hypothesized that alternate pathways are involved in the observed injury. EXPERIMENTAL DESIGN Studies were done in wild-type, Oct1/2(-/-), or p53-deficient animals, all on an FVB background, receiving cisplatin intraperitoneally at 15 mg/kg. Cisplatin metabolites were analyzed using mass spectrometry, and gene expression was assessed using Affymetrix microarrays and RT-PCR arrays. RESULTS KEGG pathway analyses on kidneys from mice exposed to cisplatin revealed that the most significantly altered genes were associated with the p53 signaling network, including Cdnk1a and Mdm2, in both wild-type (P = 2.40 × 10(-11)) and Oct1/2(-/-)mice (P = 1.92 × 10(-8)). This was confirmed by demonstrating that homozygosity for a p53-null allele partially reduced renal tubular damage, whereas loss of p53 in Oct1/2(-/-)mice (p53(-/-)/Oct1/2(-/-)) completely abolished nephrotoxicity. We found that pifithrin-α, an inhibitor of p53-dependent transcriptional activation, inhibits Oct2 and can mimic the lack of nephrotoxicity observed in p53(-/-)/Oct1/2(-/-)mice. CONCLUSIONS These findings indicate that (i) the p53 pathway plays a crucial role in the kidney in response to cisplatin treatment and (ii) clinical exploration of OCT2 inhibitors may not lead to complete nephroprotection unless the p53 pathway is simultaneously antagonized.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lie Li
- Departments of Pharmaceutical Sciences and
| | - Dorothea Zeeh
- Medizinische Klinik D, Experimentelle Nephrologie, Universitätsklinikum Münster, Münster, Germany
| | - Eberhard Schlatter
- Medizinische Klinik D, Experimentelle Nephrologie, Universitätsklinikum Münster, Münster, Germany
| | - Laura J Janke
- Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee; and
| | - Giuliano Ciarimboli
- Medizinische Klinik D, Experimentelle Nephrologie, Universitätsklinikum Münster, Münster, Germany
| | | |
Collapse
|
161
|
Yang Y, Liu H, Liu F, Dong Z. Mitochondrial dysregulation and protection in cisplatin nephrotoxicity. Arch Toxicol 2014; 88:1249-56. [PMID: 24859930 PMCID: PMC4274771 DOI: 10.1007/s00204-014-1239-1] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 04/03/2014] [Indexed: 10/25/2022]
Abstract
Nephrotoxicity is a major side effect of cisplatin in chemotherapy. Pathologically, cisplatin nephrotoxicity is characterized by cell injury and death in renal tubules. The research in the past decade has gained significant understanding of the cellular and molecular mechanisms of tubular cell death, revealing a central role of mitochondrial dysregulation. The pathological changes in mitochondria in cisplatin nephrotoxicity are mainly triggered by DNA damage response, pro-apoptotic protein attack, disruption of mitochondrial dynamics, and oxidative stress. As such, inhibitory strategies targeting these cytotoxic events may provide renal protection. Nonetheless, ideal approaches for renoprotection should not only protect kidneys but also enhance the anticancer efficacy of cisplatin in chemotherapy.
Collapse
Affiliation(s)
- Yuan Yang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hong Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fuyou Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zheng Dong
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Cellular Biology & Anatomy, Medical college of Georgia at Georgia Regents University and Charlie Norwood VA Medical Center, Augusta, Georgia
| |
Collapse
|
162
|
Li Q, Shu Y. Role of solute carriers in response to anticancer drugs. MOLECULAR AND CELLULAR THERAPIES 2014; 2:15. [PMID: 26056583 PMCID: PMC4452062 DOI: 10.1186/2052-8426-2-15] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 05/14/2014] [Indexed: 12/20/2022]
Abstract
Membrane transporters play critical roles in moving a variety of anticancer drugs across cancer cell membrane, thereby determining chemotherapy efficacy and/or toxicity. The retention of anticancer drugs in cancer cells is the result of net function of efflux and influx transporters. The ATP-binding cassette (ABC) transporters are mainly the efflux transporters expressing at cancer cells, conferring the chemo-resistance in various malignant tumors, which has been well documented over the past decades. However, the function of influx transporters, in particular the solute carriers (SLC) in cancer cells, has only been recently well recognized to have significant impact on cancer therapy. The SLC transporters not only directly bring anticancer agents into cancer cells but also serve as the uptake mediators of essential nutrients for tumor growth and survival. In this review, we concentrate on the interaction of SLC transporters with anticancer drugs and nutrients, and their impact on chemo-sensitivity or -resistance of cancer cells. The differential expression patterns of SLC transporters between normal and tumor tissues may be well utilized to achieve specific delivery of chemotherapeutic agents.
Collapse
Affiliation(s)
- Qing Li
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Baltimore, Maryland USA ; Institute of Clinical Pharmacology, Central South University, Changsha, Hunan 410078 China
| | - Yan Shu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Baltimore, Maryland USA
| |
Collapse
|
163
|
Li Q, Shu Y. Role of solute carriers in response to anticancer drugs. MOLECULAR AND CELLULAR THERAPIES 2014; 2:15. [PMID: 26056583 PMCID: PMC4452062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 05/14/2014] [Indexed: 11/21/2023]
Abstract
Membrane transporters play critical roles in moving a variety of anticancer drugs across cancer cell membrane, thereby determining chemotherapy efficacy and/or toxicity. The retention of anticancer drugs in cancer cells is the result of net function of efflux and influx transporters. The ATP-binding cassette (ABC) transporters are mainly the efflux transporters expressing at cancer cells, conferring the chemo-resistance in various malignant tumors, which has been well documented over the past decades. However, the function of influx transporters, in particular the solute carriers (SLC) in cancer cells, has only been recently well recognized to have significant impact on cancer therapy. The SLC transporters not only directly bring anticancer agents into cancer cells but also serve as the uptake mediators of essential nutrients for tumor growth and survival. In this review, we concentrate on the interaction of SLC transporters with anticancer drugs and nutrients, and their impact on chemo-sensitivity or -resistance of cancer cells. The differential expression patterns of SLC transporters between normal and tumor tissues may be well utilized to achieve specific delivery of chemotherapeutic agents.
Collapse
Affiliation(s)
- Qing Li
- />Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Baltimore, Maryland USA
- />Institute of Clinical Pharmacology, Central South University, Changsha, Hunan 410078 China
| | - Yan Shu
- />Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Baltimore, Maryland USA
| |
Collapse
|
164
|
Jang KJ, Mehr AP, Hamilton GA, McPartlin LA, Chung S, Suh KY, Ingber DE. Human kidney proximal tubule-on-a-chip for drug transport and nephrotoxicity assessment. Integr Biol (Camb) 2014; 5:1119-29. [PMID: 23644926 DOI: 10.1039/c3ib40049b] [Citation(s) in RCA: 539] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Kidney toxicity is one of the most frequent adverse events reported during drug development. The lack of accurate predictive cell culture models and the unreliability of animal studies have created a need for better approaches to recapitulate kidney function in vitro. Here, we describe a microfluidic device lined by living human kidney epithelial cells exposed to fluidic flow that mimics key functions of the human kidney proximal tubule. Primary kidney epithelial cells isolated from human proximal tubule are cultured on the upper surface of an extracellular matrix-coated, porous, polyester membrane that splits the main channel of the device into two adjacent channels, thereby creating an apical 'luminal' channel and a basal 'interstitial' space. Exposure of the epithelial monolayer to an apical fluid shear stress (0.2 dyne cm(-2)) that mimics that found in living kidney tubules results in enhanced epithelial cell polarization and primary cilia formation compared to traditional Transwell culture systems. The cells also exhibited significantly greater albumin transport, glucose reabsorption, and brush border alkaline phosphatase activity. Importantly, cisplatin toxicity and Pgp efflux transporter activity measured on-chip more closely mimic the in vivo responses than results obtained with cells maintained under conventional culture conditions. While past studies have analyzed kidney tubular cells cultured under flow conditions in vitro, this is the first report of a toxicity study using primary human kidney proximal tubular epithelial cells in a microfluidic 'organ-on-a-chip' microdevice. The in vivo-like pathophysiology observed in this system suggests that it might serve as a useful tool for evaluating human-relevant renal toxicity in preclinical safety studies.
Collapse
Affiliation(s)
- Kyung-Jin Jang
- Wyss Institute for Biologically Inspired Engineering at Harvard University, CLSB Bldg. 5th floor, 3 Blackfan Circle, Boston, MA 02115, USA.
| | | | | | | | | | | | | |
Collapse
|
165
|
Quail JF, Tsai CY, Howell SB. Characterization of a monoclonal antibody capable of reliably quantifying expression of human Copper Transporter 1 (hCTR1). J Trace Elem Med Biol 2014; 28:151-159. [PMID: 24447817 PMCID: PMC3989404 DOI: 10.1016/j.jtemb.2013.12.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 11/16/2013] [Accepted: 12/10/2013] [Indexed: 12/20/2022]
Abstract
Human copper transporter 1 (hCTR1) is the high-affinity copper influx transporter in mammalian cells that also mediates the influx of cisplatin. Loss of hCTR1 expression has been implicated in the development of resistance to this cancer chemotherapeutic agent. It has turned out to be very difficult to develop antibodies to hCTR1 and polyclonal antibodies produced by different laboratories have yielded conflicting results. We have characterized a newly-available rabbit monoclonal antibody that reacts with an epitope on the N-terminal end of hCTR1 that now permits rigorous identification and quantification of hCTR1 using Western blot analysis. Postnuclear membrane (PNM) preparations made from cells engineered to express high levels of myc-tagged hCTR1, and cells in which the expression of hCTR1 was knocked down, were used to characterize the antibody. The identity of the bands detected was confirmed by immunoprecipitation, surface biotinylation and deglycosylation of myc-tagged hCTR1. Despite the specificity expected of a monoclonal antibody, the anti-hCTR1 detected a variety of bands in whole cell lysates (WCL), which made it difficult to quantify hCTR1. This problem was overcome by isolating post-nuclear membranes and using these for further analysis. Three bands were identified using this antibody in PNM preparations that migrated at 28, 33-35 and 62-64kDa. Multiple lines of evidence presented here suggest that the 33-35 and 62-64kDa bands are hCTR1 whereas the 28kDa band is a cross-reacting protein of unknown identify. The 33-35kDa band is consistent with the expected MW of the glycosylated hCTR1 monomer. This analysis now permits rigorous identification and quantification of hCTR1.
Collapse
Affiliation(s)
- Jacob F Quail
- Moores UCSD Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093, United States
| | - Cheng-Yu Tsai
- Moores UCSD Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093, United States
| | - Stephen B Howell
- Moores UCSD Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093, United States.
| |
Collapse
|
166
|
Sprowl JA, Sparreboom A. Uptake carriers and oncology drug safety. Drug Metab Dispos 2014; 42:611-22. [PMID: 24378324 PMCID: PMC3965905 DOI: 10.1124/dmd.113.055806] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 12/30/2013] [Indexed: 02/06/2023] Open
Abstract
Members of the solute carrier (SLC) family of transporters are responsible for the cellular influx of a broad range of endogenous compounds and xenobiotics in multiple tissues. Many of these transporters are highly expressed in the gastrointestinal tract, liver, and kidney and are considered to be of particular importance in governing drug absorption, elimination, and cellular sensitivity of specific organs to a wide variety of oncology drugs. Although the majority of studies on the interaction of oncology drugs with SLC have been restricted to the use of exploratory in vitro model systems, emerging evidence suggests that several SLCs, including OCT2 and OATP1B1, contribute to clinically important phenotypes associated with those agents. Recent literature has indicated that modulation of SLC activity may result in drug-drug interactions, and genetic polymorphisms in SLC genes have been described that can affect the handling of substrates. Alteration of SLC function by either of these mechanisms has been demonstrated to contribute to interindividual variability in the pharmacokinetics and toxicity associated with several oncology drugs. In this report, we provide an update on this rapidly emerging field.
Collapse
Affiliation(s)
- Jason A Sprowl
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | |
Collapse
|
167
|
Wen X, Buckley B, McCandlish E, Goedken MJ, Syed S, Pelis R, Manautou JE, Aleksunes LM. Transgenic expression of the human MRP2 transporter reduces cisplatin accumulation and nephrotoxicity in Mrp2-null mice. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1299-308. [PMID: 24641901 DOI: 10.1016/j.ajpath.2014.01.025] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 01/06/2014] [Accepted: 01/13/2014] [Indexed: 12/22/2022]
Abstract
The chemotherapeutic drug cisplatin is actively transported into proximal tubules, leading to acute renal injury. Previous studies suggest that the multidrug resistance-associated protein 2 (Mrp2) transporter may efflux cisplatin conjugates from cells. We sought to determine whether the absence of Mrp2 alters the accumulation and toxicity of platinum in the kidneys of mice and whether transgenic expression of the human MRP2 gene could protect against cisplatin injury in vivo. Plasma, kidneys, and livers from vehicle- and cisplatin-treated wild-type and Mrp2-null mice were collected for quantification of platinum and toxicity. By 24 hours, twofold higher concentrations of platinum were detected in the kidneys and livers of Mrp2-null mice compared with wild types. Enhanced platinum concentrations in Mrp2-null mice were observed in DNA and cytosolic fractions of the kidneys. Four days after cisplatin treatment, more extensive proximal tubule injury was observed in Mrp2-null mice compared with wild-type mice. Kidneys from naive Mrp2-null mice had elevated glutathione S-transferase mRNA levels, which could increase the formation of cisplatin-glutathione conjugates that may be metabolized to toxic thiol intermediates. Transgenic expression of the human MRP2 gene in Mrp2-null mice reduced the accumulation and nephrotoxicity of cisplatin to levels observed in wild-type mice. These data suggest that deficiency in Mrp2 lowers platinum excretion and increases susceptibility to kidney injury, which can be rescued by the human MRP2 ortholog.
Collapse
Affiliation(s)
- Xia Wen
- Department of Pharmacology and Toxicology, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Brian Buckley
- Environmental and Occupational Health Sciences Institute, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Elizabeth McCandlish
- Environmental and Occupational Health Sciences Institute, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Michael J Goedken
- Department of Pharmacology and Toxicology, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Samira Syed
- Department of Pharmacology and Toxicology, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Ryan Pelis
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - José E Manautou
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut
| | - Lauren M Aleksunes
- Department of Pharmacology and Toxicology, Rutgers, The State University of New Jersey, Piscataway, New Jersey; Environmental and Occupational Health Sciences Institute, Rutgers, The State University of New Jersey, Piscataway, New Jersey.
| |
Collapse
|
168
|
ALI BH, RAMKUMAR A, MADANAGOPAL TT, WALY MI, TAGELDIN M, AL-ABRI S, FAHIM M, YASIN J, NEMMAR A. Motor and Behavioral Changes in Mice With Cisplatin-Induced Acute Renal Failure. Physiol Res 2014; 63:35-45. [DOI: 10.33549/physiolres.932585] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
We have previously shown that chronic renal failure in rats induces changes in motor activity and behavior. Similar work on the possible effects of acute renal failure (ARF) induced by cisplatin (CP) is lacking. This is the subject matter of the current work. CP was injected intraperitoneally (i.p.) at a single dose of 20 mg/kg to induce a state of ARF, and three days later, its effects on motor activity, thermal and chemical nociceptive tests, neuromuscular coordination, pentobarbitone-sleeping time, exploration activity and two depression models were investigated. The platinum concentration in the kidneys and brains of mice was also measured. The occurrence of CP-induced ARF was ascertained by standard physiological, biochemical and histo-pathological methods. CP induced all the classical biochemical, physiological and histopathological signs of ARF. The average renal platinum concentration of CP-treated mice was 5.16 ppm, but there was no measurable concentration of platinum in the whole brains. CP treatment significantly decreased motor and exploration activities, and increased immobility time in depression models, suggesting a possible depression-like state. There was also a significant decrease in neuromuscular coordination in CP-treated mice. CP, given at a nephrotoxic dose, induced several adverse motor and behavioral alterations in mice. Further behavioral tests and molecular and biochemical investigations in the brains of mice with CP-induced ARF are warranted.
Collapse
Affiliation(s)
- B. H. ALI
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Sultanate of Oman
| | | | | | | | | | | | | | | | | |
Collapse
|
169
|
Jennings P, Aschauer L, Wilmes A, Gstraunthaler G. Renal Cell Culture. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2014. [DOI: 10.1007/978-1-4939-0521-8_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
170
|
Klotho has dual protective effects on cisplatin-induced acute kidney injury. Kidney Int 2013; 85:855-70. [PMID: 24304882 PMCID: PMC3972320 DOI: 10.1038/ki.2013.489] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 09/11/2013] [Accepted: 09/19/2013] [Indexed: 02/07/2023]
Abstract
Klotho protects the kidney from ischemia-reperfusion injury, but its effect on nephrotoxins is unknown. Here we determined whether Klotho protects the kidney from cisplatin toxicity. Cisplatin increased plasma creatinine and induced tubular injury, which were exaggerated in Klotho haplosufficient (Kl/+) and ameliorated in transgenic Klotho overexpressing (Tg-Kl) mice. Neutrophil gelatinase-associated lipocalin and active caspase-3 protein and the number of apoptotic cells in the kidney were higher in Kl/+ and lower in Tg-Kl compared with wild-type mice. Klotho suppressed basolateral uptake of cisplatin by the normal rat kidney cell line (NRK), an effect similar to cimetidine, a known inhibitor of organic cation transport (OCT). A decrease in cell surface and total OCT2 protein and OCT activity by Klotho was mimicked by β-glucuronidase. The Klotho effect was attenuated by β-glucuronidase inhibition. On the other hand, OCT2 mRNA was reduced by Klotho but not by β-glucuronidase. Moreover, cimetidine inhibited OCT activity but not OCT2 expression. Unlike cimetidine, Klotho reduced cisplatin-induced apoptosis from either the basolateral or apical side and even when added after NRK cells were already loaded with cisplatin. Thus, Klotho protects the kidney against cisplatin nephrotoxicity by reduction of basolateral uptake of cisplatin by OCT2 and a direct anti-apoptotic effect independent of cisplatin uptake. Klotho may be a useful agent to prevent and treat cisplatin-induced nephrotoxicity.
Collapse
|
171
|
Liu Y, Lu X, Nguyen S, Olson JL, Webb HK, Kroetz DL. Epoxyeicosatrienoic acids prevent cisplatin-induced renal apoptosis through a p38 mitogen-activated protein kinase-regulated mitochondrial pathway. Mol Pharmacol 2013; 84:925-34. [PMID: 24092818 PMCID: PMC3834146 DOI: 10.1124/mol.113.088302] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 10/02/2013] [Indexed: 01/17/2023] Open
Abstract
Soluble epoxide hydrolase (sEH) catalyzes the conversion of epoxyeicosatrienoic acids into less active eicosanoids, and inhibitors of sEH have anti-inflammatory and antiapoptotic properties. Based on previous observations that sEH inhibition attenuates cisplatin-induced nephrotoxicity by modulating nuclear factor-κB signaling, we hypothesized that this strategy would also attenuate cisplatin-induced renal apoptosis. Inhibition of sEH with AR9273 [1-adamantan-1-yl-3-(1-methylsulfonyl-piperidin-4-yl-urea)] reduced cisplatin-induced apoptosis through mechanisms involving mitochondrial apoptotic pathways and by reducing reactive oxygen species. Renal mitochondrial Bax induction following cisplatin treatment was significantly decreased by treatment of mice with AR9273 and these antiapoptotic effects involved p38 mitogen-activated protein kinase signaling. Similar mechanisms contributed to reduced apoptosis in Ephx2(-/-) mice treated with cisplatin. Moreover, in pig kidney proximal tubule cells, cisplatin-induced mitochondrial trafficking of Bax and cytochrome c, caspase-3 activation, and oxidative stress are significantly attenuated in the presence of epoxyeicosatrienoic acids (EETs). Collectively, these in vivo and in vitro studies demonstrate a role for EETs in limiting cisplatin-induced renal apoptosis. Inhibition of sEH represents a novel therapeutic strategy for protection against cisplatin-induced renal damage.
Collapse
MESH Headings
- 8,11,14-Eicosatrienoic Acid/analogs & derivatives
- 8,11,14-Eicosatrienoic Acid/metabolism
- 8,11,14-Eicosatrienoic Acid/pharmacology
- Animals
- Antineoplastic Agents/toxicity
- Apoptosis/drug effects
- Caspase 3/metabolism
- Cell Line
- Cisplatin/toxicity
- Enzyme Activation
- Epoxide Hydrolases/antagonists & inhibitors
- Epoxide Hydrolases/metabolism
- Epoxy Compounds/metabolism
- Epoxy Compounds/pharmacology
- Kidney/drug effects
- Kidney/metabolism
- Kidney/pathology
- Kidney Tubules, Proximal/drug effects
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mitochondria/metabolism
- Reactive Oxygen Species/metabolism
- Signal Transduction
- Superoxide Dismutase/metabolism
- Swine
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Yingmei Liu
- Departments of Bioengineering and Therapeutic Sciences (Y.L., X.L., S.N., D.L.K.) and Anatomic Pathology (J.L.O.), University of California San Francisco, San Francisco, California; and Arête Therapeutics, Hayward, California (H.K.W.)
| | | | | | | | | | | |
Collapse
|
172
|
Role of CFTR in oxidative stress and suicidal death of renal cells during cisplatin-induced nephrotoxicity. Cell Death Dis 2013; 4:e817. [PMID: 24091660 PMCID: PMC3824665 DOI: 10.1038/cddis.2013.355] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 07/16/2013] [Accepted: 08/06/2013] [Indexed: 11/08/2022]
Abstract
The clinical use of the antineoplastic drug cisplatin is limited by its deleterious nephrotoxic side effect. Cisplatin-induced nephrotoxicity is associated with an increase in oxidative stress, leading ultimately to renal cell death and irreversible kidney dysfunction. Oxidative stress could be modified by the cystic fibrosis transmembrane conductance regulator protein (CFTR), a Cl− channel not only involved in chloride secretion but as well in glutathione (GSH) transport. Thus, we tested whether the inhibition of CFTR could protect against cisplatin-induced nephrotoxicity. Using a renal proximal cell line, we show that the specific inhibitor of CFTR, CFTRinh-172, prevents cisplatin-induced cell death and apoptosis by modulating the intracellular reactive oxygen species balance and the intracellular GSH concentration. This CFTRinh-172-mediated protective effect occurs without affecting cellular cisplatin uptake or the formation of platinum-DNA adducts. The protective effect of CFTRinh-172 in cisplatin-induced nephrotoxicity was also investigated in a rat model. Five days after receiving a single cisplatin injection (5 mg/kg), rats exhibited renal failure, as evidenced by the alteration of biochemical and functional parameters. Pretreatment of rats with CFTRinh-172 (1 mg/kg) prior to cisplatin injection significantly prevented these deleterious cisplatin-induced nephrotoxic effects. Finally, we demonstrate that CFTRinh-172 does not impair cisplatin-induced cell death in the cisplatin-sensitive A549 cancer cell line. In conclusion, the use of a specific inhibitor of CFTR may represent a novel therapeutic approach in the prevention of nephrotoxic side effects during cisplatin treatment without affecting its antitumor efficacy.
Collapse
|
173
|
Mazaheri S, Nematbakhsh M, Bahadorani M, Pezeshki Z, Talebi A, Ghannadi AR, Ashrafi F. Effects of Fennel Essential Oil on Cisplatin-induced Nephrotoxicity in Ovariectomized Rats. Toxicol Int 2013; 20:138-45. [PMID: 24082507 PMCID: PMC3783680 DOI: 10.4103/0971-6580.117256] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background: Cisplatin (cis-diamminedichloroplatinum II (CDDP)) is an effective drug in cancer therapy to treat solid tumors. However, the drug is accompanied by nephrotoxicity. Previous reports indicated that estrogen has no protective role against CDDP-induced nephrotoxicity, but the role of phytoestrogen as an estrogenic agent in plants is not determined yet. The major composition of fennel essential oil (FEO) is trans-anethole that has estrogenic activity; so, we used FEO as a phytoestrogen source against CDDP-induced nephrotoxicity. Materials and Methods: Fifty-four ovariectomized Wistar rats were divided into seven groups. Groups 1-3 received different doses of FEO (250, 500, and 1000 mg/kg/day, respectively) for 10 days. Group 4 received saline for 10 days plus single dose of CDDP (7 mg/kg, intraperitoneally (ip)) at day 3. Groups 5-7 received FEO similar to groups 1-3, respectively; plus a single dose of CDDP (7 mg/kg, ip) on day 3. On day 10, the animals were sacrificed for histopathological studies. Results: The serum levels of blood urea nitrogen (BUN) and creatinine (Cr), kidney tissue damage score (KTDS), and kidney weight (KW) and body weight changes in CDDP-treated groups increased significantly (P < 0.05). FEO did not reduce the levels of BUN and Cr, KTDS, and KW and body weight changes. Also, the serum and tissue levels of nitrite were not altered significantly by FEO. Conclusion: FEO, as a source of phytoestrogen, did not induce kidney damage. In addition, FEO similar to estrogen was not a nephroprotectant agent against CDDP-induced nephrotoxicity.
Collapse
Affiliation(s)
- Safoora Mazaheri
- Water and Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, Iran ; Deparment of Biology, Falavarjan Branch, Islamic Azad University, Isfahan, Iran
| | | | | | | | | | | | | |
Collapse
|
174
|
Chen R, Li J, Hu WW, Wang ML, Zou SL, Miao LY. Circadian variability of pharmacokinetics of cisplatin in patients with non-small-cell lung carcinoma: analysis with the NONMEM program. Cancer Chemother Pharmacol 2013; 72:1111-23. [DOI: 10.1007/s00280-013-2288-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 09/02/2013] [Indexed: 01/31/2023]
|
175
|
Hinai Y, Motoyama S, Niioka T, Miura M. Absence of effect of SLC22A2 genotype on cisplatin-induced nephrotoxicity in oesophageal cancer patients receiving cisplatin and 5-fluorouracil: report of results discordant with those of earlier studies. J Clin Pharm Ther 2013; 38:498-503. [PMID: 24102360 DOI: 10.1111/jcpt.12097] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 08/15/2013] [Indexed: 11/29/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Cancer patients treated with cisplatin chemotherapy frequently experience drug-induced nephrotoxicity. Clinical studies using a single chemotherapeutic regimen or large sample sizes for patients with the SLC22A2 808T allele have not been reported. Here, we examined 95 patients with oesophageal cancer who received 5-fluorouracil and cisplatin (FP) to determine whether nephrotoxicity was affected by SLC22A2 808G>T polymorphism. METHODS The change rate of the estimated glomerular filtration rate (eGFR) was used for the evaluation of cisplatin-induced nephrotoxicity and calculated for each patient according to the following formula: change rate = (prechemotherapy value - post-chemotherapy value)/prechemotherapy value. Genotyping of SLC22A2 808G>T was carried out using the polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method. RESULTS The eGFR after FP chemotherapy was significantly lower than that before chemotherapy, and the mean difference in eGFR was 25·7 mL/min (P < 0·01). There was no significant difference in the mean change rate of the eGFR following FP chemotherapy between the SLC22A2 808GG genotype (n = 70) and the 808GT+TT genotypes (n = 25) (27·9% and 27·8%, respectively). In multiple regression analyses, the change rate of eGFR following FP chemotherapy was associated with the eGFR value prior to chemotherapy (P = 0·04). WHAT IS NEW AND CONCLUSION In FP chemotherapy for oesophageal cancers, cisplatin-induced nephrotoxicity seems to be unaffected by the SLC22A2 808G>T polymorphism. The eGFR prior to chemotherapy might be an important risk factor for cisplatin-induced nephrotoxicity. Our present study was estimated with a single chemotherapeutic regimen, eGFR, and was calculated using serum creatinine, age and the sex of the patient and sample sizes of 25 patients with SLC22A2 808T allele. However, further examinations with a larger sample size to corroborate the study results might be necessary.
Collapse
Affiliation(s)
- Y Hinai
- Department of Pharmacy, Akita University Hospital, Akita, Japan
| | | | | | | |
Collapse
|
176
|
Li Q, Guo D, Dong Z, Zhang W, Zhang L, Huang SM, Polli JE, Shu Y. Ondansetron can enhance cisplatin-induced nephrotoxicity via inhibition of multiple toxin and extrusion proteins (MATEs). Toxicol Appl Pharmacol 2013; 273:100-9. [PMID: 24001450 DOI: 10.1016/j.taap.2013.08.024] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 08/12/2013] [Accepted: 08/23/2013] [Indexed: 12/23/2022]
Abstract
The nephrotoxicity limits the clinical application of cisplatin. Human organic cation transporter 2 (OCT2) and multidrug and toxin extrusion proteins (MATEs) work in concert in the elimination of cationic drugs such as cisplatin from the kidney. We hypothesized that co-administration of ondansetron would have an effect on cisplatin nephrotoxicity by altering the function of cisplatin transporters. The inhibitory potencies of ondansetron on metformin accumulation mediated by OCT2 and MATEs were determined in the stable HEK-293 cells expressing these transporters. The effects of ondansetron on drug disposition in vivo were examined by conducting the pharmacokinetics of metformin, a classical substrate for OCTs and MATEs, in wild-type and Mate1-/- mice. The nephrotoxicity was assessed in the wild-type and Mate1-/- mice received cisplatin with and without ondansetron. Both MATEs, including human MATE1, human MATE2-K, and mouse Mate1, and OCT2 (human and mouse) were subject to ondansetron inhibition, with much greater potencies by ondansetron on MATEs. Ondansetron significantly increased tissue accumulation and pharmacokinetic exposure of metformin in wild-type but not in Mate1-/- mice. Moreover, ondansetron treatment significantly enhanced renal accumulation of cisplatin and cisplatin-induced nephrotoxicity which were indicated by increased levels of biochemical and molecular biomarkers and more severe pathohistological changes in mice. Similar increases in nephrotoxicity were caused by genetic deficiency of MATE function in mice. Therefore, the potent inhibition of MATEs by ondansetron enhances the nephrotoxicity associated with cisplatin treatment in mice. Potential nephrotoxic effects of combining the chemotherapeutic cisplatin and the antiemetic 5-hydroxytryptamine-3 (5-HT3) receptor antagonists, such as ondansetron, should be investigated in patients.
Collapse
Affiliation(s)
- Qing Li
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, MD, USA; Institute of Clinical Pharmacology, Central South University, Hunan 410078, China
| | | | | | | | | | | | | | | |
Collapse
|
177
|
Signalling mechanisms involved in renal pathological changes during cisplatin-induced nephropathy. Eur J Clin Pharmacol 2013; 69:1863-74. [PMID: 23929259 DOI: 10.1007/s00228-013-1568-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 07/24/2013] [Indexed: 12/20/2022]
Abstract
CONTEXT Cisplatin, a coordination platinum complex, is used as a potential anti-neoplastic agent, having well recognized DNA-damaging property that triggers cell-cycle arrest and cell death in cancer therapy. Beneficial chemotherapeutic actions of cisplatin can be detrimental for kidneys. BACKGROUND Unbound cisplatin gets accumulated in renal tubular cells, leading to cell injury and death. This liable action of cisplatin on kidneys is mediated by altered intracellular signalling pathways such as mitogen-activated protein kinase (MAPK), extracellular regulated kinase (ERK), or C- Jun N terminal kinase/stress-activated protein kinase (JNK/SAPK). Further, these signalling alterations are responsible for release and activation of tumour necrosis factor (TNF-α), mitochondrial dysfunction, and apoptosis, which ultimately cause the renal pathogenic process. Cisplatin itself enhances the generation of reactive oxygen species (ROS) and activation of nuclear factor-κB (NF-κB), inflammation, and mitochondrial dysfunction, which further leads to renal apoptosis. Cisplatin-induced nephropathy is also mediated through the p53 and protein kinase-Cδ (PKCδ) signalling pathways. OBJECTIVE This review explores these signalling alterations and their possible role in the pathogenesis of cisplatin-induced renal injury.
Collapse
|
178
|
Maimaitiyiming H, Li Y, Cui W, Tong X, Norman H, Qi X, Wang S. Increasing cGMP-dependent protein kinase I activity attenuates cisplatin-induced kidney injury through protection of mitochondria function. Am J Physiol Renal Physiol 2013; 305:F881-90. [PMID: 23825069 DOI: 10.1152/ajprenal.00192.2013] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Cisplatin is widely used to treat malignancies. However, its major limitation is the development of dose-dependent nephrotoxicity. The precise mechanisms of cisplatin-induced kidney damage remain unclear, and the renoprotective agents during cisplatin treatment are still lacking. Here, we demonstrated that the expression and activity of cGMP-dependent protein kinase-I (PKG-I) were reduced in cisplatin-treated renal tubular cells in vitro as well as in the kidney tissues from cisplatin-treated mice in vivo. Increasing PKG activity by both pharmacological and genetic approaches attenuated cisplatin-induced kidney cell apoptosis in vitro. This was accompanied by decreased Bax/Bcl2 ratio, caspase 3 activity, and cytochrome c release. Cisplatin-induced mitochondria membrane potential loss in the tubular cells was also prevented by increased PKG activity. All of these data suggest a protective effect of PKG on mitochondria function in renal tubular cells. Importantly, increasing PKG activity pharmacologically or genetically diminished cisplatin-induced tubular damage and preserved renal function during cisplatin treatment in vivo. Mitochondria structural and functional damage in the kidney from cisplatin-treated mice was inhibited by increased PKG activity. In addition, increasing PKG activity enhanced ciaplatin-induced cell death in several cancer cell lines. Taken together, these results suggest that increasing PKG activity may be a novel option for renoprotection during cisplatin-based chemotherapy.
Collapse
Affiliation(s)
- Hasiyeti Maimaitiyiming
- Graduate Center for Nutritional Sciences, Univ. of Kentucky, Wethington Bldg. Rm. 583, 900 S. Limestone St., Lexington, KY 40536.
| | | | | | | | | | | | | |
Collapse
|
179
|
Ivy KD, Kaplan JH. A re-evaluation of the role of hCTR1, the human high-affinity copper transporter, in platinum-drug entry into human cells. Mol Pharmacol 2013; 83:1237-46. [PMID: 23543413 PMCID: PMC3657103 DOI: 10.1124/mol.113.085068] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 03/29/2013] [Indexed: 01/11/2023] Open
Abstract
Cisplatin (cDDP) is an anticancer drug used in a number of malignancies, including testicular, ovarian, cervical, bladder, lung, head, and neck cancers. Its use is limited by the development of resistance, often rationalized via effects on cellular uptake. It has been claimed that human copper transporter 1 (hCTR1), the human high-affinity copper transporter, is the major entry pathway for cDDP and related drugs via a mechanism that mimics copper. This is an unexpected property of hCTR1, a highly selective copper (I) transporter. We compared the uptake rates of copper with cDDP (and several analogs) into human embryonic kidney 293 cells overexpressing wild-type or mutant hCTR1, mouse embryonic fibroblasts that do or do not express CTR1, and human ovarian tumor cells that are sensitive or resistant to cDDP. We have also compared the effects of extracellular copper, which causes regulatory endocytosis of hCTR1, to those of cDDP. We confirm the correlation between higher hCTR1 levels and higher platinum drug uptake in tumor cells sensitive to the drug. However, we show that hCTR1 is not the major entry route of platinum drugs, and that the copper transporter is not internalized in response to extracellular drug. Our data suggest the major entry pathway for platinum drugs is not saturable at relevant concentrations and not protein-mediated. Clinical trials have been initiated that depend upon regulating membrane levels of hCTR1. If reduced drug uptake is a major factor in resistance, hCTR1 is unlikely to be a productive target in attempts to enhance efficacy, although the proteins involved in copper homeostasis may play a role.
Collapse
Affiliation(s)
- Kristin D Ivy
- Department of Biochemistry & Molecular Genetics, University of Illinois College of Medicine, Chicago, IL 60607, USA
| | | |
Collapse
|
180
|
Genetic disruption of Abl nuclear import reduces renal apoptosis in a mouse model of cisplatin-induced nephrotoxicity. Cell Death Differ 2013; 20:953-62. [PMID: 23660976 PMCID: PMC3679464 DOI: 10.1038/cdd.2013.42] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
DNA damage activates nuclear Abl tyrosine kinase to stimulate intrinsic apoptosis in cancer cell lines and mouse embryonic stem cells. To examine the in vivo function of nuclear Abl in apoptosis, we generated Abl-μNLS (μ, mutated in nuclear localization signals) mice. We show here that cisplatin-induced apoptosis is defective in the renal proximal tubule cells (RPTC) from the Ablμ/μ mice. When injected with cisplatin, we found similar levels of platinum in the Abl+/+ and the Ablμ/μ kidneys, as well as similar initial inductions of p53 and PUMAα expression. However, the accumulation of p53 and PUMAα could not be sustained in the Ablμ/μ kidneys, leading to reductions in renal apoptosis and tubule damage. Co-treatment of cisplatin with the Abl kinase inhibitor, imatinib, reduced the accumulation of p53 and PUMAα in the Abl+/+ but not in the Ablμ/μ kidneys. The residual apoptosis in the Ablμ/μ mice was not further reduced in the Ablμ/μ; p53−/− double-mutant mice, suggesting that nuclear Abl and p53 are epistatic to each other in this apoptosis response. Although apoptosis and tubule damage were reduced, cisplatin-induced increases in phospho-Stat-1 and blood urea nitrogen were similar between the Abl+/+ and the Ablμ/μ kidneys, indicating that RPTC apoptosis is not the only factor in cisplatin-induced nephrotoxicity. These results provide in vivo evidence for the pro-apoptotic function of Abl, and show that its nuclear localization and tyrosine kinase activity are both required for the sustained expression of p53 and PUMAα in cisplatin-induced renal apoptosis.
Collapse
|
181
|
Máthé C, Szénási G, Sebestény A, Blázovics A, Szentmihályi K, Hamar P, Albert M. Protective effect of CV247 against cisplatin nephrotoxicity in rats. Hum Exp Toxicol 2013; 33:789-99. [DOI: 10.1177/0960327113480972] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
CV247 (CV), an aqueous mixture of copper (Cu) and manganese (Mn) gluconates, vitamin C and sodium salicylate increased the antitumour effects of cisplatin (CDPP; cis-diamminedichloroplatinum) in vitro. We hypothesized that the antioxidant and cyclooxygenase-2 (COX-2; prostaglandin-endoperoxide synthase 2) inhibitory components of CV can protect the kidneys from CDPP nephrotoxicity in rats. CDPP (6.5 mg/kg, intraperitoneally) slightly elevated serum creatinine (Crea) and blood urea nitrogen (BUN) 12 days after treatment. Kidney histology demonstrated extensive tubular epithelial damage and COX-2 immunoreactivity increased 14 days after treatment. A large amount of platinum (Pt) accumulated in the kidney of CDPP-treated rats. Furthermore, CDPP decreased renal iron (Fe), molybdenum (Mo), zinc (Zn), Cu and Mn concentrations and increased plasma Fe and Cu concentrations. CDPP elevated plasma free radical concentration. Treatment with CV alone for 14 days (twice 3 ml/kg/day orally) did not influence these parameters. Chronic CV administration after CDPP reduced renal histological damage and slightly decreased COX-2 immunoreactivity, while failed to prevent the increase in Crea and BUN levels. Blood free radical concentration was reduced, that is, CV improved redox homeostasis. CV restored plasma Fe and renal Fe, Mo and Zn, while decreased Pt and elevated Cu and Mn concentrations in the kidney. Besides the known synergistic antitumour effects with CDPP, CV partially protected the kidneys from CDPP nephrotoxicity probably through its antioxidant effect.
Collapse
Affiliation(s)
- C. Máthé
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - G. Szénási
- Institute of Pathophysiology, Semmelweis University, Budapest, Hungary
| | - A. Sebestény
- Laboratory Animal Science Unit, Faculty of Veterinary Science, Szent István University, Budapest, Hungary
| | - A. Blázovics
- Department of Pharmacognosy, Semmelweis University, Budapest, Hungary
| | - K. Szentmihályi
- Institute of Materials and Environmental Chemistry Research Centre for Natural Sciences, Budapest, Hungary
| | - P. Hamar
- Institute of Pathophysiology, Semmelweis University, Budapest, Hungary
| | - M. Albert
- Vetmed Laboratory Ltd, Budapest, Hungary
| |
Collapse
|
182
|
Functional mechanotransduction is required for cisplatin-induced hair cell death in the zebrafish lateral line. J Neurosci 2013; 33:4405-14. [PMID: 23467357 DOI: 10.1523/jneurosci.3940-12.2013] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Cisplatin, one of the most commonly used anticancer drugs, is known to cause inner ear hair cell damage and hearing loss. Despite much investigation into mechanisms of cisplatin-induced hair cell death, little is known about the mechanism whereby cisplatin is selectively toxic to hair cells. Using hair cells of the zebrafish lateral line, we found that chemical inhibition of mechanotransduction with quinine and EGTA protected against cisplatin-induced hair cell death. Furthermore, we found that the zebrafish mutants mariner (myo7aa) and sputnik (cad23) that lack functional mechanotransduction were resistant to cisplatin-induced hair cell death. Using a fluorescent analog of cisplatin, we found that chemical or genetic inhibition of mechanotransduction prevented its uptake. These findings demonstrate that cisplatin-induced hair cell death is dependent on functional mechanotransduction in the zebrafish lateral line.
Collapse
|
183
|
Sprowl JA, Ness RA, Sparreboom A. Polymorphic Transporters and Platinum Pharmacodynamics. Drug Metab Pharmacokinet 2013; 28:19-27. [DOI: 10.2133/dmpk.dmpk-12-rv-073] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
184
|
Pezeshki Z, Nematbakhsh M, Nasri H, Talebi A, Pilehvarian AA, Safari T, Eshraghi-Jazi F, Haghighi M, Ashrafi F. Evidence against protective role of sex hormone estrogen in Cisplatin-induced nephrotoxicity in ovarectomized rat model. Toxicol Int 2013; 20:43-7. [PMID: 23833437 PMCID: PMC3702126 DOI: 10.4103/0971-6580.111568] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Cisplatin (CP) is an effective drug in cancer therapy to treat the solid tumors, but it is accompanied with nephrotoxicity. The protective effect of estrogen in cardiovascular diseases is well-documented; but its nephron-protective effect against CP-induced nephrotoxicity is not completely understood. MATERIALS AND METHODS Thirty ovarectomized Wistar rats were divided in to five groups. Groups 1-3 received different doses of estradiol valerate (0.5, 2.5 and 10 mg/kg/week) in sesame oil for 4 weeks, and at the end of week 3, a single dose of CP (7 mg/kg, intraperitoneal [IP]) was administrated. Group 4 (positive control) received the same regimen as group 1-3 without estradiol without vehicle. The negative control group (Group 5) received sesame oil during the study. The animals were sacrificed 1 week after CP injection for histopathological studies. RESULTS The serum level of blood urea nitrogen and creatinine, kidney tissue damage score (KTDS), kidney weight and percentage of body weight change in CP-treated groups significantly increased (P < 0.05), however, there were no significant differences detected between the estrogen-treated groups (Groups 1-3) and the positive control group (Group 4). Although, estradiol administration enhanced the serum level of nitrite, it was not affected by CP. Finally, significant correlation between KTDS and kidney weight was detected (r (2) = 0.63, P < 0.01). CONCLUSION Estrogen is not nephron-protective against CP-induced nephrotoxicity. Moreover, it seems that the mechanism may be related to estrogen-induced oxidative stress in the kidney, which may promote the nephrotoxicity.
Collapse
Affiliation(s)
- Zahra Pezeshki
- Water and Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Basic Sciences Isfahan University of Payam Noor, Isfahan, Iran
| | - Mehdi Nematbakhsh
- Water and Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Physiology, Isfahan University of Medical Sciences, Isfahan, Iran
- Kidney Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Nasri
- Water and Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Kidney Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ardeshir Talebi
- Water and Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Clinical Pathology, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Tahereh Safari
- Water and Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Physiology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Eshraghi-Jazi
- Water and Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Haghighi
- Water and Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farzaneh Ashrafi
- Water and Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Internal Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
185
|
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
|
186
|
Plasticity in the copper–thioether bond: Manifestation in blue Cu proteins and in synthetic analogs. J Inorg Biochem 2012; 115:182-5. [DOI: 10.1016/j.jinorgbio.2012.01.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2011] [Revised: 01/13/2012] [Accepted: 01/13/2012] [Indexed: 11/19/2022]
|
187
|
Jones QRD, Warford J, Rupasinghe HPV, Robertson GS. Target-based selection of flavonoids for neurodegenerative disorders. Trends Pharmacol Sci 2012; 33:602-10. [PMID: 22980637 DOI: 10.1016/j.tips.2012.08.002] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 08/09/2012] [Accepted: 08/15/2012] [Indexed: 11/26/2022]
Abstract
Habitual consumption of dietary flavonoids known to improve mitochondrial bioenergetics and inhibit various secondary sources of reactive oxygen species (ROS) reduces the risk for neurodegenerative disorders such as Parkinson's disease (PD), stroke, and Alzheimer's disease (AD). Combining specific dietary flavonoids selected on the basis of oral bioavailability, brain penetration, and the inhibition of multiple processes responsible for excessive ROS production may be a viable approach for the prevention and treatment of neurodegenerative disorders. Inclusion of flavonoids that raise cAMP levels in the brain may be of additional benefit by reducing the production of proinflammatory mediators and stimulating the transcriptional machinery necessary for mitochondrial biosynthesis. Preclinical models suggest that flavonoids reduce hearing loss resulting from treatment with the chemotherapeutic drug cisplatin by opposing the excessive production of ROS and proinflammatory mediators implicated in PD, stroke, and AD. Flavonoid combinations optimized for efficacy in models of cisplatin-induced hearing loss (CIHL) may therefore have therapeutic utility for neurodegenerative disorders.
Collapse
Affiliation(s)
- Quinton R D Jones
- Department of Pharmacology, Faculty of Medicine, 1459 Oxford Street, Dalhousie University, Halifax, Nova Scotia, Canada B3H 4R2
| | | | | | | |
Collapse
|
188
|
Cilastatin protects against cisplatin-induced nephrotoxicity without compromising its anticancer efficiency in rats. Kidney Int 2012; 82:652-63. [PMID: 22718191 DOI: 10.1038/ki.2012.199] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cisplatin is an anticancer agent marred by nephrotoxicity; however, limiting this adverse effect may allow the use of higher doses to improve its efficacy. Cilastatin, a small molecule inhibitor of renal dehydropeptidase I, prevents proximal tubular cells from undergoing cisplatin-induced apoptosis in vitro. Here, we explored the in vivo relevance of these findings and the specificity of protection for kidney cells in cisplatin-treated rats. Cisplatin increased serum blood urea nitrogen and creatinine levels, and the fractional excretion of sodium. Cisplatin decreased the glomerular filtration rate, promoted histological renal injury and the expression of many pro-apoptotic proteins in the renal cortex, increased the Bax/Bcl2 ratio, and oxidative stress in kidney tissue and urine. All these features were decreased by cilastatin, which preserved renal function but did not modify the pharmacokinetics of cisplatin area under the curve. The cisplatin-induced death of cervical, colon, breast, and bladder-derived cancer cell lines was not prevented by cilastatin. Thus, cilastatin has the potential to prevent cisplatin nephrotoxicity without compromising its anticancer efficacy.
Collapse
|
189
|
Kim J, Long KE, Tang K, Padanilam BJ. Poly(ADP-ribose) polymerase 1 activation is required for cisplatin nephrotoxicity. Kidney Int 2012; 82:193-203. [PMID: 22437413 DOI: 10.1038/ki.2012.64] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Apoptosis, necrosis, and inflammation are hallmarks of cisplatin nephrotoxicity; however, the role and mechanisms of necrosis and inflammation remains undefined. As poly(ADP-ribose) polymerase 1 (PARP1) inhibition or its gene deletion is renoprotective in several renal disease models, we tested whether its activation may be involved in cisplatin nephrotoxicity. Parp1 deficiency was found to reduce cisplatin-induced kidney dysfunction, oxidative stress, and tubular necrosis, but not apoptosis. Moreover, neutrophil infiltration, activation of nuclear factor-κB, c-Jun N-terminal kinases, p38 mitogen-activated protein kinase, and upregulation of proinflammatory genes were all abrogated by Parp1 deficiency. Using proximal tubule epithelial cells isolated from Parp1-deficient and wild-type mice and pharmacological inhibitors, we found evidence for a PARP1/Toll-like receptor 4/p38/tumor necrosis factor-α axis following cisplatin injury. Furthermore, pharmacological inhibition of PARP1 protected against cisplatin-induced kidney structural/functional damage and inflammation. Thus, our findings suggest that PARP1 activation is a primary signal and its inhibition/loss protects against cisplatin-induced nephrotoxicity. Targeting PARP1 may offer a potential therapeutic strategy for cisplatin nephrotoxicity.
Collapse
Affiliation(s)
- Jinu Kim
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA
| | | | | | | |
Collapse
|
190
|
dos Santos NAG, Carvalho Rodrigues MA, Martins NM, dos Santos AC. Cisplatin-induced nephrotoxicity and targets of nephroprotection: an update. Arch Toxicol 2012; 86:1233-50. [PMID: 22382776 DOI: 10.1007/s00204-012-0821-7] [Citation(s) in RCA: 264] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 02/14/2012] [Indexed: 01/15/2023]
Abstract
Cisplatin is a highly effective antitumor agent whose clinical application is limited by the inherent nephrotoxicity. The current measures of nephroprotection used in patients receiving cisplatin are not satisfactory, and studies have focused on the investigation of new possible protective strategies. Many pathways involved in cisplatin nephrotoxicity have been delineated and proposed as targets for nephroprotection, and many new potentially protective agents have been reported. The multiple pathways which lead to renal damage and renal cell death have points of convergence and share some common modulators. The most frequent event among all the described pathways is the oxidative stress that acts as both a trigger and a result. The most exploited pathways, the proposed protective strategies, the achievements obtained so far as well as conflicting data are summarized and discussed in this review, providing a general view of the knowledge accumulated with past and recent research on this subject.
Collapse
Affiliation(s)
- Neife Aparecida Guinaim dos Santos
- Department of Clinical, Toxicological Analyses and Food Sciences of School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Brazil
| | | | | | | |
Collapse
|
191
|
Pabla N, Dong Z. Curtailing side effects in chemotherapy: a tale of PKCδ in cisplatin treatment. Oncotarget 2012; 3:107-11. [PMID: 22403741 PMCID: PMC3292897 DOI: 10.18632/oncotarget.439] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2012] [Accepted: 01/30/2012] [Indexed: 12/21/2022] Open
Abstract
The efficacy of chemotherapy is often limited by side effects in normal tissues. This is exemplified by cisplatin, a widely used anti-cancer drug that may induce serious toxicity in normal tissues and organs including the kidneys. Decades of research have delineated multiple signaling pathways that lead to kidney cell injury and death during cisplatin treatment. However, the same signaling pathways may also be activated in cancer cells and be responsible for the chemotherapeutic effects of cisplatin in tumors and, as a result, blockade of these pathways is expected to reduce the side effects as well as the anti-cancer efficacy. Thus, to effectively curtail the side effects, it is imperative to elucidate and target the cell killing mechanisms that are specific to normal (and not cancer) tissues. Our recent work identified protein kinase C δ (PKCδ) as a new and critical mediator of cisplatin-induced kidney cell injury and death. Importantly, inhibition of PKCδ enhanced the chemotherapeutic effects of cisplatin in several tumor models while alleviating the side effect in kidneys, opening a new avenue for normal tissue protection during chemotherapy.
Collapse
Affiliation(s)
- Navjotsingh Pabla
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Health Sciences University and Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30912, USA
| | | |
Collapse
|
192
|
Pabla N, Dong G, Jiang M, Huang S, Kumar MV, Messing RO, Dong Z. Inhibition of PKCδ reduces cisplatin-induced nephrotoxicity without blocking chemotherapeutic efficacy in mouse models of cancer. J Clin Invest 2011; 121:2709-22. [PMID: 21633170 DOI: 10.1172/jci45586] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 04/06/2011] [Indexed: 12/21/2022] Open
Abstract
Cisplatin is a widely used cancer therapy drug that unfortunately has major side effects in normal tissues, notably nephrotoxicity in kidneys. Despite intensive research, the mechanism of cisplatin-induced nephrotoxicity remains unclear, and renoprotective approaches during cisplatin-based chemotherapy are lacking. Here we have identified PKCδ as a critical regulator of cisplatin nephrotoxicity, which can be effectively targeted for renoprotection during chemotherapy. We showed that early during cisplatin nephrotoxicity, Src interacted with, phosphorylated, and activated PKCδ in mouse kidney lysates. After activation, PKCδ regulated MAPKs, but not p53, to induce renal cell apoptosis. Thus, inhibition of PKCδ pharmacologically or genetically attenuated kidney cell apoptosis and tissue damage, preserving renal function during cisplatin treatment. Conversely, inhibition of PKCδ enhanced cisplatin-induced cell death in multiple cancer cell lines and, remarkably, enhanced the chemotherapeutic effects of cisplatin in several xenograft and syngeneic mouse tumor models while protecting kidneys from nephrotoxicity. Together these results demonstrate a role of PKCδ in cisplatin nephrotoxicity and support targeting PKCδ as an effective strategy for renoprotection during cisplatin-based cancer therapy.
Collapse
Affiliation(s)
- Navjotsingh Pabla
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Health Sciences University, Augusta, Georgia, USA
| | | | | | | | | | | | | |
Collapse
|
193
|
Moreno-Gordaliza E, Giesen C, Lázaro A, Esteban-Fernández D, Humanes B, Cañas B, Panne U, Tejedor A, Jakubowski N, Gómez-Gómez MM. Elemental bioimaging in kidney by LA-ICP-MS as a tool to study nephrotoxicity and renal protective strategies in cisplatin therapies. Anal Chem 2011; 83:7933-40. [PMID: 21913725 DOI: 10.1021/ac201933x] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS)-based methodology is presented for Pt, Cu, and Zn bioimaging on whole kidney 3 μm sagittal sections from rats treated with pharmacological doses of cisplatin, which were sacrificed once renal damage had taken place. Pt turned out to accumulate in the kidney cortex and corticomedullary junction, corresponding to areas where the proximal tubule S3 segments (the most sensitive cells to cisplatin nephrotoxicity) are located. This demonstrates the connection between platinum accumulation and renal damage proved by histological examination of HE-stained sections and evaluation of serum and urine biochemical parameters. Cu and Zn distribution maps revealed a significant displacement in cells by Pt, as compared to control tissues. A dramatic decrease in the Pt accumulation in the cortex was observed when cilastatin was coadministered with cisplatin, which can be related to its nephroprotective effect. Excellent imaging reproducibility, sensitivity (LOD 50 fg), and resolution (down to 8 μm) were achieved, demonstrating that LA-ICP-MS can be applied as a microscopic metal detector at cellular level in certain tissues. A simple and quick approach for the estimation of Pt tissue levels was proposed, based on tissue spiking.
Collapse
Affiliation(s)
- Estefanía Moreno-Gordaliza
- Department of Analytical Chemistry, Faculty of Chemistry, Universidad Complutense de Madrid, Avda. Complutense s/n, 28040, Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
194
|
Ding D, He J, Allman BL, Yu D, Jiang H, Seigel GM, Salvi RJ. Cisplatin ototoxicity in rat cochlear organotypic cultures. Hear Res 2011; 282:196-203. [PMID: 21854840 DOI: 10.1016/j.heares.2011.08.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 08/03/2011] [Accepted: 08/05/2011] [Indexed: 01/17/2023]
Abstract
Ototoxicity is a dose-limiting side effect of chemotherapeutic treatment with cisplatin. In a series of experiments on neonatal rat cochlear organotypic cultures, the extent of damage induced by a broad range of cisplatin treatment concentrations was examined. Paradoxically, it was found that hair cell loss was greater following 48 h exposure to low (10, 50 and 100 μM) versus high (400 and 1000 μM) concentrations of cisplatin; these findings indicate that hair cells possess intrinsic resistance to high levels of extracellular cisplatin. Using cisplatin conjugated to Alexa Fluor 488, it was found that cisplatin is readily taken up by hair cells at low concentrations, but is largely excluded at high concentrations. Recent studies indicate that the major influx of cisplatin into hair cells occurs via the copper transporter, Ctr1, whereas ATP7A and ATP7B are copper pumps responsible for cisplatin sequestration and efflux. Using immunolabeling procedures for these copper trafficking proteins, it was found that Ctr1 and ATP7B were localized in the hair cells, whereas ATP7A showed extensive labeling in the pillar cells in the organ of Corti. Additional experiments confirmed the protective effect of copper sulfate and cimetidine in attenuating cisplatin-induced hair cell loss. However, because neither copper sulfate nor cimetidine provided complete protection against cisplatin, and high levels of copper sulfate itself were found to be ototoxic, it is suggested that future therapeutic efforts may benefit from a combination of pharmacological treatments which seek to not only limit the uptake of cisplatin into cochlear cells but also increase its efflux.
Collapse
Affiliation(s)
- Dalian Ding
- Center for Hearing and Deafness, State University of New York at Buffalo, 137 Cary Hall, 3435 Main Street, Buffalo, NY 14214, USA
| | | | | | | | | | | | | |
Collapse
|
195
|
Sánchez-González PD, López-Hernández FJ, López-Novoa JM, Morales AI. An integrative view of the pathophysiological events leading to cisplatin nephrotoxicity. Crit Rev Toxicol 2011; 41:803-21. [DOI: 10.3109/10408444.2011.602662] [Citation(s) in RCA: 164] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
196
|
Uehara T, Yamate J, Torii M, Maruyama T. Comparative nephrotoxicity of Cisplatin and nedaplatin: mechanisms and histopathological characteristics. J Toxicol Pathol 2011; 24:87-94. [PMID: 22272048 PMCID: PMC3234610 DOI: 10.1293/tox.24.87] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 01/24/2011] [Indexed: 12/23/2022] Open
Abstract
The antineoplastic platinum complexes cisplatin and its analogues are widely used in the chemotherapy of a variety of human malignancies, and are especially active against several types of cancers. Nedaplatin is a second-generation platinum complex with reduced nephrotoxicity. However, their use commonly causes nephrotoxicity due to a lack of tumor tissue selectivity. Several recent studies have provided significant insights into the molecular and histopathological events associated with nedaplatin nephrotoxicity. In this review, we summarize findings concerning the renal histopathology and molecular pathogenesis induced by antineoplastic platinum complexes, with a particular focus on the comparative nephrotoxicity of cisplatin and nedaplatin in rats.
Collapse
Affiliation(s)
- Takeki Uehara
- Drug Safety Evaluation, Drug Developmental Research
Laboratories, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka, Osaka 561-0825,
Japan
| | - Jyoji Yamate
- Department of Veterinary Pathology, Graduate School of
Agriculture and Biological Science, Osaka Prefecture University, 1-58 Rinkuu Ourai Kita,
Izumisano, Osaka 598-8531, Japan
| | - Mikinori Torii
- Drug Safety Evaluation, Drug Developmental Research
Laboratories, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka, Osaka 561-0825,
Japan
| | - Toshiyuki Maruyama
- Drug Safety Evaluation, Drug Developmental Research
Laboratories, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka, Osaka 561-0825,
Japan
| |
Collapse
|
197
|
Burger H, Loos WJ, Eechoute K, Verweij J, Mathijssen RHJ, Wiemer EAC. Drug transporters of platinum-based anticancer agents and their clinical significance. Drug Resist Updat 2011; 14:22-34. [PMID: 21251871 DOI: 10.1016/j.drup.2010.12.002] [Citation(s) in RCA: 164] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Revised: 12/23/2010] [Accepted: 12/24/2010] [Indexed: 01/11/2023]
Abstract
Platinum-based drugs are among the most active anticancer agents and are successfully used in a wide variety of human malignancies. However, acquired and/or intrinsic resistance still represent a major limitation. Lately, in particular mechanisms leading to impaired uptake and/or decreased cellular accumulation of platinum compounds have attracted attention. In this review, we focus on the role of active platinum uptake and efflux systems as determinants of platinum sensitivity and -resistance and their contribution to platinum pharmacokinetics (PK) and pharmacodynamics (PD). First, the three mostly used platinum-based anticancer agents as well as the most promising novel platinum compounds in development are put into clinical perspective. Next, we describe the presently known potential platinum transporters--with special emphasis on organic cation transporters (OCTs)--and discuss their role on clinical outcome (i.e. efficacy and adverse events) of platinum-based chemotherapy. In addition, transporter-mediated tumour resistance, the impact of potential platinum transporter-mediated drug-drug interactions, and the role of drug transporters in the renal elimination of platinum compounds are discussed.
Collapse
Affiliation(s)
- Herman Burger
- Department of Medical Oncology, Erasmus Medical Center Rotterdam-Josephine Nefkens Institute and Daniel den Hoed Cancer Center, Rotterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
198
|
Abstract
INTRODUCTION Membrane transporters are important determinants of in vivo drug disposition, therapeutic efficacy and adverse drug reactions. Many commonly used drugs are organic cations and substrates of organic cation transporters (OCTs). These transporters have a large binding site containing partially overlapping interaction domains for different substrates and are specifically distributed around the body. Consequently, drug interactions with these transporters can result in specific toxicity. AREAS COVERED This review describes the general properties of OCT and illustrates their importance for the development of important drug toxicities using the examples of metformin and cisplatin. Additionally, this review discusses the role of OCT polymorphisms in the modulation of these toxic effects. EXPERT OPINION Understanding how drugs interact with membrane transporters is pivotally important in explaining the mechanisms of specific toxicities and also in designing new drugs or new therapeutic protective protocols by specific competition at the transporter. Defining the pharmacogenomics of these transporters will be essential to personalized medicine, enabling physicians to choose drugs for patients based on efficacy, availability, cost, safety, tolerability and convenience.
Collapse
Affiliation(s)
- Giuliano Ciarimboli
- Medizinische Klinik und Poliklinik D, Experimentelle Nephrologie, Universitätsklinikum Münster, Domagkstr. 3a, 48149 Münster, Germany.
| |
Collapse
|
199
|
Regulation of Cisplatin cytotoxicity by cu influx transporters. Met Based Drugs 2011; 2010:317581. [PMID: 21274436 PMCID: PMC3025362 DOI: 10.1155/2010/317581] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Accepted: 12/07/2010] [Indexed: 12/30/2022] Open
Abstract
Platinum drugs are an important class of cancer chemotherapeutics. However, the use of these drugs is limited by the development of resistance during treatment with decreased accumulation being a common mechanism. Both Cu transporters CTR1 and CTR2 influence the uptake and cytotoxicity of cisplatin. Although it is structurally similar to CTR1, CTR2 functions in a manner opposite to that of CTR1 with respect to Pt drug uptake. Whereas knockout of CTR1 reduces Pt drug uptake, knockdown of CTR2 enhances cisplatin uptake and cytotoxicity. CTR2 is subject to transcriptional and posttranscriptional regulation by both Cu and cisplatin; this regulation is partly dependent on the Cu chaperone ATOX1. Insight into the mechanisms by which CTR1 and CTR2 regulate sensitivity to the Pt-containing drugs has served as the basis for novel pharmacologic strategies for improving their efficacy.
Collapse
|
200
|
Yonezawa A, Inui KI. Organic cation transporter OCT/SLC22A and H(+)/organic cation antiporter MATE/SLC47A are key molecules for nephrotoxicity of platinum agents. Biochem Pharmacol 2010; 81:563-8. [PMID: 21144842 DOI: 10.1016/j.bcp.2010.11.016] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 11/25/2010] [Accepted: 11/30/2010] [Indexed: 12/19/2022]
Abstract
Platinum agents have been widely used in cancer chemotherapy for a long time. Cisplatin, carboplatin, oxaliplatin and nedaplatin have a common chemical structure consisting of platinum, carrier groups and leaving groups, and undergo the similar mechanism of cytotoxicity. However, each agent differs in its efficacy and adverse effects, although the molecular mechanism involved is unclear. Recently, it was reported that organic cation transporter OCT/SLC22A, and multidrug and toxin extrusion MATE/SLC47A play a role in the pharmacokinetics of platinum agents. Only cisplatin induces nephrotoxicity and the toxicity is kidney-specific. Kidney-specific OCT2 mediates the transport of cisplatin and is the determinant of cisplatin-induced nephrotoxicity. In addition, cisplatin and oxaliplatin are substrates for these transporters, but carboplatin and nedaplatin are not. Substrate specificity could regulate the features of platinum agents. In this commentary, we will discuss the characteristics of OCT and MATE, and demonstrate the recent topics about the relationship between the transport of platinum agents by organic cation transporters and their pharmacological characteristics.
Collapse
Affiliation(s)
- Atsushi Yonezawa
- Department of Pharmacy, Kyoto University Hospital, Faculty of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | | |
Collapse
|