1
|
Ikeda Y, Suga N, Matsuda S. Efficacy of Life Protection Probably from Newly Isolated Bacteria against Cisplatin-Induced Lethal Toxicity. Microorganisms 2023; 11:2246. [PMID: 37764090 PMCID: PMC10536890 DOI: 10.3390/microorganisms11092246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Cisplatin may be commonly used in chemotherapy against various solid tumors. However, cisplatin has a limited safety range with serious side effects, which may be one of the dose-restraining reasons for cisplatin. A favorable therapeutic approach is immediately required for ameliorating cisplatin-induced toxicity. In the present study, the potential protective effects of certain bacteria have been investigated at the lethal dosage of cisplatin in mice experimental models. Treated under the highest dosage of cisplatin, treatment of certain commensal bacteria could significantly increase the survival rate. In addition, our findings revealed that probiotic supplementation of these bacteria could result in the attenuation of the damage appearance on the kidney as well as the alteration of several antioxidant-related gene expressions, including SOD1, SOD2, SOD3, Nrf2, and/or HO-1 genes in the high dosage of cisplatin-treated mice. In short, acute kidney injury in mice was induced by a single dose of cisplatin 11 or 15 mg/kg intraperitoneally. Then, peroral administration of newly isolated bacteria could protect against the cisplatin-induced injury, probably by decreasing oxidative stress. Therefore, the data shown here might suggest that the usage of certain probiotic supplementation could contribute to the life protection of patients suffering from severe toxicity of cisplatin. However, the molecular mechanisms need to be further explored.
Collapse
Affiliation(s)
- Yuka Ikeda
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Naoko Suga
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Satoru Matsuda
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| |
Collapse
|
2
|
Wei YS, Chen YL, Li WY, Yang YY, Lin SJ, Wu CH, Yang JI, Wang TE, Yu J, Tsai PS. Antioxidant Nanoparticles Restore Cisplatin-Induced Male Fertility Defects by Promoting MDC1-53bp1-Associated Non-Homologous DNA Repair Mechanism and Sperm Intracellular Calcium Influx. Int J Nanomedicine 2023; 18:4313-4327. [PMID: 37576465 PMCID: PMC10416785 DOI: 10.2147/ijn.s408623] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 07/19/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction Cisplatin, a commonly used anticancer compound, exhibits severe off-target organ toxicity. Due to its wide application in cancer treatment, the reduction of its damage to normal tissue is an imminent clinical need. Cisplatin-induced testicular oxidative stress and damage lead to male sub- or infertility. Despite earlier studies showing that the natural polyphenol extracts honokiol serve as the free radical scavenger that reduces the accumulation of intracellular free radicals, whether honokiol exhibits direct effects on the testis and sperm is unclear. Thus, the aim of the current study is to investigate the direct effects of honokiol on testicular recovery and sperm physiology. Methods We encapsulated this polyphenol antioxidation compound into liposome-based nanoparticles (nHNK) and gave intraperitoneally to mice at a dosage of 5 mg/kg body mass every other day for consecutive 6 weeks. Results We showed that nHNK promotes MDC1-53bp1-associated non-homologous DNA double-strand break repair signaling pathway that minimizes cisplatin-induced DNA damage. This positive effect restores spermatogenesis and allows the restructuring of the multi-spermatogenic layers in the testis. By reducing mitochondrial oxidative damage, nHNK also protects sperm mitochondrial structure and maintains both testicular and sperm ATP production. By a yet-to-identify mechanism, nHNK restores sperm calcium influx at the sperm midpiece and tail, which is essential for sperm hypermotility and their interaction with the oocyte. Discussion Taken together, the nanoparticulated antioxidant counteracts cisplatin-induced male fertility defects and benefits patients undertaking cisplatin-based chemotherapy. These data may allow the reintroduction of cisplatin for systemic applications in patients at clinics with reduced testicular toxicity.
Collapse
Affiliation(s)
- Yu-Syuan Wei
- Graduate Institute of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Yu-Liang Chen
- Department of Chemical Engineering, College of Engineering, National Taiwan University, Taipei, 106, Taiwan
| | - Wei-Yun Li
- Graduate Institute of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Ya-Yi Yang
- Department of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Sung-Jan Lin
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, 10617, Taiwan
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, 10051, Taiwan
- Department of Dermatology, National Taiwan University Hospital and College of Medicine, Taipei, 10002, Taiwan
| | - Ching-Ho Wu
- Graduate Institute of Veterinary Clinical Science, School of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Jiue-In Yang
- Department of Plant Pathology and Microbiology, National Taiwan University, Taipei, 10617, Taiwan
| | - Tse-En Wang
- Graduate Institute of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Jiashing Yu
- Department of Chemical Engineering, College of Engineering, National Taiwan University, Taipei, 106, Taiwan
| | - Pei-Shiue Tsai
- Graduate Institute of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
- Department of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, 10617, Taiwan
| |
Collapse
|
3
|
Zhang Y, Xu Q, Sun Q, Kong R, Liu H, Yi X, Liang Z, Letcher RJ, Liu C. Ustiloxin A inhibits proliferation of renal tubular epithelial cells in vitro and induces renal injury in mice by disrupting structure and respiratory function of mitochondria. JOURNAL OF HAZARDOUS MATERIALS 2023; 448:130791. [PMID: 36706486 DOI: 10.1016/j.jhazmat.2023.130791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/08/2023] [Accepted: 01/12/2023] [Indexed: 06/18/2023]
Abstract
Recently, we found that Ustiloxin A (UA, a mycotoxin) was widely detected in paddy environment and rice samples from several countries, and was also detected in human urine samples from China. However, the current knowledge about the health risks of UA are limited. In this research, the cytotoxicity of UA in mice renal tubular epithelial cells (mRTECs) was evaluated, and the results indicated that UA arrested cell cycle in G2/M phase via altering cellular morphology and microtubule, and inhibited the proliferation and division of mRTECs. Furthermore, UA could inhibit mitochondrial respiration via binding to the CoQ-binding site in dihydro-orotate dehydrogenase (DHODH) protein, and resulted in mitochondrial damage. These adverse effects of UA on mitochondria might be responsible for the cytotoxicity observed in vitro. In vivo, UA at concentrations that were comparable to the realistic concentrations of human exposure induced renal insufficiency in mice, and this might be associated with the renal mitochondrial damage in mice. However, exposure to UA at those realistic concentrations did not promote the progression from renal insufficiency to renal fibrosis and chronic kidney disease was not observed in mice.
Collapse
Affiliation(s)
- Yongkang Zhang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Qiaolin Xu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Qian Sun
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Ren Kong
- School of Environmental Studies, China University of Geosciences, Wuhan 430074, China
| | - Hao Liu
- College of Plant Science & Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Xun'e Yi
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhengqi Liang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Robert J Letcher
- Departments of Chemistry and Biology, Carleton University, Ottawa K1S 5B6, ON, Canada
| | - Chunsheng Liu
- School of Environmental Studies, China University of Geosciences, Wuhan 430074, China.
| |
Collapse
|
4
|
Time dependent cisplatin dosing differences on hypoalgesia focusing on oxidative stress. Eur J Pharmacol 2023; 942:175519. [PMID: 36682481 DOI: 10.1016/j.ejphar.2023.175519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023]
Abstract
Although cisplatin is a key drug in cancer chemotherapy, it often causes sensory peripheral neuropathy, presenting as allodynia in the early stage and hypoalgesia in the serious stage. Chronotherapy has previously been shown to ameliorate cisplatin-induced peripheral neuropathy that was severe enough to cause hypoalgesia in rats. It also has adverse effects such as renal dysfunction and ototoxicity, which are induced by oxidative stress. Here, we show that oxidative stress causes severe cisplatin-induced peripheral neuropathy, and that differences in oxidative stress occur depending on the dosing time of cisplatin. Cisplatin was administered to rats at 5:00 or 17:00 every seven days for four weeks. The antioxidant agent, 1,3-Dimethylthiourea (DMTU), was administered before and after the administration of cisplatin. The hot plate test was used to assess hypoalgesia. Oxidative stress in the sciatic nerve was assessed from thiobarbituric acid reactive substances (TBARs) and superoxide dismutase (SOD) activity. Nerve apoptosis was analysed with qRT-PCR. We observed an increase in TBARs and a decrease in SOD activity with the development of cisplatin-induced hypoalgesia, which was ameliorated by DMTU treatment. Furthermore, differences in the dosing time of cisplatin caused differences in oxidative stress which were correlated with cisplatin-induced hypoalgesia. Severe oxidative stress caused cisplatin-induced hypoalgesia, and chronotherapy with cisplatin ameliorated hypoalgesia by reducing oxidative stress. In the future, chronotherapy with cisplatin may contribute to the treatment of cancer in humans.
Collapse
|
5
|
Wei M, Gao Y, Cheng D, Zhang H, Zhang W, Shen Y, Huang Q, An X, Wang B, Yu Z, Wang N, Chen H, Xu Y, Gui D. Notoginsenoside Fc ameliorates renal tubular injury and mitochondrial damage in acetaminophen-induced acute kidney injury partly by regulating SIRT3/SOD2 pathway. Front Med (Lausanne) 2023; 9:1055252. [PMID: 36714147 PMCID: PMC9875593 DOI: 10.3389/fmed.2022.1055252] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/28/2022] [Indexed: 01/13/2023] Open
Abstract
Introduction Mitochondria dysfunction is one of the primary causes of tubular injury in acute kidney injury (AKI). Notoginsenoside Fc (Fc), a new saponin isolated from Panax notoginseng, exhibited numerous pharmacological actions. However, the beneficial effects of Fc on renal tubular impairment and mitochondrial dysfunction in AKI have not been fully studied. Methods In this study, we established acetaminophen (APAP)-induced AKI model in mice to examine the therapeutic impacts of Fc on AKI. Results Our results showed that Fc could decrease the levels of the serum creatinine (Scr), blood urea nitrogen (BUN) and Cystatin C in mice with AKI. Fc also ameliorated renal histopathology, renal tubular cells apoptosis and restored expression of apoptosis-related proteins such as Bax, Bcl-2 and caspase3 (C-caspase3). Additionally, Fc increased the protein expression of SIRT3 and SOD2 in kidneys from mice with AKI. In vitro studies further showed Fc reduced the apoptosis of HK-2 cells exposure to APAP, attenuated the loss of mitochondrial membrane potential and decreased the formation of mitochondrial superoxide. Fc also partly restored the protein expression of Bax, Bcl-2, C-Caspase3, SIRT3, and SOD2 in HK-2 cells exposure to APAP. Conclusion In summary, Fc might reduce renal tubular injury and mitochondrial dysfunction in AKI partly through the regulation of SIRT3/SOD2 pathway.
Collapse
Affiliation(s)
- Miaomiao Wei
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China,Department of Nephrology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuancheng Gao
- The Third Affiliated Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Dongsheng Cheng
- Department of Nephrology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiying Zhang
- Department of Nephrology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Zhang
- Department of Nephrology, Shanghai Yangpu Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Yilan Shen
- Department of Nephrology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qunwei Huang
- Department of Nephrology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoning An
- Department of Nephrology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing Wang
- Department of Traditional Chinese Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhonghai Yu
- Department of Traditional Chinese Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Niansong Wang
- Department of Nephrology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongbo Chen
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China,*Correspondence: Hongbo Chen
| | - Youhua Xu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, China,Youhua Xu
| | - Dingkun Gui
- Department of Nephrology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,Department of Central Laboratory, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,Dingkun Gui
| |
Collapse
|
6
|
Iskander A, Yan LJ. Cisplatin-Induced Kidney Toxicity: Potential Roles of Major NAD +-Dependent Enzymes and Plant-Derived Natural Products. Biomolecules 2022; 12:1078. [PMID: 36008971 PMCID: PMC9405866 DOI: 10.3390/biom12081078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/25/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022] Open
Abstract
Cisplatin is an FDA approved anti-cancer drug that is widely used for the treatment of a variety of solid tumors. However, the severe adverse effects of cisplatin, particularly kidney toxicity, restrict its clinical and medication applications. The major mechanisms of cisplatin-induced renal toxicity involve oxidative stress, inflammation, and renal fibrosis, which are covered in this short review. In particular, we review the underlying mechanisms of cisplatin kidney injury in the context of NAD+-dependent redox enzymes including mitochondrial complex I, NAD kinase, CD38, sirtuins, poly-ADP ribosylase polymerase, and nicotinamide nucleotide transhydrogenase (NNT) and their potential contributing roles in the amelioration of cisplatin-induced kidney injury conferred by natural products derived from plants. We also cover general procedures used to create animal models of cisplatin-induced kidney injury involving mice and rats. We highlight the fact that more studies will be needed to dissect the role of each NAD+-dependent redox enzyme and its involvement in modulating cisplatin-induced kidney injury, in conjunction with intensive research in NAD+ redox biology and the protective effects of natural products against cisplatin-induced kidney injury.
Collapse
Affiliation(s)
| | - Liang-Jun Yan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
7
|
Kim JS, Han YK, Kong MJ, Park KM. Short-term control of diet affects cisplatin-induced acute kidney injury through modulation of mitochondrial dynamics and mitochondrial GSH. Physiol Rep 2022; 10:e15348. [PMID: 35748040 PMCID: PMC9226808 DOI: 10.14814/phy2.15348] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 05/22/2022] [Indexed: 04/21/2023] Open
Abstract
Obesity affects acute kidney injury (AKI) induced by various clinical settings, including transplantation and cisplatin-cancer therapy. However, the effect of short-term food intake change remains to be defined. Here, we investigated the effects of short-term high-fat diet intake and food restriction on cisplatin-induced AKI. Mice were fed either a high-fat diet (HFD) or a low-fat diet (LFD) for 11 days or were not fed for 40 hh (fasting), before cisplatin administration. Cisplatin-induced functional and structural damages to kidneys in both HFD- and LFD-fed mice, with greater damages in HFD-fed mice than LFD-fed mice. HFD decreased mitochondrial total glutathione (tGSH) level, along with increases in the plasma and kidney cholesterol levels. Cisplatin caused the increase of kidney cholesterol levels and oxidative stress, along with the decrease of mitochondrial tGSH levels. In addition, cisplatin-induced mitochondrial damage and apoptosis of tubular cells in both HFD- and LFD-fed mice. An increase of Fis1 (mitochondria fission 1 protein), whereas a decrease of Opa1 (mitochondria fusion 1 protein) occurred by cisplatin. These cisplatin effects were greater in HFD-fed mice than in LFD-fed mice. Administration of mitochondria-specific antioxidant treatment during HFD feeding inhibited these cisplatin-induced changes. Fasting for 40 h also significantly reduced the cisplatin-induced changes mentioned above. These data demonstrate that short-term HFD intake worsens cisplatin-induced oxidative stress by the reduction of mitochondrial tGSH, resulting in increased cisplatin-induced nephrotoxicity. These data newly indicate that the control of calorie intake, even for a short period, affects kidney susceptibility to injury. Although most studies described the effects of a long-term high-fat diet on the kidneys, in this study, we found that even if a high-fat diet was consumed for a short-term, physiological changes and mitochondria tGSH decrease in the kidneys, and consequently increased cisplatin-nephrotoxic susceptibility. These data suggest the association of calorie intake with kidney susceptibility to cisplatin.
Collapse
Affiliation(s)
- Ji Su Kim
- Department of Anatomy and BK21 PlusSchool of Medicine, Kyungpook National UniversityDaeguRepublic of Korea
| | - Yong Kwon Han
- Department of Anatomy and BK21 PlusSchool of Medicine, Kyungpook National UniversityDaeguRepublic of Korea
| | - Min Jung Kong
- Department of Anatomy and BK21 PlusSchool of Medicine, Kyungpook National UniversityDaeguRepublic of Korea
- Cardiovascular Research Institute, Kyungpook National UniversityDaeguRepublic of Korea
| | - Kwon Moo Park
- Department of Anatomy and BK21 PlusSchool of Medicine, Kyungpook National UniversityDaeguRepublic of Korea
- Cardiovascular Research Institute, Kyungpook National UniversityDaeguRepublic of Korea
| |
Collapse
|
8
|
The mature fruit of Lycium chinense Miller attenuates cisplatin-induced renal damage in mice via regulation of oxidant and antioxidant enzymes. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
9
|
Abouzed TK, Sherif EAE, Barakat MES, Sadek KM, Aldhahrani A, Nasr NE, Eldomany E, Khailo K, Dorghamm DA. Assessment of gentamicin and cisplatin-induced kidney damage mediated via necrotic and apoptosis genes in albino rats. BMC Vet Res 2021; 17:350. [PMID: 34784920 PMCID: PMC8594120 DOI: 10.1186/s12917-021-03023-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 09/15/2021] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Gentamicin (GM) is a low-cost, low-resistance antibiotic commonly used to treat gram-negative bacterial diseases. Cisplatin (Csp) is a platinum-derived anti-neoplastic agent. This experiment aimed to identify the early signs of gentamicin and cisplatin-induced nephrotoxicity in rats. Thirty Wistar rats were divided into three groups of 10: a control group, which received no treatment; a gentamicin group administered by a dose of (100 mg/kg, IP) for 7 consecutive days, and a cisplatin group was administered intraperitoneal in a dose of (1.5 mg/kg body weight) repeated twice a week for 3 weeks. RESULTS Both experimental groups exhibited increased levels of creatinine, urea, and uric acid, with the cisplatin-treated group showing higher levels than the gentamicin group. Experimental groups also exhibited significantly increased Malondialdehyde (MDA), reduced glutathione (GSH), and glutathione peroxidase (GSH-Px) with more pronounced effects in the cisplatin-treated group. Further, both experimental groups exhibited significant up-regulation of Tumor Necrosis Factor α (TNF-α), caspase-3, and Bax and down regulation of Bcl-2. CONCLUSION These findings confirm the use of necrotic, apoptotic genes as early biomarkers in the detection of tubular kidney damage. Further, cisplatin was shown to have a greater nephrotoxic effect than gentamicin; therefore, its use should be constrained accordingly when co-administered with gentamicin.
Collapse
Affiliation(s)
- Tarek Kamal Abouzed
- Biochemistry Department, Faculty of Veterinary Medicine Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Eman Abd Elrahman Sherif
- Biochemistry Department, Faculty of Veterinary Medicine Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Mohamed El Sayed Barakat
- Biochemistry Unit, Animal Health Research Institute, Kafrelsheikh branch. Agricultural Research Center (ARC), Kafrelsheikh, Egypt.
| | - Kadry Mohamed Sadek
- Biochemistry Department, Faculty of Veterinary Medicine Damanhour University, Damanhour, Egypt
| | - Adil Aldhahrani
- Clinical laboratory science Department, Turabah University College, Taif University, Taif, Saudi Arabia
| | - Nasr Elsayed Nasr
- Biochemistry Department, Faculty of Veterinary Medicine Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Ehab Eldomany
- Department of Biotechnology and Life science, Faculty of Postgraduate Studies for Advanced Science Beni-suef University, Beni-suef, Egypt
| | - Khaled Khailo
- Biochemistry Department, Faculty of Veterinary Medicine Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Doaa Abdallha Dorghamm
- Biochemistry Department, Faculty of Veterinary Medicine Kafrelsheikh University, Kafrelsheikh, Egypt
| |
Collapse
|
10
|
Abstract
Cisplatin has been a mainstay of cancer chemotherapy since the 1970s. Despite its broad anticancer potential, its clinical use has regularly been constrained by kidney toxicities. This review details those biochemical pathways and metabolic conversions that underlie the kidney toxicities. A wide range of redox events contribute to the eventual physiological consequences of drug activities.
Collapse
|
11
|
FEN1 Blockade for Platinum Chemo-Sensitization and Synthetic Lethality in Epithelial Ovarian Cancers. Cancers (Basel) 2021; 13:cancers13081866. [PMID: 33919707 PMCID: PMC8070745 DOI: 10.3390/cancers13081866] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/18/2021] [Accepted: 04/05/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Overall survival outcomes, despite platinum-based chemotherapy, for patients with advanced ovarian cancer remains poor. Increased DNA repair capacity is a key route to platinum resistance in ovarian cancer. In the current study, we show that FEN1, a key player in DNA repair, is overexpressed in ovarian cancer and associated with poor survival. Pre-clinically FEN1 blockade not only increased platinum sensitivity but was also synthetically lethal in BRCA2 and POLβ deficient ovarian cancer cells. Together the data provides evidence that FEN1 is a promising anti-cancer target in ovarian cancer. Abstract FEN1 plays critical roles in long patch base excision repair (LP-BER), Okazaki fragment maturation, and rescue of stalled replication forks. In a clinical cohort, FEN1 overexpression is associated with aggressive phenotype and poor progression-free survival after platinum chemotherapy. Pre-clinically, FEN1 is induced upon cisplatin treatment, and nuclear translocation of FEN1 is dependent on physical interaction with importin β. FEN1 depletion, gene inactivation, or inhibition re-sensitizes platinum-resistant ovarian cancer cells to cisplatin. BRCA2 deficient cells exhibited synthetic lethality upon treatment with a FEN1 inhibitor. FEN1 inhibitor-resistant PEO1R cells were generated, and these reactivated BRCA2 and overexpressed the key repair proteins, POLβ and XRCC1. FEN1i treatment was selectively toxic to POLβ deficient but not XRCC1 deficient ovarian cancer cells. High throughput screening of 391,275 compounds identified several FEN1 inhibitor hits that are suitable for further drug development. We conclude that FEN1 is a valid target for ovarian cancer therapy.
Collapse
|
12
|
Abdel-Daim MM, Mahmoud OM, Al Badawi MH, Alghamdi J, Alkahtani S, Salem NA. Protective effects of Citrus limonia oil against cisplatin-induced nephrotoxicity. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:41540-41550. [PMID: 32691312 DOI: 10.1007/s11356-020-10066-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/08/2020] [Indexed: 06/11/2023]
Abstract
Cisplatin is broadly used in the treatment of malignancies. However, the high incidence of nephrotoxicity following cisplatin use deters its clinical utility. Former studies have shown that the essential oils, obtained from Citrus limonia demonstrated significant anti-inflammatory and antioxidant effects. The aim of the current work was to evaluate the protective effects of Citrus limonia oil against cisplatin-induced nephrotoxicity. Thirty-two adult male mice were divided into four groups, eight mice each. The control group received distilled water, and the second group received a single intraperitoneal injection of cisplatin (20 mg/kg), while the third and fourth groups received cisplatin plus Citrus limonia oil at 100 or 200 mg/kg for 10 days, respectively. GC-MS analysis showed that the major components in Citrus limonia oil were D-limonene, 5-methyl-pentadecane, (n)-menthol, 3,7-dimethyl-(E)-2,6-octadienal, 3,7-dimethyl-2,6-octadienal, and nonadecane. Biochemical analysis showed that cisplatin intoxication was associated with significantly increased (p < 0.05) serum levels of urea and creatine and pro-inflammatory cytokines, as well as augmented renal tissue oxidative stress. Light microscopic examination showed loss of renal architecture, atrophied glomeruli, interstitial hemorrhage, dilated cortical tubules with cast formation, and excessive collagen production. Electron microscopic examination revealed compressed and karyorrhectic endothelial nuclei with chromatin condensation in the glomeruli, accumulation of mesangial matrix, and obliteration of glomerular blood capillaries. Co-administration of Citrus limonia oil attenuated these effects in renal histopathological, morphometric, and ultrastructural examinations, frequently in a dose-dependent manner. In conclusion, Citrus limonia oil can ameliorate the toxic effect of cisplatin on mice kidneys, probably through its antioxidant and anti-inflammatory effects.
Collapse
Affiliation(s)
- Mohamed M Abdel-Daim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia.
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt.
| | - Omayma M Mahmoud
- Anatomy and Embryology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Manal H Al Badawi
- Anatomy and Embryology Department, Faculty of Medicine, Helwan University, Helwan, Egypt
| | - Jawahir Alghamdi
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Saad Alkahtani
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Noha A Salem
- Anatomy and Embryology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
13
|
Abd El-Kader M, Taha RI. Comparative nephroprotective effects of curcumin and etoricoxib against cisplatin-induced acute kidney injury in rats. Acta Histochem 2020; 122:151534. [PMID: 32151374 DOI: 10.1016/j.acthis.2020.151534] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Although cisplatin (CIS) acts as potent chemotherapy, nephrotoxicity still its major life-threatening side effect. The purpose of this study was to discuss and compare the renoprotective effects of curcumin (CUR) and etoricoxib (ETB) against CIS-induced nephrotoxicity. MATERIALS & METHODS Thirty six adult female rats were divided equally into 6 groups: Group I (control), Group II (CIS) received cisplatin (7.5 mg/kg i.p), Group III (CUR) and group IV (ETB) received curcumin (200 mg/kg/day) or etoricoxib (10 mg/kg/day) respectively via gavage for seven continuous days. Group V (CIS + CUR) and Group VI (CIS + ETB) received curcumin (200 mg/kg/day) or etoricoxib (10 mg/kg/day) via gavage for seven continuous days. On the 4th day, the rats received cisplatin (7.5 mg/kg i.p) as a single injection 1 h after last curcumin or etoricoxib administration. At the assigned time, blood and tissue samples were collected for biochemical, histochemical, histopathological, immunohistochemical, and RT-PCR gene expression studies. RESULTS Curcumin administration significantly decreased CIS-induced elevation of serum creatinine and blood urea nitrogen (BUN), and reversed oxidative stress markers; glutathione (GSH) and malondialdehyde (MDA) to control level. Suppression of inflammatory and apoptotic responses by CUR co-treatment was evidenced by decreased iNOS and BAX immunohistochemical reactions, and TNF-α and Caspase3 gene expressions which were detected by RT-PCR in kidney tissues. To our knowledge, this is the first time to discuss the effect of ETB on CIS induced nephrotoxicity. Although ETB reduced the previously mentioned inflammatory and apoptotic markers, its effect was less than that of CUR. Administration of ETB couldn't modify the disturbed levels of creatinine, BUN, GSH, and MDA. CONCLUSION In conclusion, CUR provided a promising renoprotective effect against CIS induced nephrotoxicity. Further studies are recommended to approve or disapprove the protective role of ETB in CIS induced nephrotoxicity.
Collapse
Affiliation(s)
- Marwa Abd El-Kader
- Anatomy and Embryology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Reham Ismail Taha
- Anatomy and Embryology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| |
Collapse
|
14
|
Sudo S, Kajiya H, Okano S, Sasaki M, Katsumata Y, Ohno J, Ikebe T, Hiraki A, Okabe K. Cisplatin-induced programmed cell death ligand-2 expression is associated with metastasis ability in oral squamous cell carcinoma. Cancer Sci 2020; 111:1113-1123. [PMID: 32012401 PMCID: PMC7156784 DOI: 10.1111/cas.14336] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/20/2020] [Accepted: 01/24/2020] [Indexed: 12/12/2022] Open
Abstract
Programmed cell death ligands (PD‐Ls) are expressed in tumor cells where they bind to programmed cell death‐1, an immunocyte co–receptor, resulting in tumor cell evasion from the immune system. Chemotherapeutic drugs have been recently reported to induce the expression of PD‐L, such as PD‐L1, in some cancer cells. However, little is known regarding PD‐L2 expression and its role in oral squamous cell carcinoma (OSCC). In this study, we examined the effect of cisplatin on the expression and regulation of PD‐L2 in OSCC cell lines and analyzed malignant behavior in PD‐L2‐expressing cells using colony, transwell and transformation assays. In addition, we examined PD‐L2 expression in the tumor tissues of OSCC patients using cytology and tissue microarray methods. In OSCC cell lines, cisplatin treatment upregulated PD‐L2 expression, along with that of the drug efflux transporter ABCG2, via signal transducers and activator of transcription (STAT) 1/3 activation. Moreover, PD‐L2‐positive or PD‐L2‐overexpressing cells demonstrated upregulation in both invasion and transformation ability but not in proliferation compared with PD‐L2‐negative or PD‐L2‐silencing cells. PD‐L2 expression was also observed in OSCC cells of cytology samples and tissue from OSCC patients. The intensity of PD‐L2 expression was correlated with more malignant morphological features in the histological appearance and an invasive pattern. Our findings indicate that cisplatin‐upregulated PD‐L2 expression in OSCC via STAT1/3 activation and the expression of PD‐L2 are likely to be associated with malignancy in OSCC. The PD‐L2 expression in cisplatin‐resistant OSCC cells may be a critical factor in prognosis of advanced OSCC patients.
Collapse
Affiliation(s)
- Shunichi Sudo
- Department of Physiological Science and Molecular Biology, Fukuoka Dental College, Fukuoka, Japan.,Department of Oral Maxillofacial Surgery, Fukuoka Dental College, Fukuoka, Japan
| | - Hiroshi Kajiya
- Department of Physiological Science and Molecular Biology, Fukuoka Dental College, Fukuoka, Japan.,Research Center for Regenerative Medicine, Fukuoka Dental College, Fukuoka, Japan
| | - Shinji Okano
- Department of Morphological Biology, Fukuoka Dental College, Fukuoka, Japan
| | - Mina Sasaki
- Department of Oral Maxillofacial Surgery, Fukuoka Dental College, Fukuoka, Japan
| | - Yuri Katsumata
- Department of Oral Maxillofacial Surgery, Fukuoka Dental College, Fukuoka, Japan
| | - Jun Ohno
- Research Center for Regenerative Medicine, Fukuoka Dental College, Fukuoka, Japan
| | - Tetsuro Ikebe
- Department of Oral Maxillofacial Surgery, Fukuoka Dental College, Fukuoka, Japan
| | - Akimitsu Hiraki
- Department of Oral Maxillofacial Surgery, Fukuoka Dental College, Fukuoka, Japan
| | - Koji Okabe
- Department of Physiological Science and Molecular Biology, Fukuoka Dental College, Fukuoka, Japan
| |
Collapse
|
15
|
Cheng P, Liu H, Li Y, Pi P, Jiang Y, Zang S, Li X, Fu A, Ren X, Xu J, Holmgren A, Lu J. Inhibition of thioredoxin reductase 1 correlates with platinum-based chemotherapeutic induced tissue injury. Biochem Pharmacol 2020; 175:113873. [PMID: 32092292 DOI: 10.1016/j.bcp.2020.113873] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/20/2020] [Indexed: 12/14/2022]
Abstract
Platinum-containing drugs (PtDs; e.g. cisplatin, carboplatin, and oxaliplatin) have been widely used as anticancer reagents against various cancers. However, treatment with these drugs results in undesirable adverse effects with unknown mechanisms. Herein, we found a strong correlation between the inhibitory effects of PtDs on cytosolic thioredoxin reductase (TXNRD1) and tissue injury. Of the PtDs tested, cisplatin was found to be the most effective inhibitory PtD against TXRND1, causing the severest kidney injury. The initial inhibition of TXNRD1 in the kidney resulted from cisplatin-induced transcriptional activation of Nrf2-regulated genes including Txnrd1. However, the antioxidant responses in the kidney did not reverse the cisplatin-induced oxidation process. Nephrotoxicity was accompanied with an increase of protein glutathionylation and a cellular thiol redox environment oxidation. These results suggest that the changes of the cellular thiol-dependent redox environment regulated by TXNRD1 is a major event in the adverse effects of cisplatin in kidney.
Collapse
Affiliation(s)
- Ping Cheng
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Huan Liu
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Yinchuan Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Peiling Pi
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Yu Jiang
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Shaozhen Zang
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Xiaorong Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Ailing Fu
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Xiaoyuan Ren
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institute, SE-171 77 Stockholm, Sweden
| | - Jianqiang Xu
- School of Life and Pharmaceutical Sciences & Panjin Institute of Industrial Technology, Dalian University of Technology, Panjin 124221, China
| | - Arne Holmgren
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institute, SE-171 77 Stockholm, Sweden
| | - Jun Lu
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China.
| |
Collapse
|
16
|
Links between cancer metabolism and cisplatin resistance. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 354:107-164. [PMID: 32475471 DOI: 10.1016/bs.ircmb.2020.01.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cisplatin is one of the most potent and widely used chemotherapeutic agent in the treatment of several solid tumors, despite the high toxicity and the frequent relapse of patients due to the onset of drug resistance. Resistance to chemotherapeutic agents, either intrinsic or acquired, is currently one of the major problems in oncology. Thus, understanding the biology of chemoresistance is fundamental in order to overcome this challenge and to improve the survival rate of patients. Studies over the last 30 decades have underlined how resistance is a multifactorial phenomenon not yet completely understood. Recently, tumor metabolism has gained a lot of interest in the context of chemoresistance; accumulating evidence suggests that the rearrangements of the principal metabolic pathways within cells, contributes to the sensitivity of tumor to the drug treatment. In this review, the principal metabolic alterations associated with cisplatin resistance are highlighted. Improving the knowledge of the influence of metabolism on cisplatin response is fundamental to identify new possible metabolic targets useful for combinatory treatments, in order to overcome cisplatin resistance.
Collapse
|
17
|
Citrus aurantium Ameliorates Cisplatin-Induced Nephrotoxicity. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3960908. [PMID: 31886208 PMCID: PMC6925785 DOI: 10.1155/2019/3960908] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 11/11/2019] [Indexed: 01/03/2023]
Abstract
We aimed to study the effects of Citrus aurantium (C. aurantium) on renal functions in cisplatin-induced nephrotoxicity in rats. The study involved male Wistar rats weighing 250–300 g that were fed and kept under standard conditions. Rats were randomly divided into control, cisplatin administered, C. aurantium 200 mg/kg, and C. aurantium 400 mg/kg groups. Cisplatin was administered at 5 mg/kg i.p. once at the start of study to induce nephrotoxicity. Blood and urine samples were obtained at alternative days for analysis. The body weight and urine output were monitored at regular intervals. Plasma and urinary sodium, potassium, and creatinine levels were measured at the end of study duration. Absolute excretion of sodium and potassium; sodium to potassium ratio; kidney weights; fractional excretion of sodium and potassium; and absolute creatinine clearance were determined to analyze the effects of C. aurantium. Histopathological changes of kidney tissues were studied to determine relevant effects. The results indicate that cisplatin lowered the total body weights while raising the urinary output and kidney weights, reversed by C. aurantium both dose and time dependently. Similarly, C. aurantium markedly normalized plasma, urinary sodium, potassium, and creatinine levels. Cisplatin-induced absolute sodium clearance, absolute potassium clearance, absolute creatinine clearance, sodium to potassium ratio, and fractional excretion of sodium and potassium were significantly reversed by C. aurantium. Histopathological analysis showed notable improvement in C. aurantium administered groups as compared to cisplatin-induced group. Study suggests that C. aurantium possesses excellent nephroprotective effects against cisplatin-induced toxicity.
Collapse
|
18
|
Li Q, Liang X, Yang Y, Zeng X, Zhong X, Huang C. Panax notoginseng saponins ameliorate cisplatin-induced mitochondrial injury via the HIF-1α/mitochondria/ROS pathway. FEBS Open Bio 2019; 10:118-126. [PMID: 31715069 PMCID: PMC6943232 DOI: 10.1002/2211-5463.12760] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/26/2019] [Accepted: 11/12/2019] [Indexed: 01/15/2023] Open
Abstract
Cisplatin is a major antineoplastic drug that is used to treat solid tumors, but its use is restricted by its nephrotoxicity. Such cisplatin‐induced nephrotoxicity (CIN) is believed to occur primarily through mitochondrial damage and reactive oxygen species (ROS) generation. Our previous studies have indicated that Panax notoginseng saponins (PNSs) mitigate CIN by enhancing hypoxia‐inducible factor 1α (HIF‐1α)‐induced mitochondrial autophagy. In this study, the role of the HIF‐1α/mitochondria/ROS pathway in PNSs protection against CIN was investigated using a rat model. A CIN model was generated by giving rats intraperitoneal injections with cisplatin (a single dose) and then treating them with or without 2‐methoxyestradiol (HIF‐1α inhibitor) and PNSs. We then measured ROS levels, superoxide dismutase, glutathione, catalase malondialdehyde and nitric oxide (to evaluate oxidative stress) and ATP, mitochondrial membrane potential and mitochondrial permeability transition pore opening (to evaluate mitochondrial function) in kidneys at different time points. We observed that PNSs remarkably reduced the levels of ROS, malondialdehyde and nitric oxide, as well as the opening of mitochondrial permeability transition pore, which is increased by cisplatin and further increased by HIF‐1α inhibition. In addition, PNSs increased the levels of superoxide dismutase, catalase and glutathione, as well as ATP and mitochondrial membrane potential in renal tissues; these are all reduced by cisplatin and further reduced by HIF‐1α inhibition. In conclusion, we demonstrate here that PNSs protects against mitochondrial damage induced by cisplatin through HIF‐1α/mitochondria/ROS.
Collapse
Affiliation(s)
- Qingqing Li
- Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xueyan Liang
- Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yufang Yang
- Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xian Zeng
- Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaobin Zhong
- Regenerative Medicine Research Center of Guangxi Medical University, Nanning, China
| | - Chun Huang
- Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
19
|
Effect of Ficus carica L. leaves extract loaded gold nanoparticles against cisplatin-induced acute kidney injury. Colloids Surf B Biointerfaces 2019; 184:110465. [DOI: 10.1016/j.colsurfb.2019.110465] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 08/22/2019] [Accepted: 08/27/2019] [Indexed: 01/21/2023]
|
20
|
Potential use of edaravone to reduce specific side effects of chemo-, radio- and immuno-therapy of cancers. Int Immunopharmacol 2019; 77:105967. [DOI: 10.1016/j.intimp.2019.105967] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 09/30/2019] [Accepted: 10/07/2019] [Indexed: 02/06/2023]
|
21
|
Gokduman K. Sensitization of cisplatin-resistant ovarian cancer cells by magnetite iron oxide nanoparticles: an in vitro study. Nanomedicine (Lond) 2019; 14:3177-3191. [PMID: 31724481 DOI: 10.2217/nnm-2019-0126] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aim: To investigate potential of magnetite iron oxide nanoparticles (MION) to sensitize cisplatin-resistant ovarian cancer cells to cisplatin, which to the best of found knowledge has not been reported previously. Materials & methods: MION with a diameter of approximately 20 nm were synthesized, and characterized using Fourier transform infrared spectroscopy, powder x-ray diffraction and particle size analyzer. Results: The synthesized MION have increased reactive oxygen species levels and decreased glutathione levels in cisplatin-resistant ovarian cancer cells (OVCAR-3 and SKOV-3). Using MTT, capsase-3 activity and live/dead assays, capability of the synthesized MION to sensitize cisplatin-resistant ovarian cancer cells has been illustrated. Conclusion: Thus, for further investigations, the synthesized MION can be considered as a potent agent enabling much more effective cisplatin-based therapies for ovarian cancer.
Collapse
Affiliation(s)
- Kurtulus Gokduman
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.,Institute of Biomedical Engineering, Bogazici University, Istanbul 34684, Turkey
| |
Collapse
|
22
|
Sirt5 Attenuates Cisplatin-Induced Acute Kidney Injury through Regulation of Nrf2/HO-1 and Bcl-2. BIOMED RESEARCH INTERNATIONAL 2019; 2019:4745132. [PMID: 31815138 PMCID: PMC6878818 DOI: 10.1155/2019/4745132] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 08/11/2019] [Accepted: 08/29/2019] [Indexed: 12/30/2022]
Abstract
Cisplatin- (CDDP) induced acute kidney injury (AKI) limits the clinical use of cisplatin. Several sirtuin (SIRT) family proteins are involved in AKI, while the roles of Sirt5 in cisplatin-induced AKI remain unknown. In the present study, we characterized the role and mechanism of Sirt5 in cisplatin-induced apoptosis using the human kidney 2 (HK-2) cell line. CDDP treatment decreased Sirt5 expression of HK-2 cells in a dose-dependent manner. In addition, Sirt5 overexpression enhanced the metabolic activity in CDDP-treated HK-2 cells while Sirt5 siRNA attenuated it. Forced expression of Sirt5 inhibited CDDP-induced apoptosis while Sirt5 siRNA showed the opposite effects. Accordingly, Sirt5 overexpression inhibited the level of caspase 3 cleavage and cytochrome c levels. Furthermore, we found that Sirt5 increased mitochondrial membrane potentials and ameliorated intracellular ROS production. Mitotracker Red staining indicated that Sirt5 overexpression was able to maintain the mitochondrial density during CDDP treatment. We also investigated possible downstream targets of Sirt5 and found that Sirt5 increased Nrf2, HO-1, and Bcl-2 while it decreased Bax protein expression. Sirt5 siRNA showed the opposite effect on these proteins. The levels of Nrf2, HO-1, and Bcl-2 proteins in HK-2 cells were also decreased after CDDP treatment. Moreover, Nrf2 and Bcl-2 siRNA partly abolished the protecting effect of Sirt5 on CDDP-induced apoptosis and cytochrome c release. Catalase inhibitor 3-AT also abolished the cytoprotective effect of Sirt5. Together, the results demonstrated that Sirt5 attenuated cisplatin-induced apoptosis and mitochondrial injury in human kidney HK-2 cells, possibly through the regulation of Nrf2/HO-1 and Bcl-2.
Collapse
|
23
|
The Predictive Role of the Biomarker Kidney Molecule-1 (KIM-1) in Acute Kidney Injury (AKI) Cisplatin-Induced Nephrotoxicity. Int J Mol Sci 2019; 20:ijms20205238. [PMID: 31652595 PMCID: PMC6834366 DOI: 10.3390/ijms20205238] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/09/2019] [Accepted: 10/17/2019] [Indexed: 12/14/2022] Open
Abstract
Acute kidney injury (AKI) following platinum-based chemotherapeutics is a frequently reported serious side-effect. However, there are no approved biomarkers that can properly identify proximal tubular injury while routine assessments such as serum creatinine lack sensitivity. Kidney-injury-molecule 1 (KIM-1) is showing promise in identifying cisplatin-induced renal injury both in vitro and in vivo studies. In this review, we focus on describing the mechanisms of renal tubular cells cisplatin-induced apoptosis, the associated inflammatory response and oxidative stress and the role of KIM-1 as a possible biomarker used to predict cisplatin associated AKI.
Collapse
|
24
|
Li X, Wang Q, Deng G, Liu Y, Wei B, Liu X, Bao W, Wang Q, Wu S. Porous Se@SiO2 nanospheres attenuate cisplatin-induced acute kidney injury via activation of Sirt1. Toxicol Appl Pharmacol 2019; 380:114704. [DOI: 10.1016/j.taap.2019.114704] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 08/03/2019] [Accepted: 08/06/2019] [Indexed: 12/16/2022]
|
25
|
Boran T, Gunaydin A, Jannuzzi AT, Ozcagli E, Alpertunga B. Celastrol pretreatment as a therapeutic option against cisplatin-induced nephrotoxicity. Toxicol Res (Camb) 2019; 8:723-730. [PMID: 31588349 PMCID: PMC6762010 DOI: 10.1039/c9tx00141g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 07/29/2019] [Indexed: 12/31/2022] Open
Abstract
Celastrol is a natural bioactive compound extracted from the medicinal plant Tripterygium wilfordii Hook F. It exhibits immunosuppressive, anti-inflammatory, and antioxidant activities. Cisplatin is a commonly used chemotherapeutic drug in the treatment of a wide range of tumors. Although very effective therapeutically, it can cause nephrotoxicity leading to dose reduction or discontinuation of treatment. This study aims to clarify the therapeutic potential of celastrol in cisplatin-induced nephrotoxicity. The possible protective effects of celastrol pretreatment against cisplatin-induced oxidative stress and genotoxicity were investigated. A rat kidney epithelial cell line NRK-52E was pretreated with the desired concentrations of celastrol (200 nM, 100 nM, and 50 nM) for 24 h. The cells were treated with 50 μM cisplatin for a further 24 h to see whether cisplatin caused the same or less toxicity compared to the vehicle control group. Alkaline comet assay was performed for genotoxicity assessment. Genotoxicity evaluation revealed that celastrol caused a statistically significant reduction in DNA damage. Oxidative stress parameters were evaluated by measuring the glutathione (GSH) and protein carbonyl (PC) levels and also by measuring the enzyme activities of glutathione peroxidase (GPx), glutathione reductase (GR), catalase (CAT) and superoxide dismutase (SOD) enzymes. Celastrol pretreatment increased the GSH content of the cells and ameliorated the protein carbonylation level. Likewise, celastrol pretreatment improved the GR and CAT activities. However, no significant difference was observed in GPx and SOD activities. In the light of these findings, celastrol treatment could be a therapeutic option to reduce cisplatin-induced nephrotoxicity. Further studies are needed for the clarification of its therapeutic potential.
Collapse
Affiliation(s)
- Tugce Boran
- Istanbul University , Faculty of Pharmacy , Department of Pharmaceutical Toxicology , 34116 , Beyazıt , Istanbul , Turkey . ; ; Tel: +902124400000
| | - Aysenur Gunaydin
- Istanbul University , Faculty of Pharmacy , Department of Pharmaceutical Toxicology , 34116 , Beyazıt , Istanbul , Turkey . ; ; Tel: +902124400000
- Bezmialem Vakif University , Faculty of Pharmacy , Department of Pharmaceutical Toxicology , Vatan Street , 34093 , Fatih , Istanbul , Turkey
| | - Ayse Tarbin Jannuzzi
- Istanbul University , Faculty of Pharmacy , Department of Pharmaceutical Toxicology , 34116 , Beyazıt , Istanbul , Turkey . ; ; Tel: +902124400000
| | - Eren Ozcagli
- Istanbul University , Faculty of Pharmacy , Department of Pharmaceutical Toxicology , 34116 , Beyazıt , Istanbul , Turkey . ; ; Tel: +902124400000
| | - Buket Alpertunga
- Istanbul University , Faculty of Pharmacy , Department of Pharmaceutical Toxicology , 34116 , Beyazıt , Istanbul , Turkey . ; ; Tel: +902124400000
| |
Collapse
|
26
|
Genotoxic stress-triggered β-catenin/JDP2/PRMT5 complex facilitates reestablishing glutathione homeostasis. Nat Commun 2019; 10:3761. [PMID: 31434880 PMCID: PMC6704105 DOI: 10.1038/s41467-019-11696-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 07/30/2019] [Indexed: 01/17/2023] Open
Abstract
The mechanisms underlying how cells subjected to genotoxic stress reestablish reduction-oxidation (redox) homeostasis to scavenge genotoxic stress-induced reactive oxygen species (ROS), which maintains the physiological function of cellular processes and cell survival, remain unclear. Herein, we report that, via a TCF-independent mechanism, genotoxic stress induces the enrichment of β-catenin in chromatin, where it forms a complex with ATM phosphorylated-JDP2 and PRMT5. This elicits histone H3R2me1/H3R2me2s-induced transcriptional activation by the recruitment of the WDR5/MLL methyltransferase complexes and concomitant H3K4 methylation at the promoters of multiple genes in GSH-metabolic cascade. Treatment with OICR-9429, a small-molecule antagonist of the WDR5-MLL interaction, inhibits the β-catenin/JDP2/PRMT5 complex-reestablished GSH metabolism, leading to a lethal increase in the already-elevated levels of ROS in the genotoxic-agent treated cancer cells. Therefore, our results unveil a plausible role for β-catenin in reestablishing redox homeostasis upon genotoxic stress and shed light on the mechanisms of inducible chemotherapy resistance in cancer. It is known that genotoxic stress induces high levels of ROS and deplete cellular glutathione stores. Here, Cao et al. uncover a β-catenin-dependent TCF/LEF-independent mechanism that promotes histone-mediated transcriptional activation of glutathione synthesis.
Collapse
|
27
|
Qi L, Luo Q, Zhang Y, Jia F, Zhao Y, Wang F. Advances in Toxicological Research of the Anticancer Drug Cisplatin. Chem Res Toxicol 2019; 32:1469-1486. [PMID: 31353895 DOI: 10.1021/acs.chemrestox.9b00204] [Citation(s) in RCA: 247] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cisplatin is one of the most widely used chemotherapeutic agents for various solid tumors in the clinic due to its high efficacy and broad spectrum. The antineoplastic activity of cisplatin is mainly due to its ability to cross-link with DNA, thus blocking transcription and replication. Unfortunately, the clinical use of cisplatin is limited by its severe, dose-dependent toxic side effects. There are approximately 40 specific toxicities of cisplatin, among which nephrotoxicity is the most common one. Other common side effects include ototoxicity, neurotoxicity, gastrointestinal toxicity, hematological toxicity, cardiotoxicity, and hepatotoxicity. These side effects together reduce the life quality of patients and require lowering the dosage of the drug, even stopping administration, thus weakening the treatment effect. Few effective measures exist clinically against these side effects because the exact mechanisms of various side effects from cisplatin remain still unclear. Therefore, substantial effort has been made to explore the complicated biochemical processes involved in the toxicology of cisplatin, aiming to identify effective ways to reduce or eradicate its toxicity. This review summarizes and reviews the updated advances in the toxicological research of cisplatin. We anticipate to provide insights into the understanding of the mechanisms underlying the side effects of cisplatin and designing comprehensive therapeutic strategies involving cisplatin.
Collapse
Affiliation(s)
- Luyu Qi
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China.,University of Chinese Academy of Sciences , Beijing 100049 , P.R. China
| | - Qun Luo
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China.,University of Chinese Academy of Sciences , Beijing 100049 , P.R. China
| | - Yanyan Zhang
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China
| | - Feifei Jia
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China
| | - Yao Zhao
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China
| | - Fuyi Wang
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China.,University of Chinese Academy of Sciences , Beijing 100049 , P.R. China.,Basic Medical College , Shandong University of Chinese Traditional Medicine , Jinan 250355 , P.R. China
| |
Collapse
|
28
|
Thangapandiyan S, Ramesh M, Miltonprabu S, Hema T, Jothi GB, Nandhini V. Sulforaphane potentially attenuates arsenic-induced nephrotoxicity via the PI3K/Akt/Nrf2 pathway in albino Wistar rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:12247-12263. [PMID: 30835071 PMCID: PMC6476857 DOI: 10.1007/s11356-019-04502-w] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 02/05/2019] [Indexed: 05/31/2023]
Abstract
Oxidative stress plays a significant role in the pathophysiology of numerous kidney diseases, generally mediated by reactive oxygen species (ROS). Arsenic (Ar) is known to exert its toxicity through the generation of ROS and inflammation. The current study investigates the protective effects of sulforaphane (SFN) against arsenic-induced renal damage via PI3K/Akt-mediated Nrf2 pathway signaling. Thirty-two male albino Wistar rats were randomly divided into four groups of eight animals each, designated as control, arsenic (Ar), sulforaphane plus Ar (SFN+Ar), and sulforaphane alone (SFN), with oral administration of Ar (5 mg/kg BW) and SFN (80 mg/kg BW) daily for 28 days. Ar administration significantly (P < 0.05) increased the levels of ROS, OHdG, Ar accumulation, and lipid peroxidation, and decreased levels of enzymatic and nonenzymatic antioxidants. Notably, a significant (P < 0.05) increase was observed in markers of apoptosis, DNA damage, TUNEL-positive cells, and dark staining of ICAM-1 in renal tissue with decreased PI3K/Akt/Nrf2 gene expression. The biochemical findings were supported by histopathological and electron microscopy evaluation, which showed severe renal damage in rats treated with Ar. Pretreatment with SFN significantly (P < 0.05) attenuated renal ROS, OHdG, lipid peroxidation, and DNA damage, and increased phase II antioxidants via PI3K/Akt-mediated Nrf2 activation in renal tissue. These results show that dietary supplementation with SFN protects against Ar-induced nephrotoxicity via the PI3K/Akt-mediated Nrf2 signaling pathway in the rat kidney.
Collapse
Affiliation(s)
- Shanmugam Thangapandiyan
- N-PDF, Unit of Toxicology, Department of Zoology, Bharathiar University, Coimbatore, Tamilnadu, 641064, India.
| | - Mathan Ramesh
- Unit of Toxicology, Department of Zoology, Bharathiar University, Coimbatore, Tamilnadu, 641 046, India
| | | | - Tamilselvan Hema
- Unit of Toxicology, Department of Zoology, Bharathiar University, Coimbatore, Tamilnadu, 641 046, India
| | | | - Venkatesan Nandhini
- Unit of Toxicology, Department of Zoology, Bharathiar University, Coimbatore, Tamilnadu, 641 046, India
| |
Collapse
|
29
|
Renalase attenuates mitochondrial fission in cisplatin-induced acute kidney injury via modulating sirtuin-3. Life Sci 2019; 222:78-87. [DOI: 10.1016/j.lfs.2019.02.042] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/04/2019] [Accepted: 02/20/2019] [Indexed: 02/01/2023]
|
30
|
Chan S, Wang R, Man K, Nicholls J, Li H, Sun H, Chan GCF. A Novel Synthetic Compound, Bismuth Zinc Citrate, Could Potentially Reduce Cisplatin-Induced Toxicity Without Compromising the Anticancer Effect Through Enhanced Expression of Antioxidant Protein. Transl Oncol 2019; 12:788-799. [PMID: 30921749 PMCID: PMC6438849 DOI: 10.1016/j.tranon.2019.02.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 02/14/2019] [Accepted: 02/14/2019] [Indexed: 02/07/2023] Open
Abstract
Cisplatin is a common anticancer drug, but it comes with significant nephrotoxicity. Further cisplatin-induced oxidative stress contributes to the pathogenesis of the nephrotoxicity. A new compound, BiZn, can potentially prevent this complication. We verified our postulation by in vitro and in vivo models. From our findings, BiZn did not affect cisplatin-induced cytotoxicity on neuroblastoma cells under both in vitro and in vivo settings. However, BiZn significantly reduced the blood urea nitrogen and creatinine levels in cisplatin-treated mice. Under the lethal dosage of cisplatin, co-treatment of BiZn significantly increased the survival rate. BiZn stimulated antioxidant proteins metallothionein (MT) and glutathione (GSH) generation from kidney cells and minimized cisplatin-induced apoptosis. Knocking down MT-IIA and inhibiting GSH abolished such protection. In conclusion, pretreatment of BiZn decreased cisplatin-induced renal toxicity without affecting its antitumor activity. BiZn-induced antioxidant proteins MT and GSH may contribute to the renal protection effect.
Collapse
Affiliation(s)
- Shing Chan
- Department of Paediatrics & Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong
| | - Runming Wang
- Department of Chemistry, Faculty of Science, The University of Hong Kong
| | - Kwan Man
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong
| | - John Nicholls
- Department of Pathology, LKS Faculty of Medicine, The University of Hong Kong
| | - Hongyan Li
- Department of Chemistry, Faculty of Science, The University of Hong Kong
| | - Hongzhe Sun
- Department of Chemistry, Faculty of Science, The University of Hong Kong.
| | - Godfrey Chi-Fung Chan
- Department of Paediatrics & Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong.
| |
Collapse
|
31
|
Zhou YD, Hou JG, Yang G, Jiang S, Chen C, Wang Z, Liu YY, Ren S, Li W. Icariin ameliorates cisplatin-induced cytotoxicity in human embryonic kidney 293 cells by suppressing ROS-mediated PI3K/Akt pathway. Biomed Pharmacother 2019; 109:2309-2317. [DOI: 10.1016/j.biopha.2018.11.108] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 11/05/2018] [Accepted: 11/25/2018] [Indexed: 12/12/2022] Open
|
32
|
Wang QW, Jia LY, Shi DL, Wang RF, Lu LN, Xie JJ, Sun K, Feng HQ, Li X. Effects of extracellular ATP on local and systemic responses of bean (Phaseolus vulgaris L) leaves to wounding. Biosci Biotechnol Biochem 2018; 83:417-428. [PMID: 30458666 DOI: 10.1080/09168451.2018.1547623] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Wounding increased the extracellular Adenosine 5'-triphosphate (eATP) level of kidney bean leaves. Treatment with wounding or exogenous ATP increased the hydrogen peroxide (H2O2) content, activities of catalase and polyphenol oxidase, and malondialdehyde content in both the treated and systemic leaves. Pre-treatment with ATP-degrading enzyme, apyrase, to the wounded leaves reduced the wound-induced local and systemic increases in H2O2 content, activities of catalase and polyphenol oxidase, and malondialdehyde content. Application of dimethylthiourea (DMTU) and diphenylene iodonium (DPI) to the wounded and ATP-treated leaves, respectively, reduced the wound- and ATP-induced local and systemic increases in H2O2 content, activities of catalase and polyphenol oxidase, and malondialdehyde content. Moreover, the wound- and ATP-induced systemic increases of these physiological parameters were suppressed when DMTU or DPI applied to leaf petiole of the wounded and ATP-treated leaves. These results suggest that eATP at wounded sites could mediate the wound-induced local and systemic responses by H2O2-dependent signal transduction.
Collapse
Affiliation(s)
- Qing-Wen Wang
- a Department of Biology Science, College of Life Sciences , Northwest Normal University , Lanzhou , China
| | - Lin-Yun Jia
- a Department of Biology Science, College of Life Sciences , Northwest Normal University , Lanzhou , China
| | - Dai-Long Shi
- a Department of Biology Science, College of Life Sciences , Northwest Normal University , Lanzhou , China
| | - Rong-Fang Wang
- b Institute of Chemical Engineering , Qingdao University of Science and Technology , Qingdao , China
| | - Li-Na Lu
- a Department of Biology Science, College of Life Sciences , Northwest Normal University , Lanzhou , China
| | - Jia-Jia Xie
- a Department of Biology Science, College of Life Sciences , Northwest Normal University , Lanzhou , China
| | - Kun Sun
- a Department of Biology Science, College of Life Sciences , Northwest Normal University , Lanzhou , China
| | - Han-Qing Feng
- a Department of Biology Science, College of Life Sciences , Northwest Normal University , Lanzhou , China
| | - Xin Li
- c Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations , Lanzhou University , Lanzhou , China
| |
Collapse
|
33
|
Gómez-Sierra T, Eugenio-Pérez D, Sánchez-Chinchillas A, Pedraza-Chaverri J. Role of food-derived antioxidants against cisplatin induced-nephrotoxicity. Food Chem Toxicol 2018; 120:230-242. [DOI: 10.1016/j.fct.2018.07.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 06/22/2018] [Accepted: 07/06/2018] [Indexed: 12/21/2022]
|
34
|
Mitochondrial NADP +-dependent isocitrate dehydrogenase deficiency increases cisplatin-induced oxidative damage in the kidney tubule cells. Cell Death Dis 2018; 9:488. [PMID: 29695796 PMCID: PMC5916920 DOI: 10.1038/s41419-018-0537-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/22/2018] [Accepted: 03/27/2018] [Indexed: 12/18/2022]
Abstract
Mitochondrial NADP+-dependent isocitrate dehydrogenase (IDH2) plays an important role in the formation of NADPH, which is critical for the maintenance of mitochondrial redox balance. Cis-diamminedichloroplatinum II (cisplatin), an effective anticancer drug, induces oxidative stress-related nephrotoxicity, limiting its use. Therefore, we investigated whether IDH2, which is a critical enzyme in the NADPH-associated mitochondrial antioxidant system, is involved in cisplatin nephrotoxicity. Idh2 gene-deleted (Idh2−/−) mice and wild-type (Idh2+/+) littermates were treated with cisplatin, with or without 2-(2,2,6,6-tetramethylpiperidin-1-oxyl-4-ylamino)-2-oxoethyl) triphenylphosphonium chloride (Mito-T), a mitochondria-specific antioxidant. Cisplatin-induced renal functional and morphological impairments were greater in Idh2−/− mice than in Idh2+/+ mice. Mito-T mitigated those impairments in both Idh2−/− and Idh2+/+ mice and this mitigation was greater in Idh2−/− than in Idh2+/+ mice. Cisplatin impaired IDH2 function in the mitochondria, decreasing mitochondrial NADPH and GSH levels and increasing H2O2 generation; protein, lipid, and DNA oxidation; mitochondrial damage; and apoptosis. These cisplatin-induced changes were much more severe in Idh2−/− mice than in Idh2+/+ mice. Mito-T treatment attenuated cisplatin-induced alterations in both Idh2−/− and Idh2+/+ mice and this mitigation was greater in Idh2−/− than in Idh2+/+ mice. Altogether, these data demonstrate that cisplatin induces the impairment of the mitochondrial IDH2-NADPH-GSH antioxidant system and IDH2 deficiency aggravates cisplatin-induced mitochondrial oxidative damage, inducing more severe nephrotoxicity. This suggests that the mitochondrial IDH2-NADPH-GSH antioxidant system is a target for the prevention of cisplatin-induced kidney cell death.
Collapse
|
35
|
Wang TEJ, Liu HT, Lai YH, Jan TR, Nomura N, Chang HW, Chou CC, Lee YJ, Tsai PSJ. Honokiol, a Polyphenol Natural Compound, Attenuates Cisplatin-Induced Acute Cytotoxicity in Renal Epithelial Cells Through Cellular Oxidative Stress and Cytoskeleton Modulations. Front Pharmacol 2018; 9:357. [PMID: 29755347 PMCID: PMC5932397 DOI: 10.3389/fphar.2018.00357] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 03/27/2018] [Indexed: 12/30/2022] Open
Abstract
Cisplatin is a potent anti-cancer drug that has been widely used in the treatment of various cancers; however, cisplatin administration results in severe nephrotoxicity and impedes its clinical applications. In this study, we showed that honokiol, a polyphenol constituent extracted from Magnolia officinalis exhibited a short-term protective effect against cisplatin-induced damages in renal epithelial cells in vitro. The protective effects of honokiol were resulted from the combination of (1) reduced cellular oxidative stress ranging from 53 to 32% reduction during a 24-h incubation, (2) the maintenance of cellular antioxidant capacity and (3) the stabilization of cytoskeletal structure of the kidney epithelial cells. By promoting the polymerization of actin (1.6-fold increase) and tubulin (1.8-fold increase) cytoskeleton, honokiol not only maintained epithelial cell morphology, but also stabilized cellular localizations of tight junction protein Occludin and adhesion junction protein E-Cadherin. With stabilized junction protein complexes and structural polymerized cytoskeleton network, honokiol preserved epithelial cell polarity and morphology and thus reduced cisplatin-induced cell disruption and damages. Our data demonstrated for the first time that honokiol could counteract with cisplatin-induced damages in renal epithelial cells in vitro, future in vivo studies would further validate the potential clinical application of honokiol in cisplatin-based cancer treatments with reduced nephrotoxicity.
Collapse
Affiliation(s)
- Tse-En J Wang
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Hung-Ting Liu
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Hua Lai
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Tong-Rong Jan
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Naohiro Nomura
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hui-Wen Chang
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Molecular and Comparative Pathobiology, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Chi-Chung Chou
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Ya-Jane Lee
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Veterinary Clinical Science, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Pei-Shiue J Tsai
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
36
|
Tong Y, Huang X, Lu M, Yu BY, Tian J. Prediction of Drug-Induced Nephrotoxicity with a Hydroxyl Radical and Caspase Light-Up Dual-Signal Nanoprobe. Anal Chem 2018; 90:3556-3562. [DOI: 10.1021/acs.analchem.7b05454] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Yuling Tong
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Xitong Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Mi Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Bo-Yang Yu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Jiangwei Tian
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, P.R. China
| |
Collapse
|
37
|
Alabi QK, Akomolafe RO, Olukiran OS, Nafiu AO, Adefisayo MA, Owotomo OI, Omole JG, Olamilosoye KP. Combined Administration ofl-Carnitine and Ascorbic Acid Ameliorates Cisplatin-Induced Nephrotoxicity in Rats. J Am Coll Nutr 2018; 37:387-398. [DOI: 10.1080/07315724.2017.1409139] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Quadri Kunle Alabi
- Department of Physiological Sciences, Faculty of Basic Medical Sciences, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria
- Department of Haematology and Blood Transfusion, College of Medicine, Faculty of Basic Medical Sciences, Afe Babalola University, Ado Ekiti, Ekiti, State, Nigeria
| | - Rufus Ojo Akomolafe
- Department of Physiological Sciences, Faculty of Basic Medical Sciences, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria
| | - Olaoluwa Sesan Olukiran
- Department of Physiological Sciences, Faculty of Basic Medical Sciences, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria
| | - Aliyat Olajumoke Nafiu
- Department of Physiological Sciences, Faculty of Basic Medical Sciences, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria
| | - Modinat Adebukola Adefisayo
- Department of Physiological Sciences, Faculty of Basic Medical Sciences, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria
- Department of Physiology, Faculty of Basic Medical Sciences, University of Medical Sciences, Ondo State, Nigeria
| | - Olurotimi Isaac Owotomo
- Department of Haematology and Immunology, Faculty of Basic Medical Sciences, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria
| | - Joseph Gbenga Omole
- Department of Physiological Sciences, Faculty of Basic Medical Sciences, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria
| | - Kehinde Peace Olamilosoye
- Department of Physiological Sciences, Faculty of Basic Medical Sciences, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria
| |
Collapse
|
38
|
Farooqui Z, Shahid F, Khan AA, Khan F. Oral administration of Nigella sativa oil and thymoquinone attenuates long term cisplatin treatment induced toxicity and oxidative damage in rat kidney. Biomed Pharmacother 2017; 96:912-923. [PMID: 29223554 DOI: 10.1016/j.biopha.2017.12.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 11/27/2017] [Accepted: 12/04/2017] [Indexed: 01/09/2023] Open
Abstract
Cisplatin (CP) is an effective anti-cancer drug which causes remarkable toxicity to the kidney, particularly to proximal tubules, by generating reactive oxygen species. Nigella sativa (NS), commonly known as "black cumin" reduces the progression of various kidney disorders. Thymoquinone (TQ), the major bioactive constituent of NS seeds, has been credited for various pharmacological effects of NS. Since, a typical clinical CP dosing regimen involves CP administration in multiple cycles over a long time duration, hence the present study aimed to evaluate the renoprotective efficacy of NS oil and TQ against multiple dose CP treatment induced deleterious biochemical and histological alterations in rat kidney. Adult male Wistar rats were divided into six groups viz. control, CP, CPNSO, CPTQ, NSO and TQ. Animals in CPNSO and CPTQ groups were pre-administered NSO (2ml/kg bwt, orally) and TQ (1.5mg/kg bwt, orally) respectively for 14 days and were then treated with CP (3mg/kg bwt, i.p), every fourth day for 20 days while still receiving NSO/TQ. NSO and TQ administration, prior to and along with CP treatment, attenuated CP induced renal functional impairment as evident by significantly restored serum creatinine and blood urea nitrogen levels. CP treatment alone led to significant decline in the specific activities of brush border membrane (BBM) marker enzymes viz. ALP (-46.64%), GGTase (-50.24%) and LAP (-42.15%), while NSO or TQ administration to CP treated rats significantly prevented the decline in the activities of these enzymes in isolated BBM vesicles (BBMVs) as well as in the homogenates of renal cortex and medulla. Furthermore, both NSO and TQ administration also mitigated the CP induced perturbations in renal metabolic and redox status. Histological studies supported these biochemical results showing significant attenuation of CP induced kidney damage in CPNSO and CPTQ cotreated groups. Thus, NSO and TQ have excellent scope for use as functional food or combinatorial nutraceuticals in CP chemotherapy to ameliorate the accompanying nephropathy in long term cancer chemotherapy.
Collapse
Affiliation(s)
- Zeba Farooqui
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, U.P., India
| | - Faaiza Shahid
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, U.P., India
| | - Aijaz Ahmed Khan
- Department of Anatomy, Faculty of Medicine, J. N. Medical College, Aligarh Muslim University, Aligarh 202002, U.P., India
| | - Farah Khan
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, U.P., India.
| |
Collapse
|
39
|
Ma ZN, Liu Z, Wang Z, Ren S, Tang S, Wang YP, Xiao SY, Chen C, Li W. Supplementation of American ginseng berry extract mitigated cisplatin-evoked nephrotoxicity by suppressing ROS-mediated activation of MAPK and NF-κB signaling pathways. Food Chem Toxicol 2017; 110:62-73. [DOI: 10.1016/j.fct.2017.10.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 09/21/2017] [Accepted: 10/07/2017] [Indexed: 01/04/2023]
|
40
|
Noh MR, Kim KY, Han SJ, Kim JI, Kim HY, Park KM. Methionine Sulfoxide Reductase A Deficiency Exacerbates Cisplatin-Induced Nephrotoxicity via Increased Mitochondrial Damage and Renal Cell Death. Antioxid Redox Signal 2017; 27:727-741. [PMID: 28158949 DOI: 10.1089/ars.2016.6874] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
AIMS Methionine sulfoxide reductase A (MsrA), which is abundantly localized in the mitochondria, reduces methionine-S-sulfoxide, scavenging reactive oxygen species (ROS). Cisplatin, an anticancer drug, accumulates at high levels in the mitochondria of renal cells, causing mitochondrial impairment that ultimately leads to nephrotoxicity. Here, we investigated the role of MsrA in cisplatin-induced mitochondrial damage and kidney cell death using MsrA gene-deleted (MsrA-/-) mice. RESULTS Cisplatin injection resulted in increases of ROS production, methionine oxidation, and oxidative damage in the kidneys. This oxidative stress was greater in MsrA-/- mouse kidneys than in wild-type (MsrA+/+) mouse kidneys. MsrA gene deletion exacerbated cisplatin-induced reductions in the expression and activity of MsrA and MsrBs, and the expression of thioredoxin 1, glutathione peroxidase 1 and 4, mitochondrial superoxide dismutase, cystathionine-β-synthase, and cystathionine-γ-lyase. Cisplatin induced swelling, cristae loss, and fragmentation of mitochondria with increased lipid peroxidation, more so in MsrA-/- than in MsrA+/+ kidneys. The ratio of mitochondrial fission regulator (Fis1) to fusion regulator (Opa1) was higher in MsrA-/- than MsrA+/+ mice. MsrA deletion exacerbated cisplatin-induced increases in Bax to Bcl-2 ratio, cleaved caspase-3 level, and apoptosis, whereas MsrA overexpression attenuated cisplatin-induced oxidative stress and apoptosis. INNOVATION MsrA gene deletion in mice exacerbates cisplatin-induced renal injury through increases of mitochondrial susceptibility, whereas MsrA overexpression protects cells against cisplatin. CONCLUSION This study demonstrates that MsrA protects kidney cells against cisplatin-induced methionine oxidation, oxidative stress, mitochondrial damage, and apoptosis, suggesting that MsrA could be a useful target protein for the treatment of cisplatin-induced nephrotoxicity. Antioxid. Redox Signal. 27, 727-741.
Collapse
Affiliation(s)
- Mi Ra Noh
- 1 Department of Anatomy and BK21 Plus, Kyungpook National University School of Medicine , Junggu, Daegu, Republic of Korea
| | - Ki Young Kim
- 2 Department of Biochemistry and Molecular Biology, Yeungnam University College of Medicine , Namgu, Daegu, Republic of Korea
| | - Sang Jun Han
- 1 Department of Anatomy and BK21 Plus, Kyungpook National University School of Medicine , Junggu, Daegu, Republic of Korea
| | - Jee In Kim
- 3 Department of Molecular Medicine and MRC, Keimyung University School of Medicine , Dalseogu, Daegu, Republic of Korea
| | - Hwa-Young Kim
- 2 Department of Biochemistry and Molecular Biology, Yeungnam University College of Medicine , Namgu, Daegu, Republic of Korea
| | - Kwon Moo Park
- 1 Department of Anatomy and BK21 Plus, Kyungpook National University School of Medicine , Junggu, Daegu, Republic of Korea
| |
Collapse
|
41
|
Cisplatin-induced human peripheral blood mononuclear cells’ oxidative stress and nephrotoxicity in head and neck cancer patients: the influence of hydrogen peroxide. Mol Cell Biochem 2017; 440:139-145. [DOI: 10.1007/s11010-017-3162-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 08/16/2017] [Indexed: 01/09/2023]
|
42
|
Ameliorative Effect of Daidzein on Cisplatin-Induced Nephrotoxicity in Mice via Modulation of Inflammation, Oxidative Stress, and Cell Death. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3140680. [PMID: 28831294 PMCID: PMC5558675 DOI: 10.1155/2017/3140680] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 06/14/2017] [Accepted: 06/20/2017] [Indexed: 12/21/2022]
Abstract
Oxidative stress and inflammation are part and parcel of cisplatin-induced nephrotoxicity. The purpose of this work is to study the role of soy isoflavone constituent, daidzein, in cisplatin-induced renal damage. Cisplatin-induced nephrotoxicity was evident by the histological damage in proximal tubular cells and by the increase in serum neutrophil gelatinase-associated lipocalin (NGAL), blood urea nitrogen (BUN), creatinine, and urinary kidney injury molecule-1 (KIM-1). Cisplatin-induced cell death was shown by TUNEL staining and caspase-3/7 activity. Daidzin treatment reduced all kidney injury markers (NGAL, BUN, creatinine, and KIM-1) and attenuated cell death (apoptotic markers). In cisplatin-induced kidney injury, renal oxidative/nitrative stress was manifested by the increase in lipid peroxidation and protein nitration. Cisplatin induced the reactive oxygen species-generating enzyme NOX-2 and impaired antioxidant defense enzyme activities such as glutathione peroxidase (GPX) and superoxide dismutase (SOD) activities. Cisplatin-induced oxidative/nitrative stress was attenuated by daidzein treatment. Cisplatin induced CD11b-positive macrophages in kidneys and daidzein attenuated CD11b-positive cells. Daidzein attenuated cisplatin-induced inflammatory cytokines tumor necrosis factor α (TNFα), interleukin 10 (IL-10), interleukin 18 (IL-18), and monocyte chemoattractant protein-1 (MCP-1). Daidzein attenuated cell death in vitro. Our data suggested that daidzein attenuated cisplatin-induced kidney injury through the downregulation of oxidative/nitrative stress, immune cells, inflammatory cytokines, and apoptotic cell death, thus improving kidney regeneration.
Collapse
|
43
|
Huang YC, Tsai MS, Hsieh PC, Shih JH, Wang TS, Wang YC, Lin TH, Wang SH. Galangin ameliorates cisplatin-induced nephrotoxicity by attenuating oxidative stress, inflammation and cell death in mice through inhibition of ERK and NF-kappaB signaling. Toxicol Appl Pharmacol 2017; 329:128-139. [DOI: 10.1016/j.taap.2017.05.034] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 05/21/2017] [Accepted: 05/26/2017] [Indexed: 02/06/2023]
|
44
|
Cascella M, Palma G, Barbieri A, Bimonte S, Amruthraj NJ, Muzio MR, Del Vecchio V, Rea D, Falco M, Luciano A, Arra C, Cuomo A. Role of Nigella sativa and Its Constituent Thymoquinone on Chemotherapy-Induced Nephrotoxicity: Evidences from Experimental Animal Studies. Nutrients 2017; 9:625. [PMID: 28629150 PMCID: PMC5490604 DOI: 10.3390/nu9060625] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 06/14/2017] [Accepted: 06/15/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Most chemotherapeutic drugs are known to cause nephrotoxicity. Therefore, new strategies have been considered to prevent chemotherapy-induced nephrotoxicity. It is of note that Nigella sativa (NS), or its isolated compound Thymoquinone (TQ), has a potential role in combating chemotherapy-induced nephrotoxicity. AIM To analyze and report the outcome of experimental animal studies on the protective effects of NS/TQ on chemotherapy-associated kidney complications. DESIGN Standard systematic review and narrative synthesis. DATA SOURCES MEDLINE, EMBASE databases were searched for relevant articles published up to March 2017. Additionally, a manual search was performed. Criteria for a study's inclusion were: conducted in animals, systematic reviews and meta-analysis, containing data on nephroprotective effects of NS/TQ compared to a placebo or other substance. All strains and genders were included. RESULTS The database search yielded 71 studies, of which 12 (cisplatin-induced nephrotoxicity 8; methotrexate-induced nephrotoxicity 1; doxorubicin-induced nephrotoxicity 2; ifosfamide-induced nephrotoxicity 1) were included in this review. CONCLUSIONS Experimental animal studies showed the protective effect of NS, or TQ, on chemotherapy-induced nephrotoxicity. These effects are caused by decreasing lipid peroxidation and increasing activity of antioxidant enzymes in renal tissue of chemotherapy-treated animals.
Collapse
Affiliation(s)
- Marco Cascella
- Division of Anesthesia and Pain Medicine, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", Via Mariano Semmola, 80131 Naples, Italy.
| | - Giuseppe Palma
- S.S.D. Sperimentazione Animale, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", 80131 Naples, Italy.
| | - Antonio Barbieri
- S.S.D. Sperimentazione Animale, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", 80131 Naples, Italy.
| | - Sabrina Bimonte
- Division of Anesthesia and Pain Medicine, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", Via Mariano Semmola, 80131 Naples, Italy.
| | - Nagoth Joseph Amruthraj
- S.S.D. Sperimentazione Animale, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", 80131 Naples, Italy.
- Clinical, Experimental and Medical Sciences, Chair of Nephrology, Department of Cardio-Vascular Medicine, University of Study of Campania "Luigi Vanvitelli", 81100 Caserta, Italy.
| | - Maria Rosaria Muzio
- Division of Infantile Neuropsychiatry, UOMI-Maternal and Infant Health, Asl NA 3 SUD, Torre del Greco, Via Marconi, 80059 Naples, Italy.
| | - Vitale Del Vecchio
- S.S.D. Sperimentazione Animale, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", 80131 Naples, Italy.
| | - Domenica Rea
- S.S.D. Sperimentazione Animale, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", 80131 Naples, Italy.
| | - Michela Falco
- S.S.D. Sperimentazione Animale, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", 80131 Naples, Italy.
| | - Antonio Luciano
- S.S.D. Sperimentazione Animale, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", 80131 Naples, Italy.
| | - Claudio Arra
- S.S.D. Sperimentazione Animale, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", 80131 Naples, Italy.
| | - Arturo Cuomo
- Division of Anesthesia and Pain Medicine, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", Via Mariano Semmola, 80131 Naples, Italy.
| |
Collapse
|
45
|
Quintanilha JCF, de Sousa VM, Visacri MB, Amaral LS, Santos RMM, Zambrano T, Salazar LA, Moriel P. Involvement of cytochrome P450 in cisplatin treatment: implications for toxicity. Cancer Chemother Pharmacol 2017; 80:223-233. [PMID: 28612092 DOI: 10.1007/s00280-017-3358-x] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 06/07/2017] [Indexed: 12/23/2022]
Abstract
PURPOSE The aim of this study is to evaluate the relationship between the CYP450 enzyme family and cisplatin toxicity. METHODS This article examined a collection of studies suggesting that CYP450 enzymes may influence cisplatin toxicity. We performed a narrative mini-review. RESULTS The studies review showed that CYP450 enzymes have an important role in drug-induced hepatotoxicity and nephrotoxicity, mainly CYP2E1 and CYP4A11. The studies also suggested that the cisplatin and CYP2E1 interaction leads to the generation of reactive oxygen species (ROS) and other oxidants resulting in renal injury; and that ROS generated by both the use of cisplatin and by the CYP2E1 increases tissue damage, induces apoptosis, and causes liver failure. CONCLUSIONS We observed that there is an important relationship between CYP450 and cisplatin, involving increased toxicity. However, the possible mechanisms described for the involvement of CYP450 enzymes in nephrotoxicity and hepatotoxicity induced by cisplatin need to be confirmed by further studies. Therefore, there is a need for a deeper investigation focusing on cisplatin toxicity mediated by CYP450 enzymes, which would undoubtedly contribute to a better understanding of the mechanisms that have been implicated so far.
Collapse
Affiliation(s)
| | - Vanessa Marcilio de Sousa
- Faculty of Pharmaceutical Sciences, University of Campinas (UNICAMP), 200 Cândido Portinari, Campinas, 13083-871, SP, Brazil
| | | | - Laís Sampaio Amaral
- School of Medical Sciences, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | | | - Tomás Zambrano
- Center of Molecular Biology and Pharmacogenetics, Faculty of Medicine, University of La Frontera, Temuco, Chile
| | - Luis Antonio Salazar
- Center of Molecular Biology and Pharmacogenetics, Faculty of Medicine, University of La Frontera, Temuco, Chile
| | - Patricia Moriel
- School of Medical Sciences, University of Campinas (UNICAMP), Campinas, SP, Brazil.
- Faculty of Pharmaceutical Sciences, University of Campinas (UNICAMP), 200 Cândido Portinari, Campinas, 13083-871, SP, Brazil.
| |
Collapse
|
46
|
Birer SR, Lee CT, Choudhury KR, Young KH, Spasojevic I, Batinic-Haberle I, Crapo JD, Dewhirst MW, Ashcraft KA. Inhibition of the Continuum of Radiation-Induced Normal Tissue Injury by a Redox-Active Mn Porphyrin. Radiat Res 2017; 188:94-104. [PMID: 28517962 DOI: 10.1667/rr14757.1.s1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Normal tissue damage after head and neck radiotherapy involves a continuum of pathologic events to the mucosa, tongue and salivary glands. We examined the radioprotective effects of MnBuOE, a redox-active manganese porphyrin, at three stages of normal tissue damage: immediate (leukocyte endothelial cell [L/E] interactions), early (mucositis) and late (xerostomia and fibrosis) after treatment. In this study, mice received 0 or 9 Gy irradiation to the oral cavity and salivary glands ± MnBuOE treatment. Changes in leukocyte-endothelial cell interactions were measured 24 h postirradiation. At 11 days postirradiation, mucositis was assessed with a cathepsin-sensitive near-infrared optical probe. Stimulated saliva production was quantified at 11 weeks postirradiation. Finally, histological analyses were conducted to assess the extent of long-term effects in salivary glands at 12 weeks postirradiation. MnBuOE reduced oral mucositis, xerostomia and salivary gland fibrosis after irradiation. Additionally, although we have previously shown that MnBuOE does not interfere with tumor control at high doses when administered with radiation alone, most head and neck cancer patients will be treated with the combinations of radiotherapy and cisplatin. Therefore, we also evaluated whether MnBuOE would protect tumors against radiation and cisplatin using tumor growth delay as an endpoint. Using a range of radiation doses, we saw no evidence that MnBuOE protected tumors from radiation and cisplatin. We conclude that MnBuOE radioprotects normal tissue at both early and late time points, without compromising anti-tumor effects of radiation and cisplatin.
Collapse
Affiliation(s)
- Samuel R Birer
- a Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, 27710
| | - Chen-Ting Lee
- a Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, 27710
| | - Kingshuk Roy Choudhury
- b Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina, 27710
| | - Kenneth H Young
- a Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, 27710
| | - Ivan Spasojevic
- c Department of Medicine, Duke University Medical Center, Durham, North Carolina, 27710.,d Duke Cancer Institute, Pharmaceutical Research PK-PD Core Laboratory, Durham, North Carolina 27710
| | - Ines Batinic-Haberle
- a Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, 27710
| | - James D Crapo
- e Department of Medicine, National Jewish Health, Denver, Colorado 80206 and BioMimetix JV, LLC, Englewood, Colorado 80113
| | - Mark W Dewhirst
- a Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, 27710
| | - Kathleen A Ashcraft
- a Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, 27710
| |
Collapse
|
47
|
Chima UM, Abu AH, Dawuda PM, Kisani AI, Ahemen T. Effect of Storage Time on Cauda Epididymal Sperm Parameters of Nigerian Local Dogs. ACTA ACUST UNITED AC 2017. [DOI: 10.4236/ojvm.2017.711016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
48
|
Wang X, Nichols L, Grunz-Borgmann EA, Sun Z, Meininger GA, Domeier TL, Baines CP, Parrish AR. Fascin2 regulates cisplatin-induced apoptosis in NRK-52E cells. Toxicol Lett 2016; 266:56-64. [PMID: 27989596 DOI: 10.1016/j.toxlet.2016.11.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 11/01/2016] [Accepted: 11/27/2016] [Indexed: 01/09/2023]
Abstract
Previous studies have shown that the aging kidney has a marked loss of α(E)-catenin in proximal tubular epithelium. α-Catenin, a key regulator of the actin cytoskeleton, interacts with a variety of actin-binding proteins. Cisplatin-induced loss of fascin2, an actin bundling protein, was observed in cells with a stable knockdown of α(E)-catenin (C2 cells), as well as in aging (24 mon), but not young (4 mon), kidney. Fascin2 co-localized with α-catenin and the actin cytoskeleton in NRK-52E cells. Knockdown of fascin2 increased the susceptibility of tubular epithelial cells to cisplatin-induced injury. Overexpression of fascin2 in C2 cells restored actin stress fibers and attenuated the increased sensitivity of C2 cells to cisplatin-induced apoptosis. Interestingly, fascin2 overexpression attenuated cisplatin-induced mitochondrial dysfunction and oxidative stress in C2 cells. These data demonstrate that fascin2, a putative target of α(E)-catenin, may play important role in preventing cisplatin-induced acute kidney injury.
Collapse
Affiliation(s)
- Xinhui Wang
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, United States
| | - LaNita Nichols
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, United States
| | - Elizabeth A Grunz-Borgmann
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, United States
| | - Zhe Sun
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, United States; Dalton Cardiovascular Research Center, School of Medicine, University of Missouri, Columbia, MO 65212, United States
| | - Gerald A Meininger
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, United States; Dalton Cardiovascular Research Center, School of Medicine, University of Missouri, Columbia, MO 65212, United States
| | - Timothy L Domeier
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, United States
| | - Christopher P Baines
- Dalton Cardiovascular Research Center, School of Medicine, University of Missouri, Columbia, MO 65212, United States; Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65212, United States
| | - Alan R Parrish
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, United States.
| |
Collapse
|
49
|
Ohba H, Kanazawa M, Kakiuchi T, Tsukada H. Effects of acetaminophen on mitochondrial complex I activity in the rat liver and kidney: a PET study with 18F-BCPP-BF. EJNMMI Res 2016; 6:82. [PMID: 27873239 PMCID: PMC5118230 DOI: 10.1186/s13550-016-0241-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 11/15/2016] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND In the present study, 2-tert-butyl-4-chloro-5-[6-(4-18F-fluorobutoxy)-pyridin-3-ylmethoxy]-2H-pyridazin-3-one (18F-BCPP-BF), a PET probe for mitochondrial complex I (MC-I), was used to validate whether MC-I is a useful biomarker for detecting acetaminophen-induced dysfunctions in the liver and kidney. The kinetic and distribution of 18F-BCPP-BF were assessed in rats using high-resolution animal PET in vivo. The binding specificity of 18F-BCPP-BF to MC-I in the liver and kidney was confirmed by the pre-administration of rotenone, a specific MC-I inhibitor. The effects of acetaminophen on MC-I activity were assessed 2 and 24 h after the administration of vehicle or acetaminophen at a dose of 100 or 300 mg/kg. Biochemical parameters in plasma and urine were assessed 2, 6, and 24 h after the administration of vehicle or acetaminophen. RESULTS The uptake of 18F-BCPP-BF by the liver and kidney was significantly inhibited by the pre-administration of rotenone. Two and more hours after the administration of acetaminophen, the uptake of 18F-BCPP-BF was dose-dependently reduced in the liver, even at 100 mg/kg, and in the kidney at 300 mg/kg, whereas biological parameters started to be affected 6 h or later at doses of 300 mg/kg. CONCLUSIONS The present study demonstrated that 18F-BCPP-BF has potential as a PET probe for the quantitative imaging of hepatic and renal dysfunction as impaired MC-I activity in the early phase of the treatment for an overdose of acetaminophen in the living body with PET.
Collapse
Affiliation(s)
- Hiroyuki Ohba
- Central Research Laboratory, Hamamatsu Photonics K.K., 5000 Hirakuchi, Hamamatsu, Shizuoka, 434-8601, Japan
| | - Masakatsu Kanazawa
- Central Research Laboratory, Hamamatsu Photonics K.K., 5000 Hirakuchi, Hamamatsu, Shizuoka, 434-8601, Japan
| | - Takeharu Kakiuchi
- Central Research Laboratory, Hamamatsu Photonics K.K., 5000 Hirakuchi, Hamamatsu, Shizuoka, 434-8601, Japan
| | - Hideo Tsukada
- Central Research Laboratory, Hamamatsu Photonics K.K., 5000 Hirakuchi, Hamamatsu, Shizuoka, 434-8601, Japan.
| |
Collapse
|
50
|
Vuda M, Kamath A. Drug induced mitochondrial dysfunction: Mechanisms and adverse clinical consequences. Mitochondrion 2016; 31:63-74. [PMID: 27771494 DOI: 10.1016/j.mito.2016.10.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/24/2016] [Accepted: 10/17/2016] [Indexed: 01/12/2023]
Abstract
Several commonly used medications impair mitochondrial function resulting in adverse effects or toxicities. Drug induced mitochondrial dysfunction may be a consequence of increased production of reactive oxygen species, altered mitochondrial permeability transition, impaired mitochondrial respiration, mitochondrial DNA damage or inhibition of beta-oxidation of fatty acids. The clinical manifestation depends on the specific drug and its effect on mitochondria. Given the ubiquitous presence of mitochondria and its central role in cellular metabolism, drug-mitochondrial interactions may manifest clinically as hepatotoxicity, enteropathy, myelosuppression, lipodystrophy syndrome or neuropsychiatric adverse effects, to name a few. The current review focuses on specific drug groups which adversely affect mitochondria, the mechanisms involved and the clinical consequences based on the data available from experimental and clinical studies. Knowledge of these adverse drug-mitochondrial interactions may help the clinicians foresee potential issues in individual patients, prevent adverse drug reactions or alter drug regimens to enhance patient safety.
Collapse
Affiliation(s)
| | - Ashwin Kamath
- Department of Pharmacology, Kasturba Medical College, Manipal University, Mangalore, India.
| |
Collapse
|