51
|
Yu D, Gu J, Chen Y, Kang W, Wang X, Wu H. Current Strategies to Combat Cisplatin-Induced Ototoxicity. Front Pharmacol 2020; 11:999. [PMID: 32719605 PMCID: PMC7350523 DOI: 10.3389/fphar.2020.00999] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/22/2020] [Indexed: 12/13/2022] Open
Abstract
Cisplatin is widely used for the treatment of a number of solid malignant tumors. However, ototoxicity induced by cisplatin is an obstacle to effective treatment of tumors. The basis for this toxicity has not been fully elucidated. It is generally accepted that hearing loss is due to excessive production of reactive oxygen species by cells of the cochlea. In addition, recent data suggest that inflammation may trigger inner ear cell death through endoplasmic reticulum stress, autophagy, and necroptosis, which induce apoptosis. Strategies have been extensively explored by which to prevent, alleviate, and treat cisplatin-induced ototoxicity, which minimize interference with antitumor activity. Of these strategies, none have been approved by the Federal Drug Administration, although several preclinical studies have been promising. This review highlights recent strategies that reduce cisplatin-induced ototoxicity. The focus of this review is to identify candidate agents as novel molecular targets, drug administration routes, delivery systems, and dosage schedules. Animal models of cisplatin ototoxicity are described that have been used to evaluate drug efficacy and side effect prevention. Finally, clinical reports of otoprotection in patients treated with cisplatin are highlighted. For the future, high-quality studies are required to provide reliable data regarding the safety and effectiveness of pharmacological interventions that reduce cisplatin-induced ototoxicity.
Collapse
Affiliation(s)
- Dehong Yu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases (14DZ2260300), Shanghai, China
| | - Jiayi Gu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases (14DZ2260300), Shanghai, China
| | - Yuming Chen
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases (14DZ2260300), Shanghai, China
| | - Wen Kang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases (14DZ2260300), Shanghai, China
| | - Xueling Wang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases (14DZ2260300), Shanghai, China
| | - Hao Wu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases (14DZ2260300), Shanghai, China
| |
Collapse
|
52
|
Kohsaka T, Minagawa I, Morimoto M, Yoshida T, Sasanami T, Yoneda Y, Ikegaya N, Sasada H. Efficacy of relaxin for cisplatin-induced testicular dysfunction and epididymal spermatotoxicity. Basic Clin Androl 2020; 30:3. [PMID: 32166037 PMCID: PMC7061478 DOI: 10.1186/s12610-020-0101-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 01/19/2020] [Indexed: 12/12/2022] Open
Abstract
Background Cisplatin (CP) is an extremely effective anticancer agent widely used to treat various cancer types, however, the potential side effects include testicular dysfunction. This study was to investigate, using a rat model of CP-induced testicular dysfunction, the protective effects of relaxin (RLN) against oxidative stress, testicular function, histological damage, spermatogenesis, germ-cell apoptosis, and sperm output, and to explore the usefulness of RLN as a potential protective drug for use with CP in chemotherapeutic treatments. Methods Sprague-Dawley male rats were used, which were divided into three groups: sham control, CP, and CP + RLN. Porcine RLN (500 ng/h) or saline was infused for 5 days using an implanted osmotic mini-pump following intraperitoneal injection of CP (6 mg/kg). RLN dose was chosen based on previous studies showing that it resulted in serum relaxin levels comparable to those in rats at the middle of pregnancy. At 5 days after CP administration, samples were collected and assessment of testicular histopathology, germ-cell apoptosis, oxidative stress, lipid peroxidation, and sperm quality was performed as main measures. Results The testicular CP model showed reduced testis weight and significantly decreased spermatogenesis scores. Additionally, CP administration induced a 4.6-fold increase in the apoptotic index associated with a significant increase in oxidative stress and upregulation of pro-apoptotic Casp3 and downregulation of anti-apoptotic Bcl2 levels, resulting in a marked reduction in sperm concentration. However, RLN administration caused a significant reduction in CP-mediated damage by attenuating oxidative stress and cell apoptosis. RLN administration efficiently scavenged ROS via the activation of SOD, CAT, and GPx and upregulation of GSH to prevent lipid peroxidation and decreased apoptosis by altering Bcl2 and Casp3 expression, thereby reducing histopathological damage and restoring spermatogenesis. Furthermore, RLN ameliorated attenuated sperm motility in the cauda epididymis resulting from CP treatment. Conclusions This study clearly indicates that RLN exerts a protective effect against CP-induced testicular damage through attenuation of oxidative stress and suppression of apoptosis. Our findings suggest RLN as a potentially efficacious drug for use with cisplatin chemotherapy in order to ameliorate CP-induced side effects and testicular injury adversely affecting spermatogenesis, sperm quality, and oxidative-stress parameters.
Collapse
Affiliation(s)
- Tetsuya Kohsaka
- 1Department of Applied Life Sciences, Animal Reproduction & Physiology Faculty of Agriculture, Shizuoka University, 836 Ohya, Suruga-ku, Shizuoka, 422-8529 Japan
| | - Itaru Minagawa
- 1Department of Applied Life Sciences, Animal Reproduction & Physiology Faculty of Agriculture, Shizuoka University, 836 Ohya, Suruga-ku, Shizuoka, 422-8529 Japan
| | - Masashi Morimoto
- 1Department of Applied Life Sciences, Animal Reproduction & Physiology Faculty of Agriculture, Shizuoka University, 836 Ohya, Suruga-ku, Shizuoka, 422-8529 Japan
| | - Takuya Yoshida
- 2Department of Clinical Nutrition, School of Food and Nutritional Science, University of Shizuoka, Shizuoka, 422-8526 Japan
| | - Tomohiro Sasanami
- 1Department of Applied Life Sciences, Animal Reproduction & Physiology Faculty of Agriculture, Shizuoka University, 836 Ohya, Suruga-ku, Shizuoka, 422-8529 Japan
| | - Yoshitaka Yoneda
- Advanced Reproductive Medical Center, Shizuoka Ladies Clinic, Shizuoka, 420-0837 Japan
| | - Naoki Ikegaya
- Department of Medicine, Yaizu Municipal General Hospital, Shizuoka, 422-8505 Japan
| | - Hiroshi Sasada
- 5Division of Animal Science, Kitasato University School of Veterinary Medicine, Towada, 034-8628 Japan
| |
Collapse
|
53
|
Ciccarese F, Raimondi V, Sharova E, Silic-Benussi M, Ciminale V. Nanoparticles as Tools to Target Redox Homeostasis in Cancer Cells. Antioxidants (Basel) 2020; 9:antiox9030211. [PMID: 32143322 PMCID: PMC7139659 DOI: 10.3390/antiox9030211] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 02/20/2020] [Accepted: 02/29/2020] [Indexed: 12/18/2022] Open
Abstract
Reactive oxygen species (ROS) constitute a homeostatic rheostat that modulates signal transduction pathways controlling cell turnover. Most oncogenic pathways activated in cancer cells drive a sustained increase in ROS production, and cancer cells are strongly addicted to the increased activity of scavenging pathways to maintain ROS below levels that produce macromolecular damage and engage cell death pathways. Consistent with this notion, tumor cells are more vulnerable than their normal counterparts to pharmacological treatments that increase ROS production and inhibit ROS scavenging. In the present review, we discuss the recent advances in the development of integrated anticancer therapies based on nanoparticles engineered to kill cancer cells by raising their ROS setpoint. We also examine nanoparticles engineered to exploit the metabolic and redox alterations of cancer cells to promote site-specific drug delivery to cancer cells, thus maximizing anticancer efficacy while minimizing undesired side effects on normal tissues.
Collapse
Affiliation(s)
- Francesco Ciccarese
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (F.C.); (V.R.); (E.S.); (M.S.-B.)
| | - Vittoria Raimondi
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (F.C.); (V.R.); (E.S.); (M.S.-B.)
| | - Evgeniya Sharova
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (F.C.); (V.R.); (E.S.); (M.S.-B.)
| | - Micol Silic-Benussi
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (F.C.); (V.R.); (E.S.); (M.S.-B.)
| | - Vincenzo Ciminale
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy; (F.C.); (V.R.); (E.S.); (M.S.-B.)
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
- Correspondence:
| |
Collapse
|
54
|
Gonçalves MS, Silveira AFD, Murashima ADAB, Rossato M, Hippolito MA. Otoprotection Mechanisms Against Oxidative Stress Caused by Cisplatin. Int Arch Otorhinolaryngol 2020; 24:e47-e52. [PMID: 31929833 PMCID: PMC6952293 DOI: 10.1055/s-0039-1698782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/07/2019] [Indexed: 11/18/2022] Open
Abstract
Introduction
Cisplatin damages the auditory system and is related to the generation of free radicals. Glutathione peroxidase is an endogenous free radicals remover.
Objective
To investigate the mechanisms involved in otoprotection by N-acetylcysteine through the expression of glutathione peroxidase in outer hair cells from rats treated with cisplatin.
Methods
Male Wistar rats were intraperitoneally injected with cisplatin (8 mg/Kg) and/or received oral administration by gavage of N-acetylcysteine (300 mg/Kg) for 3 consecutive days. On the 4
th
day, the animals were euthanized and beheaded. The tympanic bullae were removed and prepared for scanning electron microscopy and immunofluorescence.
Results
Among the groups exposed to ototoxic doses of cisplatin, there was an increase in glutathione peroxidase immunostaining in two groups, the one exposed to cisplatin alone, and the group exposed to both cisplatin and N-acetylcysteine.
Conclusion
The expression of glutathione peroxidase in the outer hair cells of rats exposed to cisplatin showed the synthesis of this enzyme under cellular toxicity conditions.
Collapse
Affiliation(s)
| | | | - Adriana de Andrade Batista Murashima
- Department of Ophthalmology, Otorhinolaryngology and Head and Neck Surgery, Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, Ribeirao Preto, SP, Brazil
| | - Maria Rossato
- Department of Ophthalmology, Otorhinolaryngology and Head and Neck Surgery, Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, Ribeirao Preto, SP, Brazil
| | - Miguel Angelo Hippolito
- Department of Ophthalmology, Otorhinolaryngology and Head and Neck Surgery, Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, Ribeirao Preto, SP, Brazil
| |
Collapse
|
55
|
Rehman MU, Rather IA. Myricetin Abrogates Cisplatin-Induced Oxidative Stress, Inflammatory Response, and Goblet Cell Disintegration in Colon of Wistar Rats. PLANTS 2019; 9:plants9010028. [PMID: 31878169 PMCID: PMC7020155 DOI: 10.3390/plants9010028] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022]
Abstract
Cisplatin [cis-diamminedichloroplatinum II] is an extensively prescribed drug in cancer chemotherapy; it is also useful for the treatment of diverse types of malignancies. Conversely, cisplatin is associated with a range of side effects such as nephrotoxicity, hepatotoxicity, gastrointestinal toxicity, and so on. Myricetin (3,5,7-trihydroxy-2-(3,4,5-trihydroxyphenyl)-4chromenone) is a very common natural flavonoid found in fruits, tea, and plants. It has been found to have high-value pharmacological properties and strong health benefits. To examine the role of myricetin in colon toxicity induced by cisplatin, we conducted a concurrent prophylactic study in experimental animals that were treated orally with myricetin for 14 days at two doses—25 and 50 mg/kg of body weight. On the 14th day, a single intraperitoneal injection of cisplatin (7.5 mg/kg body weight) was administered in all groups except control. The effects of myricetin in cisplatin-induced toxicity in the colon were assessed in terms of antioxidant status, phase-II detoxification enzymes, the level of inflammatory markers, and goblet cell disintegration. Myricetin was found to restore the level of all the antioxidant enzymes analyzed in the study. In addition, the compound ameliorated cisplatin-induced lipid peroxidation, increase in xanthine oxidase activity, and phase-II detoxifying enzyme activity. Myricetin also attenuated deteriorative effects induced by cisplatin by regulating the level of molecular markers of inflammation (NF-κB, Nrf-2, IL-6, and TNF-α), restoring Nrf-2 levels, and controlling goblet cell disintegration. The current study reinforces the conclusion that myricetin exerts protection in colon toxicity via up-regulation of inflammatory markers, improving anti-oxidant status, and protecting tissue damage.
Collapse
Affiliation(s)
- Muneeb U. Rehman
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, P.O. Box-2457, Riyadh 11451, Saudi Arabia
- Division of Biochemistry, Faculty of Veterinary Science and Animal Husbandry, SKAUST-Kashmir, Alustang, Srinagar, J&K 190006, India
- Correspondence: (M.U.R.); (I.A.R.)
| | - Irfan A. Rather
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University (KAU) P.O. Box-80141, Jeddah 21589, Saudi Arabia
- Correspondence: (M.U.R.); (I.A.R.)
| |
Collapse
|
56
|
Raimondi V, Ciccarese F, Ciminale V. Oncogenic pathways and the electron transport chain: a dangeROS liaison. Br J Cancer 2019; 122:168-181. [PMID: 31819197 PMCID: PMC7052168 DOI: 10.1038/s41416-019-0651-y] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 10/30/2019] [Accepted: 11/05/2019] [Indexed: 02/07/2023] Open
Abstract
Driver mutations in oncogenic pathways, rewiring of cellular metabolism and altered ROS homoeostasis are intimately connected hallmarks of cancer. Electrons derived from different metabolic processes are channelled into the mitochondrial electron transport chain (ETC) to fuel the oxidative phosphorylation process. Electrons leaking from the ETC can prematurely react with oxygen, resulting in the generation of reactive oxygen species (ROS). Several signalling pathways are affected by ROS, which act as second messengers controlling cell proliferation and survival. On the other hand, oncogenic pathways hijack the ETC, enhancing its ROS-producing capacity by increasing electron flow or by impinging on the structure and organisation of the ETC. In this review, we focus on the ETC as a source of ROS and its modulation by oncogenic pathways, which generates a vicious cycle that resets ROS levels to a higher homoeostatic set point, sustaining the cancer cell phenotype.
Collapse
Affiliation(s)
| | | | - Vincenzo Ciminale
- Veneto Institute of Oncology IOV - IRCCS, Padua, Italy. .,Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy.
| |
Collapse
|
57
|
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: 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: 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
|
58
|
LKB1/AMPK Pathway and Drug Response in Cancer: A Therapeutic Perspective. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8730816. [PMID: 31781355 PMCID: PMC6874879 DOI: 10.1155/2019/8730816] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 09/10/2019] [Accepted: 09/16/2019] [Indexed: 12/25/2022]
Abstract
Inactivating mutations of the tumor suppressor gene Liver Kinase B1 (LKB1) are frequently detected in non-small-cell lung cancer (NSCLC) and cervical carcinoma. Moreover, LKB1 expression is epigenetically regulated in several tumor types. LKB1 has an established function in the control of cell metabolism and oxidative stress. Clinical and preclinical studies support a role of LKB1 as a central modifier of cellular response to different stress-inducing drugs, suggesting LKB1 pathway as a highly promising therapeutic target. Loss of LKB1-AMPK signaling confers sensitivity to energy depletion and to redox homeostasis impairment and has been associated with an improved outcome in advanced NSCLC patients treated with chemotherapy. In this review, we provide an overview of the interplay between LKB1 and its downstream targets in cancer and focus on potential therapeutic strategies whose outcome could depend from LKB1.
Collapse
|
59
|
Solid Lipid Nanoparticles Loaded with Glucocorticoids Protect Auditory Cells from Cisplatin-Induced Ototoxicity. J Clin Med 2019; 8:jcm8091464. [PMID: 31540035 PMCID: PMC6780793 DOI: 10.3390/jcm8091464] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 08/23/2019] [Accepted: 09/12/2019] [Indexed: 12/18/2022] Open
Abstract
Cisplatin is a chemotherapeutic agent that causes the irreversible death of auditory sensory cells, leading to hearing loss. Local administration of cytoprotective drugs is a potentially better option co-therapy for cisplatin, but there are strong limitations due to the difficulty of accessing the inner ear. The use of nanocarriers for the efficient delivery of drugs to auditory cells is a novel approach for this problem. Solid lipid nanoparticles (SLNs) are biodegradable and biocompatible nanocarriers with low solubility in aqueous media. We show here that stearic acid-based SLNs have the adequate particle size, polydispersity index and ζ-potential, to be considered optimal nanocarriers for drug delivery. Stearic acid-based SLNs were loaded with the fluorescent probe rhodamine to show that they are efficiently incorporated by auditory HEI-OC1 (House Ear Institute-Organ of Corti 1) cells. SLNs were not ototoxic over a wide dose range. Glucocorticoids are used to decrease cisplatin-induced ototoxicity. Therefore, to test SLNs’ drug delivery efficiency, dexamethasone and hydrocortisone were tested either alone or loaded into SLNs and tested in a cisplatin-induced ototoxicity in vitro assay. Our results indicate that the encapsulation in SLNs increases the protective effect of low doses of hydrocortisone and lengthens the survival of HEI-OC1 cells treated with cisplatin.
Collapse
|
60
|
Wang N, Song L, Xu Y, Zhang L, Wu Y, Guo J, Ji W, Li L, Zhao J, Zhang X, Zhan L. Loss of Scribble confers cisplatin resistance during NSCLC chemotherapy via Nox2/ROS and Nrf2/PD-L1 signaling. EBioMedicine 2019; 47:65-77. [PMID: 31495720 PMCID: PMC6796531 DOI: 10.1016/j.ebiom.2019.08.057] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 08/23/2019] [Accepted: 08/23/2019] [Indexed: 12/26/2022] Open
Abstract
Background Cisplatin resistance remains a major clinical obstacle to the successful treatment of non-small cell lung cancer (NSCLC). Scribble contributes to ROS-induced inflammation and cisplatin-elevated toxic reactive oxygen species (ROS) promotes cell death. However, it is unknown whether and how Scribble is involved in the cisplatin-related cell death and the underlying mechanism of Scribble in response to chemotherapies and in the process of oxidative stress in NSCLC. Methods We used two independent cohorts of NSCLC samples derived from patients treated with platinum-containing chemotherapy and xenograft modeling in vivo. We analyzed the correlation between Scribble and Nox2 or Nrf2/PD-L1 both in vivo and in vitro, and explored the role of Scribble in cisplatin-induced ROS and apoptosis. Findings Clinical analysis revealed that Scribble expression positively correlated with clinical outcomes and chemotherapeutic sensitivity in NSCLC patients. Scribble protected Nox2 protein from proteasomal degradation. Scribble knockdown induced cisplatin resistance by blocking Nox2/ROS and apoptosis in LRR domain-dependent manner. In addition, low levels of Scribble correlated with high levels of PD-L1 via activation of Nrf2 transcription in vivo and in vitro. Interpretations Our study revealed that polarity protein Scribble increased cisplatin-induced ROS generation and is beneficial to chemotherapeutic outcomes in NSCLC. Although Scribble deficiency tends to lead to cisplatin resistance by Nox2/ROS and Nrf2/PD-L1, it is still possible that Scribble deficiency-induced PD-L1 may yield benefits in immunotherapy. Fund National Key R&D Program of China, Strategic Priority Research Program of the Chinese Academy of Sciences, National Natural Science Foundation of China, China Postdoctoral Science Foundation.
Collapse
Affiliation(s)
- Na Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lele Song
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Changhai Hospital, The Second Military Medical University, Shanghai 200433, China
| | - Yi Xu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Longfu Zhang
- Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yanjun Wu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jingyu Guo
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Weiwei Ji
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Li Li
- Department of Biology, Chemistry and Environmental Studies, Molloy College, New York 11571, USA
| | - Jingya Zhao
- Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xin Zhang
- Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Lixing Zhan
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
61
|
Zhou Z, Xu J, Bao X, Shi J, Liu B, Chen Y, Li J. Nuclear Nrf2 Activity in Laryngeal Carcinoma is Regulated by SENP3 After Cisplatin-Induced Reactive Oxygen Species Stress. J Cancer 2019; 10:3427-3434. [PMID: 31293646 PMCID: PMC6603410 DOI: 10.7150/jca.30318] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 04/25/2019] [Indexed: 02/01/2023] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a nuclear transcription factor that is activated by reactive oxygen species (ROS). Recent studies reported that hyperactivation of the Nrf2 pathway creates an environment that favors the survival of normal as well as malignant cells, protecting them against oxidative stress, chemotherapeutic agents, and radiotherapy. SUMO1/sentrin/SMT3 specific peptidase 3 (SENP3) reverses sumoylation of small ubiquitin-like modifier (SUMO)-conjugates. We demonstrated that Nrf2 was detected in the nuclei of laryngeal carcinoma cells, but not in cells of tissues surrounding the cancer, which correlated with the appearance of SENP3 in the nuclei. Silencing of Nrf2 in laryngeal carcinoma cell line Hep-2 significantly reduced cell viability and enhanced apoptosis rates under cisplatin, 5-fluorouracil (5-FU) and phenethyl isothiocyanate (PEITC) exposure. Cisplatin exposure induced ROS stress in Hep-2 cells in a time-dependent manner and was accompanied by increased Nrf2 and SENP3 protein accumulations, an effect reversed by the addition of the antioxidant N-acetyl-cysteine (NAC). Silencing of SENP3 led to reduced Nrf2 protein levels, whereas overexpression of SENP3 led to concomitant enhanced transcription of the Nrf2 target genes HO-1, NQO1, GCLC and GSTM1. Immunoprecipitation showed that overexpressed Nrf2 and SENP3 could be precipitated together, indicating that they were intracellular bound to each other. Our data identified intranuclear activation of Nrf2 is triggered by cisplatin-induced ROS development through the activity of SENP3. These findings provide novel insights into the Nrf2 reduced cancer cell response to the chemotherapy of laryngeal carcinoma.
Collapse
Affiliation(s)
- Zheng Zhou
- Department of Otolaryngology, Ren ji Hospital, School of Medicine, Shanghai Jiao tong University, Shanghai, China
| | - Ji Xu
- Department of Otolaryngology, Ren ji Hospital, School of Medicine, Shanghai Jiao tong University, Shanghai, China
| | - Ximing Bao
- Department of Otolaryngology, Ren ji Hospital, School of Medicine, Shanghai Jiao tong University, Shanghai, China
| | - Jiali Shi
- Department of Otolaryngology, Ren ji Hospital, School of Medicine, Shanghai Jiao tong University, Shanghai, China
| | - Bin Liu
- Department of Otolaryngology, Ren ji Hospital, School of Medicine, Shanghai Jiao tong University, Shanghai, China
| | - Yanqing Chen
- Department of Otolaryngology, Ren ji Hospital, School of Medicine, Shanghai Jiao tong University, Shanghai, China
| | - Jiping Li
- Department of Otolaryngology, Ren ji Hospital, School of Medicine, Shanghai Jiao tong University, Shanghai, China
| |
Collapse
|
62
|
Ismail NI, Othman I, Abas F, H Lajis N, Naidu R. Mechanism of Apoptosis Induced by Curcumin in Colorectal Cancer. Int J Mol Sci 2019; 20:E2454. [PMID: 31108984 PMCID: PMC6566943 DOI: 10.3390/ijms20102454] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 04/20/2019] [Accepted: 04/26/2019] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is among the top three cancer with higher incident and mortality rate worldwide. It is estimated that about over than 1.1 million of death and 2.2 million new cases by the year 2030. The current treatment modalities with the usage of chemo drugs such as FOLFOX and FOLFIRI, surgery and radiotherapy, which are usually accompanied with major side effects, are rarely cured along with poor survival rate and at higher recurrence outcome. This trigger the needs of exploring new natural compounds with anti-cancer properties which possess fewer side effects. Curcumin, a common spice used in ancient medicine was found to induce apoptosis by targeting various molecules and signaling pathways involved in CRC. Disruption of the homeostatic balance between cell proliferation and apoptosis could be one of the promoting factors in colorectal cancer progression. In this review, we describe the current knowledge of apoptosis regulation by curcumin in CRC with regard to molecular targets and associated signaling pathways.
Collapse
Affiliation(s)
- Nor Isnida Ismail
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway Darul Ehsan, Malaysia.
- UniKL MESTECH, A1-1 Jalan TKS1, Taman Kajang Sentral, 43000 Kajang, Malaysia.
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway Darul Ehsan, Malaysia.
| | - Faridah Abas
- Laboratory of Natural Products, Faculty of Science, University Putra Malaysia, UPM, 43400 Serdang, Malaysia.
- Department of Food Science, Faculty of Food Science and Technology, University Putra Malaysia, UPM, 434000 Serdang, Malaysia.
| | - Nordin H Lajis
- Laboratory of Natural Products, Faculty of Science, University Putra Malaysia, UPM, 43400 Serdang, Malaysia.
| | - Rakesh Naidu
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway Darul Ehsan, Malaysia.
| |
Collapse
|
63
|
Üstün Bezgin S, Uygur KK, Gökdoğan Ç, Elmas Ç, Göktaş G. The Effects of Riluzole on Cisplatin-induced Ototoxicity. Int Arch Otorhinolaryngol 2019; 23:e267-e275. [PMID: 31360245 PMCID: PMC6660296 DOI: 10.1055/s-0038-1676654] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 10/21/2018] [Indexed: 01/06/2023] Open
Abstract
Introduction
Riluzole (2-amino-6-trifluoromethoxy benzothiazole) is known as a neuroprotective, antioxidant, antiapoptotic agent. It may have beneficial effects on neuronal cell death due to cisplatin-induced ototoxicity.
Objective
To evaluate the effect of riluzole on cisplatin-induced ototoxicity in guinea pigs.
Methods
Twenty-four guinea pigs, studied in three groups, underwent auditory brainstem response evaluation using click and 8 kHz tone burst stimuli. Subsequently, 5 mg/kg of cisplatin were administered to all animals for 3 days intraperitoneally (i.p.) to induce ototoxicity. Half an hour prior to cisplatin, groups 1, 2 and 3 received 2 ml of saline i.p., 6 mg/kg of riluzole hydrochloride i.p., and 8 mg/kg of riluzole hydrochloride i.p., respectively, for 3 days. The auditory brainstem responses were repeated 24 hours after the last drug administration. The cochleae were analyzed by transmission electron microscopy (TEM).
Results
After drug administiration, for 8,000 Hz stimulus, group 1 had significantly higher threshold shifts when compared with groups 2 (
p
< 0.05) and 3 (
p
< 0.05), and there was no significant difference in threshold shifts between groups 2 and 3 (
p
> 0.05). Transmission electron microscopy findings demonstrated the protective effect of riluzole on the hair cells and the stria vascularis, especially in the group treated with 8 mg/kg of riluzole hydrochloride.
Conclusion
We can say that riluzole may have a protective effect on cisplatin- induced ototoxicity. However, additional studies are needed to confirm these results and the mechanisms of action of riluzole.
Collapse
Affiliation(s)
- Selin Üstün Bezgin
- Department of Otorhinolaryngology, Kanuni Sultan Süleyman Education and Research Hospital, İstanbul, Turkey
| | - Kadir Kemal Uygur
- Department of Otorhinolaryngology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Çağıl Gökdoğan
- Department of Otorhinolaryngology, Muğla Sıtkı Koçman University Faculty of Medicine, Muğla, Turkey
| | - Çiğdem Elmas
- Department of Histology and Embryology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Güleser Göktaş
- Department of Histology and Embryology, Lokman Hekim University, Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
64
|
Zhao X, Cai A, Peng Z, Liang W, Xi H, Li P, Chen G, Yu J, Chen L. JS-K induces reactive oxygen species-dependent anti-cancer effects by targeting mitochondria respiratory chain complexes in gastric cancer. J Cell Mol Med 2019; 23:2489-2504. [PMID: 30672108 PMCID: PMC6433691 DOI: 10.1111/jcmm.14122] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 11/08/2018] [Accepted: 12/06/2018] [Indexed: 12/12/2022] Open
Abstract
As a nitric oxide (NO) donor prodrug, JS‐K inhibits cancer cell proliferation, induces the differentiation of human leukaemia cells, and triggers apoptotic cell death in various cancer models. However, the anti‐cancer effect of JS‐K in gastric cancer has not been reported. In this study, we found that JS‐K inhibited the proliferation of gastric cancer cells in vitro and in vivo and triggered mitochondrial apoptosis. Moreover, JS‐K induced a significant accumulation of reactive oxygen species (ROS), and the clearance of ROS by antioxidant reagents reversed JS‐K‐induced toxicity in gastric cancer cells and subcutaneous xenografts. Although JS‐K triggered significant NO release, NO scavenging had no effect on JS‐K‐induced toxicity in vivo and in vitro. Therefore, ROS, but not NO, mediated the anti‐cancer effects of JS‐K in gastric cancer. We also explored the potential mechanism of JS‐K‐induced ROS accumulation and found that JS‐K significantly down‐regulated the core proteins of mitochondria respiratory chain (MRC) complex I and IV, resulting in the reduction of MRC complex I and IV activity and the subsequent ROS production. Moreover, JS‐K inhibited the expression of antioxidant enzymes, including copper‐zinc‐containing superoxide dismutase (SOD1) and catalase, which contributed to the decrease of antioxidant enzymes activity and the subsequent inhibition of ROS clearance. Therefore, JS‐K may target MRC complex I and IV and antioxidant enzymes to exert ROS‐dependent anti‐cancer function, leading to the potential usage of JS‐K in the prevention and treatment of gastric cancer.
Collapse
Affiliation(s)
- Xudong Zhao
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Aizhen Cai
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zheng Peng
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Wenquan Liang
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Hongqing Xi
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Peiyu Li
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Guozhu Chen
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Jiyun Yu
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Lin Chen
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
65
|
Hazman Ö, Bozkurt MF, Fidan AF, Uysal FE, Çelik S. The Effect of Boric Acid and Borax on Oxidative Stress, Inflammation, ER Stress and Apoptosis in Cisplatin Toxication and Nephrotoxicity Developing as a Result of Toxication. Inflammation 2018; 41:1032-1048. [PMID: 29500724 DOI: 10.1007/s10753-018-0756-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The development of treatment protocols that can reduce side effects in chemotherapy applications is extremely important in terms of cancer treatment. In this context, it was aimed to investigate the effects of boric acid and borax on cisplatin toxicity (nephrotoxicity) in rats. In the experimental phase, eight groups were formed from rats. Boric acid and borax were given to the treatment groups with three different doses using gavage. On the fifth day of the study, cisplatin (10 mg/kg) was administered to all rats except the control group. At the end of the study, oxidative stress-related (GSH, MDA, PCO, GPx, 8-OHdG), inflammation-related (TNF-α, IL-1β, IL-18, MCP-1, ICAM, TGF-β), apoptosis-related (p53, caspase 1, 3, 8, 12, bcl-2, bcl-xL, NFkB), and ER stress-related (GRP78, ATF-6, PERK) basic parameters were analyzed in serum, erythrocyte, and kidney tissues. Kidney tissues were also examined by histopathological and immunohistochemical methods. Borax and boric acid at different doses decreased inflammation and oxidative stress caused by cisplatin toxicity and increased ER stress. As a result of the treatments applied to experimental animals, it was determined that boric acid and borax reduced apoptotic damage in kidney tissue, but the decrease was statistically significant only in 200 mg/kg boric acid-administered group. In the study, low anti-apoptotic effects of borate doses with the anti-inflammatory and antioxidant effect may be due to increased ER stress at the relevant doses. Further studies on the effects of boron compounds on ER stress and apoptotic mechanisms may clarify this issue. Thus, possible side effects or if there are new usage areas of borone compounds which have many usage areas in clinics can be detected.
Collapse
Affiliation(s)
- Ömer Hazman
- Department of Chemistry, Biochemistry Division, Faculty of Science and Arts, Afyon Kocatepe University, Afyonkarahisar, Turkey.
| | - Mehmet Fatih Bozkurt
- Department of Pathology, Faculty of Veterinary, Afyon Kocatepe University, Afyonkarahisar, Turkey
| | - Abdurrahman Fatih Fidan
- Department of Biochemistry, Faculty of Veterinary, Afyon Kocatepe University, Afyonkarahisar, Turkey
| | - Fadime Erkan Uysal
- Department of Chemistry, Biochemistry Division, Faculty of Science and Arts, Afyon Kocatepe University, Afyonkarahisar, Turkey
| | - Sefa Çelik
- Department of Biochemistry, Faculty of Medicine, Afyon Kocatepe University, Afyonkarahisar, Turkey
| |
Collapse
|
66
|
Lactobacillus supplementation prevents cisplatin-induced cardiotoxicity possibly by inflammation inhibition. Cancer Chemother Pharmacol 2018; 82:999-1008. [DOI: 10.1007/s00280-018-3691-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 09/20/2018] [Indexed: 12/18/2022]
|
67
|
Monroe JD, Millay MH, Patty BG, Smith ME. The curcuminoid, EF-24, reduces cisplatin-mediated reactive oxygen species in zebrafish inner ear auditory and vestibular tissues. J Clin Neurosci 2018; 57:152-156. [PMID: 30243600 DOI: 10.1016/j.jocn.2018.09.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 09/10/2018] [Indexed: 12/12/2022]
Abstract
Cisplatin is a widely used chemotherapy drug that can damage auditory and vestibular tissue and cause hearing and balance loss through the intracellular release of reactive oxygen species (ROS). Curcumin has anticancer efficacy and can also counteract cisplatin's damaging effect against sensory tissue by scavenging intracellular ROS, but curcumin's applicability is limited due to its low bioavailability. EF-24 is a synthetic curcumin analog that is more bioavailable than curcumin and can target cancer, but its effects against cisplatin-mediated ROS in auditory and vestibular tissue is currently unknown. In this study, we employed a novel zebrafish inner ear tissue culture system to determine if EF-24 counteracted cisplatin-mediated ROS release in two sensory endorgans, the saccule and the utricle. The zebrafish saccule is associated with auditory function and the utricle with vestibular function. Trimmed endorgans were placed in tissue culture media with a fluorescent reactive oxygen species indicator dye, and intracellular ROS release was measured using a spectrophotometer. We found that cisplatin treatment significantly increased ROS compared to controls, but that EF-24 treatment did not alter or even decreased ROS. Importantly, when equimolar cisplatin and EF-24 treatments are combined, ROS did not increase compared to controls. This suggests that EF-24 may be able to prevent intracellular ROS caused by cisplatin treatment in inner ear tissue.
Collapse
Affiliation(s)
- Jerry D Monroe
- Department of Biology, Western Kentucky University, 1906 College Heights Boulevard, #11080, Bowling Green, KY 42101-1080, United States
| | - Matthew H Millay
- Department of Biology, Western Kentucky University, 1906 College Heights Boulevard, #11080, Bowling Green, KY 42101-1080, United States
| | - Blaine G Patty
- Department of Biology, Western Kentucky University, 1906 College Heights Boulevard, #11080, Bowling Green, KY 42101-1080, United States
| | - Michael E Smith
- Department of Biology, Western Kentucky University, 1906 College Heights Boulevard, #11080, Bowling Green, KY 42101-1080, United States.
| |
Collapse
|
68
|
LKB1 loss is associated with glutathione deficiency under oxidative stress and sensitivity of cancer cells to cytotoxic drugs and γ-irradiation. Biochem Pharmacol 2018; 156:479-490. [PMID: 30222967 DOI: 10.1016/j.bcp.2018.09.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 09/13/2018] [Indexed: 01/25/2023]
Abstract
The liver kinase B1 (LKB1) gene is a tumor suppressor associated with the hereditary Peutz-Jeghers syndrome and frequently mutated in non-small cell lung cancer and in cervical cancer. Previous studies showed that the LKB1/AMPK axis is involved in regulation of cell death and survival under metabolic stress. By using isogenic pairs of cancer cell lines, we report here that the genetic loss of LKB1 was associated with increased intracellular levels of total choline containing metabolites and, under oxidative stress, it impaired maintenance of glutathione (GSH) levels. This resulted in markedly increased intracellular reactive oxygen species (ROS) levels and sensitivity to ROS-induced cell death. These effects were rescued by re-expression of LKB1 or pre-treatment with the anti-oxidant and GSH replenisher N-acetyl cysteine. This role of LKB1 in response to ROS-inducing agents was largely AMPK-dependent. Finally, we observed that LKB1 defective cells are highly sensitive to cisplatin and γ-irradiation in vitro, suggesting that LKB1 mutated tumors could be targeted by oxidative stress-inducing therapies.
Collapse
|
69
|
Liu Jun Zi Tang-A Potential, Multi-Herbal Complementary Therapy for Chemotherapy-Induced Neurotoxicity. Int J Mol Sci 2018; 19:ijms19041258. [PMID: 29690597 PMCID: PMC5979528 DOI: 10.3390/ijms19041258] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 04/10/2018] [Accepted: 04/18/2018] [Indexed: 12/25/2022] Open
Abstract
Liu Jun Zi Tang (LJZT) has been used to treat functional dyspepsia and depression, suggesting its effects on gastrointestinal and neurological functions. LJZT is currently used as a complementary therapy to attenuate cisplatin-induced side effects, such as dyspepsia. However, its effect on chemotherapy-induced neuropathic pain or neurotoxicity has rarely been studied. Thus, we explored potential mechanisms underlying LJZT protection against cisplatin-induced neurotoxicity. We observed that LJZT attenuated cisplatin-induced thermal hyperalgesia in mice and apoptosis in human neuroblastoma SH-SY5Y cells. Furthermore, it also attenuated cisplatin-induced cytosolic and mitochondrial free radical formation, reversed the cisplatin-induced decrease in mitochondrial membrane potential, and increased the release of mitochondrial pro-apoptotic factors. LJZT not only activated the peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) promoter region, but also attenuated the cisplatin-induced reduction of PGC-1α expression. Silencing of the PGC-1α gene counteracted the protection of LJZT. Taken together, LJZT mediated, through anti-oxidative effect and mitochondrial function regulation, to prevent cisplatin-induced neurotoxicity.
Collapse
|
70
|
Abbas HHK, Alhamoudi KMH, Evans MD, Jones GDD, Foster SS. MTH1 deficiency selectively increases non-cytotoxic oxidative DNA damage in lung cancer cells: more bad news than good? BMC Cancer 2018; 18:423. [PMID: 29661172 PMCID: PMC5903006 DOI: 10.1186/s12885-018-4332-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 04/04/2018] [Indexed: 12/18/2022] Open
Abstract
Background Targeted therapies are based on exploiting cancer-cell-specific genetic features or phenotypic traits to selectively kill cancer cells while leaving normal cells unaffected. Oxidative stress is a cancer hallmark phenotype. Given that free nucleotide pools are particularly vulnerable to oxidation, the nucleotide pool sanitising enzyme, MTH1, is potentially conditionally essential in cancer cells. However, findings from previous MTH1 studies have been contradictory, meaning the relevance of MTH1 in cancer is still to be determined. Here we ascertained the role of MTH1 specifically in lung cancer cell maintenance, and the potential of MTH1 inhibition as a targeted therapy strategy to improve lung cancer treatments. Methods Using siRNA-mediated knockdown or small-molecule inhibition, we tested the genotoxic and cytotoxic effects of MTH1 deficiency on H23 (p53-mutated), H522 (p53-mutated) and A549 (wildtype p53) non-small cell lung cancer cell lines relative to normal MRC-5 lung fibroblasts. We also assessed if MTH1 inhibition augments current therapies. Results MTH1 knockdown increased levels of oxidatively damaged DNA and DNA damage signaling alterations in all lung cancer cell lines but not normal fibroblasts, despite no detectable differences in reactive oxygen species levels between any cell lines. Furthermore, MTH1 knockdown reduced H23 cell proliferation. However, unexpectedly, it did not induce apoptosis in any cell line or enhance the effects of gemcitabine, cisplatin or radiation in combination treatments. Contrastingly, TH287 and TH588 MTH1 inhibitors induced apoptosis in H23 and H522 cells, but only increased oxidative DNA damage levels in H23, indicating that they kill cells independently of DNA oxidation and seemingly via MTH1-distinct mechanisms. Conclusions MTH1 has a NSCLC-specific p53-independent role for suppressing DNA oxidation and genomic instability, though surprisingly the basis of this may not be reactive-oxygen-species-associated oxidative stress. Despite this, overall our cell viability data indicates that targeting MTH1 will likely not be an across-the-board effective NSCLC therapeutic strategy; rather it induces non-cytotoxic DNA damage that could promote cancer heterogeneity and evolution. Electronic supplementary material The online version of this article (10.1186/s12885-018-4332-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hussein H K Abbas
- Department of Genetics and Genome Biology, University of Leicester, Leicester, Leicestershire, LE1 7RH, UK.,Department of Pathology and Forensic Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Kheloud M H Alhamoudi
- Department of Genetics and Genome Biology, University of Leicester, Leicester, Leicestershire, LE1 7RH, UK
| | - Mark D Evans
- Faculty of Health and Life Sciences, De Montfort University, Leicester, Leicestershire, LE1 9BH, UK
| | - George D D Jones
- Department of Genetics and Genome Biology, University of Leicester, Leicester, Leicestershire, LE1 7RH, UK.
| | - Steven S Foster
- Department of Genetics and Genome Biology, University of Leicester, Leicester, Leicestershire, LE1 7RH, UK.
| |
Collapse
|
71
|
Chemotherapeutic-Induced Cardiovascular Dysfunction: Physiological Effects, Early Detection-The Role of Telomerase to Counteract Mitochondrial Defects and Oxidative Stress. Int J Mol Sci 2018. [PMID: 29534446 PMCID: PMC5877658 DOI: 10.3390/ijms19030797] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Although chemotherapeutics can be highly effective at targeting malignancies, their ability to trigger cardiovascular morbidity is clinically significant. Chemotherapy can adversely affect cardiovascular physiology, resulting in the development of cardiomyopathy, heart failure and microvascular defects. Specifically, anthracyclines are known to cause an excessive buildup of free radical species and mitochondrial DNA damage (mtDNA) that can lead to oxidative stress-induced cardiovascular apoptosis. Therefore, oncologists and cardiologists maintain a network of communication when dealing with patients during treatment in order to treat and prevent chemotherapy-induced cardiovascular damage; however, there is a need to discover more accurate biomarkers and therapeutics to combat and predict the onset of cardiovascular side effects. Telomerase, originally discovered to promote cellular proliferation, has recently emerged as a potential mechanism to counteract mitochondrial defects and restore healthy mitochondrial vascular phenotypes. This review details mechanisms currently used to assess cardiovascular damage, such as C-reactive protein (CRP) and troponin levels, while also unearthing recently researched biomarkers, including circulating mtDNA, telomere length and telomerase activity. Further, we explore a potential role of telomerase in the mitigation of mitochondrial reactive oxygen species and maintenance of mtDNA integrity. Telomerase activity presents a promising indicator for the early detection and treatment of chemotherapy-derived cardiac damage.
Collapse
|
72
|
Ferreira Penêda J, Barros Lima N, Ribeiro L, Helena D, Domingues B, Condé A. Cochleotoxicity monitoring protocol. ACTA OTORRINOLARINGOLOGICA ESPANOLA 2018; 69:105-109. [DOI: 10.1016/j.otorri.2017.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/20/2017] [Accepted: 03/24/2017] [Indexed: 10/19/2022]
|
73
|
Cochleotoxicity monitoring protocol. ACTA OTORRINOLARINGOLOGICA ESPANOLA 2018. [DOI: 10.1016/j.otoeng.2018.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
74
|
Agarwal A, Kasinathan A, Ganesan R, Balasubramanian A, Bhaskaran J, Suresh S, Srinivasan R, Aravind KB, Sivalingam N. Curcumin induces apoptosis and cell cycle arrest via the activation of reactive oxygen species-independent mitochondrial apoptotic pathway in Smad4 and p53 mutated colon adenocarcinoma HT29 cells. Nutr Res 2018; 51:67-81. [PMID: 29673545 DOI: 10.1016/j.nutres.2017.12.011] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 12/31/2017] [Accepted: 12/31/2017] [Indexed: 11/25/2022]
Abstract
Curcumin is a natural dietary polyphenol compound that has various pharmacological activities such as antiproliferative and cancer-preventive activities on tumor cells. Indeed, the role reactive oxygen species (ROS) generated by curcumin on cell death and cell proliferation inhibition in colon cancer is poorly understood. In the present study, we hypothesized that curcumin-induced ROS may promote apoptosis and cell cycle arrest in colon cancer. To test this hypothesis, the apoptosis-inducing potential and cell cycle inhibition effect of ROS induced by curcumin was investigated in Smd4 and p53 mutated HT-29 colon adenocarcinoma cells. We found that curcumin treatment significantly increased the level of ROS in HT-29 cells in a dose- and time-dependent manner. Furthermore, curcumin treatment markedly decreased the cell viability and proliferation potential of HT-29 cells in a dose- and time-dependent manner. Conversely, generation of ROS and inhibitory effect of curcumin on HT-29 cells were abrogated by N-acetylcysteine treatment. In addition, curcumin treatment did not show any cytotoxic effects on HT-29 cells. Furthermore, curcumin-induced ROS generation caused the DNA fragmentation, chromatin condensation, and cell nuclear shrinkage and significantly increased apoptotic cells in a dose- and time-dependent manner in HT-29 cells. However, pretreatment of N-acetylcysteine inhibited the apoptosis-triggering effect of curcumin-induced ROS in HT-29 cells. In addition, curcumin-induced ROS effectively mediated cell cycle inhibition in HT-29 cells. In conclusion, our data provide the first evidence that curcumin induces ROS independent apoptosis and cell cycle arrest in colon cancer cells that carry mutation on Smad4 and p53.
Collapse
Affiliation(s)
- Ayushi Agarwal
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamilnadu, India
| | - Akiladdevi Kasinathan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamilnadu, India
| | - Ramamoorthi Ganesan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamilnadu, India
| | - Akhila Balasubramanian
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamilnadu, India
| | - Jahnavi Bhaskaran
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamilnadu, India
| | - Samyuktha Suresh
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamilnadu, India
| | - Revanth Srinivasan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamilnadu, India
| | - K B Aravind
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamilnadu, India
| | - Nageswaran Sivalingam
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamilnadu, India.
| |
Collapse
|
75
|
Hany E, Sobh MA, ElKhier MTA, ElSabaa HM, Zaher AR. The Effect of Different Routes of Injection of Bone Marrow Mesenchymal Stem Cells on Parotid Glands of Rats Receiving Cisplatin: A Comparative Study. Int J Stem Cells 2017; 10:169-178. [PMID: 28844126 PMCID: PMC5741198 DOI: 10.15283/ijsc17022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2017] [Indexed: 12/24/2022] Open
Abstract
Background and Objectives Cisplatin is a powerful antitumor chemotherapeutic agent that is widely used in the treatment of many cancers but it has many side effects on many organs including salivary glands. Bone marrow is considered to be a rich environment that comprises many types of stem cells of which BMSCs (Bone marrow mesenchymal stem cells) are the most studied with potentiality to differentiate into many cell types. This study was conducted to evaluate the effect of different routes of injection of BMSCs on parotid glands of rats receiving cisplatin. Methods and Results Sprague-Dawley rats were divided into 3 groups: a negative control group receiving phosphate buffered saline, a positive control group receiving cisplatin, and an experimental group where rats received cisplatin and then received iron oxide-labeled BMSCs by either intravenous or intraparotid routes or both. Animals were sacrificed at periods of 3,6,10 and 15 days after cisplatin injection, then histological, ultrastructural and immunohistochemical studies were done. The experimental stem cell treated group showed better histological features and increased PCNA proliferation index when compared to the control. The systemic and combination groups showed better results than the local group. Iron oxide-labeled cells were detected with Prussian blue stain. Conclusions This study proved that BMSCs can improve cisplatin induced cytotoxicity in parotid glands. Systemic administration showed to have a better effect than local intraparotid administration and comparable effect to combined administration.
Collapse
Affiliation(s)
- Eman Hany
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, Egypt
| | - Mohammed A Sobh
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Mazen T Abou ElKhier
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, Egypt
| | - Heba M ElSabaa
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, Egypt.,Department of Oral Biology, School of Dentistry, Badr University, Cairo, Egypt
| | - Ahmed R Zaher
- Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura, Egypt
| |
Collapse
|
76
|
Martín-Saldaña S, Palao-Suay R, Aguilar MR, García-Fernández L, Arévalo H, Trinidad A, Ramírez-Camacho R, San Román J. pH-sensitive polymeric nanoparticles with antioxidant and anti-inflammatory properties against cisplatin-induced hearing loss. J Control Release 2017; 270:53-64. [PMID: 29197586 DOI: 10.1016/j.jconrel.2017.11.032] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 11/17/2017] [Accepted: 11/20/2017] [Indexed: 11/28/2022]
Abstract
Polymeric nanoparticles (NPs) based on smart synthetic amphiphilic copolymers are used to transport and controlled release dexamethasone in the inner ear to protect against the ototoxic effect of cisplatin. The NPs were based on a mixture of two pseudo-block polymer drugs obtained by free radical polymerization: poly(VI-co-HEI) and poly(VP-co-MVE) or poly(VP-co-MTOS), being VI 1-vinylimidazole, VP N-vinylpyrrolidone, and HEI, MVE and MTOS the methacrylic derivatives of ibuprofen, α-tocopherol and α-tocopheryl succinate, respectively. The NPs were obtained by nanoprecipitation with appropriate hydrodynamic properties, and isoelectric points that matched the pH of inflamed tissue. The NPs were tested both in vitro (using HEI-OC1 cells) and in vivo (using a murine model) with good results. Although the concentration of dexamethasone administered in the NPs is around two orders of magnitude lower that the conventional treatment for intratympanic administration, the NPs protected from the cytotoxic effect of cisplatin when the combination of the appropriate properties in terms of size, zeta potential, encapsulation efficiency and isoelectric point were achieved. To the best of our knowledge this is the first time that pH sensitive NPs are used to protect from cisplatin-induced hearing loss by intratympanic administration.
Collapse
Affiliation(s)
- Sergio Martín-Saldaña
- Grupo de Biomateriales, Departamento de Nanomateriales Poliméricos y Biomateriales, Instituto de Ciencia y Tecnología de Polímeros, ICTP-CSIC, C/Juan de la Cierva, 3, 28006 Madrid, Spain; Ear Research Group, Hospital UniversitarioPuerta de Hierro Majadahonda, Health Research Institute Puerta de Hierro, Madrid, Spain
| | - Raquel Palao-Suay
- Grupo de Biomateriales, Departamento de Nanomateriales Poliméricos y Biomateriales, Instituto de Ciencia y Tecnología de Polímeros, ICTP-CSIC, C/Juan de la Cierva, 3, 28006 Madrid, Spain; Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Spain
| | - María Rosa Aguilar
- Grupo de Biomateriales, Departamento de Nanomateriales Poliméricos y Biomateriales, Instituto de Ciencia y Tecnología de Polímeros, ICTP-CSIC, C/Juan de la Cierva, 3, 28006 Madrid, Spain; Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Spain.
| | - Luis García-Fernández
- Grupo de Biomateriales, Departamento de Nanomateriales Poliméricos y Biomateriales, Instituto de Ciencia y Tecnología de Polímeros, ICTP-CSIC, C/Juan de la Cierva, 3, 28006 Madrid, Spain; Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Spain
| | - Humberto Arévalo
- Grupo de Biomateriales, Departamento de Nanomateriales Poliméricos y Biomateriales, Instituto de Ciencia y Tecnología de Polímeros, ICTP-CSIC, C/Juan de la Cierva, 3, 28006 Madrid, Spain
| | - Almudena Trinidad
- Ear Research Group, Hospital UniversitarioPuerta de Hierro Majadahonda, Health Research Institute Puerta de Hierro, Madrid, Spain
| | - Rafael Ramírez-Camacho
- Ear Research Group, Hospital UniversitarioPuerta de Hierro Majadahonda, Health Research Institute Puerta de Hierro, Madrid, Spain
| | - Julio San Román
- Grupo de Biomateriales, Departamento de Nanomateriales Poliméricos y Biomateriales, Instituto de Ciencia y Tecnología de Polímeros, ICTP-CSIC, C/Juan de la Cierva, 3, 28006 Madrid, Spain; Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Spain
| |
Collapse
|
77
|
Naples J, Cox R, Bonaiuto G, Parham K. Prestin as an Otologic Biomarker of Cisplatin Ototoxicity in a Guinea Pig Model. Otolaryngol Head Neck Surg 2017; 158:541-546. [DOI: 10.1177/0194599817742093] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Objective To evaluate (1) whether changes in serum prestin aid in early detection of cisplatin ototoxicity, (2) the role of diltiazem as an otoprotectant, and (3) whether prestin levels are sensitive to effects of diltiazem. Study Design Experimental animal study. Setting Translational research laboratory. Subjects Twenty female guinea pigs. Methods Two groups of 10 guinea pigs were used. The relationship between serum prestin levels and auditory brainstem response (ABR) thresholds was compared between the groups. All animals had baseline blood draws and ABR thresholds recorded prior to cisplatin administration. Intraperitoneal cisplatin bolus (8 mg/kg) was administered followed by 5 consecutive days of intratympanic (IT) diltiazem (2 mg/kg) or sham IT-saline injection. Serum prestin levels and ABR thresholds were measured at days 1, 2, 3, 7, and 14 postcisplatin. Results In sham, IT-saline–treated animals, mean prestin levels were elevated above baseline on days 1 to 7. The prestin levels were significantly elevated from baseline on day 1 ( P < .001), while significant ABR threshold elevations did not occur until day 2 ( P = .028) for click-evoked ABRs and day 3 ( P = .041) for tones. In diltiazem-treated animals, prestin levels were not elevated above baseline but ABR thresholds were elevated on days 1 to 3. However, the thresholds returned toward baseline on days 7 and 14. Conclusion Changes in serum prestin levels were detectable prior to shifts in ABR thresholds in a guinea pig cisplatin ototoxicity model. These changes did not occur in diltiazem-treated animals. Prestin may serve as a biomarker of cochlear injury that is sensitive to therapeutic interventions in cisplatin ototoxicity.
Collapse
Affiliation(s)
- James Naples
- Division of Otolaryngology–Head & Neck Surgery, Department of Surgery, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Robert Cox
- University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Gregory Bonaiuto
- Division of Otolaryngology–Head & Neck Surgery, Department of Surgery, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Kourosh Parham
- Division of Otolaryngology–Head & Neck Surgery, Department of Surgery, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| |
Collapse
|
78
|
Ju SM, Jo YS, Jeon YM, Pae HO, Kang DG, Lee HS, Bae JS, Jeon BH. Phosphorylation of eIF2α suppresses cisplatin-induced p53 activation and apoptosis by attenuating oxidative stress via ATF4-mediated HO-1 expression in human renal proximal tubular cells. Int J Mol Med 2017; 40:1957-1964. [PMID: 29039478 DOI: 10.3892/ijmm.2017.3181] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 09/07/2017] [Indexed: 11/06/2022] Open
Abstract
Cisplatin is one of the most widely used chemotherapeutic agents for the treatment of human cancers. However, the nephrotoxicity of cisplatin limits its use as a therapeutic agent. It has been suggested that oxidative stress and p53 activation play important roles in cisplatin-induced nephrotoxicity. It has been demonstrated that the eukaryotic translation initiation factor 2α (eIF2α) may protect HK-2 human renal proximal tubular cells against cisplatin-induced apoptosis through inhibition of reactive oxygen species (ROS)‑mediated p53 activation. The aim of the present study was to investigate the effects of siRNA‑mediated knockdown of the PKR-like endoplasmic reticulum kinase (PERK) gene, which induces the phosphorylation of eIF2α, or Sal003, a selective inhibitor of eIF2α dephosphorylation, on cisplatin‑induced apoptosis in HK-2 cells. Cisplatin induced eIF2α phosphorylation as well as p53 activation. In particular, inhibition of p53 by pifithrin‑α, and upregulation of eIF2α phosphorylation by Sal003, reduced cisplatin-induced apoptosis. Of note, Sal003‑mediated upregulation of eIF2α phosphorylation suppressed cisplatin‑induced p53 activation. Furthermore, reduction of eIF2α phosphorylation by PERK knockdown enhanced cisplatin-induced p53 activation and apoptosis. In addition, the ROS scavenger N-acetyl-L-cysteine inhibited eIF2α phosphorylation as well as p53 activation in HK-2 cells treated with cisplatin, suggesting that oxidative stress induced by cisplatin may lead to apoptosis through p53 activation; furthermore, this stress may confer resistance to apoptosis via eIF2α phosphorylation, which was further supported by the finding that cisplatin‑induced ROS generation was attenuated by Sal003, whereas it was enhanced by PERK knockdown. Furthermore, cisplatin induced the expression of activating transcription factor 4 (ATF4) and heme oxygenase-1 (HO-1) that were enhanced by Sal003 and reduced by PERK knockdown. Taken together, these results suggest that phosphorylation of eIF2α suppresses cisplatin‑induced p53 activation and apoptosis by attenuating oxidative stress via ATF4-mediated HO-1 expression in HK-2 cells, as ATF4 expression is usually dependent on the phosphorylation of eIF2α and may also transcriptionally induce the expression of HO-1 in response to oxidative stress. Therefore, regulation of eIF2α phosphorylation may play an important role in alleviating cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Sung-Min Ju
- Department of Pathology, College of Korean Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Yong-Seok Jo
- Department of Pathology, College of Korean Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Yoo-Min Jeon
- Department of Pathology, College of Korean Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Hyun-Ock Pae
- Department of Microbiology and Immunology, School of Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Dae-Gill Kang
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Ho-Sub Lee
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| | - Jun-Sang Bae
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine and Institute for Medical Sciences, Jeonju, Jeonbuk 54907, Republic of Korea
| | - Byung-Hun Jeon
- Department of Pathology, College of Korean Medicine, Wonkwang University, Iksan, Jeonbuk 54538, Republic of Korea
| |
Collapse
|
79
|
Tian L, Zhang J, Ren X, Liu X, Gao W, Zhang C, Sun Y, Liu M. Overexpression of miR-26b decreases the cisplatin-resistance in laryngeal cancer by targeting ATF2. Oncotarget 2017; 8:79023-79033. [PMID: 29108284 PMCID: PMC5668017 DOI: 10.18632/oncotarget.20784] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 08/09/2017] [Indexed: 01/09/2023] Open
Abstract
Cisplatin is a common used chemotherapeutic drug for the treatment of laryngeal cancer. However, drug-resistance is a major obstacle in platinum-based chemotherapy for laryngeal cancer. Recent studies have demonstrated that dysregulation of microRNAs (miRNAs) is responsible for chemoresistance in multiple cancers including laryngeal cancer, but the potential mechanisms are required to be explored. In the present study, we constantly exposed the laryngeal cancer cell line Hep-2 with cisplatin to establish a cisplatin-resistant laryngeal cancer cell model (Hep-2/R). We found that Hep-2/R cells exhibited obvious resistance to cisplatin compared to the Hep-2 cells. However, overexpression of miR-26b significantly decreased the half maximal inhibitory concentration (IC50) of cisplatin to Hep-2/R. Mechanically, miR-26b in Hep-2/R decreased the expression of ATF2, and thus inhibiting the phosphorylation of ATF2 and formation of cellular ATF2-c-Jun complex induced by cisplatin. As the results, Hep-2/R cells failed to overexpress the Bcl-xl which is a key anti-apoptotic protein under the cisplatin treatment. Therefore, overexpression of miR-26b was found to be able to promote mitochondrial apoptosis induced by cisplatin.
Collapse
Affiliation(s)
- Linli Tian
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China, 150086
| | - Jiarui Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China, 150086
| | - Xiuxia Ren
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China, 150086
| | - Xinyu Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China, 150086
| | - Wei Gao
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China, 150086
| | - Chen Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China, 150086
| | - Yanan Sun
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China, 150086
| | - Ming Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, The Second Affiliated Hospital, Harbin Medical University, Harbin, China, 150086
| |
Collapse
|
80
|
Zheng Y, Deng Z, Yin J, Wang S, Lu D, Wen X, Li X, Xiao D, Hu C, Chen X, Zhang W, Zhou H, Liu Z. The association of genetic variations in DNA repair pathways with severe toxicities in NSCLC patients undergoing platinum‐based chemotherapy. Int J Cancer 2017; 141:2336-2347. [PMID: 28791697 DOI: 10.1002/ijc.30921] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 06/25/2017] [Accepted: 07/25/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Yi Zheng
- Department of Clinical PharmacologyXiangya Hospital, Central South UniversityChangsha410008 People's Republic of China
- Hunan Key Laboratory of PharmacogeneticsInstitute of Clinical Pharmacology, Central South UniversityChangsha410078 People's Republic of China
- Key Laboratory of Hunan Province for Traditional Chinese Medicine in Obstetrics and Gynecology Research, Hunan Provincial Maternal and Child Health Care HospitalChangsha410008 People's Republic of China
| | - Zheng Deng
- Department of Respiratory MedicineXiangya Hospital, Central South UniversityChangsha Hunan410008 People's Republic of China
| | - Jiye Yin
- Department of Clinical PharmacologyXiangya Hospital, Central South UniversityChangsha410008 People's Republic of China
- Hunan Key Laboratory of PharmacogeneticsInstitute of Clinical Pharmacology, Central South UniversityChangsha410078 People's Republic of China
| | - Shiming Wang
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary AnthropologyInstitute of Genetics, School of Life Sciences, Fudan UniversityShanghai20000 People's Republic of China
| | - Daru Lu
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary AnthropologyInstitute of Genetics, School of Life Sciences, Fudan UniversityShanghai20000 People's Republic of China
| | - Xiaoke Wen
- Key Laboratory of Hunan Province for Traditional Chinese Medicine in Obstetrics and Gynecology Research, Hunan Provincial Maternal and Child Health Care HospitalChangsha410008 People's Republic of China
| | - Xiangping Li
- Department of PharmacyXiangya Hospital, Central South UniversityChangsha410008 People's Republic of China
| | - Di Xiao
- Department of PharmacyXiangya Hospital, Central South UniversityChangsha410008 People's Republic of China
| | - Chengping Hu
- Department of Respiratory MedicineXiangya Hospital, Central South UniversityChangsha Hunan410008 People's Republic of China
| | - Xiang Chen
- Department of DermatologyXiangya Hospital, Central South UniversityChangsha Hunan410008 People's Republic of China
| | - Wei Zhang
- Department of Clinical PharmacologyXiangya Hospital, Central South UniversityChangsha410008 People's Republic of China
- Hunan Key Laboratory of PharmacogeneticsInstitute of Clinical Pharmacology, Central South UniversityChangsha410078 People's Republic of China
| | - Honghao Zhou
- Department of Clinical PharmacologyXiangya Hospital, Central South UniversityChangsha410008 People's Republic of China
- Hunan Key Laboratory of PharmacogeneticsInstitute of Clinical Pharmacology, Central South UniversityChangsha410078 People's Republic of China
| | - Zhaoqian Liu
- Department of Clinical PharmacologyXiangya Hospital, Central South UniversityChangsha410008 People's Republic of China
- Hunan Key Laboratory of PharmacogeneticsInstitute of Clinical Pharmacology, Central South UniversityChangsha410078 People's Republic of China
| |
Collapse
|
81
|
Yu Z, Cowan JA. Catalytic Metallodrugs: Substrate-Selective Metal Catalysts as Therapeutics. Chemistry 2017; 23:14113-14127. [PMID: 28688119 DOI: 10.1002/chem.201701714] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Zhen Yu
- Department of Chemistry and Biochemistry; The Ohio State University; 100 West 18th Avenue Columbus OH 43210 USA
| | - James A. Cowan
- Department of Chemistry and Biochemistry; The Ohio State University; 100 West 18th Avenue Columbus OH 43210 USA
| |
Collapse
|
82
|
FTY720 Induces Autophagy-Associated Apoptosis in Human Oral Squamous Carcinoma Cells, in Part, through a Reactive Oxygen Species/Mcl-1-Dependent Mechanism. Sci Rep 2017; 7:5600. [PMID: 28717222 PMCID: PMC5514089 DOI: 10.1038/s41598-017-06047-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 06/07/2017] [Indexed: 11/14/2022] Open
Abstract
In this study, we interrogated the mechanism by which the immunosuppressant FTY720 mediates anticancer effects in oral squamous cell carcinoma (OSCC) cells. FTY720 differentially suppressed the viability of the OSCC cell lines SCC4, SCC25, and SCC2095 with IC50 values of 6.1, 6.3, and 4.5 μM, respectively. This antiproliferative effect was attributable to the ability of FTY720 to induce caspase-dependent apoptosis. Mechanistic evidence suggests that FTY720-induced apoptosis was associated with its ability to inhibit Akt-NF-κB signaling, to facilitate the proteasomal degradation of the antiapoptotic protein Mcl-1, and to increase reactive oxygen species (ROS) generation. Both overexpression of Mcl-1 and inhibition of ROS partially protected cells from FTY720-induced caspase-9 activation, PARP cleavage and cytotoxicity. In addition, FTY720 induced autophagy in OSCC cells, as manifested by LC3B-II conversion, decreased p62 expression, and accumulation of autophagosomes. Inhibition of autophagy by bafilomycin A1 protected cells from FTY720-induced apoptosis. Together, these findings suggest an intricate interplay between autophagy and apoptosis in mediating the tumor-suppressive effect in OSCC cells, which underlies the translational potential of FTY720 in fostering new therapeutic strategies for OSCC.
Collapse
|
83
|
El-Shitany NA, Eid B. Proanthocyanidin protects against cisplatin-induced oxidative liver damage through inhibition of inflammation and NF-κβ/TLR-4 pathway. ENVIRONMENTAL TOXICOLOGY 2017; 32:1952-1963. [PMID: 28371137 DOI: 10.1002/tox.22418] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/02/2017] [Accepted: 03/05/2017] [Indexed: 06/07/2023]
Abstract
Although cisplatin (CIS) is a highly effective anticancer drug, hepatotoxicity is one of the most common adverse effects associated with its use. Recently, reactive oxygen species (ROS) and inflammation are suggested to be key factors in the pathophysiology of CIS-induced acute liver damage. The aim of this study is to investigate the possible protective effect of proanthocyanidin (PRO) against CIS-induced acute hepatotoxicity. Rats were divided into four groups: 1, Control; 2, PRO; 3, CIS; and 4, PRO + CIS. Biochemical studies and histopathology were used to assess liver damage. ROS, inflammatory cytokines, nuclear factor kappa beta (NF-κβ), inducible cyclooxygenase enzyme (COX-2), inducible nitric oxide synthase (iNOS), toll-like receptor-4 (TLR-4) gene expression, and apoptotic markers were also assessed. PRO pretreatment protected the liver against CIS-induced toxicity as indicated by decreased plasma levels of liver function enzymes and the normal liver histopathology observed in the PRO + CIS group. PRO pretreatment also diminished indicators of oxidative stress in the liver, including nitric oxide (NO) and malondialdehyde (MDA). It also increased the antioxidants, reduced glutathione (GSH), glutathione peroxidase (GPx), superoxide dismutase (SOD), and catalase (CAT) in the liver. Plasma interleukin-1 beta (IL-1β), IL-6, and tumor necrosis factor-alpha (TNF-α) were all reduced. Liver gene expression of NF-κβ, COX-2, iNOS, and TLR-4 were all downregulated. Furthermore, PRO administration downregulated the liver expression of the apoptotic marker, Bax, while upregulated the antiapoptotic marker, Bcl2. In conclusion, our results revealed that PRO may protect against CIS-induced acute liver damage mainly through inhibition of ROS, inflammation, and apoptosis.
Collapse
Affiliation(s)
- Nagla A El-Shitany
- Department of Pharmacology and Toxicolog, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Basma Eid
- Department of Pharmacology and Toxicolog, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
84
|
Naples JG. Calcium-channel blockers as therapeutic agents for acquired sensorineural hearing loss. Med Hypotheses 2017; 104:121-125. [PMID: 28673569 DOI: 10.1016/j.mehy.2017.05.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 05/28/2017] [Accepted: 05/31/2017] [Indexed: 11/27/2022]
Abstract
Acquired sensorineural hearing loss represents a challenging clinical scenario. Currently, there are few approved therapies for treating this type of hearing loss, and diagnosis is often made after permanent damage has occurred. There are numerous etiologies for acquired hearing loss, with complex mechanisms underlying each cause. Despite these complexities, apoptosis of the structures within the inner ear, is a theme common to many forms of acquired hearing loss. Apoptosis is a calcium-dependent process, and within the inner ear, L- and T-type calcium channels are believed to contribute to calcium availability during this process. There are few studies limited to animal models evaluating the role of calcium-channel blockers (CCBs) as otoprotective agents in the setting of acquired hearing loss. Here, I hypothesize that CCBs will provide utility as a therapy against acquired forms of sensorineural hearing loss by preventing calcium influx that occurs during inner ear cellular apoptosis.
Collapse
Affiliation(s)
- James G Naples
- University of Connecticut Health Center, 263 Farmington Ave, Farmington, CT 06030, United States.
| |
Collapse
|
85
|
Shang D, Wu J, Guo L, Xu Y, Liu L, Lu J. Metformin increases sensitivity of osteosarcoma stem cells to cisplatin by inhibiting expression of PKM2. Int J Oncol 2017; 50:1848-1856. [PMID: 28393220 DOI: 10.3892/ijo.2017.3950] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 03/20/2017] [Indexed: 12/17/2022] Open
Abstract
Multiple drug resistance is reported to be a major obstacle in treatment of osteosarcoma (OS). Research has demonstrated that small subsets of cells called cancer stem cells (CSCs) are responsible for multiple drug resistance. CSCs are potential targets for reversing chemoresistance. In the present study, we compared cisplatin sensitivity between OS stem cells and OS non-stem cells. We confirmed that OS stem cells showed significant cisplatin-resistance compared with the OS non-CSCs. Mechanically, we proved that overexpression of the pyruvate kinase isoenzyme M2 (PKM2) was responsible for the resistance to cisplatin in OS stem cells. As a potential strategy, we found that co-treatment with metformin significantly decreased the half maximal inhibitory concentration (IC50) of cisplatin to HOS OS stem cells by downregulating the expression of PKM2. PKM2 downregulation resulted in, metformin inhibited glucose uptake, lactate production and ATP production in HOS CSCs. Therefore, metformin impaired the resistance of HOS CSCs to cisplatin and promoted cisplatin-induced apoptosis. In addition, antitumor effects of other chemotherapeutic drugs such as doxorubicin and 5-fluorouracil were proved to be enhanced by metformin on OS stem cells.
Collapse
Affiliation(s)
- Depeng Shang
- Department of Orthopedics, Zhongshan Hospital Affiliated of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Ju Wu
- Department of General Surgery, Zhongshan Hospital Affiliated of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Lianyi Guo
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Yanju Xu
- Department of Orthopedics, Zhongshan Hospital Affiliated of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Lezi Liu
- Department of Orthopedics, Zhongshan Hospital Affiliated of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Jianmin Lu
- Department of Orthopedics, Zhongshan Hospital Affiliated of Dalian University, Dalian, Liaoning 116001, P.R. China
| |
Collapse
|
86
|
Gentile F, Arcaro A, Pizzimenti S, Daga M, Cetrangolo GP, Dianzani C, Lepore A, Graf M, Ames PRJ, Barrera G. DNA damage by lipid peroxidation products: implications in cancer, inflammation and autoimmunity. AIMS GENETICS 2017; 4:103-137. [PMID: 31435505 PMCID: PMC6690246 DOI: 10.3934/genet.2017.2.103] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 04/12/2017] [Indexed: 12/19/2022]
Abstract
Oxidative stress and lipid peroxidation (LPO) induced by inflammation, excess metal storage and excess caloric intake cause generalized DNA damage, producing genotoxic and mutagenic effects. The consequent deregulation of cell homeostasis is implicated in the pathogenesis of a number of malignancies and degenerative diseases. Reactive aldehydes produced by LPO, such as malondialdehyde, acrolein, crotonaldehyde and 4-hydroxy-2-nonenal, react with DNA bases, generating promutagenic exocyclic DNA adducts, which likely contribute to the mutagenic and carcinogenic effects associated with oxidative stress-induced LPO. However, reactive aldehydes, when added to tumor cells, can exert an anticancerous effect. They act, analogously to other chemotherapeutic drugs, by forming DNA adducts and, in this way, they drive the tumor cells toward apoptosis. The aldehyde-DNA adducts, which can be observed during inflammation, play an important role by inducing epigenetic changes which, in turn, can modulate the inflammatory process. The pathogenic role of the adducts formed by the products of LPO with biological macromolecules in the breaking of immunological tolerance to self antigens and in the development of autoimmunity has been supported by a wealth of evidence. The instrumental role of the adducts of reactive LPO products with self protein antigens in the sensitization of autoreactive cells to the respective unmodified proteins and in the intermolecular spreading of the autoimmune responses to aldehyde-modified and native DNA is well documented. In contrast, further investigation is required in order to establish whether the formation of adducts of LPO products with DNA might incite substantial immune responsivity and might be instrumental for the spreading of the immunological responses from aldehyde-modified DNA to native DNA and similarly modified, unmodified and/or structurally analogous self protein antigens, thus leading to autoimmunity.
Collapse
Affiliation(s)
- Fabrizio Gentile
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
| | - Alessia Arcaro
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
| | - Stefania Pizzimenti
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Martina Daga
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | | | - Chiara Dianzani
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Alessio Lepore
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Maria Graf
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Paul R. J. Ames
- CEDOC, NOVA Medical School, Universidade NOVA de Lisboa, Lisboa, Portugal, and Department of Haematology, Dumfries Royal Infirmary, Dumfries, Scotland, UK
| | - Giuseppina Barrera
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| |
Collapse
|
87
|
Martín-Saldaña S, Palao-Suay R, Aguilar MR, Ramírez-Camacho R, San Román J. Polymeric nanoparticles loaded with dexamethasone or α-tocopheryl succinate to prevent cisplatin-induced ototoxicity. Acta Biomater 2017; 53:199-210. [PMID: 28213099 DOI: 10.1016/j.actbio.2017.02.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 01/23/2017] [Accepted: 02/11/2017] [Indexed: 12/20/2022]
Abstract
The aim of this work is the development of highly protective agents to be administered locally within the middle ear to avoid cisplatin-induced ototoxicity, which affects to 100% of the clinical patients at ultra-high concentrations (16mg/kg). The protective agents are based on polymeric nanoparticles loaded with dexamethasone or α-tocopheryl succinate as anti-inflammarory and anti-apoptotic molecules. Dexamethasone and α-tocopheryl succinate are poorly soluble in water and present severe side effects when systemic administered during long periods of time. Their incorporation in the hydrophobic core of nanoparticles with the appropriate hydrodynamic properties provides the desired effects in vitro (lower cisplatin-induced toxicity, decreasing of caspase 3/7 activity, and lower IL-1β release) and in vivo (reducing the hearing loss at the local level). The local administration of the nanoparticles by bullostomy provides an adequate dose of drug without systemic interference with the chemotherapeutic effect of cisplatin. STATEMENT OF SIGNIFICANCE 100% of the cancer patients receiving ultra-high doses of CDDP (16mg/kg) suffer severe hearing loss, being a limiting factor in antineoplastic treatments. In this paper we describe the application of polymeric nanoparticles loaded with dexamethasone or α-tocopheryl succinate to palliate the cisplatin ototoxicity derived from chemotherapy treatment. These new nanoparticles, that encapsulate, transport, and deliver dexamethasone or α-tocopheryl succinate in the middle ear, are able to partially prevent ototoxicity derived from high doses of CDDP. This is an interdisciplinary study in which in vitro and in vivo experiments are described and extensively discussed. The importance of the results opens an excellent opportunity to the translation to the clinic.
Collapse
Affiliation(s)
- Sergio Martín-Saldaña
- Department of Otorhinolaryngology, Puerta de Hierro Majadahonda University Hospital, C/ Manuel de Falla, 1, 28222 Majadahonda, Spain; Group of Biomaterials, Department of Polymeric Nanomaterials and Biomaterials, Institute of Polymer Science and Technology, CSIC, C/ Juan de la Cierva, 3, 28006 Madrid, Spain
| | - Raquel Palao-Suay
- Group of Biomaterials, Department of Polymeric Nanomaterials and Biomaterials, Institute of Polymer Science and Technology, CSIC, C/ Juan de la Cierva, 3, 28006 Madrid, Spain; Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Spain
| | - María Rosa Aguilar
- Group of Biomaterials, Department of Polymeric Nanomaterials and Biomaterials, Institute of Polymer Science and Technology, CSIC, C/ Juan de la Cierva, 3, 28006 Madrid, Spain; Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Spain.
| | - Rafael Ramírez-Camacho
- Department of Otorhinolaryngology, Puerta de Hierro Majadahonda University Hospital, C/ Manuel de Falla, 1, 28222 Majadahonda, Spain; Universidad Autónoma de Madrid, Cantoblanco Campus University, 28049 Madrid, Spain
| | - Julio San Román
- Group of Biomaterials, Department of Polymeric Nanomaterials and Biomaterials, Institute of Polymer Science and Technology, CSIC, C/ Juan de la Cierva, 3, 28006 Madrid, Spain; Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Spain
| |
Collapse
|
88
|
Kisselev PA, Panibrat OV, Sysa AR, Anisovich MV, Zhabinskii VN, Khripach VA. Flow-cytometric analysis of reactive oxygen species in cancer cells under treatment with brassinosteroids. Steroids 2017; 117:11-15. [PMID: 27343978 DOI: 10.1016/j.steroids.2016.06.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 06/14/2016] [Accepted: 06/20/2016] [Indexed: 12/19/2022]
Abstract
To explore the underlying mechanism of cancer cell growth inhibition by brassinosteroids (BS), reactive oxygen species (ROS) generation under treatment with 28-homocastasterone and its synthetic derivatives (22S,23S)-28-homocastasterone was measured in A549 human lung adenocarcinoma cells. BS induced ROS generation in A549 cells and their growth in a time and dose-dependent manner. The maximal effect was observed for (22S,23S)-28-homocastasterone which at 30μM concentration showed a 6-fold increase of ROS generation in comparison with the control.
Collapse
Affiliation(s)
- Pyotr A Kisselev
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Kuprevich str., 5/2, 220141 Minsk, Belarus
| | - Olesya V Panibrat
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Kuprevich str., 5/2, 220141 Minsk, Belarus
| | - Aliaksei R Sysa
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Kuprevich str., 5/2, 220141 Minsk, Belarus
| | - Marina V Anisovich
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Kuprevich str., 5/2, 220141 Minsk, Belarus
| | - Vladimir N Zhabinskii
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Kuprevich str., 5/2, 220141 Minsk, Belarus.
| | - Vladimir A Khripach
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Kuprevich str., 5/2, 220141 Minsk, Belarus
| |
Collapse
|
89
|
Youn CK, Kim J, Jo ER, Oh J, Do NY, Cho SI. Protective Effect of Tempol against Cisplatin-Induced Ototoxicity. Int J Mol Sci 2016; 17:ijms17111931. [PMID: 27869744 PMCID: PMC5133926 DOI: 10.3390/ijms17111931] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 11/01/2016] [Accepted: 11/15/2016] [Indexed: 01/22/2023] Open
Abstract
One of the major adverse effects of cisplatin chemotherapy is hearing loss. Cisplatin-induced ototoxicity hampers treatment because it often necessitates dose reduction, which decreases cisplatin efficacy. This study was performed to investigate the effect of Tempol on cisplatin-induced ototoxicity in an auditory cell line, House Ear Institute-Organ of Corti 1 (HEI-OC1). Cultured HEI-OC1 cells were exposed to 30 μM cisplatin for 24 h with or without a 2 h pre-treatment with Tempol. Cell viability was determined using 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay and apoptotic cells were identified using terminal deoxynucleotidyl transferase dUTP nick end labeling of nuclei (TUNEL) assay and flow cytometry. The effects of Tempol on cisplatin-induced cleaved poly(ADP-ribose) polymerase, cleaved caspase, and mitochondrial inducible nitric oxide synthase expression were evaluated using western blot analysis. Levels of intracellular reactive oxygen species (ROS) were measured to assess the effects of Tempol on cisplatin-induced ROS accumulation. Mitochondria were evaluated by confocal microscopy, and the mitochondrial membrane potential was measured to investigate whether Tempol protected against cisplatin-induced mitochondrial dysfunction. Cisplatin treatment decreased cell viability, and increased apoptotic features and markers, ROS accumulation, and mitochondrial dysfunction. Tempol pre-treatment before cisplatin exposure significantly inhibited all these cisplatin-induced effects. These results demonstrate that Tempol inhibits cisplatin-induced cytotoxicity in HEI-OC1, and could play a preventive role against cisplatin-induced ototoxicity.
Collapse
Affiliation(s)
- Cha Kyung Youn
- Department of Otolaryngology-Head and Neck Surgery, Chosun University School of Medicine, Gwangju 61453, Korea.
- Division of Natural Medical Science, Chosun University School of Medicine, Gwangju 61452, Korea.
| | - Jun Kim
- Department of Otolaryngology-Head and Neck Surgery, Chosun University School of Medicine, Gwangju 61453, Korea.
| | - Eu-Ri Jo
- Department of Otolaryngology-Head and Neck Surgery, Chosun University School of Medicine, Gwangju 61453, Korea.
| | - Jeonghyun Oh
- Department of Otolaryngology-Head and Neck Surgery, Chosun University School of Medicine, Gwangju 61453, Korea.
| | - Nam Yong Do
- Department of Otolaryngology-Head and Neck Surgery, Chosun University School of Medicine, Gwangju 61453, Korea.
| | - Sung Il Cho
- Department of Otolaryngology-Head and Neck Surgery, Chosun University School of Medicine, Gwangju 61453, Korea.
| |
Collapse
|
90
|
Tian Y, Wu K, Liu Q, Han N, Zhang L, Chu Q, Chen Y. Modification of platinum sensitivity by KEAP1/NRF2 signals in non-small cell lung cancer. J Hematol Oncol 2016; 9:83. [PMID: 27601007 PMCID: PMC5012055 DOI: 10.1186/s13045-016-0311-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 08/25/2016] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND The objective of this study was to evaluate the effect of platinum-based drugs on nuclear-factor erythroid2 like 2 (NRF2) signaling in non-small cell lung cancer cell lines with or without Kelch-like ECH-associated protein 1 (KEAP1) mutations and to determine the role of NRF2 and KEAP1 on platinum-based drug treatment. METHODS We used real-time PCR to assess relative mRNA expression and used western blotting and immunofluorescence assays to assess protein expression. Small interfering RNA and shuttle plasmids were used to modulate the expression of NRF2, wild-type KEAP1, and mutant KEAP1. Drug sensitivity to platinum-based drugs was evaluated with Cell Count Kit-8. RESULTS We found that platinum-based therapies modified the NRF2 signaling pathway differently in KEAP1-mutated non-small cell lung cancer (NSCLC) cell lines compared with wild-type KEAP1 cell lines. The reactive degree of NRF2 signaling also varies between nedaplatin and cisplatin. The modification of NRF2 or KEAP1 expression in NSCLC cell lines disrupted downstream gene expression and cell sensitivity to platinum-based drugs. Finally, gene expression data retrieved from The Cancer Genome Atlas (TCGA) consortium indicated that KEAP1 mutation significantly affects NRF2 signaling activity in patients with NSCLC. CONCLUSIONS Our findings suggest that NRF2 signaling plays an indispensable role in NSCLC cell sensitivity to platinum-based treatments and provides a rationale for using NRF2 as a specific biomarker for predicting which patients will be most likely to benefit from platinum-based treatment.
Collapse
Affiliation(s)
- Yijun Tian
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Qian Liu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Na Han
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Li Zhang
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China.
| | - Yuan Chen
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China.
| |
Collapse
|
91
|
Spontaneous Cannabinoid Receptor 2 (CB2) Expression in the Cochlea of Adult Albino Rat and Its Up-Regulation after Cisplatin Treatment. PLoS One 2016; 11:e0161954. [PMID: 27564061 PMCID: PMC5001640 DOI: 10.1371/journal.pone.0161954] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/15/2016] [Indexed: 12/22/2022] Open
Abstract
We provide evidence for the presence of cannabinoid CB2 receptors in some cellular types of the cochlea of the adult albino rat. Cannabinoids and their receptors are increasingly being studied because of their high potential for clinical use. As a hyperspecialized portion of the peripheral nervous system, study of the expression and function of cannabinoid receptors in the hearing organ is of high interest. Stria vascularis and inner hair cells express CB2 receptor, as well as neurites and cell bodies of the spiral ganglion. Cellular types such as supporting cells and outer hair cells, in which the expression of other types of functional receptors has been reported, do not significantly express CB2 receptors in this study. An up-regulation of CB2 gene expression was detected after an ototoxic event such as cisplatin treatment, probably due to pro-inflammatory events triggered by the drug. That fact suggests promising potential of CB2 receptor as a therapeutic target for new treatments to palliate cisplatin-induced hearing loss and other ototoxic events which triggers inflammatory pathways.
Collapse
|
92
|
Kuhnt T, Stang A, Wienke A, Vordermark D, Schweyen R, Hey J. Potential risk factors for jaw osteoradionecrosis after radiotherapy for head and neck cancer. Radiat Oncol 2016; 11:101. [PMID: 27473433 PMCID: PMC4967325 DOI: 10.1186/s13014-016-0679-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 07/23/2016] [Indexed: 01/04/2023] Open
Abstract
INTRODUCTION To identify potential risk factors for the development of jaw osteoradionecrosis (ORN) after 3D-conformal radiotherapy (3D-CRT) and intensity-modulated radiotherapy (IMRT) among patients with newly diagnosed head and neck cancer. MATERIAL AND METHODS This study included 776 patients who underwent 3D-CRT or IMRT for head and neck cancer at the Department of Radiotherapy at the University Hospital Halle-Wittenberg between 2003 and 2013. Sex, dental status prior to radiotherapy, tumor site, bone surgery during tumor resection, concomitant chemotherapy, and the development of advanced ORN were documented for each patient. ORN was classified as grade 3, 4, or 5 according to the Radiation Therapy Oncology Group/European Organization for Research and Treatment of Cancer classification or grade 3 or 4 according to the late effects in normal tissues/subjective, objective, management, and analytic scale. The cumulative incidence of ORN was estimated. Cox regression analysis was used to identify prognostic risk factors for the development of ORN. RESULTS Fifty-one patients developed advanced ORN (relative frequency 6.6 %, cumulative incidence 12.4 %). The highest risk was found in patients who had undergone primary bone surgery during tumor resection (hazard ratio [HR] = 5.87; 95 % confidence interval [CI]: 3.09-11.19) and in patients with tumors located in the oral cavity (HR = 4.69; 95 % CI: 1.33-16.52). Sex, dentition (dentulous vs. edentulous), and chemotherapy had no clinically relevant influence. DISCUSSION AND CONCLUSION In contrast to most previous studies, we noted a low cumulative incidence of advanced ORN. Patients with tumors located in the oral cavity and those who undergo bone surgery during tumor resection prior to RT may be considered a high-risk group for the development of ORN.
Collapse
Affiliation(s)
- Thomas Kuhnt
- Department of Diagnostic Imaging and Radiation Medicine, University Clinic, University Leipzig, Leipzig, Germany
| | - Andreas Stang
- Department of Medical Informatics, Biometry and Epidemiology, University Clinic Essen, Essen, Germany
| | - Andreas Wienke
- Department of Medical Epidemiology, Biometry and Computer Science Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Dirk Vordermark
- Department of Radiotherapy, University Clinic, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Ramona Schweyen
- Department of Prosthetic Dentistry, University School of Dental Medicine, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Jeremias Hey
- Department of Prosthetic Dentistry, University School of Dental Medicine, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
93
|
Yan L, Hu R, Tu S, Cheng WJ, Zheng Q, Wang JW, Kan WS, Ren YJ. Emodin mitigates the oxidative stress induced by cisplatin in osteosarcoma MG63 cells. Oncol Lett 2016; 12:1981-1985. [PMID: 27602124 DOI: 10.3892/ol.2016.4902] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 10/12/2015] [Indexed: 12/15/2022] Open
Abstract
Previously, the application of cisplatin in chemotherapy was limited due to the significant side effects on normal cell growth. In the present study, the concomitant application of emodin with cisplatin was demonstrated to ameliorate cisplatin-induced oxidative stress and markedly suppress tumor cell proliferation for the first time. Human osteosarcoma MG-63 cells were treated with cisplatin alone or in combination with emodin. The cell viability was determined by MTS assays and the augmentation of reactive oxygen species were determined by fluorogenic probes; in addition, a stable MG-63 subline bearing antioxidant response element (ARE)-driven luciferase expression was developed to monitor the activation of the nuclear factor erythroid 2-related factor 2 (Nrf2)-ARE signaling pathway. The results indicated that cisplatin or emodin may inhibit MG-63 cell proliferation in a time- or dose-dependent manner, respectively. Concomitant treatment with cisplatin and emodin demonstrated synergic anti-tumor effects. Cisplatin augmented reactive oxygen species in the MG-63 cells, followed by the translocation of Nrf2 from the cytoplasm into the nucleus, which triggered ARE-driven luciferase expression. The addition of emodin diminished the previously described phenomenon, resulting in decreased ROS augmentation, translocation of Nrf2 and ARE-driven luciferase activity. In conclusion, emodin could ameliorate cisplatin-induced oxidative stress and protect the cells from oxidative stress-induced damage. The findings of the present study provide a novel strategy for the treatment of osteosarcoma using emodin and cisplatin.
Collapse
Affiliation(s)
- Li Yan
- Department of Reparative and Reconstructive Surgery of Orthopedics, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, P.R. China
| | - Rui Hu
- Department of Reparative and Reconstructive Surgery of Orthopedics, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, P.R. China
| | - Song Tu
- Department of Reparative and Reconstructive Surgery of Orthopedics, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, P.R. China
| | - Wen-Jun Cheng
- Department of Reparative and Reconstructive Surgery of Orthopedics, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, P.R. China
| | - Qiong Zheng
- Department of Reparative and Reconstructive Surgery of Orthopedics, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, P.R. China
| | - Jun-Wen Wang
- Department of Reparative and Reconstructive Surgery of Orthopedics, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, P.R. China
| | - Wu-Sheng Kan
- Department of Reparative and Reconstructive Surgery of Orthopedics, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, P.R. China
| | - Yi-Jun Ren
- Department of Reparative and Reconstructive Surgery of Orthopedics, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, P.R. China
| |
Collapse
|
94
|
Roldán-Fidalgo A, Martín Saldaña S, Trinidad A, Olmedilla-Alonso B, Rodríguez-Valiente A, García-Berrocal JR, Ramírez-Camacho R. In vitro and in vivo effects of lutein against cisplatin-induced ototoxicity. ACTA ACUST UNITED AC 2016; 68:197-204. [PMID: 26850526 DOI: 10.1016/j.etp.2016.01.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 01/05/2016] [Accepted: 01/14/2016] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Cisplatin is a commonly prescribed drug that produces ototoxicity as a side effect. Lutein is a carotenoid with antioxidant and anti-inflammatory properties previously tested for eye, heart and skin diseases but not evaluated to date in ear diseases. AIM To evaluate the protective effects of lutein on HEI-OC1 auditory cell line and in a Wistar rat model of cisplatin ototoxicity. MATERIALS AND METHODS In vitro study: Culture HEI-OC1 cells were exposed to lutein (2.5-100 μM) and to 25 μM cisplatin for 24h. In vivo study: Twenty eight female Wistar rats were randomized into three groups. Group A (n=8) received intratympanic lutein (0.03 mL) (1mg/mL) in the right ear and saline solution in the left one to determine the toxicity of lutein. Group B (n=8) received also intraperitoneal cisplatin (10mg/kg) to test the efficacy of lutein against cisplatin ototoxicity. Group C (n=12) received intratympanic lutein (0.03 mL) (1mg/mL) to quantify lutein in cochlear fluids (30 min, 1h and 5 days after treatment). Hearing function was evaluated by means of Auditory Steady-State Responses before the procedure and 5 days after (groups A and B). Morphological changes were studied by confocal laser scanning microscopy. RESULTS In vitro study: Lutein significantly reduced the cisplatin-induced cytotoxicity in the HEI-OC1 cells when they were pre-treated with lutein concentrations of 60 and 80 μM. In vivo study: Intratympanic lutein (1mg/mL) application showed no ototoxic effects. However it did not achieve protective effect against cisplatin-induced ototoxicity in Wistar rats. CONCLUSIONS Although lutein has shown beneficial effects in other pathologies, the present study only obtained protection against cisplatin ototoxicity in culture cells, but not in the in vivo model. The large molecule size, the low dose administered, and restriction to diffusion in the inner ear could account for this negative result.
Collapse
Affiliation(s)
- A Roldán-Fidalgo
- Department of Otorhinolaryngology, Hospital Puerta de Hierro Majadahonda, Universidad Autónoma de Madrid, Madrid, Spain.
| | - S Martín Saldaña
- Foundation for Biomedical Research, Puerta de Hierro Hospital, Madrid, Spain
| | - A Trinidad
- Department of Otorhinolaryngology, Hospital Puerta de Hierro Majadahonda, Universidad Autónoma de Madrid, Madrid, Spain
| | - B Olmedilla-Alonso
- Institute of Food Science, Technology and Nutrition, CSIC, Madrid, Spain
| | - A Rodríguez-Valiente
- Department of Otorhinolaryngology, Hospital Puerta de Hierro Majadahonda, Universidad Autónoma de Madrid, Madrid, Spain
| | - J R García-Berrocal
- Department of Otorhinolaryngology, Hospital Puerta de Hierro Majadahonda, Universidad Autónoma de Madrid, Madrid, Spain
| | - R Ramírez-Camacho
- Department of Otorhinolaryngology, Hospital Puerta de Hierro Majadahonda, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
95
|
Interstitial renal fibrosis due to multiple cisplatin treatments is ameliorated by semicarbazide-sensitive amine oxidase inhibition. Kidney Int 2016; 89:374-85. [DOI: 10.1038/ki.2015.327] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 08/31/2015] [Accepted: 09/03/2015] [Indexed: 02/07/2023]
|
96
|
Ferreira JA, Peixoto A, Neves M, Gaiteiro C, Reis CA, Assaraf YG, Santos LL. Mechanisms of cisplatin resistance and targeting of cancer stem cells: Adding glycosylation to the equation. Drug Resist Updat 2016; 24:34-54. [DOI: 10.1016/j.drup.2015.11.003] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 11/09/2015] [Accepted: 11/18/2015] [Indexed: 02/06/2023]
|
97
|
Ma C, Liu X, Cheng S, Li Q, Zhang R. A novel 4-fluorobenzenetelluronic trimethyltin ester: synthesis, characterization and in vitro cytotoxicity assessment. NEW J CHEM 2016. [DOI: 10.1039/c6nj00288a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A novel 4-fluorobenzenetelluronic trimethyltin ester (Me3Sn)4[p-F-PhTe(μ-O)(OH)O2]2 (1) has been prepared and structurally characterized, and apoptotic cytotoxicity assessments for A549 cells are made.
Collapse
Affiliation(s)
- Chunlin Ma
- School of Chemistry and Chemical Engineering
- Liaocheng University
- Liaocheng 252059
- P. R. China
| | - Xinglong Liu
- School of Chemistry and Chemical Engineering
- Liaocheng University
- Liaocheng 252059
- P. R. China
| | - Shuang Cheng
- School of Chemistry and Chemical Engineering
- Liaocheng University
- Liaocheng 252059
- P. R. China
| | - Qianli Li
- School of Chemistry and Chemical Engineering
- Liaocheng University
- Liaocheng 252059
- P. R. China
| | - Rufen Zhang
- School of Chemistry and Chemical Engineering
- Liaocheng University
- Liaocheng 252059
- P. R. China
| |
Collapse
|
98
|
Cisplatin-Induced Hearing Loss. TRANSLATIONAL RESEARCH IN AUDIOLOGY, NEUROTOLOGY, AND THE HEARING SCIENCES 2016. [DOI: 10.1007/978-3-319-40848-4_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
99
|
Teitz T, Goktug AN, Chen T, Zuo J. Development of Cell-Based High-Throughput Chemical Screens for Protection Against Cisplatin-Induced Ototoxicity. Methods Mol Biol 2016; 1427:419-30. [PMID: 27259939 DOI: 10.1007/978-1-4939-3615-1_22] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Various compounds have been tested in recent years for protection against cisplatin-induced hearing loss, but no compound has yet been FDA approved for clinical use in patients. Towards this goal, we developed an unbiased, high-throughput, mammalian cochlear cell-based chemical screen that allowed quantification of the protection ability of bioactive compounds and ranked them for future testing ex vivo in cochlear explant cultures and in vivo in animal models. In our primary screens, protection in the HEI-OC1 organ of Corti immortalized cell line was measured by the ability of each compound to inhibit caspase-3/7 activity triggered by cisplatin treatment (50 μM cisplatin for 22 h). A total of 4385 unique bioactive compounds were tested in a single dose of 8 μM and promising compounds were validated by dose response curves covering ten, 1:3 serial diluted concentrations. Primary hits were defined as having more than 60 % inhibition of the caspase-3/7 activity. Toxicity of the top compounds was measured by a CellTiter-Glo (CTG) assay that measured the viability of the cells in the presence of compound alone in similar dose responsive analysis. A combination of the caspase-3/7 inhibition activity assay (as measured by IC50) and the CTG viability assay (as determined by LD50) identified the top protective compounds in the HEI-OC1 cells. In the future, the top hits in our screens will be tested for their protective ability ex vivo in mouse cochlear explants and in vivo in animal models. Our mammalian cochlear cell-based, high-throughput chemical screening assays described here can be further modified and represent an initial successful step towards therapeutic intervention of hearing disorders, an unmet medical need of our society.
Collapse
Affiliation(s)
- Tal Teitz
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Asli N Goktug
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Jian Zuo
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
| |
Collapse
|
100
|
Otoprotective properties of 6α-methylprednisolone-loaded nanoparticles against cisplatin: In vitro and in vivo correlation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 12:965-976. [PMID: 26733264 DOI: 10.1016/j.nano.2015.12.367] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 12/07/2015] [Accepted: 12/09/2015] [Indexed: 11/24/2022]
Abstract
UNLABELLED 6α-Methylprednisolone-loaded surfactant-free nanoparticles have been developed to palliate cisplatin ototoxicity. Nanoparticles were based on two different amphiphilic pseudo-block copolymers obtained by free radical polymerization and based on N-vinyl pyrrolidone and a methacrylic derivative of α-tocopheryl succinate or α-tocopherol. Copolymers formed spherical nanoparticles by nanoprecipitation in aqueous media that were able to encapsulate 6α-methylprednisolone in their inner core. The obtained nanovehicles were tested in vitro using HEI-OC1 cells and in vivo in a murine model. Unloaded nanoparticles were not able to significantly reduce the cisplatin ototoxicity. Loaded nanoparticles reduced cisplatin-ototoxicity in vitro being more active those based on the methacrylic derivative of vitamin E, due to their higher encapsulation efficiency. This formulation was able to protect hair cells in the base of the cochlea, having a positive effect in the highest frequencies tested in a murine model. A good correlation between the in vitro and the in vivo experiments was found. FROM THE CLINICAL EDITOR Cisplatin is a commonly used chemotherapeutic agent against many cancers clinically. However, one of the significant side-effects remains ototoxicity. Here, the authors presented their data on using 6α-methylprednisolone-loaded nanoparticles in the reduction of ototoxicity in in-vitro and in-vivo experiments. Early promising results should enable further refinement of adopting this new approach in future experiments.
Collapse
|