1
|
Villa J, Cury J, Kessler L, Tan X, Richter CP. Enhancing biocompatibility of the brain-machine interface: A review. Bioact Mater 2024; 42:531-549. [PMID: 39308547 PMCID: PMC11416625 DOI: 10.1016/j.bioactmat.2024.08.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/05/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
In vivo implantation of microelectrodes opens the door to studying neural circuits and restoring damaged neural pathways through direct electrical stimulation and recording. Although some neuroprostheses have achieved clinical success, electrode material properties, inflammatory response, and glial scar formation at the electrode-tissue interfaces affect performance and sustainability. Those challenges can be addressed by improving some of the materials' mechanical, physical, chemical, and electrical properties. This paper reviews materials and designs of current microelectrodes and discusses perspectives to advance neuroprosthetics performance.
Collapse
Affiliation(s)
- Jordan Villa
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
| | - Joaquin Cury
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
| | - Lexie Kessler
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
| | - Xiaodong Tan
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
- The Hugh Knowles Center, Department of Communication Sciences and Disorders, Northwestern University, USA
| | - Claus-Peter Richter
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
- The Hugh Knowles Center, Department of Communication Sciences and Disorders, Northwestern University, USA
- Department of Communication Sciences and Disorders, Northwestern University, USA
- Department of Biomedical Engineering, Northwestern University, USA
| |
Collapse
|
2
|
Kessler L, Koo C, Richter CP, Tan X. Hearing loss during chemotherapy: prevalence, mechanisms, and protection. Am J Cancer Res 2024; 14:4597-4632. [PMID: 39417180 PMCID: PMC11477841 DOI: 10.62347/okgq4382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 08/23/2024] [Indexed: 10/19/2024] Open
Abstract
Ototoxicity is an often-underestimated sequela for cancer patients undergoing chemotherapy, with an incidence rate exceeding 50%, affecting approximately 4 million individuals worldwide each year. Despite the nearly 2,000 publications on chemotherapy-related ototoxicity in the past decade, the understanding of its prevalence, mechanisms, and preventative or therapeutic measures remains ambiguous and subject to debate. To date, only one drug, sodium thiosulfate, has gained FDA approval for treating ototoxicity in chemotherapy. However, its utilization is restricted. This review aims to offer clinicians and researchers a comprehensive perspective by thoroughly and carefully reviewing available data and current evidence. Chemotherapy-induced ototoxicity is characterized by four primary symptoms: hearing loss, tinnitus, vertigo, and dizziness, originating from both auditory and vestibular systems. Hearing loss is the predominant symptom. Amongst over 700 chemotherapeutic agents documented in various databases, only seven are reported to induce hearing loss. While the molecular mechanisms of the hearing loss caused by the two platinum-based drugs are extensively explored, the pathways behind the action of the other five drugs are primarily speculative, rooted in their therapeutic properties and side effects. Cisplatin attracts the majority of attention among these drugs, encompassing around two-thirds of the literature regarding ototoxicity in chemotherapy. Cisplatin ototoxicity chiefly manifests through the loss of outer hair cells, possibly resulting from damages directly by cisplatin uptake or secondary effects on the stria vascularis. Both direct and indirect influences contribute to cisplatin ototoxicity, while it is still debated which path is dominant or where the primary target of cisplatin is located. Candidates for hearing protection against cisplatin ototoxicity are also discussed, with novel strategies and methods showing promise on the horizon.
Collapse
Affiliation(s)
- Lexie Kessler
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern UniversityChicago, Illinois 60611, USA
| | - Chail Koo
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern UniversityChicago, Illinois 60611, USA
| | - Claus-Peter Richter
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern UniversityChicago, Illinois 60611, USA
- Hugh Knowles Center for Clinical and Basic Science in Hearing and Its Disorders, Northwestern UniversityEvanston, Illinois 60208, USA
- Department of Biomedical Engineering, Northwestern UniversityEvanston, Illinois 60208, USA
- Department of Communication Sciences and Disorders, Northwestern UniversityEvanston, Illinois 60208, USA
| | - Xiaodong Tan
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern UniversityChicago, Illinois 60611, USA
- Hugh Knowles Center for Clinical and Basic Science in Hearing and Its Disorders, Northwestern UniversityEvanston, Illinois 60208, USA
| |
Collapse
|
3
|
Bovee S, Klump GM, Köppl C, Pyott SJ. The Stria Vascularis: Renewed Attention on a Key Player in Age-Related Hearing Loss. Int J Mol Sci 2024; 25:5391. [PMID: 38791427 PMCID: PMC11121695 DOI: 10.3390/ijms25105391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Age-related hearing loss (HL), or presbycusis, is a complex and heterogeneous condition, affecting a significant portion of older adults and involving various interacting mechanisms. Metabolic presbycusis, a type of age-related HL, is characterized by the dysfunction of the stria vascularis, which is crucial for maintaining the endocochlear potential necessary for hearing. Although attention on metabolic presbycusis has waned in recent years, research continues to identify strial pathology as a key factor in age-related HL. This narrative review integrates past and recent research, bridging findings from animal models and human studies, to examine the contributions of the stria vascularis to age-related HL. It provides a brief overview of the structure and function of the stria vascularis and then examines mechanisms contributing to age-related strial dysfunction, including altered ion transport, changes in pigmentation, inflammatory responses, and vascular atrophy. Importantly, this review outlines the contribution of metabolic mechanisms to age-related HL, highlighting areas for future research. It emphasizes the complex interdependence of metabolic and sensorineural mechanisms in the pathology of age-related HL and highlights the importance of animal models in understanding the underlying mechanisms. The comprehensive and mechanistic investigation of all factors contributing to age-related HL, including cochlear metabolic dysfunction, remains crucial to identifying the underlying mechanisms and developing personalized, protective, and restorative treatments.
Collapse
Affiliation(s)
- Sonny Bovee
- Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany; (S.B.); (G.M.K.); (C.K.)
| | - Georg M. Klump
- Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany; (S.B.); (G.M.K.); (C.K.)
- Cluster of Excellence “Hearing4all”, Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany
- Research Centre Neurosensory Science, Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany
| | - Christine Köppl
- Department of Neuroscience, School of Medicine and Health Science, Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany; (S.B.); (G.M.K.); (C.K.)
- Cluster of Excellence “Hearing4all”, Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany
- Research Centre Neurosensory Science, Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany
| | - Sonja J. Pyott
- Department of Otorhinolaryngology/Head and Neck Surgery, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
- The Research School of Behavioural and Cognitive Neurosciences, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
| |
Collapse
|
4
|
Teraoka M, Hato N, Inufusa H, You F. Role of Oxidative Stress in Sensorineural Hearing Loss. Int J Mol Sci 2024; 25:4146. [PMID: 38673731 PMCID: PMC11050000 DOI: 10.3390/ijms25084146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/27/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Hearing is essential for communication, and its loss can cause a serious disruption to one's social life. Hearing loss is also recognized as a major risk factor for dementia; therefore, addressing hearing loss is a pressing global issue. Sensorineural hearing loss, the predominant type of hearing loss, is mainly due to damage to the inner ear along with a variety of pathologies including ischemia, noise, trauma, aging, and ototoxic drugs. In addition to genetic factors, oxidative stress has been identified as a common mechanism underlying several cochlear pathologies. The cochlea, which plays a major role in auditory function, requires high-energy metabolism and is, therefore, highly susceptible to oxidative stress, particularly in the mitochondria. Based on these pathological findings, the potential of antioxidants for the treatment of hearing loss has been demonstrated in several animal studies. However, results from human studies are insufficient, and future clinical trials are required. This review discusses the relationship between sensorineural hearing loss and reactive oxidative species (ROS), with particular emphasis on age-related hearing loss, noise-induced hearing loss, and ischemia-reperfusion injury. Based on these mechanisms, the current status and future perspectives of ROS-targeted therapy for sensorineural hearing loss are described.
Collapse
Affiliation(s)
- Masato Teraoka
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Ehime University, Toon 791-0295, Ehime, Japan;
| | - Naohito Hato
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Ehime University, Toon 791-0295, Ehime, Japan;
| | - Haruhiko Inufusa
- Division of Anti-Oxidant Research, Life Science Research Center, Gifu University, Yanagito 1-1, Gifu 501-1194, Japan; (H.I.); (F.Y.)
| | - Fukka You
- Division of Anti-Oxidant Research, Life Science Research Center, Gifu University, Yanagito 1-1, Gifu 501-1194, Japan; (H.I.); (F.Y.)
| |
Collapse
|
5
|
Nassauer L, Staecker H, Huang P, Renslo B, Goblet M, Harre J, Warnecke A, Schott JW, Morgan M, Galla M, Schambach A. Protection from cisplatin-induced hearing loss with lentiviral vector-mediated ectopic expression of the anti-apoptotic protein BCL-XL. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102157. [PMID: 38450280 PMCID: PMC10915631 DOI: 10.1016/j.omtn.2024.102157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 02/15/2024] [Indexed: 03/08/2024]
Abstract
Cisplatin is a highly effective chemotherapeutic agent, but it can cause sensorineural hearing loss (SNHL) in patients. Cisplatin-induced ototoxicity is closely related to the accumulation of reactive oxygen species (ROS) and subsequent death of hair cells (HCs) and spiral ganglion neurons (SGNs). Despite various strategies to combat ototoxicity, only one therapeutic agent has thus far been clinically approved. Therefore, we have developed a gene therapy concept to protect cochlear cells from cisplatin-induced toxicity. Self-inactivating lentiviral (LV) vectors were used to ectopically express various antioxidant enzymes or anti-apoptotic proteins to enhance the cellular ROS scavenging or prevent apoptosis in affected cell types. In direct comparison, anti-apoptotic proteins mediated a stronger reduction in cytotoxicity than antioxidant enzymes. Importantly, overexpression of the most promising candidate, Bcl-xl, achieved an up to 2.5-fold reduction in cisplatin-induced cytotoxicity in HEI-OC1 cells, phoenix auditory neurons, and primary SGN cultures. BCL-XL protected against cisplatin-mediated tissue destruction in cochlear explants. Strikingly, in vivo application of the LV BCL-XL vector improved hearing and increased HC survival in cisplatin-treated mice. In conclusion, we have established a preclinical gene therapy approach to protect mice from cisplatin-induced ototoxicity that has the potential to be translated to clinical use in cancer patients.
Collapse
Affiliation(s)
- Larissa Nassauer
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Hinrich Staecker
- Department of Otolaryngology-Head and Neck Surgery, University of Kansas School of Medicine, Kansas City, KS 66160, USA
| | - Peixin Huang
- Department of Otolaryngology-Head and Neck Surgery, University of Kansas School of Medicine, Kansas City, KS 66160, USA
| | - Bryan Renslo
- Department of Otolaryngology-Head and Neck Surgery, University of Kansas School of Medicine, Kansas City, KS 66160, USA
| | - Madeleine Goblet
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, 30625 Hannover, Germany
- Cluster of Excellence “Hearing4all”, Hannover Medical School, 30625 Hannover, Germany
| | - Jennifer Harre
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, 30625 Hannover, Germany
- Cluster of Excellence “Hearing4all”, Hannover Medical School, 30625 Hannover, Germany
| | - Athanasia Warnecke
- Department of Otorhinolaryngology, Head and Neck Surgery, Hannover Medical School, 30625 Hannover, Germany
- Cluster of Excellence “Hearing4all”, Hannover Medical School, 30625 Hannover, Germany
| | - Juliane W. Schott
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Michael Morgan
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Melanie Galla
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
6
|
Kishimoto-Urata M, Urata S, Fujimoto C, Yamasoba T. Role of Oxidative Stress and Antioxidants in Acquired Inner Ear Disorders. Antioxidants (Basel) 2022; 11:1469. [PMID: 36009187 PMCID: PMC9405327 DOI: 10.3390/antiox11081469] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 01/27/2023] Open
Abstract
Oxygen metabolism in the mitochondria is essential for biological activity, and reactive oxygen species (ROS) are produced simultaneously in the cell. Once an imbalance between ROS production and degradation (oxidative stress) occurs, cells are damaged. Sensory organs, especially those for hearing, are constantly exposed during daily life. Therefore, almost all mammalian species are liable to hearing loss depending on their environment. In the auditory pathway, hair cells, spiral ganglion cells, and the stria vascularis, where mitochondria are abundant, are the main targets of ROS. Excessive generation of ROS in auditory sensory organs is widely known to cause sensorineural hearing loss, and mitochondria-targeted antioxidants are candidates for treatment. This review focuses on the relationship between acquired hearing loss and antioxidant use to provide an overview of novel antioxidants, namely medicines, supplemental nutrients, and natural foods, based on clinical, animal, and cultured-cell studies.
Collapse
Affiliation(s)
| | | | | | - Tatsuya Yamasoba
- Department of Otolaryngology, Graduate School of Medicine, The University of Tokyo, Tokyo 1138655, Japan; (M.K.-U.); (S.U.); (C.F.)
| |
Collapse
|
7
|
Domingo IK, Latif A, Bhavsar AP. Pro-Inflammatory Signalling PRRopels Cisplatin-Induced Toxicity. Int J Mol Sci 2022; 23:7227. [PMID: 35806229 PMCID: PMC9266867 DOI: 10.3390/ijms23137227] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023] Open
Abstract
Cisplatin is a platinum-based chemotherapeutic that has long since been effective against a variety of solid-cancers, substantially improving the five-year survival rates for cancer patients. Its use has also historically been limited by its adverse drug reactions, or cisplatin-induced toxicities (CITs). Of these reactions, cisplatin-induced nephrotoxicity (CIN), cisplatin-induced peripheral neuropathy (CIPN), and cisplatin-induced ototoxicity (CIO) are the three most common of several CITs recognised thus far. While the anti-cancer activity of cisplatin is well understood, the mechanisms driving its toxicities have only begun to be defined. Most of the literature pertains to damage caused by oxidative stress that occurs downstream of cisplatin treatment, but recent evidence suggests that the instigator of CIT development is inflammation. Cisplatin has been shown to induce pro-inflammatory signalling in CIN, CIPN, and CIO, all of which are associated with persisting markers of inflammation, particularly from the innate immune system. This review covered the hallmarks of inflammation common and distinct between different CITs, the role of innate immune components in development of CITs, as well as current treatments targeting pro-inflammatory signalling pathways to conserve the use of cisplatin in chemotherapy and improve long-term health outcomes of cancer patients.
Collapse
Affiliation(s)
| | | | - Amit P. Bhavsar
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1, Canada; (I.K.D.); (A.L.)
| |
Collapse
|
8
|
Chen CH, Huang CY, Lin HYH, Wang MC, Chang CY, Cheng YF. Association of Sodium Thiosulfate With Risk of Ototoxic Effects From Platinum-Based Chemotherapy: A Systematic Review and Meta-analysis. JAMA Netw Open 2021; 4:e2118895. [PMID: 34338793 PMCID: PMC8329743 DOI: 10.1001/jamanetworkopen.2021.18895] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
IMPORTANCE Platinum-induced ototoxic effects are a significant issue because platinum-based chemotherapy is one of the most commonly used therapeutic medications. Sodium thiosulfate (STS) is considered a potential otoprotectant for the prevention of platinum-induced ototoxic effects that functions by binding the platinum-based agent, but its administration raises concerns regarding the substantial attenuation of the antineoplastic outcome associated with platinum. OBJECTIVE To evaluate the association between concurrent STS and reduced risk of ototoxic effects among patients undergoing platinum-based chemotherapy and to evaluate outcomes, including event-free survival, overall survival, and adverse outcomes. DATA SOURCES From inception through November 7, 2020, databases, including the Cochrane Library, PubMed, Embase, Web of Science, and Scopus, were searched. STUDY SELECTION Studies enrolling patients with cancer who were undergoing platinum-based chemotherapy that compared ototoxic effects development between patients who received STS and patients who did not and provided adequate information for meta-analysis were regarded as eligible. This study followed the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines. DATA EXTRACTION AND SYNTHESIS The data were extracted by 2 reviewers independently. A random-effects model was used to explore objectives. MAIN OUTCOMES AND MEASURES Relative risks (RRs) for ototoxic effects development and hemopoietic event development comparing the experimental group and the control group were estimated. Secondary outcomes were hazard ratios (HRs) for event-free survival and overall survival. Sensitivity analysis and trial sequential analysis were conducted to further consolidate pooled results. RESULTS Among 4 eligible studies that were included, there were 3 randomized clinical trials and 1 controlled study. A total of 278 patients were allocated to the experimental group (ie, platinum-based chemotherapy plus STS; 158 patients, including 13 patients using contralatral ears of the control group as samples) or the control group (ie, chemotherapy; 133 patients, including 13 patients using contralateral ears of the experimental group as samples). Overall, patients who received STS had a statistically significantly decreased risk of ototoxic effects during the course of platinum-based chemotherapy (RR, 0.61; 95% CI, 0.49-0.77; P < .001; I2 = 5.0%) without a statistically significant increase in the risk of poor event-free survival (HR, 1.13; 95% CI, 0.70-1.82; P = .61; I2 = 0%) or overall survival (HR, 1.90; 95% CI, 0.90-4.03; P = .09; I2 = 0%). In the trial sequential analysis of event-free survival (z = -0.52) and overall survival (z = -1.68), although the cumulative z curves did not surpass the traditional significance boundary (-1.96 to 1.96 for both) or sequential monitoring boundary (event-free survival: -8.0 to 8.0; overall survival boundary not renderable in the analysis because the information size was too small) of the adjusted CI, they did not reach the required information size. CONCLUSIONS AND RELEVANCE This meta-analysis found that concurrent STS delivery was associated with a decreased risk of platinum-induced ototoxic effects among patients treated with platinum-induced chemotherapy. These findings suggest that concurrent STS for protection against ototoxic effects should be considered for patients indicated for platinum-based chemotherapy.
Collapse
Affiliation(s)
- Chih-Hao Chen
- Department of Otolaryngology-Head and Neck Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chii-Yuan Huang
- Department of Otolaryngology-Head and Neck Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Heng-Yu Haley Lin
- Department of Otolaryngology-Head and Neck Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Medical Education, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Mao-Che Wang
- Department of Otolaryngology-Head and Neck Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Yu Chang
- Department of Anesthesiology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Yen-Fu Cheng
- Department of Otolaryngology-Head and Neck Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
9
|
Tang Q, Wang X, Jin H, Mi Y, Liu L, Dong M, Chen Y, Zou Z. Cisplatin-induced ototoxicity: Updates on molecular mechanisms and otoprotective strategies. Eur J Pharm Biopharm 2021; 163:60-71. [PMID: 33775853 DOI: 10.1016/j.ejpb.2021.03.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/20/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023]
Abstract
Cisplatin is a highly effective antitumor drug generally used in the treatment of solid malignant tumors. However, cisplatin causes severe side effects such as bone marrow depression, nephrotoxicity, and ototoxicity, thus limiting its clinical application. The incidence of ototoxicity induced by cisplatin ranges from 20% to 70%, and it usually manifests as a progressive, bilateral and irreversible hearing loss. Although the etiology of cisplatin-induced ototoxicity remains unclear, an increasing body of evidence suggests that the ototoxicity of cisplatin is mainly related to the production of reactive oxygen species and activation of apoptotic pathway in cochlear tissues. Many drugs have been well proved to protect cisplatin-induced hearing loss in vitro and in vivo. However, the anti-tumor effect of cisplatin is also weakened by systemic administration of those drugs for hearing protection, especially antioxidants. Therefore, establishing a local administration strategy contributes to the otoprotection without affecting the effect of cisplatin. This review introduces the pathology of ototoxicity caused by cisplatin, and focuses on recent developments in the mechanisms and protective strategies of cisplatin-induced ototoxicity.
Collapse
Affiliation(s)
- Qing Tang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China; Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Xianren Wang
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Huan Jin
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China; Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Yanjun Mi
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research and Thoracic Tumor Diagnosis & Treatment, The First Affiliated Hospital of Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, China
| | - Lingfeng Liu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China; Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Mengyuan Dong
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China; Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Yibing Chen
- Genetic and Prenatal Diagnosis Center, Department of Gynecology and Obstetrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China.
| | - Zhengzhi Zou
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China; Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou 510631, China; Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
10
|
Cortés Fuentes IA, Burotto M, Retamal MA, Frelinghuysen M, Caglevic C, Gormaz JG. Potential use of n-3 PUFAs to prevent oxidative stress-derived ototoxicity caused by platinum-based chemotherapy. Free Radic Biol Med 2020; 160:263-276. [PMID: 32827639 DOI: 10.1016/j.freeradbiomed.2020.07.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023]
Abstract
Platinum-based compounds are widely used for the treatment of different malignancies due to their high effectiveness. Unfortunately, platinum-based treatment may lead to ototoxicity, an often-irreversible side effect without a known effective treatment and prevention plan. Platinum-based compound-related ototoxicity results mainly from the production of toxic levels of reactive oxygen species (ROS) rather than DNA-adduct formation, which has led to test strategies based on direct ROS scavengers to ameliorate hearing loss. However, favorable clinical results have been associated with several complications, including potential interactions with chemotherapy efficacy. To understand the contribution of the different cytotoxic mechanisms of platinum analogues on malignant cells and auditory cells, the particular susceptibility and response of both kinds of cells to molecules that potentially interfere with these mechanisms, is fundamental to develop innovative strategies to prevent ototoxicity without affecting antineoplastic effects. The n-3 long-chain polyunsaturated fatty acids (n-3 PUFAs) have been tried in different clinical settings, including with cancer patients. Nevertheless, their use to decrease cisplatin-induced ototoxicity has not been explored to date. In this hypothesis paper, we address the mechanisms of platinum compounds-derived ototoxicity, focusing on the differences between the effects of these compounds in neoplastic versus auditory cells. We discuss the basis for a strategic use of n-3 PUFAs to potentially protect auditory cells from platinum-derived injury without affecting neoplastic cells and chemotherapy efficacy.
Collapse
Affiliation(s)
- Ignacio A Cortés Fuentes
- Otorhinolaryngology Service, Hospital Barros Luco-Trudeau, San Miguel, Santiago, Chile; Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Mauricio Burotto
- Oncology Department, Clínica Universidad de Los Andes, Santiago, Chile; Bradford Hill, Clinical Research Center, Santiago, Chile
| | - Mauricio A Retamal
- Universidad Del Desarrollo, Centro de Fisiología Celular e Integrativa, Facultad de Medicina Clínica Alemana, Santiago, Chile.
| | | | - Christian Caglevic
- Cancer Research Department, Fundación Arturo López Pérez, Santiago, Chile
| | - Juan G Gormaz
- Faculty of Medicine, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
11
|
Wu J, Ye J, Kong W, Zhang S, Zheng Y. Programmed cell death pathways in hearing loss: A review of apoptosis, autophagy and programmed necrosis. Cell Prolif 2020; 53:e12915. [PMID: 33047870 PMCID: PMC7653260 DOI: 10.1111/cpr.12915] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/23/2020] [Accepted: 09/09/2020] [Indexed: 02/05/2023] Open
Abstract
Programmed cell death (PCD)—apoptosis, autophagy and programmed necrosis—is any pathological form of cell death mediated by intracellular processes. Ototoxic drugs, ageing and noise exposure are some common pathogenic factors of sensorineural hearing loss (SNHL) that can induce the programmed death of auditory hair cells through different pathways, and eventually lead to the loss of hair cells. Furthermore, several mutations in apoptotic genes including DFNA5, DFNA51 and DFNB74 have been suggested to be responsible for the new functional classes of monogenic hearing loss (HL). Therefore, in this review, we elucidate the role of these three forms of PCD in different types of HL and discuss their guiding significance for HL treatment. We believe that further studies of PCD pathways are necessary to understand the pathogenesis of HL and guide scientists and clinicians to identify new drug targets for HL treatment.
Collapse
Affiliation(s)
- Junhao Wu
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Ye
- College of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Weili Kong
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Shouyue Zhang
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Yun Zheng
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Cisplatin-Induced Stria Vascularis Damage Is Associated with Inflammation and Fibrosis. Neural Plast 2020; 2020:8851525. [PMID: 33029120 PMCID: PMC7527906 DOI: 10.1155/2020/8851525] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 08/05/2020] [Accepted: 09/03/2020] [Indexed: 12/20/2022] Open
Abstract
The stria vascularis (SV) generates the endocochlear potential (EP) in the inner ear and is necessary for proper hair cell (HC) mechanotransduction and hearing. Cell junctions are indispensable for the establishment of compositionally distinct fluid compartments in the inner ear. Ototoxic drug cisplatin can damage SV and cause sensorineural hearing loss; however, the underlying mechanisms behind such injury are unclear. In this study, after the intraperitoneal injection of cisplatin (3 mg/kg/day for 7 days) in mice, we determined the auditory function by EP recording and auditory brainstem response (ABR) analysis, observed the ultrastructure of SV by transmission electron microscopy (TEM), and examined the expression and distribution of cell junction proteins by western blot, PCR, and immunofluorescence staining. We discovered that the EP was significantly reduced while ABR thresholds were significantly elevated in cisplatin-treated mice; cisplatin induced ultrastructural changes in marginal cells (MCs), endothelial cells (ECs), pericytes, etc. We found that cisplatin insulted auditory function not only by reducing the expression of zonula occludens protein-1 (ZO-1) in MCs of the SV but also by decreasing the expression of connexin 26 (Cx26) and connexin 43 (Cx43) in MCs and basal cells (BCs). More importantly, cisplatin induced activations of perivascular-resident macrophage-like melanocytes (PVM/Ms) and interleukin-1beta (IL-1β) as well as increased expressions of profibrotic proteins such as laminin and collagen IV in SV. Thus, our results firstly showed that cisplatin induced fibrosis, inflammation, and the complex expression change of cell junctions in SV.
Collapse
|
13
|
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: 8.4] [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
|
14
|
Cai J, Wu X, Li X, Ma C, Xu L, Guo X, Li J, Wang H, Han Y. Allicin Protects against Cisplatin-Induced Stria Vascularis Damage: Possible Relation to Inhibition of Caspase-3 and PARP-1-AIF-Mediated Apoptotic Pathways. ORL J Otorhinolaryngol Relat Spec 2019; 81:202-214. [DOI: 10.1159/000500557] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 04/23/2019] [Indexed: 11/19/2022]
|
15
|
Sheth S, Sheehan K, Dhukhwa A, Al Aameri RFH, Mamillapalli C, Mukherjea D, Rybak LP, Ramkumar V. Oral Administration of Caffeine Exacerbates Cisplatin-Induced Hearing Loss. Sci Rep 2019; 9:9571. [PMID: 31267026 PMCID: PMC6606569 DOI: 10.1038/s41598-019-45964-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/14/2019] [Indexed: 12/31/2022] Open
Abstract
Adenosine A1 receptors (A1AR) are well characterized for their role in cytoprotection. Previous studies have demonstrated the presence of these receptors in the cochlea where their activation were shown to suppress cisplatin-induced inflammatory response and the resulting ototoxicity. Inhibition of A1AR by caffeine, a widely consumed psychoactive substance, could antagonize the endogenous protective role of these receptors in cochlea and potentiate cisplatin-induced hearing loss. This hypothesis was tested in a rat model of cisplatin ototoxicity following oral administration of caffeine. We report here that single-dose administration of caffeine exacerbates cisplatin-induced hearing loss without increasing the damage to outer hair cells (OHCs), but increased synaptopathy and inflammation in the cochlea. These effects of caffeine were mediated by its blockade of A1AR, as co-administration of R-PIA, an A1AR agonist, reversed the detrimental actions of caffeine and cisplatin on hearing loss. Multiple doses of caffeine exacerbated cisplatin ototoxicity which was associated with damage to OHCs and cochlear synaptopathy. These findings highlight a possible drug-drug interaction between caffeine and cisplatin for ototoxicity and suggest that caffeine consumption should be cautioned in cancer patients treated with a chemotherapeutic regimen containing cisplatin.
Collapse
Affiliation(s)
- Sandeep Sheth
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Kelly Sheehan
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Asmita Dhukhwa
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Raheem F H Al Aameri
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Chaitanya Mamillapalli
- Department of Internal Medicine (Division of Endocrinology), Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Debashree Mukherjea
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Leonard P Rybak
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Vickram Ramkumar
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States.
| |
Collapse
|
16
|
Fitzakerley JL, Trachte GJ. Genetics of guanylyl cyclase pathways in the cochlea and their influence on hearing. Physiol Genomics 2018; 50:780-806. [PMID: 29958079 DOI: 10.1152/physiolgenomics.00056.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Although hearing loss is the most common sensory deficit in Western societies, there are no successful pharmacological treatments for this disorder. Recent experiments have demonstrated that manipulation of intracellular cyclic guanosine monophosphate (cGMP) concentrations can have both beneficial and harmful effects on hearing. In this review, we will examine the role of cGMP as a key second messenger involved in many aspects of cochlear function and discuss the known functions of downstream effectors of cGMP in sound processing. The nitric oxide-stimulated soluble guanylyl cyclase system (sGC) and the two natriuretic peptide-stimulated particulate GCs (pGCs) will be more extensively covered because they have been studied most thoroughly. The cochlear GC systems are attractive targets for medical interventions that improve hearing while simultaneously representing an under investigated source of sensorineural hearing loss.
Collapse
Affiliation(s)
- Janet L Fitzakerley
- Department of Biomedical Sciences, University of Minnesota Medical School , Duluth, Minnesota
| | - George J Trachte
- Department of Biomedical Sciences, University of Minnesota Medical School , Duluth, Minnesota
| |
Collapse
|
17
|
Sheehan K, Sheth S, Mukherjea D, Rybak LP, Ramkumar V. Trans-Tympanic Drug Delivery for the Treatment of Ototoxicity. J Vis Exp 2018. [PMID: 29608150 DOI: 10.3791/56564] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The systemic administration of protective agents to treat drug-induced ototoxicity is limited by the possibility that these protective agents could interfere with the chemotherapeutic efficacy of the primary drugs. This is especially true for the drug cisplatin, whose anticancer actions are attenuated by antioxidants which provide adequate protection against hearing loss. Other current or potential otoprotective agents could pose a similar problem, if administered systemically. The application of various biologicals or protective agents directly to the cochlea would allow for high levels of these agents locally with limited systemic side effects. In this report, we demonstrate a trans-tympanic method of delivery of various drugs or biological reagents to the cochlea, which should enhance basic science research on the cochlea and provide a simple way of directing the use of otoprotective agents in the clinics. This report details a method of trans-tympanic drug delivery and provides examples of how this technique has been used successfully in experimental animals to treat cisplatin ototoxicity.
Collapse
Affiliation(s)
- Kelly Sheehan
- Department of Surgery, Division of Otolaryngology, Southern Illinois University School of Medicine
| | - Sandeep Sheth
- Department of Pharmacology, Southern Illinois University School of Medicine
| | - Debashree Mukherjea
- Department of Surgery, Division of Otolaryngology, Southern Illinois University School of Medicine
| | - Leonard P Rybak
- Department of Surgery, Division of Otolaryngology, Southern Illinois University School of Medicine; Department of Pharmacology, Southern Illinois University School of Medicine
| | - Vickram Ramkumar
- Department of Pharmacology, Southern Illinois University School of Medicine;
| |
Collapse
|
18
|
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
|
19
|
Eroğlu O, Karlıdağ T, Kuloğlu T, Keleş E, Kaygusuz İ, Yalçın Ş. The Protective Effect of Cortexin on Cisplatin-Induced Ototoxicity. J Int Adv Otol 2017; 14:27-33. [PMID: 29092803 DOI: 10.5152/iao.2017.3825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE The aim of this report is to evaluate whether cortexin provides any protective activity against ototoxicity of cisplatin. MATERIALS AND METHODS The study was performed on 30 healthy adult Wistar Albino rats, and rats were randomly divided into three groups of ten. Group I (Control group) was given intraperitoneal (ip) saline solution 1 mL/day. Group II (Cisplatin group) was given ip cisplatin for 2 days at doses of 10 mg/kg. Group III (Cisplatin + Cortexin group) was given ip cisplatin for 2 days at same doses with ip cortexin 2 mg/day for 7 days. Before and on the fourth day of the study, all subjects underwent auditory brainstem response (ABR) and distortion product otoacoustic emissions (DPOAE) tests. At the end of fourth day, half of the subjects in all three groups were decapitated, and their cochlea were removed for histopathologic examination. On the eighth day, tests of the remaining subjects and histopathological examinations were repeated. RESULTS ABR tests on the fourth and eighth days showed elevations in the mean hearing thresholds of Groups II and III compared to Group I (p < 0.05). DPOAE tests revealed a loss in emission values on the fourth and eighth days of the study compared to the baseline in Groups II and III. Comparison of Groups II with III showed that emission loss was higher in Group II at both time points, and the difference was more pronounced on the eighth day. Histopathological findings supported these tests. CONCLUSION Cortexin provide protective activity against cisplatin-induced ototoxicity.
Collapse
Affiliation(s)
- Orkun Eroğlu
- Department of Otorhinolaryngology, Fırat University School of Medicine, Elazığ, Turkey
| | - Turgut Karlıdağ
- Department of Otorhinolaryngology, Fırat University School of Medicine, Elazığ, Turkey
| | - Tuncay Kuloğlu
- Department of Histology and Embryology, Fırat University School of Medicine, Elazığ, Turkey
| | - Erol Keleş
- Department of Otorhinolaryngology, Fırat University School of Medicine, Elazığ, Turkey
| | - İrfan Kaygusuz
- Department of Otorhinolaryngology, Fırat University School of Medicine, Elazığ, Turkey
| | - Şinasi Yalçın
- Department of Otorhinolaryngology, Fırat University School of Medicine, Elazığ, Turkey
| |
Collapse
|
20
|
Ramaswamy B, Roy S, Apolo AB, Shapiro B, Depireux DA. Magnetic Nanoparticle Mediated Steroid Delivery Mitigates Cisplatin Induced Hearing Loss. Front Cell Neurosci 2017; 11:268. [PMID: 28955202 PMCID: PMC5601400 DOI: 10.3389/fncel.2017.00268] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 08/21/2017] [Indexed: 11/13/2022] Open
Abstract
Cisplatin (cis-diamminedichloroplatinum) is widely used as a chemotherapeutic drug for genitourinary, breast, lung and head and neck cancers. Though effective in inducing apoptosis in cancer cells, cisplatin treatment causes severe hearing loss among patients. Steroids have been shown to mitigate cisplatin-induced hearing loss. However, steroids may interfere with the anti-cancer properties of cisplatin if administered systemically, or are rapidly cleared from the middle and inner ear and hence lack effectiveness when administered intra-tympanically. In this work, we deliver prednisolone-loaded nanoparticles magnetically to the cochlea of cisplatin-treated mice. This magnetic delivery method substantially reduced hearing loss in treated animals at high frequency compared to control animals or animals that received intra-tympanic methylprednisolone. The method also protected the outer hair cells from cisplatin-mediated ototoxicity.
Collapse
Affiliation(s)
- Bharath Ramaswamy
- Fischell Department of Bioengineering, University of MarylandCollege Park, MD, United States.,Pfizer Inc.New York, NY, United States
| | - Soumen Roy
- Sensory Cell Biology, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health (NIH)Bethesda, MD, United States
| | - Andrea B Apolo
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH)Bethesda, MD, United States
| | - Benjamin Shapiro
- Fischell Department of Bioengineering, University of MarylandCollege Park, MD, United States.,Institute for Systems Research, University of MarylandCollege Park, MD, United States.,OtomagneticsRockville, MD, United States
| | - Didier A Depireux
- Institute for Systems Research, University of MarylandCollege Park, MD, United States.,OtomagneticsRockville, MD, United States.,Department of Otorhinolaryngology/Head and Neck Surgery, University of Maryland School of MedicineBaltimore, MD, United States
| |
Collapse
|
21
|
Scasso F, Sprio AE, Canobbio L, Scanarotti C, Manini G, Berta GN, Bassi AM. Dietary supplementation of coenzyme Q10 plus multivitamins to hamper the ROS mediated cisplatin ototoxicity in humans: A pilot study. Heliyon 2017; 3:e00251. [PMID: 28239674 PMCID: PMC5318271 DOI: 10.1016/j.heliyon.2017.e00251] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/18/2017] [Accepted: 02/08/2017] [Indexed: 11/26/2022] Open
Abstract
Oxidative stress exerts major role in the pathogenesis of side effects of many antineoplastic drugs, including ototoxicity of cisplatin. In particular, increased levels of reactive oxygen species (ROS) represent one of the molecular mechanisms underlying the apoptosis of different types of hearing cells. Antioxidants and ROS scavengers may thus represent potential therapeutic options to prevent platinum-associated ototoxicity. The aim of this preliminary case-control study was to explore the efficacy of a dietary antioxidant supplement, in order to hamper the occurrences of ototoxicity in patients undergoing cisplatin chemotherapy. As results, a significant protection against cochlear toxic damage was demonstrated in patients who took the antioxidant supplement, which furthermore prevented the occurrence of hearing disorders and tinnitus. These clinical evidences were corroborated by the oxidative status of patients. After cisplatin chemotherapy, the plasma derivatives of reactive oxygen metabolites (d-ROMs) content rapidly increased in control patients, but it was maintained in those under dietary supplementation, likely because of a higher anti-ROMs potential. Indeed, an increment in rapid anti-ROMs was detected in supplemented patients, though no differences were highlighted in terms of slow anti-ROMs. In conclusion, in this preliminary report we demonstrated the feasibility of a dietary antioxidant supplementation in order to prevent the cisplatin induced hearing damage.
Collapse
Affiliation(s)
- Felice Scasso
- Department of Otorhinolaryngology, P.A. Micone Hospital, ASL n. 3 Genovese, Genoa - Sestri Ponente, Italy
| | - Andrea Elio Sprio
- Department of Clinical and Biological Sciences, Pharmacological Unit, University of Turin, c/o Ospedale San Luigi Gonzaga, Orbassano (TO), Italy
| | - Luciano Canobbio
- Medical Oncology Department, P.A. Micone Hospital, ASL n. 3 Genovese, Genoa - Sestri Ponente, Italy
| | - Chiara Scanarotti
- Department of Experimental Medicine (DIMES) - General Pathology Sect. University of Genoa, Genoa, Italy
| | | | - Giovanni Nicolao Berta
- Department of Clinical and Biological Sciences, Pharmacological Unit, University of Turin, c/o Ospedale San Luigi Gonzaga, Orbassano (TO), Italy
| | - Anna Maria Bassi
- Department of Experimental Medicine (DIMES) - General Pathology Sect. University of Genoa, Genoa, Italy
| |
Collapse
|
22
|
Reactive oxygen species, apoptosis, and mitochondrial dysfunction in hearing loss. BIOMED RESEARCH INTERNATIONAL 2015; 2015:617207. [PMID: 25874222 PMCID: PMC4385658 DOI: 10.1155/2015/617207] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 09/10/2014] [Indexed: 12/20/2022]
Abstract
Reactive oxygen species (ROS) production is involved in several apoptotic and necrotic cell death pathways in auditory tissues. These pathways are the major causes of most types of sensorineural hearing loss, including age-related hearing loss, hereditary hearing loss, ototoxic drug-induced hearing loss, and noise-induced hearing loss. ROS production can be triggered by dysfunctional mitochondrial oxidative phosphorylation and increases or decreases in ROS-related enzymes. Although apoptotic cell death pathways are mostly activated by ROS production, there are other pathways involved in hearing loss that do not depend on ROS production. Further studies of other pathways, such as endoplasmic reticulum stress and necrotic cell death, are required.
Collapse
|
23
|
Sharawy N, Rashed L, Youakim MF. Evaluation of multi-neuroprotective effects of erythropoietin using cisplatin induced peripheral neurotoxicity model. ACTA ACUST UNITED AC 2015; 67:315-22. [PMID: 25758589 DOI: 10.1016/j.etp.2015.02.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 11/12/2014] [Accepted: 02/20/2015] [Indexed: 12/24/2022]
Abstract
Cisplatin (CDDP) is severely neurotoxic anti-neoplastic drug that causes peripheral neuropathies with clinical signs known as chemotherapy-induced peripheral neurotoxicity. The ameliorating effects of erythropoeitin on cisplatin-induced neuropathy, which seem to be mediated by enhancing the cell resistance to side effects of cisplatin rather than by influencing the formation or repair rates of cisplatin-induced cross-links in the nuclear DNA, had been previously reported. The main objective of our study is to investigate the roles of nitro-oxidative stress, nuclear factor kappa B (NFκB) gene expressions and TNF levels on the previous reported erythropoietin anti-apoptotic neuroprotective effects during cisplatin induced neurotoxicity. The present study compared the effects of erythropoietin (50 μg/kg/d thrice weekly) on cisplatin (2mg/kg/d i.p. twice weekly for 4 weeks) induced neurophysiologic changes and the associated changes in the inflammatory mediators (TNF alpha and NFKB), oxidative stress (malondialdehyde (MDA), superoxide dismutases (SOD) and glutathione) and gene expression of both neuronal nitric oxide synthase (nNOS) and inducible nitric oxide synthase (iNOS). In addition, sciatic nerve pro-apoptotic and anti-apoptotic indicators (Bcl, Bax, Caspase 3) were measured. We found that concomitant administration of erythropoietin significantly reversed the cisplatin induced nitro-oxidative stress - with significant increases in sciatic nerve glutathione and superoxide dismutase antioxidant enzyme levels and a significant decrease in iNOS gene expression. We conclude that erythropoietin anti-apoptotic neuro-protective effects could partially contribute to observed antioxidant effects of erthropoietin.
Collapse
Affiliation(s)
- Nivin Sharawy
- Department of Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt.
| | - Laila Rashed
- Department of Biochemistry, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | |
Collapse
|
24
|
Protective effect of metformin against cisplatin-induced ototoxicity in an auditory cell line. J Assoc Res Otolaryngol 2013; 15:149-58. [PMID: 24297263 DOI: 10.1007/s10162-013-0431-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 11/18/2013] [Indexed: 12/16/2022] Open
Abstract
Metformin, an antidiabetic drug with potent anticancer activity, is known to prevent oxidative stress-induced cell death in several cell types through a mechanism dependent on the mitochondria. In the present study, we investigated the influence of metformin on cisplatin ototoxicity in an auditory cell line. Cell viability was determined using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromide (Sigma, St. Louis, MO, USA) cell proliferation assay. Oxidative stress and apoptosis were assessed by flow cytometry analysis, Hoechst 33258 staining, reactive oxygen species (ROS) measurement, and western blotting. Intracellular calcium concentration changes were detected using calcium imaging. Pretreatment with 1 mM metformin prior to the application of 20 μM cisplatin significantly decreased the frequency of late apoptosis in HEI-OC1 cells and also significantly attenuated the cisplatin-induced increase in ROS. In addition, metformin inhibited the activation of caspase-3 and levels of poly-ADP-ribose polymerase (PARP). Pretreatment with metformin prevented the cisplatin-induced elevation in intracellular calcium concentrations. We propose that metformin protects against cisplatin-induced ototoxicity by inhibiting the increase in intracellular calcium levels, preventing apoptosis, and limiting ROS production.
Collapse
|
25
|
Hughes AL, Hussain N, Pafford R, Parham K. Dexamethasone otoprotection in a multidose cisplatin ototoxicity mouse model. Otolaryngol Head Neck Surg 2013; 150:115-20. [PMID: 24233060 DOI: 10.1177/0194599813511948] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To develop a murine model for multidose administration of cisplatin that produces significant hearing threshold elevations and to use this model to assess the protective properties of intratympanic (IT) dexamethasone against cisplatin ototoxicity. STUDY DESIGN Controlled repeated measures design. SETTING Translational research laboratory. SUBJECTS AND METHODS Intraperitoneal (IP) cisplatin, 2 or 3 mg/kg/day, was administered for a total of 5 or 10 days in young CBA/J mice. Pure-tone evoked auditory brainstem response (ABR) thresholds were performed on days 7, 14, 21, and 28 to evaluate hearing threshold shifts. After development of the optimal dosing regimen, 15 mice received IT dexamethasone (24 mg/ml) in one ear and IT saline in the contralateral ear. RESULTS Significant threshold elevations were obtained for the 2 and 3 mg/kg/day 10 day groups, but both had high mortality rates and were excluded as potential multidose murine models. The 3 mg/kg/day 5 day group had a lower mortality rate and significant ABR threshold elevations for all frequencies on days 7, 14, 21, and 28. Using this dosing model, no statistically significant difference between IT dexamethasone and saline treated ears was found. CONCLUSIONS Unlike previous single dose models, IT dexamethasone did not have an otoprotective effect in a multidose murine model of cisplatin ototoxicity.
Collapse
Affiliation(s)
- Amy Lawrason Hughes
- University of Connecticut Health Center, Department of Surgery, Division of Otolaryngology-Head & Neck Surgery, Farmington, Connecticut, USA
| | | | | | | |
Collapse
|
26
|
Dalian D, Haiyan J, Yong F, Yongqi L, Salvi R, Someya S, Tanokura M. Ototoxic Model of Oxaliplatin and Protection from Nicotinamide Adenine Dinucleotide. J Otol 2013; 8:63-71. [PMID: 25419212 DOI: 10.1016/s1672-2930(13)50009-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Oxaliplatin, an anticancer drug commonly used to treat colorectal cancer and other tumors, has a number of serious side effects, most notably neuropathy and ototoxicity. To gain insights into its ototoxic profile, oxaliplatin was applied to rat cochlear organ cultures. Consistent with it neurotoxic propensity, oxaliplatin selectively damaged nerve fibers at a very low dose 1 μM. In contrast, the dose required to damage hair cells and spiral ganglion neurons was 50 fold higher (50 μM). Oxailiplatin-induced cochlear lesions initially increased with dose, but unexpectedly decreased at very high doses. This non-linear dose response could be related to depressed oxaliplatin uptake via active transport mechanisms. Previous studies have demonstrated that axonal degeneration involves biologically active processes which can be greatly attenuated by nicotinamide adenine dinucleotide (NAD+). To determine if NAD+ would protect spiral ganglion axons and the hair cells from oxaliplatin damage, cochlear cultures were treated with oxaliplatin alone at doses of 10 μM or 50 μM respectively as controls or combined with 20 mM NAD+. Treatment with 10 μM oxaliplatin for 48 hours resulted in minor damage to auditory nerve fibers, but spared cochlear hair cells. However, when cochlear cultures were treated with 10 μM oxaliplatin plus 20 mM NAD+, most auditory nerve fibers were intact. 50 μM oxaliplatin destroyed most of spiral ganglion neurons and cochlear hair cells with apoptotic characteristics of cell fragmentations. However, 50 μM oxaliplatin plus 20 mM NAD+ treatment greatly reduced neuronal degenerations and hair cell missing. The results suggested that NAD+ provides significant protection against oxaliplatin-induced neurotoxicity and ototoxicity, which may be due to its actions of antioxidant, antiapoptosis, and energy supply.
Collapse
Affiliation(s)
- Ding Dalian
- Center for Hearing and Deafness, State University of New York at Buffalo, USA ; Sixth People's Hospital, Shanghai Oriental Otolaryngology Institute, Shanghai Jiao Tong University, China ; Xiangya Hospital, Central South University, China ; Department of Applied Biological Chemistry, University of Tokyo, Japan
| | - Jiang Haiyan
- Center for Hearing and Deafness, State University of New York at Buffalo, USA
| | - Fu Yong
- The First Officiated Hospital, College of Medicine, Zhejiang University
| | - Li Yongqi
- The Third Affiliated Hospital of Sun Yat-Sen University
| | - Richard Salvi
- Center for Hearing and Deafness, State University of New York at Buffalo, USA
| | | | - Masaru Tanokura
- Department of Applied Biological Chemistry, University of Tokyo, Japan
| |
Collapse
|
27
|
Mechanisms of cisplatin ototoxicity: theoretical review. The Journal of Laryngology & Otology 2013; 127:536-41. [DOI: 10.1017/s0022215113000947] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
AbstractIntroduction:Cisplatin is an effective chemotherapeutic agent commonly used in the treatment of malignant tumours, but ototoxicity is a significant side effect.Objectives:To discuss the mechanisms of cisplatin ototoxicity and subsequent cell death, and to present the results of experimental studies.Material and methods:We conducted a systematic search for data published in national and international journals and books, using the Medline, SciELO, Bireme, LILACS and PubMed databases.Results:The nicotinamide adenine dinucleotide phosphate oxidase 3 isoform (also termed NOX3) seems to be the main source of reactive oxygen species in the cochlea. These reactive oxygen species react with other molecules and trigger processes such as lipid peroxidation of the plasma membrane and increases in expression of the transient vanilloid receptor potential 1 ion channel.Conclusion:Cisplatin ototoxicity proceeds via the formation of reactive oxygen species in cochlear tissue, with apoptotic cell death as a consequence.
Collapse
|
28
|
Shafik AG, Elkabarity RH, Thabet MT, Soliman NB, Kalleny NK. Effect of intratympanic dexamethasone administration on cisplatin-induced ototoxicity in adult guinea pigs. Auris Nasus Larynx 2013; 40:51-60. [DOI: 10.1016/j.anl.2012.05.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Revised: 05/15/2012] [Accepted: 05/23/2012] [Indexed: 12/13/2022]
|
29
|
Kopecky BJ, Decook R, Fritzsch B. N-Myc and L-Myc are essential for hair cell formation but not maintenance. Brain Res 2012; 1484:1-14. [PMID: 23022312 DOI: 10.1016/j.brainres.2012.09.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 08/31/2012] [Accepted: 09/13/2012] [Indexed: 01/01/2023]
Abstract
Sensorineural hearing loss results from damage to the hair cells of the organ of Corti and is irreversible in mammals. While hair cell regeneration may prove to be the ideal therapy after hearing loss, prevention of initial hair cell loss could provide even more benefit at a lower cost. Previous studies have shown that the deletion of Atoh1 results in embryonic loss of hair cells while the absence of Barhl1, Gfi1, and Pou4f3 leads to the progressive loss of hair cells in newborn mice. We recently reported that in the early embryonic absence of N-Myc (using Pax2-Cre), hair cells in the organ of Corti develop and remain until at least seven days after birth, with subsequent progressive loss. Thus, N-Myc plays a role in hair cell viability; however, it is unclear if this is due to its early expression in hair cell precursors and throughout the growing otocyst as it functions through proliferation or its late expression exclusively in differentiated hair cells. Furthermore, the related family member L-Myc is mostly co-expressed in the ear, including in differentiated hair cells, but its function has not been studied and could be partially redundant to N-Myc. To test for a long-term function of the Mycs in differentiated hair cells, we generated nine unique genotypes knocking out N-Myc and/or L-Myc after initial formation of hair cells using the well-characterized Atoh1-Cre. We tested functionality of the auditory and vestibular systems at both P21 and four months of age and under the administration of the ototoxic drug cisplatin. We conclude that neither N-Myc nor L-Myc is likely to play important roles in long-term hair cell maintenance. Therefore, it is likely that the late-onset loss of hair cells resulting from early deletion of the Mycs leads to an unsustainable developmental defect.
Collapse
|
30
|
Reactive oxygen species in apoptosis induced by cisplatin: review of physiopathological mechanisms in animal models. Eur Arch Otorhinolaryngol 2012; 269:2455-9. [DOI: 10.1007/s00405-012-2029-0] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Accepted: 04/20/2012] [Indexed: 12/20/2022]
|
31
|
Rybak LP, Mukherjea D, Jajoo S, Kaur T, Ramkumar V. siRNA-mediated knock-down of NOX3: therapy for hearing loss? Cell Mol Life Sci 2012; 69:2429-34. [PMID: 22562580 DOI: 10.1007/s00018-012-1016-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 04/18/2012] [Accepted: 04/20/2012] [Indexed: 11/27/2022]
Abstract
Cisplatin is a widely used chemotherapeutic agent that causes significant hearing loss. Previous studies have shown that cisplatin exposure is associated with increase in reactive oxygen species (ROS) in the cochlea. The inner ear expresses a unique isoform of NADPH oxidase, NOX3. This enzyme may be the primary source of ROS generation in the cochlea. The knockdown of NOX3 by pretreatment with siRNA prevented cisplatin ototoxicity, as demonstrated by preservation of hearing thresholds and inner ear sensory cells. Trans-tympanic NOX3 siRNA reduced the expression of NOX3 and biomarkers of cochlear damage, including transient receptor vanilloid 1 (TRPV1) channel and kidney injury molecule-1 (KIM-1) in cochlear tissues. In addition, siRNA against NOX3 reduced apoptosis as demonstrated by TUNEL staining, and prevented the increased expression of Bax and abrogated the decrease in Bcl2 expression following cisplatin administration. Trans-tympanic administration of siRNA directed against NOX3 may provide a useful method of attenuating cisplatin ototoxicity. In this paper, we review recent publications dealing with the role of NOX3 in ototoxicity and the effects of siRNA against cisplatin-induced hearing loss.
Collapse
Affiliation(s)
- Leonard P Rybak
- Department of Surgery, Division of Otolaryngology, Southern Illinois University, School of Medicine, Springfield, IL 62794-9649, USA.
| | | | | | | | | |
Collapse
|
32
|
Harris MS, Gilbert JL, Lormore KA, Musunuru SA, Fritsch MH. Cisplatin ototoxicity affecting cochlear implant benefit. Otol Neurotol 2011; 32:969-72. [PMID: 21730884 PMCID: PMC3144856 DOI: 10.1097/mao.0b013e3182255893] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Report a case of loss of cochlear implant benefit after cisplatin therapy to treat osteosarcoma. Examine the implications for the loci of cisplatin-associated cochleotoxicity. STUDY DESIGN Retrospective case review. SETTING Tertiary referral center. PATIENTS Single case study. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Cochlear implant programming levels. RESULTS Increase in cochlear implant programming T- and C-levels after cisplatin therapy. CONCLUSION Cisplatin therapy likely affects spiral ganglion cells. It seems that auditory cells other than outer hair cells in the organ of Corti are affected by cisplatin because the hearing sensitivity of this patient with nonfunctioning outer hair cells declined after receiving chemotherapy. Implications of these findings are discussed.
Collapse
Affiliation(s)
- Michael S Harris
- DeVault Otologic Research Laboratory, Department of Otolaryngology-Head and Neck Surgery, Indiana, USA.
| | | | | | | | | |
Collapse
|
33
|
Oh GS, Kim HJ, Choi JH, Shen A, Kim CH, Kim SJ, Shin SR, Hong SH, Kim Y, Park C, Lee SJ, Akira S, Park R, So HS. Activation of lipopolysaccharide-TLR4 signaling accelerates the ototoxic potential of cisplatin in mice. THE JOURNAL OF IMMUNOLOGY 2010; 186:1140-50. [PMID: 21148032 DOI: 10.4049/jimmunol.1002183] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Dysfunction in immune surveillance during anticancer chemotherapy of patients often causes weakness of the host defense system and a subsequent increase in microbial infections. However, the deterioration of organ-specific function related to microbial challenges in cisplatin-treated patients has not yet been elucidated. In this study, we investigated cisplatin-induced TLR4 expression and its binding to LPS in mouse cochlear tissues and the effect of this interaction on hearing function. Cisplatin increased the transcriptional and translational expression of TLR4 in the cochlear tissues, organ of Corti explants, and HEI-OC1 cells. Furthermore, cisplatin increased the interaction between TLR4 and its microbial ligand LPS, thereby upregulating the production of proinflammatory cytokines, such as TNF-α, IL-1β, and IL-6, via NF-κB activation. In C57BL/6 mice, the combined injection of cisplatin and LPS caused severe hearing impairment compared with that in the control, cisplatin-alone, or LPS-alone groups, whereas this hearing dysfunction was completely suppressed in both TLR4 mutant and knockout mice. These results suggest that hearing function can be easily damaged by increased TLR expression and microbial infections due to the weakened host defense systems of cancer patients receiving therapy comprising three to six cycles of cisplatin alone or cisplatin combined with other chemotherapeutic agents. Moreover, such damage can occur even though patients may not experience ototoxic levels of cumulative cisplatin concentration.
Collapse
Affiliation(s)
- Gi-Su Oh
- Vestibulocochlear Research Center, Wonkwang University School of Medicine, Iksan, Jeonbuk 570-749, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
García-Berrocal JR, Nevado J, González-García JA, Sánchez-Rodríguez C, Sanz R, Trinidad A, España P, Citores MJ, Ramírez-Camacho R. Heat shock protein 70 and cellular disturbances in cochlear cisplatin ototoxicity model. J Laryngol Otol 2010; 124:599-609. [PMID: 20307356 DOI: 10.1017/s0022215110000496] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Exposure to cisplatin leads to cochlear cell death by apoptosis; these changes are most marked on the seventh day after exposure. Heat shock proteins are induced in inner ear cells in response to a variety of stimuli. This study examined the role of heat shock protein 70 in cisplatin-induced cochlear cell death. METHODS Fifty-six Sprague-Dawley rats were involved. Some were injected with cisplatin (5 mg/kg body weight), some with cisplatin plus the caspase inhibitor Z-Asp(OMe)-Glu(OMe)-Val-Asp(OME)-fluoromethylketone (5 mg/kg body weight) and others were left as controls (being injected only with saline). Seven days later, we examined the expression of heat shock protein 70 and several other apoptosis-related proteins within the rat cochlear cells; we also assessed total superoxide dismutase activity, auditory brainstem response and auditory steady state response. RESULTS Seven days after cisplatin injection, significantly increased expression of heat shock protein 70 was found within the rat cochleae. This correlated with increased executioner caspase levels, total superoxide dismutase activity and auditory brainstem response thresholds, and a significant elevation in auditory steady state response thresholds. Inhibition of caspase-3 activity significantly reduced cochlear heat shock protein 70 expression and total superoxide dismutase activity, and improved auditory brainstem response and auditory steady state response thresholds. CONCLUSIONS Seven days after cisplatin exposure, we found disturbances of the cochlear cellular machinery involving heat shock protein 70, other apoptotic proteins and total superoxide dismutase.
Collapse
|
35
|
Roles of NADPH oxidases in cisplatin-induced reactive oxygen species generation and ototoxicity. J Neurosci 2010; 30:3933-46. [PMID: 20237264 DOI: 10.1523/jneurosci.6054-09.2010] [Citation(s) in RCA: 216] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In our previous study, we clearly demonstrated the roles of pro-inflammatory cytokines, including tumor necrosis factor-alpha, interleukin-1beta (IL-1beta), and IL-6, and subsequent reactive oxygen species (ROS) generation on the pathogenesis of cisplatin ototoxicity in vitro and in vivo. ROS generation in cisplatin-treated HEI-OC1 auditory cells was also correlated with changing mitochondrial membrane potential. However, the roles of NADPH oxidase in cisplatin-induced ROS generation and ototoxicity have not been fully elucidated. Herein, immunohistochemical studies demonstrated that treatment of cisplatin induced the expression of NADPH oxidase isoforms NOX-1 and NOX-4 in HEI-OC1 auditory cells. Expression of mRNA for NOX-1, NOX-4, NOXO1, NOXA1, p47(phox), and p67(phox) was also increased. Inhibition of NADPH oxidase with diphenyleniodonium chloride or apocynin abolished ROS production and the subsequent apoptotic cell death in cisplatin-treated cells. Furthermore, suppression of NOX1 and NOX4 expression by small interfering RNA transfection markedly abolished the cytotoxicity and ROS generation by cisplatin. Together, our data suggest that ROS generated, in part, through the activation of NADPH oxidase plays an essential role in cisplatin ototoxicity.
Collapse
|
36
|
Rybak LP, Mukherjea D, Jajoo S, Ramkumar V. Cisplatin ototoxicity and protection: clinical and experimental studies. TOHOKU J EXP MED 2009; 219:177-86. [PMID: 19851045 PMCID: PMC2927105 DOI: 10.1620/tjem.219.177] [Citation(s) in RCA: 259] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Cisplatin is a chemotherapeutic agent that is widely used to treat a variety of malignant tumors. Serious dose-limiting side effects like ototoxicity, nephrotoxicity and neurotoxicity occur with the use of this agent. This review summarizes recent important clinical and experimental investigations of cisplatin ototoxicity. It also discusses the utility of protective agents employed in patients and in experimental animals. The future strategies for limiting cisplatin ototoxicity will need to avoid interference with the therapeutic effect of cisplatin in order to enhance the quality of life of patients receiving this important anti-tumor agent.
Collapse
Affiliation(s)
- Leonard P Rybak
- Department of Surgery, Southern Illinois University School of Medicine, IL, USA.
| | | | | | | |
Collapse
|
37
|
Effect of the chronic combined administration of cisplatin and paclitaxel in a rat model of peripheral neurotoxicity. Eur J Cancer 2008; 45:656-65. [PMID: 19091544 DOI: 10.1016/j.ejca.2008.10.038] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2008] [Revised: 10/14/2008] [Accepted: 10/24/2008] [Indexed: 11/22/2022]
Abstract
We have characterised for the first time the general and neurological side effects experienced when using a series of chronic non-lethal cisplatin + paclitaxel schedules in Wistar rats, selected according to our previous experience and the animals' maximum tolerated dose. At the pathological level, the use of combination schedules was definitely more toxic at the kidney and sternal bone marrow level than the single-agent schedules. At the neurophysiological examination based on the assessment of the nerve conduction velocity measurement in the tail nerve, we identified only one combination schedule that was more neurotoxic than the similar schedules based on single-agent administration. This observation was confirmed by the neuropathological examination performed on the sciatic nerve, dorsal root ganglia, ventral and dorsal roots. Our study supports the hypothesis that the general and, to a lesser extent, neurological effects of a combination of cisplatin and paclitaxel are different from those of the administration of both drugs as single agents. We believe that these models may be useful for testing neuroprotective strategies.
Collapse
|
38
|
García-Berrocal JR, Nevado J, Ramírez-Camacho R, Sanz R, González-García JA, Sánchez-Rodríguez C, Cantos B, España P, Verdaguer JM, Trinidad Cabezas A. The anticancer drug cisplatin induces an intrinsic apoptotic pathway inside the inner ear. Br J Pharmacol 2007; 152:1012-20. [PMID: 17906689 PMCID: PMC2095105 DOI: 10.1038/sj.bjp.0707405] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND AND PURPOSE Ototoxicity is a known adverse effect of cisplatin (CDDP). Since apoptosis is involved in the development of some pathological conditions associated with the administration of anticancer drugs, we examined, using immunohistochemical and electrophysiological techniques, the apoptotic changes in the cochlea of Sprague-Dawley (SD) rats after an injection of CDDP (5 mgkg(-1) body weight). EXPERIMENTAL APPROACH Luciferase assays were used to determine the different caspase activities and ATP levels in protein extracts of whole cochleae. The expression of several apoptotic-related proteins was measured by means of Western blotting. These analyses were performed 2, 7 and 30 days after the CDDP injection. The auditory brain stem response was obtained before and at the different times after the injection of CDDP, before the animals were killed. KEY RESULTS CDDP significantly increased the levels of caspase-3/7 activity and active caspase-3 protein expression and caspase-3 immunofluorescence staining, caspase-9 activity, and Bax protein expression but decreased Bcl-2 protein expression within the rat cochleae. Threshold shifts were significantly elevated 2 days after CDDP treatment. CONCLUSIONS AND IMPLICATIONS These findings support the hypothesis that cisplatin-related apoptosis evokes an intrinsic pathway of pro-apoptotic signalling within the rat cochleae. Thus, selective inhibition of the sequence of events involved in the intrinsic apoptotic pathway could provide a strategy to minimize cisplatin-induced ototoxicity.
Collapse
Affiliation(s)
- J R García-Berrocal
- Servicio de Otorrinolaringología, Hospital Universitario Puerta de Hierro, Madrid, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Gross J, Machulik A, Amarjargal N, Moller R, Ungethüm U, Kuban RJ, Fuchs FU, Andreeva N, Fuchs J, Henke W, Pohl EE, Szczepek AJ, Haupt H, Mazurek B. Expression of apoptosis-related genes in the organ of Corti, modiolus and stria vascularis of newborn rats. Brain Res 2007; 1162:56-68. [PMID: 17612509 DOI: 10.1016/j.brainres.2007.05.061] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2006] [Revised: 04/19/2007] [Accepted: 05/21/2007] [Indexed: 11/17/2022]
Abstract
Cell death in the inner ear tissues is an important mechanism leading to hearing impairment. Here, using microarrays and real-time RT-PCR we analyzed expression of selected apoptosis-related genes in rat's inner ear. We determined the gene expression in tissues freshly isolated from neonatal rats (3-5 days old) and compared it to that of explants cultured for 24 h under normoxic or hypoxic conditions. For the analyses, we used pooled samples of the organ of Corti (OC), modiolus (MOD) and stria vascularis (SV), respectively. We observed region-specific changes in gene expression between the fresh tissues and the normoxic culture. In the OC, expression of the proapoptotic genes caspase-2, caspase-3, caspase-6 and calpain-1 was downregulated. In the MOD, the antioxidative defense SOD-2 and SOD-3 were upregulated. In the SV, caspase-2, caspase-6, calpain-1 and SOD-3 were downregulated and SOD-2 upregulated. We speculate that these changes could reflect survival shift in transcriptome of inner ear explants tissues under in vitro conditions. With the exception of SOD-2, hypoxic culture conditions induced the same changes in gene expression as the normoxic conditions indicating that culture preparation is likely the dominating factor, which modifies the gene expression pattern. We conclude that various culture conditions induce different expression pattern of apoptosis-related genes in the organotypic cochlear cultures, as compared to fresh tissues. This transcriptional pattern may reflect the survival ability of specific tissues and could become a tempting target for a pharmacological intervention in inner ear diseases.
Collapse
Affiliation(s)
- Johann Gross
- Dept. Othorhinolaryngology, Charité-Universitätsmedizin Berlin, Molecular Biology Research Laboratory, Charitéplatz 1, 10117-Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Wang J, Pignol B, Chabrier PE, Saido T, Lloyd R, Tang Y, Lenoir M, Puel JL. A novel dual inhibitor of calpains and lipid peroxidation (BN82270) rescues the cochlea from sound trauma. Neuropharmacology 2007; 52:1426-37. [PMID: 17449343 DOI: 10.1016/j.neuropharm.2007.02.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2006] [Revised: 01/24/2007] [Accepted: 02/08/2007] [Indexed: 10/23/2022]
Abstract
Free radical and calcium buffering mechanisms are implicated in cochlear cell damage that has been induced by sound trauma. Thus in this study we evaluated the therapeutic effect of a novel dual inhibitor of calpains and of lipid peroxidation (BN 82270) on the permanent hearing and hair cell loss induced by sound trauma. Perfusion of BN 82270 into the scala tympani of the guinea pig cochlea prevented the formation of calpain-cleaved fodrin, translocation of cytochrome c, DNA fragmentation and hair cell degeneration caused by sound trauma. This was confirmed by functional tests in vivo, showing a clear dose-dependent reduction of permanent hearing loss (ED50 = 4.07 microM) with almost complete protection at 100 microM. Furthermore, BN82270 still remained effective even when applied onto the round window membrane after sound trauma had occurred, within a therapeutic window of 24 h. This indicates that BN 82270 may be of potential therapeutic value in treating the cochlea after sound trauma.
Collapse
MESH Headings
- Action Potentials/drug effects
- Action Potentials/physiology
- Animals
- Apoptosis/drug effects
- Calpain/antagonists & inhibitors
- Carrier Proteins/metabolism
- Cochlea/enzymology
- Cochlea/injuries
- Cochlea/pathology
- Cysteine Proteinase Inhibitors/pharmacology
- Cytochromes c/metabolism
- DNA Fragmentation/drug effects
- Dipeptides/pharmacology
- Electrophysiology
- Female
- Guinea Pigs
- Hair Cells, Auditory/drug effects
- Hair Cells, Auditory/pathology
- Hair Cells, Auditory/physiology
- Hearing Loss, Noise-Induced/pathology
- Hearing Loss, Noise-Induced/prevention & control
- Immunohistochemistry
- Lipid Peroxidation/drug effects
- Microfilament Proteins/metabolism
- Microscopy, Electron, Scanning
- Microscopy, Electron, Transmission
- Round Window, Ear/pathology
- Tympanic Membrane/drug effects
Collapse
Affiliation(s)
- Jing Wang
- INSERM U583, Laboratoire de Physiopathologie et Thérapie des Déficits Sensoriels et Moteurs, Montpellier, France
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Laurell G, Ekborn A, Viberg A, Canlon B. Effects of a single high dose of cisplatin on the melanocytes of the stria vascularis in the guinea pig. Audiol Neurootol 2007; 12:170-8. [PMID: 17259704 DOI: 10.1159/000099020] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2006] [Accepted: 10/18/2006] [Indexed: 11/19/2022] Open
Abstract
The antineoplastic drug cisplatin is known to cause a reduction in endocochlear potential. The hypothesis to be tested was whether a single high dose of cisplatin affects the melanocytes by altering the expression of melanin. Pigmented guinea pigs received a bolus injection of cisplatin (8 mg/kg as a 15-second intravenous infusion). Auditory brainstem response (ABR) thresholds and morphological analysis of the hair cells and the stria vascularis were made 96 h after injection. ABR thresholds were elevated (15-40 dB) at 12-30 kHz and a significant loss of outer hair cells in the more basal regions was found. Cisplatin caused a significantly lower density of melanin in the intermediate cells in the basal region without any signs of apoptosis. Changes in melanin content were not noted in the middle or apical cochlear regions. Significant correlations were found between melanin density, ABR threshold shifts and outer hair cell loss in the region corresponding to 30 kHz. The findings reported here further support the multiple cytotoxic effect of cisplatin on the inner ear.
Collapse
Affiliation(s)
- Göran Laurell
- Department of Clinical Sciences, Umeå University, Umeå, Sweden.
| | | | | | | |
Collapse
|
42
|
Rybak LP, Whitworth CA, Mukherjea D, Ramkumar V. Mechanisms of cisplatin-induced ototoxicity and prevention. Hear Res 2006; 226:157-67. [PMID: 17113254 DOI: 10.1016/j.heares.2006.09.015] [Citation(s) in RCA: 403] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2006] [Revised: 09/09/2006] [Accepted: 09/24/2006] [Indexed: 11/27/2022]
Abstract
Cisplatin is a widely used chemotherapeutic agent to treat malignant disease. Unfortunately, ototoxicity occurs in a large percentage of patients treated with higher dose regimens. In animal studies and in human temporal bone investigations, several areas of the cochlea are damaged, including outer hair cells in the basal turn, spiral ganglion cells and the stria vascularis, resulting in hearing impairment. The mechanisms appear to involve the production of reactive oxygen species (ROS), which can trigger cell death. Approaches to chemoprevention include the administration of antioxidants to protect against ROS at an early stage in the ototoxic pathways and the application of agents that act further downstream in the cell death cascade to prevent apoptosis and hearing loss. This review summarizes recent data that shed new light on the mechanisms of cisplatin ototoxicity and its prevention.
Collapse
Affiliation(s)
- Leonard P Rybak
- Department of Surgery, Division of Otolaryngology, Southern Illinois University, School of Medicine, P.O. Box 19653, Springfield, IL 62794-9653, USA.
| | | | | | | |
Collapse
|
43
|
Mukherjea D, Whitworth CA, Nandish S, Dunaway GA, Rybak LP, Ramkumar V. Expression of the kidney injury molecule 1 in the rat cochlea and induction by cisplatin. Neuroscience 2006; 139:733-40. [PMID: 16464536 DOI: 10.1016/j.neuroscience.2005.12.044] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2005] [Revised: 12/16/2005] [Accepted: 12/22/2005] [Indexed: 11/29/2022]
Abstract
Cisplatin is a widely used chemotherapeutic agent whose dose-limiting side effects include ototoxicity and nephrotoxicity. Recent evidence indicates that cisplatin induces the expression of a novel protein, kidney injury molecule-1, in the renal proximal tubular epithelium to aid in regeneration. In this study, we determined whether kidney injury molecule-1 is expressed in the cochlea and is induced by cisplatin. Using reverse transcriptase polymerase chain reaction techniques, we have now identified kidney injury molecule-1 in the rat cochlea and in three different mouse transformed hair cell lines. Administration of cisplatin to rats produced hearing loss and induced kidney injury molecule-1 mRNA in the rat cochlea. Pretreatment of rats with lipoic acid, a scavenger of reactive oxygen species, significantly reduced cisplatin-induced hearing loss and kidney injury molecule-1 expression. Cisplatin also increased the expression of cochlear NOX3 mRNA, a member of the superoxide generating NADPH oxidase family of proteins recently identified in the cochlea, inhibition of which decreased kidney injury molecule-1 expression. Polymerase chain reaction performed on different regions of the cochlea indicated the presence of kidney injury molecule-1 mRNA in the lateral wall, organ of Corti and spiral ganglion. This distribution was confirmed by immunocytochemistry. Taken together, these data identify kidney injury molecule-1 as a novel cochlear injury molecule, whose expression is regulated by reactive oxygen species generated via the NADPH oxidase pathway.
Collapse
Affiliation(s)
- D Mukherjea
- Department of Pharmacology, Southern Illinois University School of Medicine, PO Box 19629, Springfield, IL 62794-9626, USA
| | | | | | | | | | | |
Collapse
|
44
|
Cheng PW, Liu SH, Hsu CJ, Lin-Shiau SY. Correlation of increased activities of Na+, K+-ATPase and Ca2+-ATPase with the reversal of cisplatin ototoxicity induced by d-methionine in guinea pigs. Hear Res 2005; 205:102-9. [PMID: 15953519 DOI: 10.1016/j.heares.2005.03.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2005] [Accepted: 03/08/2005] [Indexed: 11/23/2022]
Abstract
Na(+), K(+)-ATPase and Ca(2+)-ATPase in the cochlear lateral wall play an important role in maintaining ionic homeostasis and physiologic function of the cochlea. The present study was designed to test whether the changes of Na(+), K(+)-ATPase and Ca(2+)-ATPase activities of the cochlear lateral wall and the brainstem of guinea pigs after receiving cisplatin for seven consecutive days were correlated with the altered auditory brainstem responses (ABR). Furthermore, whether a chemoprotective agent, D-methionine reversed the increased ABR threshold induced by cisplatin accompanied with the increased ATPase activities was also evaluated. The results obtained showed that cisplatin exposure caused not only a significant increase of threshold but also altered various absolute wave and interwave latencies of ABR. In addition, cisplatin significantly decreased the Na(+), K(+)-ATPase and Ca(2+)-ATPase activities in the cochlear lateral wall with a good dose-response relationship. Regression analysis indicated that an increase of ABR threshold was well correlated with a decrease of both Na(+), K(+)-ATPase and Ca(2+)-ATPase activities in the cochlear lateral wall. A chemoprotectant, D-methionine indeed reversed both abnormalities of ABR and ATPase activities in a well correlation function. The selectivity of these observed changes induced by cisplatin and D-methionine was revealed by the findings that cisplatin-treated guinea pigs had normal III-V interwave latency of ABR and no reduction of Na(+), K(+)-ATPase and Ca(2+)-ATPase specific activities in the brainstem, which is in accordance with the nonpenetrable cisplatin across the blood brain barrier. Taken all together, the present findings suggest that biochemical damage and ionic disturbance may contribute to cisplatin-induced ototoxicity to some extent, which can be reversed by d-methionine.
Collapse
Affiliation(s)
- Po-Wen Cheng
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | |
Collapse
|