1
|
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] [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
|
2
|
Tan WJT, Vlajkovic SM. Molecular Characteristics of Cisplatin-Induced Ototoxicity and Therapeutic Interventions. Int J Mol Sci 2023; 24:16545. [PMID: 38003734 PMCID: PMC10671929 DOI: 10.3390/ijms242216545] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Cisplatin is a commonly used chemotherapeutic agent with proven efficacy in treating various malignancies, including testicular, ovarian, cervical, breast, bladder, head and neck, and lung cancer. Cisplatin is also used to treat tumors in children, such as neuroblastoma, osteosarcoma, and hepatoblastoma. However, its clinical use is limited by severe side effects, including ototoxicity, nephrotoxicity, neurotoxicity, hepatotoxicity, gastrointestinal toxicity, and retinal toxicity. Cisplatin-induced ototoxicity manifests as irreversible, bilateral, high-frequency sensorineural hearing loss in 40-60% of adults and in up to 60% of children. Hearing loss can lead to social isolation, depression, and cognitive decline in adults, and speech and language developmental delays in children. Cisplatin causes hair cell death by forming DNA adducts, mitochondrial dysfunction, oxidative stress, and inflammation, culminating in programmed cell death by apoptosis, necroptosis, pyroptosis, or ferroptosis. Contemporary medical interventions for cisplatin ototoxicity are limited to prosthetic devices, such as hearing aids, but these have significant limitations because the cochlea remains damaged. Recently, the U.S. Food and Drug Administration (FDA) approved the first therapy, sodium thiosulfate, to prevent cisplatin-induced hearing loss in pediatric patients with localized, non-metastatic solid tumors. Other pharmacological treatments for cisplatin ototoxicity are in various stages of preclinical and clinical development. This narrative review aims to highlight the molecular mechanisms involved in cisplatin-induced ototoxicity, focusing on cochlear inflammation, and shed light on potential antioxidant and anti-inflammatory therapeutic interventions to prevent or mitigate the ototoxic effects of cisplatin. We conducted a comprehensive literature search (Google Scholar, PubMed) focusing on publications in the last five years.
Collapse
Affiliation(s)
- Winston J. T. Tan
- Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1023, New Zealand;
- Eisdell Moore Centre, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1023, New Zealand
| | - Srdjan M. Vlajkovic
- Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1023, New Zealand;
- Eisdell Moore Centre, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1023, New Zealand
| |
Collapse
|
3
|
Abstract
Hearing loss is often caused by death of sensory hair cells (HCs) in the inner ear. HCs are vulnerable to some ototoxic drugs, such as aminoglycosides(AGs) and the cisplatin.The most predominant form of drug-induced cell death is apoptosis. Many efforts have been made to protect HCs from cell death after ototoxic drug exposure. These mechanisms and potential targets of HCs protection will be discussed in this review.And we also propose further investigation in the field of HCs necrosis and regeneration, as well as future clinical utilization.
Collapse
|
4
|
Babolmorad G, Latif A, Domingo IK, Pollock NM, Delyea C, Rieger AM, Allison WT, Bhavsar AP. Toll-like receptor 4 is activated by platinum and contributes to cisplatin-induced ototoxicity. EMBO Rep 2021; 22:e51280. [PMID: 33733573 PMCID: PMC8097357 DOI: 10.15252/embr.202051280] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 02/18/2021] [Accepted: 02/23/2021] [Indexed: 12/12/2022] Open
Abstract
Toll-like receptor 4 (TLR4) recognizes bacterial lipopolysaccharide (LPS) and can also be activated by some Group 9/10 transition metals, which is believed to mediate immune hypersensitivity reactions. In this work, we test whether TLR4 can be activated by the Group 10 metal platinum and the platinum-based chemotherapeutic cisplatin. Cisplatin is invaluable in childhood cancer treatment but its use is limited due to a permanent hearing loss (cisplatin-induced ototoxicity, CIO) adverse effect. We demonstrate that platinum and cisplatin activate pathways downstream of TLR4 to a similar extent as the known TLR4 agonists LPS and nickel. We further show that TLR4 is required for cisplatin-induced inflammatory, oxidative, and cell death responses in hair cells in vitro and for hair cell damage in vivo. Finally, we identify a TLR4 small molecule inhibitor able to curtail cisplatin toxicity in vitro. Thus, our findings indicate that TLR4 is a promising therapeutic target to mitigate CIO.
Collapse
Affiliation(s)
- Ghazal Babolmorad
- Department of Medical Microbiology and ImmunologyFaculty of Medicine & DentistryUniversity of AlbertaEdmontonABCanada
| | - Asna Latif
- Department of Medical Microbiology and ImmunologyFaculty of Medicine & DentistryUniversity of AlbertaEdmontonABCanada
| | - Ivan K Domingo
- Department of Medical Microbiology and ImmunologyFaculty of Medicine & DentistryUniversity of AlbertaEdmontonABCanada
| | - Niall M Pollock
- Department of Biological SciencesFaculty of ScienceUniversity of AlbertaEdmontonABCanada
| | - Cole Delyea
- Department of Medical Microbiology and ImmunologyFaculty of Medicine & DentistryUniversity of AlbertaEdmontonABCanada
| | - Aja M Rieger
- Department of Medical Microbiology and ImmunologyFaculty of Medicine & DentistryUniversity of AlbertaEdmontonABCanada
| | - W Ted Allison
- Department of Biological SciencesFaculty of ScienceUniversity of AlbertaEdmontonABCanada
- Department of Medical GeneticsFaculty of Medicine & DentistryUniversity of AlbertaEdmontonABCanada
| | - Amit P Bhavsar
- Department of Medical Microbiology and ImmunologyFaculty of Medicine & DentistryUniversity of AlbertaEdmontonABCanada
- Department of Medical GeneticsFaculty of Medicine & DentistryUniversity of AlbertaEdmontonABCanada
| |
Collapse
|
5
|
Ibrahim MA, Albahlol IA, Wani FA, Abd-Eltawab Tammam A, Kelleni MT, Sayeed MU, Abd El-Fadeal NM, Mohamed AA. Resveratrol protects against cisplatin-induced ovarian and uterine toxicity in female rats by attenuating oxidative stress, inflammation and apoptosis. Chem Biol Interact 2021; 338:109402. [PMID: 33587916 DOI: 10.1016/j.cbi.2021.109402] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 01/02/2021] [Accepted: 01/31/2021] [Indexed: 12/20/2022]
Abstract
Cisplatin is an important antineoplastic drug used in multiple chemotherapeutic regimens but unfortunately causes serious toxic effects as ovarian and uterine toxicity. This study aimed to investigate the potential protective effect of resveratrol (RSV) against cisplatin-induced ovarian and uterine toxicity in female rats. Thirty-two female Wistar rats were divided randomly into four groups (n = 8 in each). Control group received oral normal saline for 28 days; RSV group received RSV (10 mg/kg; daily) via oral gavage; CIS group received a single dose of CIS (7 mg/kg; i.p.) on the 21st day; (CIS + RSV) group received both RSV and CIS by the same schedules and doses of RSV and CIS groups, respectively. Results demonstrated a significant decrease in MDA level and a significant increase in both glutathione content and activity of the antioxidant enzymes GPx, SOD, and CAT in the tissues of the ovary and uterus of CIS + RSV group in comparison to that of CIS group (P<0.05), also there are significantly decreased tissue levels of the proinflammatory cytokines and enzymes (NF-κB, IL-1β, IL-6, TNF-α, COX-2, and iNOS), increased estradiol, progesterone, prolactin and decreased FSH serum levels in CIS + RSV group compared to CIS group (P < 0.05). Moreover, there is downregulation of tissues Cleaved Caspase-3, NF-κB and Cox-2 proteins as shown in Western blot analysis, also apoptosis was significantly inhibited, evidenced by downregulation of Bax and upregulation of Bcl-2 proteins, and the ovarian and uterine histological architecture and integrity were maintained in CIS + RSV group compared to CIS group. In conclusion, these findings indicate that RSV has beneficial effects in ameliorating cisplatin-induced oxidative stress, inflammation, and apoptosis in the ovarian and uterine tissues of female rats.
Collapse
Affiliation(s)
- Mahrous Abdelbasset Ibrahim
- Forensic Medicine and Clinical Toxicology, College of Medicine, Jouf University, Aljouf, Saudi Arabia; Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Suez Canal University (SCU), Ismailia, 41522, Egypt.
| | - Ibrahim Abdelkhalek Albahlol
- Obstetrics and Gynecology Department, College of Medicine, Jouf University, Aljouf, Saudi Arabia; Obstetrics and Gynecology Department, Faculty of Medicine, Mansoura University, Egypt.
| | - Farooq Ahmed Wani
- Pathology Department, College of Medicine, Jouf University, Aljouf, Saudi Arabia.
| | - Ahmed Abd-Eltawab Tammam
- Physiology Department, College of Medicine, Jouf University, Aljouf, Saudi Arabia; Physiology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt.
| | - Mina Thabet Kelleni
- Pharmacology Department, Faculty of Medicine, Minia University, Minia, Egypt.
| | | | - Noha M Abd El-Fadeal
- Medical Biochemistry and Molecular Biology, Faculty of Medicine, Suez Canal University (SCU), Ismailia, Egypt.
| | - Alaa Abdelhamid Mohamed
- Medical Biochemistry Division, Pathology Department, College of Medicine, Jouf University, Aljouf, Saudi Arabia; Medical Biochemistry Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt.
| |
Collapse
|
6
|
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: 55] [Impact Index Per Article: 18.3] [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
|
7
|
Gentilin E, Simoni E, Candito M, Cazzador D, Astolfi L. Cisplatin-Induced Ototoxicity: Updates on Molecular Targets. Trends Mol Med 2019; 25:1123-1132. [DOI: 10.1016/j.molmed.2019.08.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/26/2019] [Accepted: 08/02/2019] [Indexed: 12/20/2022]
|
8
|
Choi MJ, Kang H, Lee YY, Choo OS, Jang JH, Park SH, Moon JS, Choi SJ, Choung YH. Cisplatin-Induced Ototoxicity in Rats Is Driven by RIP3-Dependent Necroptosis. Cells 2019; 8:E409. [PMID: 31052605 PMCID: PMC6562419 DOI: 10.3390/cells8050409] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/25/2019] [Accepted: 04/30/2019] [Indexed: 12/22/2022] Open
Abstract
Cisplatin-induced early-onset ototoxicity is linked to hearing loss. The mechanism by which cisplatin causes ototoxicity remains unclear. The purpose of this study was to identify the involvement of receptor-interacting protein kinase (RIP)3-dependent necroptosis in cisplatin-induced ototoxicity in vitro and in vivo. Sprague-Dawley rats (SD, 8 week) were treated via intraperitoneal (i.p.) injection with cisplatin (16 mg/kg for 1 day), and their hearing thresholds were measured by the auditory brainstem response (ABR) method. Hematoxylin and eosin (H & E) staining, immunohistochemistry, and western blots were performed to determine the effect of cisplatin-induced ototoxicity on cochlear morphology. Inhibitor experiments with necrostatin 1 (Nec-1) and Z-VAD were also performed in HEI-OC1 cell line. H&E stains revealed that the necroptotic changes were increased in the organ of Corti (OC) and spiral ganglion neurons (SGNs). Moreover, immunohistochemistry and western blot analysis showed that cisplatin treatment increased the protein levels of RIP3 in both OCs and SGNs. The treatment of Nec-1, a selective RIP1 inhibitor, resulted in markedly suppression of cisplatin-induced cell death in HEI-OC1 cells, whereas Z-VAD treatment did not change the cisplatin-induced cell death. Our results suggest that RIP3-dependent necroptosis was substantial in cisplatin-induced ototoxicity; inner cochlear regions, the OCs, and SGNs were especially sensitive to necroptosis.
Collapse
Affiliation(s)
- Mi-Jin Choi
- Department of Otolaryngology, Ajou University School of Medicine, Suwon 16499, Korea.
- Bk21 Plus Research Center for Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Korea.
| | - Hyunsook Kang
- Department of Otolaryngology-Head and Neck Surgery, Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan 31151, Korea.
| | - Yun Yeong Lee
- Department of Otolaryngology, Ajou University School of Medicine, Suwon 16499, Korea.
- Bk21 Plus Research Center for Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Korea.
| | - Oak-Sung Choo
- Department of Otolaryngology, Ajou University School of Medicine, Suwon 16499, Korea.
- Bk21 Plus Research Center for Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Korea.
| | - Jeong Hun Jang
- Department of Otolaryngology, Ajou University School of Medicine, Suwon 16499, Korea.
- Bk21 Plus Research Center for Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Korea.
| | - Sung-Hee Park
- Department of Otolaryngology, Ajou University School of Medicine, Suwon 16499, Korea.
- Bk21 Plus Research Center for Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Korea.
| | - Jong-Seok Moon
- Soonchunhyang Institute of Medi-bio Science, Soonchunhyang University, Cheonan 31151, Korea.
| | - Seong Jun Choi
- Department of Otolaryngology-Head and Neck Surgery, Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan 31151, Korea.
| | - Yun-Hoon Choung
- Department of Otolaryngology, Ajou University School of Medicine, Suwon 16499, Korea.
- Bk21 Plus Research Center for Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Korea.
| |
Collapse
|
9
|
Englinger B, Pirker C, Heffeter P, Terenzi A, Kowol CR, Keppler BK, Berger W. Metal Drugs and the Anticancer Immune Response. Chem Rev 2018; 119:1519-1624. [DOI: 10.1021/acs.chemrev.8b00396] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Bernhard Englinger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Christine Pirker
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Alessio Terenzi
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Christian R. Kowol
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Bernhard K. Keppler
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
| |
Collapse
|
10
|
Hazlitt RA, Min J, Zuo J. Progress in the Development of Preventative Drugs for Cisplatin-Induced Hearing Loss. J Med Chem 2018; 61:5512-5524. [PMID: 29361217 PMCID: PMC6043375 DOI: 10.1021/acs.jmedchem.7b01653] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
![]()
Cisplatin
is a highly effective treatment for malignant cancers
and has become a cornerstone in chemotherapeutic regimens. Unfortunately,
its use in the clinic is often coupled with a high incidence of severe
hearing loss. Over the past few decades, enormous effort has been
put forth to find protective agents that selectively protect against
the ototoxic side effects of cisplatin and do not interfere with its
antitumoral activity. Many therapies have been successful in preclinical
work, but only a few have shown any protection in the clinic, and
none have been approved by the FDA. This review summarizes the clinical
and preclinical studies of the most effective small-molecule candidates
currently in clinical trials, while also detailing their molecular
mechanisms of action, to gain insight for future drug development
in the field.
Collapse
|
11
|
Sheth S, Mukherjea D, Rybak LP, Ramkumar V. Mechanisms of Cisplatin-Induced Ototoxicity and Otoprotection. Front Cell Neurosci 2017; 11:338. [PMID: 29163050 PMCID: PMC5663723 DOI: 10.3389/fncel.2017.00338] [Citation(s) in RCA: 221] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/12/2017] [Indexed: 12/20/2022] Open
Abstract
Evidence of significant hearing loss during the early days of use of cisplatin as a chemotherapeutic agent in cancer patients has stimulated research into the causes and treatment of this side effect. It has generally been accepted that hearing loss is produced by excessive generation of reactive oxygen species (ROS) in cell of the cochlea, which led to the development of various antioxidants as otoprotective agents. Later studies show that ROS could stimulate cochlear inflammation, suggesting the use of anti-inflammatory agents for treatment of hearing loss. In this respect, G-protein coupled receptors, such as adenosine A1 receptor and cannabinoid 2 receptors, have shown efficacy in the treatment of hearing loss in experimental animals by increasing ROS scavenging, suppressing ROS generation, or by decreasing inflammation. Inflammation could be triggered by activation of transient receptor potential vanilloid 1 (TRPV1) channels in the cochlea and possibly other TRP channels. Targeting TRPV1 for knockdown has also been shown to be a useful strategy for ensuring otoprotection. Cisplatin entry into cochlear hair cells is mediated by various transporters, inhibitors of which have been shown to be effective for treating hearing loss. Finally, cisplatin-induced DNA damage and activation of the apoptotic process could be targeted for cisplatin-induced hearing loss. This review focuses on recent development in our understanding of the mechanisms underlying cisplatin-induced hearing loss and provides examples of how drug therapies have been formulated based on these mechanisms.
Collapse
Affiliation(s)
- Sandeep Sheth
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Debashree Mukherjea
- Department of Surgery (Otolaryngology), Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Leonard P Rybak
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States.,Department of Surgery (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
|
12
|
Ryals M, Pak K, Jalota R, Kurabi A, Ryan AF. A kinase inhibitor library screen identifies novel enzymes involved in ototoxic damage to the murine organ of Corti. PLoS One 2017; 12:e0186001. [PMID: 29049311 PMCID: PMC5648133 DOI: 10.1371/journal.pone.0186001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 09/22/2017] [Indexed: 11/25/2022] Open
Abstract
Ototoxicity is a significant side effect of a number of drugs, including the aminoglycoside antibiotics and platinum-based chemotherapeutic agents that are used to treat life-threatening illnesses. Although much progress has been made, the mechanisms that lead to ototoxic loss of inner ear sensory hair cells (HCs) remains incompletely understood. Given the critical role of protein phosphorylation in intracellular processes, including both damage and survival signaling, we screened a library of kinase inhibitors targeting members of all the major families in the kinome. Micro-explants from the organ of Corti of mice in which only the sensory cells express GFP were exposed to 200 μM of the ototoxic aminoglycoside gentamicin with or without three dosages of each kinase inhibitor. The loss of sensory cells was compared to that seen with gentamicin alone, or without treatment. Of the 160 inhibitors, 15 exhibited a statistically significant protective effect, while 3 significantly enhanced HC loss. The results confirm some previous studies of kinase involvement in HC damage and survival, and also highlight several novel potential kinase pathway contributions to ototoxicity.
Collapse
Affiliation(s)
- Matthew Ryals
- Department of Surgery/Otolaryngology, University of California, San Diego, School of Medicine, La Jolla, California, United States of America
| | - Kwang Pak
- Department of Surgery/Otolaryngology, University of California, San Diego, School of Medicine, La Jolla, California, United States of America
| | - Rahul Jalota
- Department of Surgery/Otolaryngology, University of California, San Diego, School of Medicine, La Jolla, California, United States of America
| | - Arwa Kurabi
- Department of Surgery/Otolaryngology, University of California, San Diego, School of Medicine, La Jolla, California, United States of America
| | - Allen F. Ryan
- Department of Surgery/Otolaryngology, University of California, San Diego, School of Medicine, La Jolla, California, United States of America
- Research Service, Veterans Administration Medical Center, San Diego, California, United States of America
- * E-mail:
| |
Collapse
|
13
|
Loss of STAT1 protects hair cells from ototoxicity through modulation of STAT3, c-Jun, Akt, and autophagy factors. Cell Death Dis 2015; 6:e2019. [PMID: 26673664 PMCID: PMC4720895 DOI: 10.1038/cddis.2015.362] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 11/06/2015] [Accepted: 11/10/2015] [Indexed: 12/20/2022]
Abstract
Hair cell damage is a side effect of cisplatin and aminoglycoside use. The inhibition or attenuation of this process is a target of many investigations. There is growing evidence that STAT1 deficiency decreases cisplatin-mediated ototoxicity; however, the role of STAT function and the molecules that act in gentamicin-mediated toxicity have not been fully elucidated. We used mice lacking STAT1 to investigate the effect of STAT1 ablation in cultured organs treated with cisplatin and gentamicin. Here we show that ablation of STAT1 decreased cisplatin toxicity and attenuated gentamicin-mediated hair cell damage. More TUNEL-positive hair cells were observed in explants of wild-type mice than that of STAT1−/− mice. Although cisplatin increased serine phosphorylation of STAT1 in wild-type mice and diminished STAT3 expression in wild-type and STAT1−/− mice, gentamicin increased tyrosine phosphorylation of STAT3 in STAT1−/− mice. The early inflammatory response was manifested in the upregulation of TNF-α and IL-6 in cisplatin-treated explants of wild-type and STAT1−/− mice. Expression of the anti-inflammatory cytokine IL-10 was altered in cisplatin-treated explants, upregulated in wild-type explants, and downregulated in STAT1−/− explants. Cisplatin and gentamicin triggered the activation of c-Jun. Activation of Akt was observed in gentamicin-treated explants from STAT1−/− mice. Increased levels of the autophagy proteins Beclin-1 and LC3-II were observed in STAT1−/− explants. These data suggest that STAT1 is a central player in mediating ototoxicity. Gentamicin and cisplatin activate different downstream factors to trigger ototoxicity. Although cisplatin and gentamicin triggered inflammation and activated apoptotic factors, the absence of STAT1 allowed the cells to overcome the effects of these drugs.
Collapse
|
14
|
Callejo A, Sedó-Cabezón L, Juan ID, Llorens J. Cisplatin-Induced Ototoxicity: Effects, Mechanisms and Protection Strategies. TOXICS 2015; 3:268-293. [PMID: 29051464 PMCID: PMC5606684 DOI: 10.3390/toxics3030268] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Revised: 07/08/2015] [Accepted: 07/09/2015] [Indexed: 12/11/2022]
Abstract
Cisplatin is a highly effective chemotherapeutic agent that is widely used to treat solid organ malignancies. However, serious side effects have been associated with its use, such as bilateral, progressive, irreversible, dose-dependent neurosensory hearing loss. Current evidence indicates that cisplatin triggers the production of reactive oxygen species in target tissues in the inner ear. A variety of agents that protect against cisplatin-induced ototoxicity have been successfully tested in cell culture and animal models. However, many of them interfere with the therapeutic effect of cisplatin, and therefore are not suitable for systemic administration in clinical practice. Consequently, local administration strategies, namely intratympanic administration, have been developed to achieve otoprotection, without reducing the antitumoral effect of cisplatin. While a considerable amount of pre-clinical information is available, clinical data on treatments to prevent cisplatin ototoxicity are only just beginning to appear. This review summarizes clinical and experimental studies of cisplatin ototoxicity, and focuses on understanding its toxicity mechanisms, clinical repercussions and prevention strategies.
Collapse
Affiliation(s)
- Angela Callejo
- Unitat Funcional d'Otorrinolaringologia i Al·lèrgia, Institut Universtiari Quirón Dexeus, 08028 Barcelona, Catalonia, Spain.
| | - Lara Sedó-Cabezón
- Departament de Ciències Fisiològiques II, Universitat de Barcelona, 08907 L'Hospitalet de Llobregat, Catalonia, Spain.
| | - Ivan Domènech Juan
- Unitat Funcional d'Otorrinolaringologia i Al·lèrgia, Institut Universtiari Quirón Dexeus, 08028 Barcelona, Catalonia, Spain.
- Servei d'Otorrinolaringologia, Hospital Universitario de Bellvitge, 08907 L'Hospitalet de Llobregat, Catalonia, Spain.
| | - Jordi Llorens
- Departament de Ciències Fisiològiques II, Universitat de Barcelona, 08907 L'Hospitalet de Llobregat, Catalonia, Spain.
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), 08907 L'Hospitalet de Llobregat, Catalonia, Spain.
| |
Collapse
|
15
|
Gunewardene N, Guo CX, Wong ACY, Thorne PR, Vlajkovic SM. Adenosine amine congener ameliorates cisplatin-induced hearing loss. World J Otorhinolaryngol 2013; 3:100-107. [DOI: 10.5319/wjo.v3.i3.100] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 06/27/2013] [Accepted: 07/25/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate a novel pharmacological intervention to mitigate cisplatin ototoxicity using a selective adenosine A1 receptor agonist adenosine amine congener (ADAC).
METHODS: Male Wistar rats (8-10 wk) were exposed to a two-cycle cisplatin treatment similar to clinical course of cancer chemotherapy. Each cycle comprised 4 d of intraperitoneal cisplatin injections (1 mg/kg twice daily) separated by 10 d of rest. ADAC (100 μg/kg) or drug vehicle solution (control) was administered intraperitoneally for 5 d at 24 h intervals during the second cisplatin cycle (Regime 1), or upon completion of the cisplatin treatment (Regime 2). Hearing thresholds were measured using auditory brainstem responses (ABR) before cisplatin administration (baseline) and 7 d after the end of cisplatin treatment. Histological analysis of cochlear tissues included hair cell counting and qualitative assessment of apoptosis using terminal deoxynucleotidyl transferase mediated dUTP nick end labelling (TUNEL) staining.
RESULTS: ABR threshold shifts in cisplatin-treated Wistar rats ranged from 5-29 dB across the frequency range used in the study (4-24 kHz). Higher frequencies (16-24 kHz) were mostly affected by cisplatin ototoxicity (mean threshold shift 25-29 dB). ADAC treatment during the second cisplatin cycle reduced cisplatin-induced threshold shifts by 12-16 dB (P < 0.01) at higher frequencies compared to control vehicle-treated rats. However, the treatment was ineffective if ADAC administration was delayed until after the completion of the cisplatin regime. Functional recovery was supported by increased survival of hair cells in the cochlea. Qualitative analysis using TUNEL staining demonstrated reduced apoptosis of the outer hair cells and marginal cells in the stria vascularis in animals treated with ADAC during the second cisplatin cycle.
CONCLUSION: A1 adenosine receptor agonist ADAC mitigates cisplatin-induced cochlear injury and hearing loss, however its potential interference with antineoplastic effects of cisplatin needs to be established.
Collapse
|
16
|
Godwin P, Baird AM, Heavey S, Barr MP, O'Byrne KJ, Gately K. Targeting nuclear factor-kappa B to overcome resistance to chemotherapy. Front Oncol 2013; 3:120. [PMID: 23720710 PMCID: PMC3655421 DOI: 10.3389/fonc.2013.00120] [Citation(s) in RCA: 208] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Accepted: 04/28/2013] [Indexed: 12/29/2022] Open
Abstract
Intrinsic or acquired resistance to chemotherapeutic agents is a common phenomenon and a major challenge in the treatment of cancer patients. Chemoresistance is defined by a complex network of factors including multi-drug resistance proteins, reduced cellular uptake of the drug, enhanced DNA repair, intracellular drug inactivation, and evasion of apoptosis. Pre-clinical models have demonstrated that many chemotherapy drugs, such as platinum-based agents, antracyclines, and taxanes, promote the activation of the NF-κB pathway. NF-κB is a key transcription factor, playing a role in the development and progression of cancer and chemoresistance through the activation of a multitude of mediators including anti-apoptotic genes. Consequently, NF-κB has emerged as a promising anti-cancer target. Here, we describe the role of NF-κB in cancer and in the development of resistance, particularly cisplatin. Additionally, the potential benefits and disadvantages of targeting NF-κB signaling by pharmacological intervention will be addressed.
Collapse
Affiliation(s)
- P Godwin
- Department of Clinical Medicine, Thoracic Oncology Research Group, Trinity College Dublin, St. James's Hospital Ireland Dublin, Ireland
| | | | | | | | | | | |
Collapse
|
17
|
Waissbluth S, Daniel SJ. Cisplatin-induced ototoxicity: transporters playing a role in cisplatin toxicity. Hear Res 2013; 299:37-45. [PMID: 23467171 DOI: 10.1016/j.heares.2013.02.002] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 01/16/2013] [Accepted: 02/07/2013] [Indexed: 12/13/2022]
Abstract
Cisplatin is a potent antineoplastic agent widely used for a variety of cancer types. Unfortunately, its use leads to dose limiting side effects such as ototoxicity. Up to 93% of patients receiving cisplatin chemotherapy will develop progressive and irreversible sensorineural hearing loss which leads to a decreased quality of life in cancer survivors. No treatment is currently available for cisplatin-induced ototoxicity. It appears that cisplatin causes apoptosis by binding DNA, activating the inflammatory cascade as well as generating oxidative stress in the cell. Various studies have aimed to assess the potential protective effects of compounds such as antioxidants, anti-inflammatories, caspase inhibitors, anti-apoptotic agents and calcium channel blockers against the toxicity caused by cisplatin in the inner ear with variable degrees of protection. Nevertheless, the pathophysiology of cisplatin-induced ototoxicity remains unclear. This review summarizes all of the known transporters that could play a role in cisplatin influx, leading to cisplatin-induced ototoxicity. The following were evaluated: copper transporters, organic cation transporters, the transient receptor potential channel family, calcium channels, multidrug resistance associated proteins, mechanotransduction channels and chloride channels.
Collapse
Affiliation(s)
- Sofia Waissbluth
- Department of Otolaryngology, The Montreal Children's Hospital, Quebec, Canada
| | | |
Collapse
|
18
|
Park HY, Lee MH, Kang SU, Hwang HS, Park K, Choung YH, Kim CH. Nitric oxide mediates TNF-α-induced apoptosis in the auditory cell line. Laryngoscope 2012; 122:2256-64. [PMID: 22815072 DOI: 10.1002/lary.23444] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Revised: 03/30/2012] [Accepted: 04/26/2012] [Indexed: 12/15/2022]
Abstract
OBJECTIVES/HYPOTHESIS Tumor necrosis factor-alpha (TNF-α) is released in a variety of pathological states in the inner ear. Inducible nitric oxide synthase (iNOS) can be induced by cytokines and other inflammatory factors, and is generally thought to be associated with inflammation and other pathological processes in the cochlea. The purpose of the present study was to reveal that TNF-α could induce apoptosis in the auditory cell line and to investigate the role of nitric oxide (NO) in TNF-α-induced auditory cell death. STUDY DESIGN Experimental study. METHODS UB-OC1 cells and zebrafish were exposed to TNF-α. Flow cytometry, terminal deoxynucleotidyl transferase (TdT)-mediated dUTP-biotin nick end labeling (TUNEL) assay, assay of mitochondrial membrane potential (MMP), and electron microscopy were used to show that TNF-α could induce apoptosis. Western blot was used to measure iNOS expression and mitogen-activated protein kinase pathway. RESULTS Flow cytometric analysis, TUNEL assay, MMP, and electron microscopy all demonstrated that TNF-α could induce apoptosis in UB-OC1 cells. TNF-α significantly increased NO generation and iNOS expression. Pretreatment with iNOS blocker NG-methyl-L-arginine (NMA) attenuated TNF-α-induced cell death and caspase-3 activation. Also, TNF-α treatment increased p-p38 and p-ERK, and pretreatment of NMA reduced this increased expression of p-p38 and p-ERK. CONCLUSIONS TNF-α can induce apoptosis in the auditory cell line, and NO production in response to TNF-α is essential for apoptosis.
Collapse
Affiliation(s)
- Hun Yi Park
- Department of OtolaryngologyAjou University School of Medicine, Suwon, South Korea
| | | | | | | | | | | | | |
Collapse
|
19
|
Dinh CT, Bas E, Chan SS, Dinh JN, Vu L, Van De Water TR. Dexamethasone treatment of tumor necrosis factor-alpha challenged organ of Corti explants activates nuclear factor kappa B signaling that induces changes in gene expression that favor hair cell survival. Neuroscience 2011; 188:157-67. [PMID: 21571041 DOI: 10.1016/j.neuroscience.2011.04.061] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Revised: 04/27/2011] [Accepted: 04/29/2011] [Indexed: 12/20/2022]
Abstract
The objective was to determine the role of nuclear factor kappa B (NFκB) in dexamethasone base (DXMb) protection of auditory hair cells from tumor necrosis factor-alpha (TNFα)-induced loss on gene expression and cell signaling levels. Organ of Corti (OC) explants from 3-day-old rats were cultured under one of the following conditions: (1) media only--no treatment; (2) media+TNFα; (3) media+TNFα+DXMb; (4) media+TNFα+DXMb+NFκB-Inhibitor (NFκB-I); or (5) media+TNFα+DXMb+NFκBI-Scrambled control (NFκBI-C). A total of 60 organ of Corti explants (OC) were stained with FITC-Phalloidin after 96 h in culture (conditions 1-5) for hair cell counts and imaging of surface characteristics. A total of 108 OC were used for gene expression studies (i.e. B-actin, Bax, Bcl-2, Bcl-xl, and TNFR1) after 0, 24, or 48 h in vitro (conditions 1-4). A total of 86 OC were cultured (conditions 1-3) for 48 h, 36 of which were used for phosphorylated NFκB (p-NFκB) ELISA studies and 50 for whole mount anti-p-NFκB immunostain experiments. TNFα+DXMb exposed cultures demonstrated significant upregulation in anti-apoptotic Bcl-2 and Bcl-xl genes and downregulation in pro-apoptotic Bax gene expression; DXMb treatment of TNFα explants also lowered the Bax/Bcl-2 ratio and inhibited TNFR1 upregulation. After inhibiting NFκB activity with NFκB-I, the gene expression profile following TNFα+DXMb treatment now mimics that of TNFα-challenged OC explants. The levels of p-NFκB and the degree of nuclear translocation are significantly greater in TNFα+DXMb exposed OC explants than observed in the TNFα and control groups in the middle+basal turns of OC explants. These findings were supported by the results of the hair cell counts and the imaging results obtained from the whole mount OC specimens. DXMb protects against TNFα-induced apoptosis of auditory hair cells in vitro via activation of NFκB signaling in hair cell nuclei, and regulation of the expression levels of anti- and pro-apoptotic genes and a pro-inflammatory gene.
Collapse
Affiliation(s)
- C T Dinh
- Cochlear Implant Research Program, University of Miami Ear Institute, Department of Otolaryngology, University of Miami, Miller School of Medicine, 1600 NW 10th Avenue, RMSB 3160, Miami, FL 33136-1015, USA
| | | | | | | | | | | |
Collapse
|
20
|
Expression of the apoptosis-related proteins caspase-3 and NF-kappaB in the hippocampus of Tg2576 mice. Neurosci Bull 2010; 26:37-46. [PMID: 20101271 DOI: 10.1007/s12264-010-6122-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVE To investigate the relations between neuroapoptosis and the onset and development of Alzheimer's disease (AD), especially the role of NF-kappaB in the regulation of neuroapoptosis. METHODS Caspase-3 and NF-kappaB (p50) expressions in the CA3 region of the hippocampus in APPswe Tg2576 transgenic mice were studied from postnatal day 0-180, using Nissl staining, immunohistochemistry and RT-PCR methods. RESULTS Both neuronal apoptosis and NF-kappaB activity decreased gradually with the increase of age in wild type and Tg2576 mice. However, the number of caspase-3-positive or NF-kappaB-positive pyramidal cells in Tg2576 mice was greater than that in age-matched wild type mice, with significant differences after postnatal day 14 (P < 0.01 or P < 0.05). Linear regression analyses of caspase-3 and NF-kappaB expression demonstrated a correlation between neuroapoptosis and activity of NF-kappaB. CONCLUSION The process of neuroapoptosis is consistent with the onset and development of AD. Furthermore, the observed correlation between neuroapoptosis and NF-kappaB activity suggests a role of NF-kappaB in hippocampal neuroapoptosis.
Collapse
|
21
|
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: 255] [Impact Index Per Article: 17.0] [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
|
22
|
Haake SM, Dinh CT, Chen S, Eshraghi AA, Van De Water TR. Dexamethasone protects auditory hair cells against TNFα-initiated apoptosis via activation of PI3K/Akt and NFκB signaling. Hear Res 2009; 255:22-32. [DOI: 10.1016/j.heares.2009.05.003] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Revised: 05/01/2009] [Accepted: 05/04/2009] [Indexed: 12/13/2022]
|
23
|
Adams JC, Seed B, Lu N, Landry A, Xavier RJ. Selective activation of nuclear factor kappa B in the cochlea by sensory and inflammatory stress. Neuroscience 2009; 160:530-9. [PMID: 19285117 DOI: 10.1016/j.neuroscience.2009.02.073] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2009] [Revised: 02/22/2009] [Accepted: 02/24/2009] [Indexed: 02/07/2023]
Abstract
Damage response pathways triggered by mechanical stress might reasonably be expected to be conserved throughout evolution. However, using a nuclear factor kappa B (NF-kappaB) reporter mouse we show here that this phylogenetically recent transcription factor plays a major role in the response to mechanosensory stress in the mammalian inner ear. The protective action of NF-kappaB is exerted in neither sensory nor non-sensory epithelial cells, but rather in connective tissue cells within the spiral ligament and spiral limbus. In the spiral ligament, predominantly type I fibrocytes are activated following noise exposure, whereas type II fibrocytes are activated following systemic inflammatory stress. Immune-mediated and acoustic trauma-mediated hearing loss syndromes in humans may in part result from the vulnerability of type II and type I fibrocytes to systemic inflammatory stress and acoustic trauma, respectively. Unexpected cell-specific and stress-specific NF-kappaB activation found in the inner ear in this in vivo study suggest that this approach may have wide applications in demonstrating similar specializations of stress responses in other tissues, including the brain.
Collapse
Affiliation(s)
- J C Adams
- Department of Otology and Laryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, 243 Charles Street, Boston, MA 02114, USA.
| | | | | | | | | |
Collapse
|
24
|
Dinh C, Haake S, Chen S, Hoang K, Nong E, Eshraghi A, Balkany T, Van De Water T. Dexamethasone protects organ of corti explants against tumor necrosis factor-alpha–induced loss of auditory hair cells and alters the expression levels of apoptosis-related genes. Neuroscience 2008; 157:405-13. [DOI: 10.1016/j.neuroscience.2008.09.012] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Revised: 07/17/2008] [Accepted: 09/06/2008] [Indexed: 12/19/2022]
|