1
|
Cho YJ, Han BS, Ko S, Park MS, Lee YJ, Kim SE, Lee P, Go HG, Park S, Lee H, Kim S, Park ER, Jung KH, Hong SS. Repositioning of aripiprazole, an anti‑psychotic drug, to sensitize the chemotherapy of pancreatic cancer. Int J Mol Med 2025; 55:17. [PMID: 39540370 PMCID: PMC11573310 DOI: 10.3892/ijmm.2024.5458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy with limited therapeutic options. Cisplatin is a primary chemotherapeutic agent utilized in combination with other drugs or radiotherapy for PDAC treatment. However, the severe side effects of cisplatin often necessitate discontinuation of therapy and drug resistance in tumor cells poses significant clinical challenges. Therefore, the development of effective therapeutic strategies is imperative. The present study investigated whether repositioning of the antipsychotic drug aripiprazole could sensitize the anticancer activity of cisplatin in pancreatic cancer at doses calculated by the combination index. The findings indicated that aripiprazole combined with cisplatin to suppress pancreatic cancer cell growth. Notably, the combination notably increased the expression of apoptosis markers, including cleaved caspase‑3, compared with cisplatin alone. Additionally, this combination effectively decreased XIAP and MCL‑1 expression via mitochondrial membrane potential change as revealed by JC‑1 assay, thereby inducing apoptosis. Furthermore, in fluid shear stress assay, the combination of aripiprazole and cisplatin notably inhibited cell adhesion and tumor spheroid formation. Mechanistically, phospho‑kinase array profiles showed that the enhanced anticancer efficacy of the combination treatment could be attributed to the inhibition of STAT3 signaling, which led to a significant reduction in tumor growth in a pancreatic cancer animal model. The results showed that the repositioning of aripiprazole inhibits cancer cell growth by blocking the STAT3 signaling pathway and effectively enhancing cisplatin‑induced apoptosis, thereby suggesting that the combination of aripiprazole and cisplatin may be a potent chemotherapeutic strategy for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Ye Jin Cho
- Department of Biomedical Sciences, College of Medicine and Program in Biomedical Science and Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Beom Seok Han
- Department of Biomedical Sciences, College of Medicine and Program in Biomedical Science and Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Soyeon Ko
- Department of Biomedical Sciences, College of Medicine and Program in Biomedical Science and Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Min Seok Park
- Department of Biomedical Sciences, College of Medicine and Program in Biomedical Science and Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Yun Ji Lee
- Department of Biomedical Sciences, College of Medicine and Program in Biomedical Science and Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Sang Eun Kim
- Department of Biomedical Sciences, College of Medicine and Program in Biomedical Science and Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Pureunchowon Lee
- Department of Biomedical Sciences, College of Medicine and Program in Biomedical Science and Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Han Gyeol Go
- Department of Biomedical Sciences, College of Medicine and Program in Biomedical Science and Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Shinyoung Park
- Anti-cancer Strategy Research Institute, VSPharmTech, Inc., Seoul Technopark, Seoul 01811, Republic of Korea
| | - Hyunho Lee
- Anti-cancer Strategy Research Institute, VSPharmTech, Inc., Seoul Technopark, Seoul 01811, Republic of Korea
| | - Sohee Kim
- Anti-cancer Strategy Research Institute, VSPharmTech, Inc., Seoul Technopark, Seoul 01811, Republic of Korea
| | - Eun-Ran Park
- Anti-cancer Strategy Research Institute, VSPharmTech, Inc., Seoul Technopark, Seoul 01811, Republic of Korea
| | - Kyung Hee Jung
- Department of Biomedical Sciences, College of Medicine and Program in Biomedical Science and Engineering, Inha University, Incheon 22332, Republic of Korea
| | - Soon-Sun Hong
- Department of Biomedical Sciences, College of Medicine and Program in Biomedical Science and Engineering, Inha University, Incheon 22332, Republic of Korea
| |
Collapse
|
2
|
Elmorsy EA, Saber S, Hamad RS, Abdel-Reheim MA, El-Kott AF, AlShehri MA, Morsy K, Salama SA, Youssef ME. Advances in understanding cisplatin-induced toxicity: Molecular mechanisms and protective strategies. Eur J Pharm Sci 2024; 203:106939. [PMID: 39423903 DOI: 10.1016/j.ejps.2024.106939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/30/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
Cisplatin, a widely used chemotherapeutic agent, has proven efficacy against various malignancies. However, its clinical utility is hampered by its dose-limiting toxicities, including nephrotoxicity, ototoxicity, neurotoxicity, and myelosuppression. This review aims to provide a comprehensive overview of cisplatin toxicity, encompassing its underlying mechanisms, risk factors, and emerging therapeutic strategies. The mechanisms of cisplatin toxicity are multifactorial and involve oxidative stress, inflammation, DNA damage, and cellular apoptosis. Various risk factors contribute to the interindividual variability in susceptibility to cisplatin toxicity. The risk of developing cisplatin-induced toxicity could be related to pre-existing conditions, including kidney disease, hearing impairment, neuropathy, impaired liver function, and other comorbidities. Additionally, this review highlights the emerging therapeutic strategies that could be applied to minimize cisplatin-induced toxicities and aid in optimizing cisplatin treatment regimens, improving patient outcomes, and enhancing the overall quality of cancer care.
Collapse
Affiliation(s)
- Elsayed A Elmorsy
- Department of Pharmacology and Therapeutics, College of Medicine, Qassim University, Buraidah, 51452, Saudi Arabia.
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt.
| | - Rabab S Hamad
- Biological Sciences Department, College of Science, King Faisal University, Al Ahsa 31982, Saudi Arabia; Central Laboratory, Theodor Bilharz Research Institute, Giza 12411, Egypt.
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef 62521, Egypt.
| | - Attalla F El-Kott
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia; Department of Zoology, Faculty of Science, Damanhour University, Egypt
| | - Mohammed A AlShehri
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Kareem Morsy
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia; Department of Zoology, Faculty of Science, Cairo University, Cairo, Egypt
| | - Salama A Salama
- Department of Zoology, Faculty of Science, Damanhour University, Egypt; Department of Biology, College of Science, Jazan University, Jazan 45142, Saudi Arabia
| | - Mahmoud E Youssef
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt
| |
Collapse
|
3
|
Heinrich J, Siddiqui E, Eckstein H, Naumann M, Kulak N. Ascorbate: a forgotten component in the cytotoxicity of Cu(II) ATCUN peptide complexes. J Biol Inorg Chem 2024; 29:801-809. [PMID: 39527272 PMCID: PMC11638278 DOI: 10.1007/s00775-024-02083-9] [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: 05/24/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024]
Abstract
In 1983, Linus Pauling and colleagues reported about enhanced antitumor activity of the Cu(II) complex of the simplest ATCUN (amino terminal Cu(II) and Ni(II)-binding motif) peptide (NH2-Gly-Gly-His-COOH, GGH) in the presence of ascorbate as an additive. In the following 4 decades, structural modifications of this complex were implemented, however, anticancer activity could not be significantly increased. This has led to neglecting the ATCUN motif and its Cu(II) complexes as potential chemotherapeutic agents. Furthermore, the addition of ascorbate with its positive effect on the anticancer activity has fallen into oblivion. In this work, we compared Cu(II) GGH with Cu(II) ATCUN peptides bearing β-Ala instead of Gly at the 2nd position of the peptide sequence regarding their in vitro complex stability and cytotoxicity (MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) and annexin V-FITC (fluorescein isothiocyanate) apoptosis assay) towards three cancer cell lines (AGS, HeLa and NCI-N87). Such an exchange of amino acids led to an up to three-fold higher cytotoxic effect in the presence of ascorbate. We thus achieved a significant increase in the otherwise moderate cytotoxicity of Cu(II) ATCUN-like complexes. Lipophilicity assays (n-octanol/water coefficient, log P values) of the studied complexes were used to evaluate differences in the antiproliferative activity.
Collapse
Affiliation(s)
- Julian Heinrich
- Institute of Chemistry, University of Potsdam, Karl-Liebknecht-Straße 24-25, 14476, Potsdam, Germany
- Institute of Chemistry, Otto von Guericke University, Universitätsplatz 2, 39106, Magdeburg, Germany
| | - Elisa Siddiqui
- Institute of Experimental Internal Medicine, Otto von Guericke University, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Henrike Eckstein
- Institute of Experimental Internal Medicine, Otto von Guericke University, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Otto von Guericke University, Leipziger Straße 44, 39120, Magdeburg, Germany.
| | - Nora Kulak
- Institute of Chemistry, University of Potsdam, Karl-Liebknecht-Straße 24-25, 14476, Potsdam, Germany.
- Institute of Chemistry, Otto von Guericke University, Universitätsplatz 2, 39106, Magdeburg, Germany.
| |
Collapse
|
4
|
Lu C, Chen C, Xu Y, Dai D, Sun C, Li Q. Activation of Wnt/β-catenin signaling to increase B lymphoma Moloney murine leukemia virus insertion region 1 by lithium chloride attenuates the toxicity of cisplatin in the HEI-OC1 auditory cells. Toxicol Lett 2024; 403:50-65. [PMID: 39608515 DOI: 10.1016/j.toxlet.2024.11.009] [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: 02/24/2024] [Revised: 10/31/2024] [Accepted: 11/26/2024] [Indexed: 11/30/2024]
Abstract
Cisplatin is widely used in anti-tumor therapy, but the ototoxicity caused by high-dose cisplatin often limits its efficacy, and the specific mechanism of cisplatin-induced cochlear damage is still not perfect. The Wnt/β-catenin signaling pathway is closely related to aging, embryonic development, and apoptosis. Meanwhile, B lymphoma Moloney murine leukemia virus insertion region 1 (BMI1) plays a certain role in the evolution and development of the inner ear and the occurrence and development of inner ear-related diseases. Our study intends to explore the role and specific mechanism of the Wnt/β-catenin signaling pathway and BMI1 in improving cisplatin ototoxicity. The appropriate experimental concentrations for each drug were selected by CCK-8 cell proliferation assay and Western Blot to detect apoptosis. The lentivirus transfection of HEI-OC1 cochlear hair cells was used to overexpress BMI1. Western Blot, qPCR, and immunofluorescence detected the activation of each component of BMI1 and Wnt/β-catenin signaling pathway in each experimental model. Wnt/β-catenin signaling pathway and BMI1 are jointly involved in cisplatin-induced cell injury. Low lithium chloride (LiCl) concentrations activated the Wnt/β-catenin pathway, increased BMI1 expression, and reduced cisplatin-induced hair cell injury. In contrast, overexpression of BMI1 inhibited the Wnt/β-catenin pathway and reduced hair cell injury. Meanwhile, the increased cisplatin-induced damage to hair cells by inhibiting BMI1 could not be rescued by LiCl. In conclusion, LiCl can ameliorate cisplatin ototoxicity by elevating BMI1 expression through activation of the Wnt/β-catenin pathway. Overexpression of BMI1 inhibits the Wnt/β-catenin pathway and reduces cisplatin-induced hair cell damage.
Collapse
Affiliation(s)
- Chen Lu
- Department of ENT, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Chao Chen
- Department of ENT, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Yingpeng Xu
- Department of ENT, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Dingyuan Dai
- Department of ENT, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Chen Sun
- Department of ENT, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China.
| | - Qi Li
- Department of ENT, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China; Medical School of Nanjing University, Nanjing, Jiangsu, PR China.
| |
Collapse
|
5
|
Akhlaghipour I, Moghbeli M. Matrix metalloproteinases as the critical regulators of cisplatin response and tumor cell invasion. Eur J Pharmacol 2024; 982:176966. [PMID: 39216742 DOI: 10.1016/j.ejphar.2024.176966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/10/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Cisplatin (CDDP) as one of the most common first-line chemotherapy drugs plays a vital role in the treatment of a wide range of malignant tumors. Nevertheless, CDDP resistance is observed as a therapeutic challenge in a large number of cancer patients. Considering the CDDP side effects in normal tissues, predicting the CDDP response of cancer patients can significantly help to choose the appropriate therapeutic strategy. In this regard, investigating the molecular mechanisms involved in CDDP resistance can lead to the introduction of prognostic markers in cancer patients. Matrix metalloproteinases (MMPs) have critical roles in tissue remodeling and cell migration through extracellular matrix degradation. Therefore, defects in MMPs functions can be associated with tumor metastasis and chemo resistance. In the present review, we discussed the role of MMPs in CDDP response and tumor cell invasion. PubMed, Scopus, Google Scholar, and Web of Science were searched using "MMP", "cisplatin", and "cancer" keywords for data retrieval that was limited to Apr 20, 2024. It has been reported that MMPs can increase CDDP resistance in tumor cells as the effectors of PI3K/AKT, MAPK, and NF-κB signaling pathways or independently through the regulation of structural proteins, autophagy, and epithelial-to-mesenchymal transition (EMT) process. This review has an effective role in introducing MMPs as the prognostic markers and therapeutic targets in CDDP-resistant cancer patients.
Collapse
Affiliation(s)
- Iman Akhlaghipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Abdelrasol H, Chopra A, Shvachiy L, Beutner D, Outeiro TF, Setz C. Stress granules formation in HEI-OC1 auditory cells and in H4 human neuroglioma cells secondary to cisplatin exposure. Cell Stress 2024; 8:83-98. [PMID: 39575153 PMCID: PMC11580520 DOI: 10.15698/cst2024.10.299] [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: 02/17/2024] [Revised: 08/27/2024] [Accepted: 09/04/2024] [Indexed: 11/24/2024] Open
Abstract
Stress granules (SGs) are highly dynamic micromolecular membraneless condensates that generate in cells subjected to stress. Formed from pools of untranslating messenger ribonucleoproteins (RNP), SGs dynamics constitute vital processes essential for cell survival. Here, we investigate whether established cytotoxic agents, such as the platinum-based chemotherapeutic agent cisplatin and the aminoglycoside antibiotic gentamicin, elicit SG formation in the House Ear Institute-Organ of Corti-1 (HEI-OC1) auditory cell line, H4 human neuroglioma cells and HEK-293T human embryonic kidney cells. Cells were treated with cisplatin or gentamicin for specific durations at designated concentrations. SG formation was assessed using immunocytochemistry and live cell imaging. Levels of essential proteins involved in SG assembly were evaluated using immunoblotting. We observed cisplatin-associated SG assembly in HEI-OC1 and H4 cells via confocal microscopy through antibody colabeling of G3BP1 with PABP or Caprin1. While maintaining an unchanged pattern of expression of main constituent SG proteins, cisplatin-related SGs in H4 cells persisted for at least 12 h after drug removal. Cells subjected to gentamicin exposure did not exhibit SGs. Our findings offer insights into subcellular mechanisms related to cisplatin-associated cytotoxicity, highlighting the need for future studies to further investigate this stress-response mechanism.
Collapse
Affiliation(s)
- Hebatallah Abdelrasol
- University Medical Center Göttingen, Department of Experimental Neurodegeneration, Center for Biostructural Imaging of NeurodegenerationGöttingenGermany
| | - Avika Chopra
- University Medical Center Göttingen, Department of Experimental Neurodegeneration, Center for Biostructural Imaging of NeurodegenerationGöttingenGermany
| | - Liana Shvachiy
- University Medical Center Göttingen, Department of Experimental Neurodegeneration, Center for Biostructural Imaging of NeurodegenerationGöttingenGermany
| | - Dirk Beutner
- University Medical Center Göttingen, Department of Otolaryngology-Head and Neck Surgery, InnerEarLabGöttingenGermany
| | - Tiago F Outeiro
- University Medical Center Göttingen, Department of Experimental Neurodegeneration, Center for Biostructural Imaging of NeurodegenerationGöttingenGermany
- Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle UniversityNewcastle upon TyneUnited Kingdom
- Scientific employee with an honorary contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) - German Center for Neurodegenerative Diseases, Göttingen, Germany.
| | - Cristian Setz
- University Medical Center Göttingen, Department of Experimental Neurodegeneration, Center for Biostructural Imaging of NeurodegenerationGöttingenGermany
- University Medical Center Göttingen, Department of Otolaryngology-Head and Neck Surgery, InnerEarLabGöttingenGermany
| |
Collapse
|
7
|
Fernandez K, Hoetink A, Konrad-Martin D, Berndtson D, Clark K, Dreisbach L, Geller JI, Goffi-Gomez MV, Grosnik A, Jamis C, Knight K, Lee DS, Lee J, Liberman PHP, Milnes T, Meijer AJM, Ortiz CE, Rooker J, Sanchez VA, van den Heuvel-Eibrink MM, Brewer CC, Poling GL. Roadmap to a Global Template for Implementation of Ototoxicity Management for Cancer Treatment. Ear Hear 2024:00003446-990000000-00345. [PMID: 39261989 DOI: 10.1097/aud.0000000000001592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Ototoxicity is among the adverse events related to cancer treatment that can have far-reaching consequences and negative impacts on quality-of-life for cancer patients and survivors of all ages. Ototoxicity management (OtoM) comprises the prevention, diagnosis, monitoring, and treatment, including rehabilitation and therapeutic intervention, of individuals who experience hearing loss, tinnitus, or balance/vestibular difficulties following exposures to ototoxic agents, including platinum chemotherapy (cisplatin, carboplatin) and cranial radiation. Despite the well-established physical, socioeconomic, and psychological consequences of hearing and balance dysfunction, there are no widely adopted standards for clinical management of cancer treatment-related ototoxicity. Consensus recommendations and a roadmap are needed to guide development of effective and feasible OtoM programs, direct research efforts, address the needs of caregivers and patients at all stages of cancer care and survivorship. Here we review current evidence and propose near-term to longer-term goals to advance OtoM in five strategic areas: (1) beneficiary awareness, empowerment, and engagement, (2) workforce enhancement, (3) program development, (4) policy, funding, and sustainability, and (5) research and evaluation. The goal is to identify needs and establish a roadmap to guide worldwide adoption of standardized OtoM for cancer treatment and improved outcomes for patients and survivors.
Collapse
Affiliation(s)
- Katharine Fernandez
- Division of Intramural Research, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA
| | - Alex Hoetink
- Department of Otorhinolaryngology and Head & Neck Surgery, University Medical Center Utrecht, Utrecht, the Netherlands
- Audiology, Utrecht Medical Center Utrecht Brain Center, Utrecht, the Netherlands
- Audiology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Audiology, Wilhelmina Children's Hospital, Utrecht, the Netherlands
| | - Dawn Konrad-Martin
- National Center for Rehabilitative Auditory Research, Veterans Affairs Rehabilitation Research and Development, Veterans Affairs Portland Health Care System, Portland, Oregon, USA
- Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, Portland, Oregon, USA
| | - Deborah Berndtson
- International Ototoxicity Management Group, Cancer Survivor and Advocate, Vienna, Virginia, USA
| | - Khaya Clark
- National Center for Rehabilitative Auditory Research, Veterans Affairs Rehabilitation Research and Development, Veterans Affairs Portland Health Care System, Portland, Oregon, USA
- Department of Otolaryngology/Head & Neck Surgery, Oregon Health & Science University, Portland, Oregon, USA
- Veterans Affairs Health Services Research & Development Center of Innovation, Center to Improve Veteran Involvement in Care, VA Portland Health Care System (R&D 66), Portland, Oregon, USA
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon, USA
| | - Laura Dreisbach
- School of Speech, Language, and Hearing Sciences, San Diego State University, San Diego, California, USA
| | - James I Geller
- Division of Audiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | | | - Amy Grosnik
- Division of Audiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Carmen Jamis
- Head and Neck Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Kristin Knight
- Department of Audiology, Doernbecher Children's Hospital, Oregon Health & Science University, Portland, Oregon, USA
| | - David S Lee
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - John Lee
- Division of Intramural Research, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Trisha Milnes
- Department of Audiology, Veterans Affairs Augusta Health Care System, Augusta, Georgia, USA
| | - Annelot J M Meijer
- Audiology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Audiology, Wilhelmina Children's Hospital, Utrecht, the Netherlands
| | - Candice E Ortiz
- Capital Institute of Hearing & Balance, Silver Spring, Maryland, USA
| | - Jennessa Rooker
- College of Nursing, University of South Florida, Tampa, Florida, USA
| | - Victoria A Sanchez
- Department of Otolaryngology-Head & Neck Surgery, University of South Florida, Tampa, Florida, USA
| | - Mary M van den Heuvel-Eibrink
- Audiology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Audiology, Wilhelmina Children's Hospital, Utrecht, the Netherlands
| | - Carmen C Brewer
- Division of Intramural Research, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA
| | - Gayla L Poling
- Department of Otolaryngology-Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
8
|
Santos JAV, Silva D, Marques MPM, Batista de Carvalho LAE. Platinum-based chemotherapy: trends in organic nanodelivery systems. NANOSCALE 2024; 16:14640-14686. [PMID: 39037425 DOI: 10.1039/d4nr01483a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Despite the investment in platinum drugs research, cisplatin, carboplatin and oxaliplatin are still the only Pt-based compounds used as first line treatments for several cancers, with a few other compounds being approved for administration in some Asian countries. However, due to the severe and worldwide impact of oncological diseases, there is an urge for improved chemotherapeutic approaches. Furthermore, the pharmaceutical application of platinum complexes is hindered by their inherent toxicity and acquired resistance. Nanodelivery systems rose as a key strategy to overcome these challenges, with recognized versatility and ability towards improving the safety, bioavailability and efficacy of the available drugs. Among the known nanocarriers, organic systems have been widely applied, taking advantage of their potential as drug vehicles. Researchers have mainly focused on the development of lipidic and polymeric carriers, including supramolecular structures, with an overall improvement of encapsulated platinum complexes. Herein, an overview of recent trends and strategies is presented, with the main focus on the encapsulation of platinum compounds into organic nanocarriers, showcasing the evolution in the design and development of these promising systems. This comprehensive review highlights formulation methods as well as characterization procedures, providing insights that may be helpful for the development of novel platinum nanocarriers aiming at future pharmaceutical applications.
Collapse
Affiliation(s)
- João A V Santos
- Molecular Physical-Chemistry R&D Unit, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal.
| | - Daniela Silva
- Molecular Physical-Chemistry R&D Unit, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal.
| | - Maria Paula M Marques
- Molecular Physical-Chemistry R&D Unit, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal.
- Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal
| | - Luís A E Batista de Carvalho
- Molecular Physical-Chemistry R&D Unit, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal.
| |
Collapse
|
9
|
Araujo AGFS, Silva JC, Trindade Filho EM, Silva JC, Gama KDA, Bandeira MAM, Silva WA, Mousinho KC. Cisplatin and ototoxicity in childhood: the perspective of supporting otoprotective agentes. BRAZ J BIOL 2024; 84:e279117. [PMID: 39140499 DOI: 10.1590/1519-6984.279117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 06/05/2024] [Indexed: 08/15/2024] Open
Abstract
Cisplatin is an antineoplastic medicine used in the treatment for various types of cancer. Among its side effects is ototoxicity, which may result in a bilateral and irreversible hearing loss. The ototoxic effect in the pediatric population has a bigger impact as it compromises language acquisition. The discovery of drugs with otoprotective effects and the optimal way to administer them have become significant challenges in minimizing the impact of cisplatin regarding auditory function. The objective was to understand otoprotective drugs and their relevance in the preventive treatment to cisplatin-induced ototoxicity in childhood. An integrative review was conducted by consulting databases including PubMed, Bireme, MedLine, LILACS, SciELO, and ClinicalTrials.gov. The search strategy was performed by crossing descriptors (DeCS and MeSH) and free terms. Studies published in English, Spanish, and Portuguese were selected, with no publication year restrictions. Subsequently, articles were selected according to inclusion and exclusion criteria. A total of 736 articles were found in PubMed, 431 in Bireme, 425 in MedLine, 6 in LILACS, 0 in SciELO, and 4 in ClinicalTrials.gov. After document analysis, 12 articles were selected for full analysis. Evidence was found for 8 substances with potential otoprotective effects when used with cisplatin, which tend to minimize the impact of cisplatin regarding auditory function. The substances found were: Amifostine, Dexamethasone, Genistein, Ginkgo Biloba, Lycopene, N-acetylcysteine, Polydatin also Sodium Thiosulfate. In general, these drugs are applied before, during, or after cisplatin infusion, depending on the chosen drug, via intravenous, oral, or transtympanic injections, acting as antioxidant therapy. The biochemical effects of these substances are relevant to their potential otoprotective properties, including the inactivation of oxygen free radicals and electrophilic platinum species. The use of these substances can reduce ototoxicity, decreasing cisplatin-induced hearing loss and improving the confort of life, especially for children.
Collapse
Affiliation(s)
- A G F S Araujo
- Universidade Estadual de Ciências da Saúde de Alagoas - UNCISAL, Maceió, AL, Brasil
| | - J C Silva
- Universidade Estadual de Ciências da Saúde de Alagoas - UNCISAL, Maceió, AL, Brasil
| | - E M Trindade Filho
- Universidade Estadual de Ciências da Saúde de Alagoas - UNCISAL, Maceió, AL, Brasil
| | - J C Silva
- Universidade Estadual de Ciências da Saúde de Alagoas - UNCISAL, Maceió, AL, Brasil
| | - K D A Gama
- Centro Universitário CESMAC, Maceió, AL, Brasil
| | - M A M Bandeira
- Universidade Federal do Ceará - UFC, Fortaleza, CE, Brasil
| | - W A Silva
- Universidade Federal do Ceará - UFC, Fortaleza, CE, Brasil
| | - K C Mousinho
- Universidade Estadual de Ciências da Saúde de Alagoas - UNCISAL, Maceió, AL, Brasil
- Centro Universitário CESMAC, Maceió, AL, Brasil
| |
Collapse
|
10
|
Niapour A, Abdollahzadeh M, Ghaheri Fard S, Saadati H. The therapeutic potential of 1, 25-dihydroxy vitamin D3 on cisplatin-affected neurological functions is associated with the regulation of oxidative stress and inflammatory markers as well as levels of MMP2/9. Metab Brain Dis 2024; 39:1189-1200. [PMID: 39017968 DOI: 10.1007/s11011-024-01382-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 07/05/2024] [Indexed: 07/18/2024]
Abstract
Calcitriol as a biologically active form of vitamin D3 has beneficial effects on all body systems. This vitamin has a potent neuroprotective effect via several independent mechanisms against brain insults induced by anticancer drugs. The present study was designed to examine the neuroprotective effects of calcitriol against neurotoxicity induced by cisplatin. Induction of neurotoxicity was done with cisplatin administration (5 mg/kg/week) for 5 successive weeks in male Wistar rats. The neuroprotective influence of calcitriol supplementation (100ng/kg/day for 5 weeks) was assessed through behavioral, electrophysiological, and molecular experiments. Cisplatin administration impaired spatial learning and memory and decreased prefrontal brain-derived neurotrophic factor (BDNF). Peripheral sensory neuropathy was induced through cisplatin administration. Cisplatin also reduced the amplitudes of the compound action potential of sensory nerves in electrophysiological studies. Cisplatin treatment elevated MDA levels and reduced anti-oxidant (SOD and GPx) enzymes. Pro-inflammatory cytokines (IL-1β and TNF-α) and metalloproteinase-2 and 9 (MMP-2/9) were augmented through treatment with cisplatin. Learning and memory impairments along with BDNF changes caused by cisplatin were amended with calcitriol supplementation. Reduced sensory nerve conduction velocity in the cisplatin-treated group was improved by calcitriol. Calcitriol partially improved redox imbalance and diminished the pro-inflammatory cytokines and MMP-2/9 levels. Our findings showed that calcitriol supplementation can relieve cisplatin-induced peripheral neurotoxicity. Calcitriol can be regarded as a promising new neuroprotective agent.
Collapse
Affiliation(s)
- Ali Niapour
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Maryam Abdollahzadeh
- Department of Physiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Safa Ghaheri Fard
- Department of Physiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hakimeh Saadati
- Department of Physiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
11
|
Sanchez VA, Dinh PC, Monahan PO, Althouse S, Rooker J, Sesso HD, Dolan ME, Weinzerl M, Feldman DR, Fung C, Einhorn LH, Frisina RD, Travis LB. Comprehensive Audiologic Analyses After Cisplatin-Based Chemotherapy. JAMA Oncol 2024; 10:912-922. [PMID: 38842797 PMCID: PMC11157440 DOI: 10.1001/jamaoncol.2024.1233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 12/22/2023] [Indexed: 06/07/2024]
Abstract
Importance Cisplatin is highly ototoxic but widely used. Evidence is lacking regarding cisplatin-related hearing loss (CRHL) in adult-onset cancer survivors with comprehensive audiologic assessments (eg, Words-in-Noise [WIN] tests, full-spectrum audiometry, and additional otologic measures), as well as the progression of CRHL considering comorbidities, modifiable factors associated with risk, and cumulative cisplatin dose. Objective To assess CRHL with comprehensive audiologic assessments, including the WIN, evaluate the longitudinal progression of CRHL, and identify factors associated with risk. Design, Setting, and Participants The Platinum Study is a longitudinal study of cisplatin-treated testicular cancer survivors (TCS) enrolled from 2012 to 2018 with follow-up ongoing. Longitudinal comprehensive audiologic assessments at Indiana University and Memorial Sloan Kettering Cancer Center included 100 participants without audiometrically defined profound hearing loss (HL) at baseline and at least 3.5 years from their first audiologic assessment. Data were analyzed from December 2013 to December 2022. Exposures Factors associated with risk included cumulative cisplatin dose, hypertension, hypercholesterolemia, diabetes, tobacco use, physical inactivity, body mass index, family history of HL, cognitive dysfunction, psychosocial symptoms, and tinnitus. Main Outcomes and Measures Main outcomes were audiometrically measured HL defined as combined-ears high-frequency pure-tone average (4-12 kHz) and speech-recognition in noise performance measured with WIN. Multivariable analyses evaluated factors associated with risk for WIN scores and progression of audiometrically defined HL. Results Median (range) age of 100 participants at evaluation was 48 (25-67) years; median (range) time since chemotherapy: 14 (4-31) years. At follow-up, 78 (78%) TCS had audiometrically defined HL; those self-reporting HL had 2-fold worse hearing than TCS without self-reported HL (48 vs 24 dB HL; P < .001). A total of 54 (54%) patients with self-reported HL showed clinically significant functional impairment on WIN testing. Poorer WIN performance was associated with hypercholesterolemia (β = 0.88; 95% CI, 0.08 to 1.69; P = .03), lower-education (F1 = 5.95; P = .004), and severity of audiometrically defined HL (β̂ = 0.07; 95% CI, 0.06 to 0.09; P < .001). CRHL progression was associated with hypercholesterolemia (β̂ = -4.38; 95% CI, -7.42 to -1.34; P = .01) and increasing age (β̂ = 0.33; 95% CI, 0.15 to 0.50; P < .001). Importantly, relative to age-matched male normative data, audiometrically defined CRHL progression significantly interacted with cumulative cisplatin dose (F1 = 5.98; P = .02); patients given 300 mg/m2 or less experienced significantly less progression, whereas greater temporal progression followed doses greater than 300 mg/m2. Conclusions and Relevance Follow-up of cisplatin-treated cancer survivors should include strict hypercholesterolemia control and regular audiological assessments. Risk stratification through validated instruments should include querying hearing concerns. CRHL progression relative to age-matched norms is likely associated with cumulative cisplatin dose; investigation over longer follow-up is warranted.
Collapse
Affiliation(s)
- Victoria A. Sanchez
- Department of Otolaryngology–Head and Neck Surgery, University of South Florida, Tampa
| | - Paul C. Dinh
- Department of Medical Oncology, Indiana University, Indianapolis
| | - Patrick O. Monahan
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis
| | - Sandra Althouse
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis
| | | | - Howard D. Sesso
- Division of Preventive Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - M. Eileen Dolan
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Mandy Weinzerl
- Rehabilitation Services, Indiana University Health, Indianapolis
| | - Darren R. Feldman
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Chunkit Fung
- Department of Medical Oncology, J.P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York
| | | | - Robert D. Frisina
- Department of Medical Engineering, University of South Florida, Tampa
| | - Lois B. Travis
- Department of Medical Oncology, Indiana University, Indianapolis
| |
Collapse
|
12
|
Cicek B, Danisman B, Bolat I, Kiliclioglu M, Kuzucu M, Suleyman H, Tsarouhas K, Tsatsakis A, Taghizadehghalehjoughi A. Effect of tangeretin on cisplatin-induced oxido-inflammatory brain damage in rats. J Cell Mol Med 2024; 28:e18565. [PMID: 39044287 PMCID: PMC11265995 DOI: 10.1111/jcmm.18565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 06/04/2024] [Accepted: 07/09/2024] [Indexed: 07/25/2024] Open
Abstract
Cisplatin (CIS) is a platinum-derived chemotherapeutic agent commonly utilized in the treatment of various malignant tumours. However, anticancer doses of the drug cause serious damage to the brain. This study aimed to determine the potential protective effects of tangeretin, which has antioxidant and anti-inflammatory properties, in cisplatin-induced neurotoxicity on BALB/c mice brains. Male BALB/c mice were randomized and separated into four groups. Tangeretin was given for 10 days by gavage. CIS was injected as a single dose of 10 mg/kg intraperitoneally (ip) on the 10th day. Brain tissues, malondialdehyde (MDA), total glutathione (tGSH), glutathione peroxidase (GPx), superoxide dismutase (SOD), catalase (CAT) and nitric oxide (NO) levels were measured to determine oxidative damage and myeloperoxidase, tumour necrosis factor-alpha (TNF-α), interleukin 1 beta (IL-1β), IL-6 and IL-10 were measured to determine inflammatory activity. In addition, 8-OHdG and caspase-3 were analysed by immunofluorescence methods. While CIS administration remarkably elevated reactive oxygen species, MDA, and NO levels in brain tissue compared to the control, tGSH, GPx, SOD and CAT levels were significantly decreased. Also, it has been detected that TNF-α, IL-1β and IL-6 obtained in CIS-treated groups increased as well as IL-10 decreased, thereby elevating the inflammatory response. In addition, 8-OHdG and caspase-3 immunoreactivity in neurons increased with CIS administration. Treatment with tangeretin ameliorated the deterioration in oxidant/antioxidant status, overpowered neuroinflammation and ameliorated neurotoxicity-induced apoptosis. This study shows that tangeretin has beneficial effects on CIS-induced neurodegeneration. Possible mechanisms underlying these beneficial effects include the antioxidant and anti-inflammatory properties of tangeretin.
Collapse
Affiliation(s)
- Betul Cicek
- Department of Physiology, Faculty of MedicineErzincan Binali Yildirim UniversityErzincanTurkey
| | - Betul Danisman
- Department of Biophysics, Faculty of MedicineAtaturk UniversityErzurumTurkey
| | - Ismail Bolat
- Department of Pathology, Faculty of VeterinaryAtatürk UniversityErzurumTurkey
| | - Metin Kiliclioglu
- Department of Pathology, Faculty of VeterinaryAtatürk UniversityErzurumTurkey
| | - Mehmet Kuzucu
- Department of Biology, Faculty of Arts and SciencesErzincan Binali Yildirim UniversityErzincanTurkey
| | - Halis Suleyman
- Department of Medical Pharmacology, Faculty of MedicineErzincan Binali Yildirim UniversityErzincanTurkey
| | - Konstantinos Tsarouhas
- Department of CardiologyUniversity General Hospital of Larissa, Terma MazourloLarissaGreece
| | - Aristidis Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of MedicineUniversity of CreteHeraklionGreece
| | | |
Collapse
|
13
|
Yamamoto H, Ishida Y, Zhang S, Osako M, Nosaka M, Kuninaka Y, Ishigami A, Iwahashi Y, Aragane M, Matsumoto L, Kimura A, Kondo T. Protective roles of thrombomodulin in cisplatin-induced nephrotoxicity through the inhibition of oxidative and endoplasmic reticulum stress. Sci Rep 2024; 14:14004. [PMID: 38890434 PMCID: PMC11189513 DOI: 10.1038/s41598-024-64619-y] [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: 08/30/2023] [Accepted: 06/11/2024] [Indexed: 06/20/2024] Open
Abstract
Cisplatin is an effective chemotherapeutic agent widely used for the treatment of various solid tumors. However, cisplatin has an important limitation in its use; currently, there is no method to ameliorate cisplatin-induced acute kidney injury (AKI). Thrombomodulin (TM) is well known not only for its role as a cofactor in the clinically important natural anticoagulation pathway but also for its anti-inflammatory properties. Here, we investigated the effects of TM in cisplatin-induced AKI. In mice intraperitoneally injected with 15 mg/kg cisplatin, TM (10 mg/kg) or PBS was administered intravenously at 24 h after cisplatin injection. TM significantly attenuated cisplatin-induced nephrotoxicity with the suppressed elevation of blood urea nitrogen and serum creatinine, and reduced histological damages. Actually, TM treatment significantly alleviated oxidative stress-induced apoptosis by reducing reactive oxygen species (ROS) levels in cisplatin-treated renal proximal tubular epithelial cells (RPTECs) in vitro. Furthermore, TM clarified cisplatin-induced apoptosis by reducing caspase-3 levels. In addition, TM attenuated the endoplasmic reticulum (ER) stress signaling pathway in both renal tissues and RPTECs to protect the kidneys from cisplatin-induced AKI. These findings suggest that TM is a potential protectant against cisplatin-induced nephrotoxicity through suppressing ROS generation and ER stress in response to cisplatin.
Collapse
Affiliation(s)
- Hiroki Yamamoto
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Yuko Ishida
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan.
| | - Siying Zhang
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Miyu Osako
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Mizuho Nosaka
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Yumi Kuninaka
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Akiko Ishigami
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Yuya Iwahashi
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
- Department of Urology, Wakayama Medical University, Wakayama, Japan
| | - Miki Aragane
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Lennon Matsumoto
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Akihiko Kimura
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Toshikazu Kondo
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan.
| |
Collapse
|
14
|
Magdy O, Eshra M, Rashed L, Maher M, Hosny SA, ShamsEldeen AM. Amelioration of cisplatin-induced neurodegenerative changes in rats and restoration of mitochondrial biogenesis by 6-bromoindirubin-3'-oxime: The implication of the GSK-3β/PGC1-α axis. Tissue Cell 2024; 88:102393. [PMID: 38705086 DOI: 10.1016/j.tice.2024.102393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/12/2024] [Accepted: 04/18/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND The cognitive deficits observed after treatment with chemotherapeutic drugs are obvious clinical problems. For treating chemotherapy-induced cognitive deficits (CICD), the treatment modalities must target its underlying mechanisms. Specifically, cisplatin may activate glycogen synthase kinase-3β (GSK-3β), thereby enhancing neuronal apoptosis. 6-bromoindirubin-3'-oxime (6BIO) was not investigated previously in a model of CICD. Therefore, this investigation aimed to address the impacts of GSK3 inhibition on regulating cell signaling, which contributes to neurodegeneration and cognitive impairment. METHODS Thirty adult male Wistar rats were randomly allocated into control groups, while two experimental groups were exposed to repeated cisplatin injections (2 mg/kg intraperitoneally (ip), twice weekly, nine injections), termed chemobrain groups. The rats in the two experimental groups were equally divided into the chemobrain group (untreated) and the chemobrain-6BIO group (treated with 6BIO at a dose of 8.5 μg/kg ip every two days, started after the last dose of cisplatin and continued for two weeks). RESULTS Repeated exposure to cisplatin led to a marked decline in cognitive functions. GSK3 inhibition exerted neuroprotection by decreasing the expression of p-tau and amyloid β, thereby improving cognition. 6BIO, the GSK-3β inhibitor, restored mitochondrial biogenesis by augmenting the protein levels of PGC1-α and increasing the number of mitochondria in the cerebral cortex and hippocampus. CONCLUSION 6BIO provided neuroprotection and exhibited anti-apoptotic and anti-oxidative effects in a rat model of chemobrain.
Collapse
Affiliation(s)
- Ola Magdy
- Department of Physiology, Faculty of Medicine, Cairo University, Egypt
| | - Mohammed Eshra
- Department of Physiology, Faculty of Medicine, Cairo University, Egypt
| | - Laila Rashed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Egypt
| | - Muhammed Maher
- Department of Physiology, Faculty of Medicine, Cairo University, Egypt
| | - Sara Adel Hosny
- Department of Histology, Faculty of Medicine, Cairo University, Egypt
| | | |
Collapse
|
15
|
Dong W, Jiang Y, Yao Q, Xu M, Jin Y, Dong L, Li Z, Yu D. Inhibition of CISD1 attenuates cisplatin-induced hearing loss in mice via the PI3K and MAPK pathways. Biochem Pharmacol 2024; 223:116132. [PMID: 38492782 DOI: 10.1016/j.bcp.2024.116132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/20/2024] [Accepted: 03/14/2024] [Indexed: 03/18/2024]
Abstract
Cisplatin is an effective chemotherapeutic drug for different cancers, but it also causes severe and permanent hearing loss. Oxidative stress and mitochondrial dysfunction in cochlear hair cells (HCs) have been shown to be important in the pathogenesis of cisplatin-induced hearing loss (CIHL). CDGSH iron sulfur domain 1 (CISD1, also known as mitoNEET) plays a critical role in mitochondrial oxidative capacity and cellular bioenergetics. Targeting CISD1 may improve mitochondrial function in various diseases. However, the role of CISD1 in cisplatin-induced ototoxicity is unclear. Therefore, this study was performed to assess the role of CISD1 in cisplatin-induced ototoxicity. We found that CISD1 expression was significantly increased after cisplatin treatment in both HEI-OC1 cells and cochlear HCs. Moreover, pharmacological inhibition of CISD1 with NL-1 inhibited cell apoptosis and reduced mitochondrial reactive oxygen species accumulation in HEI-OC1 cells and cochlear explants. Inhibition of CISD1 with small interfering RNA in HEI-OC1 cells had similar protective effects. Furthermore, NL-1 protected against CIHL in adult C57 mice, as evaluated by the auditory brainstem response and immunofluorescent staining. Mechanistically, RNA sequencing revealed that NL-1 attenuated CIHL via the PI3K and MAPK pathways. Most importantly, NL-1 did not interfere with the antitumor efficacy of cisplatin. In conclusion, our study revealed that targeting CISD1 with NL-1 reduced reactive oxygen species accumulation, mitochondrial dysfunction, and apoptosis via the PI3K and MAPK pathways in HEI-OC1 cell lines and mouse cochlear explants in vitro, and it protected against CIHL in adult C57 mice. Our study suggests that CISD1 may serve as a novel target for the prevention of CIHL.
Collapse
Affiliation(s)
- Wenqi Dong
- Shanghai Key Laboratory of Sleep Disordered Breathing, Department of Otolaryngology-Head and Neck Surgery, Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yumeng Jiang
- Shanghai Key Laboratory of Sleep Disordered Breathing, Department of Otolaryngology-Head and Neck Surgery, Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingxiu Yao
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Maoxiang Xu
- Department of ORL-HNS, Shanghai Fourth People's Hospital, and School of Medicine, Tongji University, Shanghai, China
| | - Yuchen Jin
- Shanghai Key Laboratory of Sleep Disordered Breathing, Department of Otolaryngology-Head and Neck Surgery, Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingkang Dong
- Shanghai Key Laboratory of Sleep Disordered Breathing, Department of Otolaryngology-Head and Neck Surgery, Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhuangzhuang Li
- Shanghai Key Laboratory of Sleep Disordered Breathing, Department of Otolaryngology-Head and Neck Surgery, Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Dongzhen Yu
- Shanghai Key Laboratory of Sleep Disordered Breathing, Department of Otolaryngology-Head and Neck Surgery, Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
16
|
Vaghari-Tabari M, Jafari-Gharabaghlou D, Mohammadi M, Hashemzadeh MS. Zinc Oxide Nanoparticles and Cancer Chemotherapy: Helpful Tools for Enhancing Chemo-sensitivity and Reducing Side Effects? Biol Trace Elem Res 2024; 202:1878-1900. [PMID: 37639166 DOI: 10.1007/s12011-023-03803-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/05/2023] [Indexed: 08/29/2023]
Abstract
Cancer chemotherapy is still a serious challenge. Chemo-resistance and destructive side effects of chemotherapy drugs are the most critical limitations of chemotherapy. Chemo-resistance is the leading cause of chemotherapy failure. Chemo-resistance, which refers to the resistance of cancer cells to the anticancer effects of chemotherapy drugs, is caused by various reasons. Among the most important of these reasons is the increase in the efflux of chemotherapy drugs due to the rise in the expression and activity of ABC transporters, the weakening of apoptosis, and the strengthening of stemness. In the last decade, a significant number of studies focused on the application of nanotechnology in cancer treatment. Considering the anti-cancer properties of zinc, zinc oxide nanoparticles have received much attention in recent years. Some studies have indicated that zinc oxide nanoparticles can target the critical mechanisms of cancer chemo-resistance and enhance the effectiveness of chemotherapy drugs. These studies have shown that zinc oxide nanoparticles can reduce the activity of ABC transporters, increase DNA damage and apoptosis, and attenuate stemness in cancer cells, leading to enhanced chemo-sensitivity. Some other studies have also shown that zinc oxide nanoparticles in low doses can be helpful in minimizing the harmful side effects of chemotherapy drugs. In this article, after a brief overview of the mechanisms of chemo-resistance and anticancer effects of zinc, we will review all these studies in detail.
Collapse
Affiliation(s)
- Mostafa Vaghari-Tabari
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Davoud Jafari-Gharabaghlou
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mozafar Mohammadi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
17
|
Shahlaei M, Asl SM, Derakhshani A, Kurek L, Karges J, Macgregor R, Saeidifar M, Kostova I, Saboury AA. Platinum-based drugs in cancer treatment: Expanding horizons and overcoming resistance. J Mol Struct 2024; 1301:137366. [DOI: 10.1016/j.molstruc.2023.137366] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
18
|
Hassan MAM, Wahdan SA, El-Naga RN, Abdelghany TM, El-Demerdash E. Ondansetron attenuates cisplatin-induced behavioral and cognitive impairment through downregulation of NOD-like receptor inflammasome pathway. Toxicol Appl Pharmacol 2024; 485:116875. [PMID: 38437957 DOI: 10.1016/j.taap.2024.116875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/21/2024] [Accepted: 02/27/2024] [Indexed: 03/06/2024]
Abstract
Cisplatin is an effective and commonly used chemotherapeutic drug; however, its use is accompanied by several adverse effects, including chemobrain. Ondansetron is a 5-HT3 antagonist, commonly used in prophylactic against chemotherapy-induced nausea and vomiting. Moreover, it has been identified as a novel neuroprotective agent in different animal models. However, its protective role against chemotherapy-induced chemobrain has not been investigated. The current study was the first study that explored the potential neuroprotective effect of ondansetron against cisplatin-induced chemobrain in rats. Cisplatin (5 mg/Kg) was injected intraperitoneally, once weekly, for 4 weeks with the daily administration of ondansetron (0.5 and 1 mg/Kg). Compared to the cisplatin-treated group, ondansetron administration showed a significant decrease in the latency time and a significant increase in ambulation, rearing, and grooming frequency in the open field test (OFT). Moreover, a significant improvement in the latency time in the rotarod and passive avoidance tests, following ondansetron administration. In addition, ondansetron treatment increased the percentage of alternation in the Y-maze test. Also, ondansetron showed a remarkable enhancement in the biochemical parameters in the hippocampus. It increased the acetylcholine (Ach) level and decreased the level of the acetylcholine esterase enzyme (AchE). Ondansetron significantly decreased interleukin-1β (Il-1β), tumor necrosis factor-alpha (TNF-α), toll-like receptor-4 (TLR-4), NOD-like receptor-3 (NLRP3) inflammasome as well as caspase-1 and caspase-3 levels. Furthermore, ondansetron significantly decreased the levels of copper transporter-1(CTR1) expression in the hippocampus. Collectively, these findings suggest that ondansetron may exhibit a neuroprotective and therapeutic activity against cisplatin-induced chemobrain.
Collapse
Affiliation(s)
- Mennat-Allah M Hassan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Sara A Wahdan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Egypt
| | - Reem N El-Naga
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Egypt
| | - Tamer M Abdelghany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr city, Cairo 11884, Egypt
| | - Ebtehal El-Demerdash
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Egypt.
| |
Collapse
|
19
|
Zahedi K, Barone S, Brooks M, Stewart TM, Foley JR, Nwafor A, Casero RA, Soleimani M. Polyamine Catabolism and Its Role in Renal Injury and Fibrosis in Mice Subjected to Repeated Low-Dose Cisplatin Treatment. Biomedicines 2024; 12:640. [PMID: 38540254 PMCID: PMC10968664 DOI: 10.3390/biomedicines12030640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 02/29/2024] [Accepted: 03/07/2024] [Indexed: 11/03/2024] Open
Abstract
Cisplatin, a chemotherapeutic agent, can cause nephrotoxic and ototoxic injuries. Using a mouse model of repeated low dose cisplatin (RLDC), we compared the kidneys of cisplatin- and vehicle-treated mice on days 3 (early injury phase) and 35 (late injury/recovery phase) after the final treatment. RNA-seq analyses revealed increases in the expression of markers of kidney injury (e.g., lipocalin 2 and kidney injury molecule 1) and fibrosis (e.g., collagen 1, fibronectin, and vimentin 1) in RLDC mice. In addition, we observed increased expression of polyamine catabolic enzymes (spermidine/spermine N1-acetyltransferase, Sat1, and spermine oxidase, Smox) and decreased expression of ornithine decarboxylase (Odc1), a rate-limiting enzyme in polyamine synthesis in mice subjected to RLDC. Upon confirmation of the RNA-seq results, we tested the hypothesis that enhanced polyamine catabolism contributes to the onset of renal injury and development of fibrosis. To test our hypothesis, we compared the severity of RLDC-induced renal injury and fibrosis in wildtype (WT), Sat1-KO, and Smox-KO mice. Our results suggest that the ablation of polyamine catabolic enzymes reduces the severity of renal injury and that modulation of the activity of these enzymes may protect against kidney damage and fibrosis caused by cisplatin treatment.
Collapse
Affiliation(s)
- Kamyar Zahedi
- Division of Nephrology, Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
- Research Services, New Mexico Veterans Health Care Center, Albuquerque, NM 87108, USA
| | - Sharon Barone
- Division of Nephrology, Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
- Research Services, New Mexico Veterans Health Care Center, Albuquerque, NM 87108, USA
| | - Marybeth Brooks
- Division of Nephrology, Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
- Research Services, New Mexico Veterans Health Care Center, Albuquerque, NM 87108, USA
| | - Tracy Murray Stewart
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jackson R. Foley
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Ashley Nwafor
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Robert A. Casero
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Manoocher Soleimani
- Division of Nephrology, Department of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
- Research Services, New Mexico Veterans Health Care Center, Albuquerque, NM 87108, USA
| |
Collapse
|
20
|
Qin H, Yuan Y, Yuan M, Yi S, Yang Y, Zhang Y. Tanshinone IIA ameliorates cisplatin-induced toxicology and cisplatin resistance via regulating SLC7A11 expression. ENVIRONMENTAL TOXICOLOGY 2024; 39:1429-1441. [PMID: 37987512 DOI: 10.1002/tox.24049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/04/2023] [Accepted: 11/09/2023] [Indexed: 11/22/2023]
Abstract
Cisplatin, a potent chemotherapy agent, is highly effective against various cancers but is hindered by resistance and toxicities. This study aims to investigate the roles of SLC7A11, a cystine/glutamate transporter, in cisplatin resistance, and explored Tanshinone IIA as a therapeutic option. Cisplatin reduced SLC7A11 in renal cells, worsening toxicity. Cisplatin-resistant gastric cancer cells show increased SLC7A11, driving resistance, while SLC7A11 knockdown curbed resistance. Tanshinone IIA showed promise in alleviating cisplatin toxicity by enhancing SLC7A11 expression and reducing associated adverse effects, while it effectively reversed cisplatin resistance in gastric cancer cells by suppressing SLC7A11. Additionally, Tanshinone IIA counteracted cisplatin resistance by inhibiting PIAS4-mediated SUMOylation of SLC7A11. Simultaneously, overexpressing miR-375, which has been shown to target SLC7A11, exacerbated cisplatin toxicity via SLC7A11 downregulation, which Tanshinone IIA attenuates. In summary, our study unveils complex SLC7A11 regulation in cisplatin resistance and toxicity. Tanshinone IIA emerges as a promising modulator of SLC7A11 through individual pathways, offering novel insights into overcoming cisplatin resistance and reducing toxicities in cancer therapy.
Collapse
Affiliation(s)
- Hai Qin
- Department of Clinical Laboratory, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang City, Guizhou Province, China
| | - Yaqin Yuan
- Microbiological Laboratory, Guizhou Center For Medical Device Testing, Guiyang City, Guizhou Province, China
| | - Manqin Yuan
- Department of Clinical Laboratory Medicine, Guizhou Medical University, Guiyang City, Guizhou Province, China
| | - Siyi Yi
- Department of Clinical Laboratory, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang City, Guizhou Province, China
| | - Yonghong Yang
- Department of Clinical Laboratory, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang City, Guizhou Province, China
| | - Yujie Zhang
- Department of Clinical Laboratory, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang City, Guizhou Province, China
| |
Collapse
|
21
|
Chen X, Xiang W, Li L, Xu K. Copper Chaperone Atox1 Protected the Cochlea From Cisplatin by Regulating the Copper Transport Family and Cell Cycle. Int J Toxicol 2024; 43:134-145. [PMID: 37859596 DOI: 10.1177/10915818231206665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
Antioxidant 1 copper chaperone (Atox1) may contribute to preventing DDP cochlear damage by regulating copper transport family and cell cycle proteins. A rat model of cochlear damage was developed by placing gelatin sponges treated with DDP in the cochlea. HEI-OC1 cells were treated with 133 μM DDP as a cell model. DDP-induced ototoxicity in rats was confirmed by immunofluorescence (IF) imaging. The damage of DDP to HEI-OC1 cells was assessed by using CCK-8, TUNEL, and flow cytometry. The relationship between Atox1, a member of the copper transport protein family, and the damage to in vivo/vitro models was explored by qRT-PCR, western blot, CCK-8, TUNEL, and flow cytometry. DDP had toxic and other side effects causing cochlear damage and promoted HEI-OC1 cell apoptosis and cell cycle arrest. The over-expression of Atox1 (oe-Atox1) was accomplished by transfecting lentiviral vectors into in vitro/vivo models. We found that oe-Atox1 increased the levels of Atox1, copper transporter 1 (CTR1), and SOD3 in HEI-OC1 cells and decreased the expression levels of ATPase copper transporting α (ATP7A) and ATPase copper transporting β (ATP7B). In addition, the transfection of oe-Atox1 decreased cell apoptosis rate and the number of G2/M stage cells. Similarly, the expression of myosin VI and phalloidin of cochlea cells in vivo decreased. Atox1 ameliorated DDP-induced damage to HEI-OC1 cells or rats' cochlea by regulating the levels of members of the copper transport family.
Collapse
Affiliation(s)
- Xubo Chen
- Department of Otolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Weiren Xiang
- Department of Otolaryngology, Head and Neck Surgery, Jiu Jiang No.1 People's Hospital, Jiujiang, China
| | - Lihua Li
- Department of Otolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Kai Xu
- Department of Otolaryngology, Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
22
|
Tang D, Wang X, Wu J, Li Y, Li C, Qiao X, Fan L, Chen Y, Zhu H, Zhang Z, He Y. Cinchonine and cinchonidine alleviate cisplatin-induced ototoxicity by regulating PI3K-AKT signaling. CNS Neurosci Ther 2024; 30:e14403. [PMID: 37577804 PMCID: PMC10848099 DOI: 10.1111/cns.14403] [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: 02/07/2023] [Revised: 07/09/2023] [Accepted: 07/31/2023] [Indexed: 08/15/2023] Open
Abstract
AIM Cinchonine (CN) and its isomer cinchonidine (CD), two of the common cinchona alkaloids, are wildly used as antimalarial drugs. However, the effects of CN and CD on the auditory system are unknown. METHODS Molecular docking and molecular dynamics (MD) simulation were used for predicting effective drugs. The CCK-8 assay was conducted for assessing cell viability in House Ear Institute-Organ of Corti 1 (HEI-OC1) cells. MitoSox Red staining revealed reactive oxygen species (ROS) amounts. TMRM staining was used to assess the mitochondrial membrane potential (ΔΨm). Immunofluorescence staining of myosin 7a was used to examine hair cells (HCs) in cisplatin-treated neonatal mouse cochlear explants, while TUJ-1 immunostaining was used for the detection of spiral ganglion neurons (SGNs). Cleaved caspase-3 and TUNEL immunostaining were utilized for apoptosis assessment. Immunoblot was carried out to detect PI3K-AKT signaling effectors. RESULTS Pretreatment with CN or CD significantly increased cell viability and reduced mitochondrial dysfunction and ROS accumulation in cisplatin-treated HEI-OC1 cells. Immunofluorescent staining of cochlear explants showed that CN and CD attenuated cisplatin-induced damage to SGNs and HCs. Immunoblot revealed that CN and CD downregulated the expression of cleaved caspase-3 and activated PI3K-AKT signaling in cisplatin-injured HEI-OC1 cells. CONCLUSION CD and CN can reduce ototoxicity caused by cisplatin and might help treat cisplatin-associated hearing loss.
Collapse
Affiliation(s)
- Dongmei Tang
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, NHC Key Laboratory of Hearing MedicineFudan UniversityShanghaiChina
| | - Xue Wang
- Department of Otorhinolaryngology‐Head and Neck SurgeryFirst Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Jingfang Wu
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, NHC Key Laboratory of Hearing MedicineFudan UniversityShanghaiChina
| | - Yimeng Li
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, NHC Key Laboratory of Hearing MedicineFudan UniversityShanghaiChina
| | - Cai Li
- Department of Otorhinolaryngology‐Head and Neck SurgeryFirst Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Xiangyun Qiao
- Department of Otorhinolaryngology Head and Neck SurgeryThe Second Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Li Fan
- Department of Otorhinolaryngology‐Head and Neck SurgeryFirst Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Yutao Chen
- Department of Otorhinolaryngology‐Head and Neck SurgeryFirst Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Huanhuan Zhu
- Department of Otorhinolaryngology‐Head and Neck SurgeryFirst Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Zhiyuan Zhang
- Department of Otorhinolaryngology‐Head and Neck SurgeryFirst Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Yingzi He
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, NHC Key Laboratory of Hearing MedicineFudan UniversityShanghaiChina
| |
Collapse
|
23
|
Becker G, Atuati SF, Oliveira SM. G Protein-Coupled Receptors and Ion Channels Involvement in Cisplatin-Induced Peripheral Neuropathy: A Review of Preclinical Studies. Cancers (Basel) 2024; 16:580. [PMID: 38339331 PMCID: PMC10854671 DOI: 10.3390/cancers16030580] [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: 12/20/2023] [Revised: 01/23/2024] [Accepted: 01/27/2024] [Indexed: 02/12/2024] Open
Abstract
Cisplatin is a platinum-based chemotherapy drug widely used to treat various solid tumours. Although it is effective in anti-cancer therapy, many patients develop peripheral neuropathy during and after cisplatin treatment. Peripheral neuropathy results from lesions or diseases in the peripheral somatosensory nervous system and is a significant cause of debilitation and suffering in patients. In recent years, preclinical studies have been conducted to elucidate the mechanisms involved in chemotherapy-induced peripheral neuropathic pain, as well as to promote new therapeutic targets since current treatments are ineffective and are associated with adverse effects. G-protein coupled receptors and ion channels play a significant role in pain processing and may represent promising targets for improving the management of cisplatin-induced neuropathic pain. This review describes the role of G protein-coupled receptors and ion channels in cisplatin-induced pain, analysing preclinical experimental studies that investigated the role of each receptor subtype in the modulation of cisplatin-induced pain.
Collapse
|
24
|
Zavala-Valencia AC, Velasco-Hidalgo L, Martínez-Avalos A, Castillejos-López M, Torres-Espíndola LM. Effect of N-Acetylcysteine on Cisplatin Toxicity: A Review of the Literature. Biologics 2024; 18:7-19. [PMID: 38250216 PMCID: PMC10799624 DOI: 10.2147/btt.s438150] [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: 10/11/2023] [Accepted: 12/08/2023] [Indexed: 01/23/2024]
Abstract
N-acetylcysteine (NAC) is a membrane-permeable cysteine precursor capable of enhancing the intracellular cysteine pool, enhancing cellular glutathione (GSH) synthesis, and thus potentiating the endogenous antioxidant mechanism. Late administration of NAC after cisplatin has been shown in different in vivo studies to reduce the side effects caused by various toxicities at different levels without affecting the antitumor efficacy of platinum, improving total and enzymatic antioxidant capacity and decreasing oxidative stress markers. These characteristics provide NAC with a rationale as a potentially effective chemo protectant in cisplatin-based therapeutic cycles. NAC represents a potential candidate as a chemoprotective agent to decrease toxicities secondary to cisplatin treatment. It suggests that it could be used in clinical trials, whereby the effective dose, timing, and route should be adjusted to optimize chemoprotection. This review provides an overview of the effect of NAC on cisplatin toxicity, a drug widely used in the clinic in adults and children.
Collapse
Affiliation(s)
- Angeles Citlali Zavala-Valencia
- Laboratory of Pharmacology, National Institute of Pediatrics, Mexico City, Mexico
- Iztacala Faculty of Higher Studies, Tlalnepantla, México
| | | | | | - Manuel Castillejos-López
- Hospital Epidemiology and Infectology Unit, National Institute of Respiratory Diseases Ismael Cosío Villegas, Mexico City, Mexico
| | | |
Collapse
|
25
|
Li X, Shi J, Teng Y, Liu Z. The preventative effect of Baihe Gujin Pill on cisplatin-induced acute kidney injury by activating the PI3K/AKT and suppressing the NF-κB/MAPK pathways. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:117071. [PMID: 37619855 DOI: 10.1016/j.jep.2023.117071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/31/2023] [Accepted: 08/20/2023] [Indexed: 08/26/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Baihe Gujin Pill (BHGJP) is a traditional Chinese medicine (TCM) derived from the "Collection of Medical Formulas". BHGJP is applied to treat lung and kidney deficiency by nourishing yin and clearing heat. However, the role and preventative mechanism of BHGJP in cisplatin induced acute kidney injury (CIAKI) are poorly understood. AIM OF THE STUDY The preventative effect of BHGJP on CIAKI by the in vitro and in vivo experiments based on network pharmacology was investigated. METHODS Network pharmacology was used to predict the protective effect of BHGJP on CIAKI. The effect and mechanism of BHGJP against CIAKI were detected and verified by the in vitro kidney cells 293T and HK-2 as well as the in vivo mice model established by a single injection of cisplatin. RESULTS Network pharmacology predicted that BHGJP prevented CIAKI by regulating PI3K/AKT and NF-κB/MAPK signaling pathways. BHGJP could reverse the reduced cell viability of HK-2 and 293T cells caused by cisplatin without decreasing its cytotoxic effects on H460, H1299, and A549 cells. Meanwhile, BHGJP effectively controlled kidney injury in the CIAKI model. Moreover, cisplatin induced cell apoptosis and accumulation of reactive oxygen species (ROS) were downregulated after treatment with BHGJP. The changes of oxidative stress indexes of GSH, MDA, and SOD as well as the inflammatory factors of TNF-α, IL-6, and IL-1β in the CIAKI model were recovered to normal state when BHGJP treatment. Furthermore, BHGJP activated PI3K/AKT pathway and suppressed the NF-κB/MAPK pathway in the CIAKI model. CONCLUSION The study found that BHGJP prevented CIAKI by inhibiting apoptosis, oxidative stress, and inflammation via regulating PI3K/AKT and NF-κB/MAPK pathways, providing new efficacy and clinical applications for BHGJP.
Collapse
Affiliation(s)
- Xinran Li
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Jieya Shi
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Yuou Teng
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China.
| | - Zhen Liu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industry Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China.
| |
Collapse
|
26
|
Hsieh CY, Tsai CY, Chou YF, Hsu CJ, Wu HP, Wu CC. Otoprotection against aminoglycoside- and cisplatin-induced ototoxicity focusing on the upstream drug uptake pathway. J Chin Med Assoc 2024; 87:17-24. [PMID: 37962398 DOI: 10.1097/jcma.0000000000001023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2023] Open
Abstract
Aminoglycoside- and cisplatin-induced ototoxicity, which is a significant issue owing to the widespread use of these drugs in clinical practice, involves the entry of aminoglycosides and cisplatin into the endolymph and hair cells via specific channels or transporters, followed by reactive oxygen species (ROS) generation and hair cells apoptosis. Current strategies focalize primarily on interference with downstream ROS effects; however, recent evidence has demonstrated that inhibiting the uptake of aminoglycosides and cisplatin by hair cells is another promising strategy for tackling the upstream drug uptake pathway. With advances in structural biology, the conformations of certain aminoglycoside and cisplatin channels and transporters, such as the mechanoelectrical transduction channel and organic cation transporter-2, have been largely elucidated. These channels and transporters may become potential targets for the introduction of new otoprotective strategies. This review focuses on the strategies for inhibiting ototoxic drugs uptake by auditory hair cells and provides potential targets for recent developments in the field of otoprotection. Molecular dynamics (MD) simulations of these proteins could help identify the molecules that inhibit the uptake of aminoglycosides and cisplatin by hair cells. Integrating upstream drug uptake pathway targets and MD simulations may help dissect molecular mechanisms and develop novel otoprotective strategies for aminoglycoside- and cisplatin-induced ototoxicity.
Collapse
Affiliation(s)
- Cheng-Yu Hsieh
- Department of Otolaryngology Head and Neck Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan, ROC
- Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan, ROC
| | - Cheng-Yu Tsai
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, Taiwan, ROC
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Yi-Fan Chou
- Department of Otolaryngology Head and Neck Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan, ROC
- School of Medicine, Tzu Chi University, Hualien, Taiwan, ROC
| | - Chuan-Jen Hsu
- Department of Otolaryngology Head and Neck Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan, ROC
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Hung-Pin Wu
- Department of Otolaryngology Head and Neck Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan, ROC
- School of Medicine, Tzu Chi University, Hualien, Taiwan, ROC
| | - Chen-Chi Wu
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan, ROC
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan, ROC
- Department of Medical Research, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan, ROC
| |
Collapse
|
27
|
Tian J, Mu Y, Ma L. Chemerin/CMKLR1 pathway exacerbates cisplatin-induced spiral ganglion neuron injury. Toxicol Res 2024; 40:73-81. [PMID: 38223664 PMCID: PMC10786799 DOI: 10.1007/s43188-023-00205-0] [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: 01/31/2023] [Revised: 07/19/2023] [Accepted: 08/02/2023] [Indexed: 01/16/2024] Open
Abstract
This study investigated whether chemerin/chemokine-like receptor 1 (CMKLR1) pathway participate in cisplatin-induced spiral ganglion neuron (SGN) damage. Middle cochlear turn was collected from C57BL/6 mice and the SGNs were cultured. Cisplatin, 2-(anaphthoyl) ethyltrimethylammonium iodide (α-NETA), or recombinant mouse chemerin was added into the medium for the treatment. Relative mRNA and protein expression was determined by RT-PCR, ELISA and Western blot, respectively. In cultured mouse cochlear SGNs, the treatment of cisplatin enhanced the secretion of chemerin and CMKLR1. Recombinant chemerin promoted but α-NETA inhibited chemerin/CMKLR1 pathway in cisplatin stimulated SGNs. Cisplatin-induced apoptosis and inflammation response in SGNs were enhanced by recombinant chemerin while inhibited by α-NETA. Recombinant chemerin promoted but α-NETA inhibited NF-κB signal in cisplatin stimulated SGNs. In conclusion, chemerin/CMKLR1 pathway regulated apoptosis and inflammation response in cisplatin-induced SGN injury through NF-κB signaling pathway. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-023-00205-0.
Collapse
Affiliation(s)
- Jie Tian
- Department of Otology, Zibo Central Hospital, No. 54, Gongqingtuan West Road, Zhangdian District, Zibo, 255036 Shandong China
| | - Ying Mu
- Department of Emergency Medicine, Zibo Central Hospital, No. 54 Gongqingtuan West Road, Zibo, 255036 Shandong China
| | - Lili Ma
- Department of Neurology, Zibo Central Hospital, No. 54 Gongqingtuan West Road, Zibo, 255036 Shandong China
| |
Collapse
|
28
|
Li Q, Chen S, Wang X, Cai J, Huang H, Tang S, He D. Cisplatin-Based Combination Therapy for Enhanced Cancer Treatment. Curr Drug Targets 2024; 25:473-491. [PMID: 38591210 DOI: 10.2174/0113894501294182240401060343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/04/2024] [Accepted: 03/15/2024] [Indexed: 04/10/2024]
Abstract
Cisplatin, a primary chemotherapeutic drug, is of great value in the realm of tumor treatment. However, its clinical efficacy is strictly hindered by issues, such as drug resistance, relapse, poor prognosis, and toxicity to normal tissue. Cisplatin-based combination therapy has garnered increasing attention in both preclinical and clinical cancer research for its ability to overcome resistance, reduce toxicity, and enhance anticancer effects. This review examines three primary co-administration strategies of cisplatin-based drug combinations and their respective advantages and disadvantages. Additionally, seven types of combination therapies involving cisplatin are discussed, focusing on their main therapeutic effects, mechanisms in preclinical research, and clinical applications. This review also discusses future prospects and challenges, aiming to offer guidance for the development of optimal cisplatin-based combination therapy regimens for improved cancer treatment.
Collapse
Affiliation(s)
- Qi Li
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
| | - Siwei Chen
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
| | - Xiao Wang
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
| | - Jia Cai
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
| | - Hongwu Huang
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
| | - Shengsong Tang
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, Hunan University of Medicine, Huaihua, China
| | - Dongxiu He
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
| |
Collapse
|
29
|
Lateef Al-Awsi GR, Arshed U, Arif A, Ramírez-Coronel AA, Alhassan MS, Mustafa YF, Rahman FF, Zabibah RS, Gupta J, Iqbal MS, Iswanto AH, Farhood B. The Chemoprotective Potentials of Alpha-lipoic Acid against Cisplatin-induced Ototoxicity: A Systematic Review. Curr Med Chem 2024; 31:3588-3603. [PMID: 37165582 DOI: 10.2174/0929867330666230509162513] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 03/08/2023] [Accepted: 04/05/2023] [Indexed: 05/12/2023]
Abstract
PURPOSE Ototoxicity is one of the major adverse effects of cisplatin therapy which restrict its clinical application. Alpha-lipoic acid administration may mitigate cisplatin-induced ototoxicity. In the present study, we reviewed the protective potentials of alpha-lipoic acid against the cisplatin-mediated ototoxic adverse effects. METHODS Based on the PRISMA guideline, we performed a systematic search for the identification of all relevant studies in various electronic databases up to June 2022. According to the inclusion and exclusion criteria, the obtained articles (n=59) were screened and 13 eligible articles were finally included in the present study. RESULTS The findings of in-vitro experiments showed that cisplatin treatment significantly reduced the auditory cell viability in comparison with the control group; nevertheless, the alpha-lipoic acid co-administration protected the cells against the reduction of cell viability induced by cisplatin treatment. Moreover, the in-vivo results of the auditory brainstem response (ABR) and distortion product otoacoustic emission (DPOAE) tests revealed a decrease in DPOAE and an increase in ABR threshold of cisplatin-injected animals; however, it was shown that alpha-lipoic acid co-treatment had an opposite pattern on the evaluated parameters. Other findings demonstrated that cisplatin treatment could significantly induce the biochemical and histopathological alterations in inner ear cells/tissue; in contrast, alpha-lipoic acid co-treatment ameliorated the cisplatin-mediated biochemical and histological changes. CONCLUSION The findings of audiometry, biochemical parameters, and histological evaluation showed that alpha-lipoic acid co-administration alleviates the cisplatin-induced ototoxicity. The protective role of alpha-lipoic acid against the cisplatin-induced ototoxicity can be due to different mechanisms of anti-oxidant, anti-apoptotic, anti-inflammatory activities, and regulation of cell cycle progression.
Collapse
Affiliation(s)
| | - Uzma Arshed
- Gujranwala Medical College, Gujranwala, Pakistan
| | - Anam Arif
- Gujranwala Medical College, Gujranwala, Pakistan
| | | | - Muataz S Alhassan
- Division of Advanced Nanomaterial Technologies, Scientific Research Center, Al-Ayen University, Thi-Qar, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul-41001, Iraq
| | - Ferry Fadzlul Rahman
- Public Health Department, Universitas Muhammadiyah Kalimantan Timur, Samarinda, Indonesia
| | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Pin Code 281406, U.P., India
| | - Muhammad Shahid Iqbal
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Acim Heri Iswanto
- Public Health Department, Faculty of Health Science, University of Pembangunan Nasional Veteran Jakarta, Jakarta, Indonesia
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
30
|
Sharma H, Sharma N, An SSA. Unique Bioactives from Zombie Fungus ( Cordyceps) as Promising Multitargeted Neuroprotective Agents. Nutrients 2023; 16:102. [PMID: 38201932 PMCID: PMC10780653 DOI: 10.3390/nu16010102] [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: 11/17/2023] [Revised: 12/08/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Cordyceps, also known as "zombie fungus", is a non-poisonous mushroom that parasitizes insects for growth and development by manipulating the host system in a way that makes the victim behave like a "zombie". These species produce promising bioactive metabolites, like adenosine, β-glucans, cordycepin, and ergosterol. Cordyceps has been used in traditional medicine due to its immense health benefits, as it boosts stamina, appetite, immunity, longevity, libido, memory, and sleep. Neuronal loss is the typical feature of neurodegenerative diseases (NDs) (Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS)) and neurotrauma. Both these conditions share common pathophysiological features, like oxidative stress, neuroinflammation, and glutamatergic excitotoxicity. Cordyceps bioactives (adenosine, N6-(2-hydroxyethyl)-adenosine, ergosta-7, 9 (11), 22-trien-3β-ol, active peptides, and polysaccharides) exert potential antioxidant, anti-inflammatory, and anti-apoptotic activities and display beneficial effects in the management and/or treatment of neurodegenerative disorders in vitro and in vivo. Although a considerable list of compounds is available from Cordyceps, only a few have been evaluated for their neuroprotective potential and still lack information for clinical trials. In this review, the neuroprotective mechanisms and safety profile of Cordyceps extracts/bioactives have been discussed, which might be helpful in the identification of novel potential therapeutic entities in the future.
Collapse
Affiliation(s)
| | - Niti Sharma
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 461-701, Gyeonggi-do, Republic of Korea;
| | - Seong Soo A. An
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 461-701, Gyeonggi-do, Republic of Korea;
| |
Collapse
|
31
|
Sone H, Lee TJ, Lee BR, Heo D, Oh S, Kwon SH. MicroRNA-mediated attenuation of branched-chain amino acid catabolism promotes ferroptosis in chronic kidney disease. Nat Commun 2023; 14:7814. [PMID: 38016961 PMCID: PMC10684653 DOI: 10.1038/s41467-023-43529-z] [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: 05/22/2023] [Accepted: 11/13/2023] [Indexed: 11/30/2023] Open
Abstract
Chronic kidney disease can develop from kidney injury incident to chemotherapy with cisplatin, which complicates the prognosis of cancer patients. MicroRNAs regulate gene expression by pairing with specific sets of messenger RNAs. Therefore, elucidating direct physical interactions between microRNAs and their target messenger RNAs can help decipher crucial biological processes associated with cisplatin-induced kidney injury. Through intermolecular ligation and transcriptome-wide sequencing, we here identify direct pairs of microRNAs and their target messenger RNAs in the kidney of male mice injured by cisplatin. We find that a group of cisplatin-induced microRNAs can target select messenger RNAs that affect the mitochondrial metabolic pathways in the injured kidney. Specifically, a cisplatin-induced microRNA, miR-429-3p, suppresses the pathway that catabolizes branched-chain amino acids in the proximal tubule, leading to cell death dependent on lipid peroxidation, called ferroptosis. Identification of miRNA-429-3p-mediated ferroptosis stimulation suggests therapeutic potential for modulating the branched-chain amino acid pathway in ameliorating cisplatin-induced kidney injury.
Collapse
Affiliation(s)
- Hisakatsu Sone
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Tae Jin Lee
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, 30912, USA
| | - Byung Rho Lee
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Dan Heo
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Sekyung Oh
- Department of Medical Science, Catholic Kwandong University College of Medicine, Incheon, 22711, South Korea
| | - Sang-Ho Kwon
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
32
|
Lu D, Yang Y, Du Y, Zhang L, Yang Y, Tibenda JJ, Nan Y, Yuan L. The Potential of Glycyrrhiza from "Medicine Food Homology" in the Fight against Digestive System Tumors. Molecules 2023; 28:7719. [PMID: 38067451 PMCID: PMC10708138 DOI: 10.3390/molecules28237719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/14/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
Glycyrrhiza has a long history of applications and a wide range of pharmacological effects. It is known as the "king of all herbs". Glycyrrhiza is effective in clearing heat, detoxifying, relieving cough, and tonifying qi and has good bioactivity in multiple inflammatory, immune, and tumor diseases. This review aims to summarize the origin, distribution, and anti-digestive system tumor mechanism of glycyrrhiza and its homologous applications in medicine and food. The active compounds include triterpenoids, flavonoids, and coumarins, which are widely used in clinical treatments, disease prevention, and daily foods because of their "enhancement of efficacy" and "reduction of toxicity" against digestive system tumors. This paper reviews the use of glycyrrhiza in digestive system tumors and provides an outlook on future research and clinical applications.
Collapse
Affiliation(s)
- Doudou Lu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, China;
| | - Yating Yang
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, China;
| | - Yuhua Du
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, China; (Y.D.); (J.J.T.)
| | - Lei Zhang
- Key Laboratory of Hui Ethnic Medicine Modernization of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China;
| | - Yi Yang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China;
| | - Joanna Japhet Tibenda
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, China; (Y.D.); (J.J.T.)
| | - Yi Nan
- Key Laboratory of Hui Ethnic Medicine Modernization of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China;
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical University, Yinchuan 750004, China; (Y.D.); (J.J.T.)
| |
Collapse
|
33
|
Guo Y, Li X, Yuan R, Ren J, Huang Y, Tian H. Compound 5 alleviated acute kidney injury without affecting the antitumor effect after cisplatin treatment. Biochem Biophys Res Commun 2023; 680:177-183. [PMID: 37742346 DOI: 10.1016/j.bbrc.2023.09.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/26/2023]
Abstract
Despite being a powerful weapon against cancer cells, cisplatin's therapeutic potential is hampered by numerous adverse reactions, including acute kidney injury (AKI). Compound 5 has 3-SH fragments at the end of the vertical short alkyl side chain, which is an ROS scavenger synthesized. In this study, we evaluated the protective effect of compound 5 on the kidney after cisplatin administration and its mechanism. The results founded that compound 5 can alleviate serum urea nitrogen and serum creatinine induced by cisplatin administration in vivo. In addition, histopathological analysis of the kidneys showed that compound 5 significantly reduced cisplatin-induced (Cis-induced) renal toxicity compared with the cisplatin group. A mechanism study showed that compound 5 significantly reduces NOX4 levels, improves the activity of antioxidant enzymes (SOD and GSH-Px), reduces Malondialdehyde (MDA) levels, increases the total antioxidant level, reduces oxidative stress, and thus reduces kidney tissue damage. At the same time, compound 5 activated the Nrf2 signaling pathway. In addition, it can increase the expression of Bax, reduce the expression of Bcl-2 and caspase-3, a marker of apoptosis, which is beneficial to the survival of kidney cells. Additionally, compound 5 did not interfere with the antitumor effects of cisplatin in in vivo xenotransplantation models.
Collapse
Affiliation(s)
- Yuying Guo
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Xuejiao Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Renbin Yuan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Jingming Ren
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Yichi Huang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Hongqi Tian
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China.
| |
Collapse
|
34
|
Jia Y, Li X, Meng X, Lei J, Xia Y, Yu L. Anticancer perspective of 6-shogaol: anticancer properties, mechanism of action, synergism and delivery system. Chin Med 2023; 18:138. [PMID: 37875983 PMCID: PMC10594701 DOI: 10.1186/s13020-023-00839-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/18/2023] [Indexed: 10/26/2023] Open
Abstract
Cancer is a malignant disease that has plagued human beings all the time, but the treatment effect of commonly used anticancer drugs in clinical practice is not ideal by reason of their drug tolerance and Strong adverse reactions to patients. Therefore, it is imperative to find effective and low-toxic anticancer drugs. Many research works have shown that natural products in Chinese herbal medicine have great anticancer potential, such as 6-shogaol, a monomer composition obtained from Chinese herbal ginger, which has been confirmed by numerous in vitro or vivo studies to be an excellent anti-cancer active substance. In addition, most notably, 6-shogaol has different selectivity for normal and cancer cells during treatment, which makes it valuable for further research and clinical development. Therefore, this review focus on the anti-cancer attributes, the mechanism and the regulation of related signaling pathways of 6-shogaol. In addition, its synergy with commonly used anticancer drugs, potential drug delivery systems and prospects for future research are discussed. This is the first review to comprehensively summarize the anti-cancer mechanism of 6-shogaol, hoping to provide a theoretical basis and guiding significance for future anti-cancer research and clinical development of 6-shogaol.
Collapse
Affiliation(s)
- Yaoxia Jia
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Chengdu, 611137, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
| | - Xing Li
- Jianyang Chinese Medicine Hospital, Chengdu, 641400, China
| | - Xiangqi Meng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Chengdu, 611137, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
| | - Jinjie Lei
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Chengdu, 611137, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
| | - Yangmiao Xia
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Chengdu, 611137, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China
| | - Lingying Yu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Chengdu, 611137, China.
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, China.
| |
Collapse
|
35
|
Ali FEM, Hassanein EHM, El-Bahrawy AH, Hemeda MS, Atwa AM. Neuroprotective effect of lansoprazole against cisplatin-induced brain toxicity: Role of Nrf2/ARE and Akt/P53 signaling pathways. J Chem Neuroanat 2023; 132:102299. [PMID: 37271475 DOI: 10.1016/j.jchemneu.2023.102299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/06/2023]
Abstract
Cisplatin is a chemotherapeutic agent usually used in treating different patterns of malignancies. One of the significant apparent complications of cisplatin chemotherapy is brain toxicity. The present study was conducted to evaluate the protective effects of lansoprazole on cisplatin-induced cortical intoxication. Thirty-two rats were allocated into four groups (8 rats/group); group I: received only a vehicle for 10 days, group II: lansoprazole was administered (50 mg/kg) via oral gavage for 10 days, group III: On 5th day of the experiment, rats were given cisplatin (10 mg/kg) i.p. once to induce cortical injury. Group IV: rats were given lansoprazole for 5 days before cisplatin and 5 days afterward. Lansoprazole administration significantly improved cisplatin-induced behavioral changes, as evidenced by decreasing the immobility time in forced swimming and open field tests. Besides, lansoprazole improved cortical histological changes, restored cortical redox balance, enhanced Nrf2/ARE expression, cisplatin-induced neuronal apoptosis, and dampened cisplatin inflammation. In addition, lansoprazole modulated cortical Akt/p53 signal. The present work was the first to show that lansoprazole co-administration reduced cortical toxicity in cisplatin-treated rats via multiple signaling pathways. The current findings provided crucial information for developing novel protective strategies to reduce cisplatin cortical toxicity.
Collapse
Affiliation(s)
- Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt.
| | - Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| | - Ali H El-Bahrawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| | - Mohamed S Hemeda
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Port Said University, Port Said, Egypt
| | - Ahmed M Atwa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| |
Collapse
|
36
|
Aleksandrova Y, Neganova M. Deciphering the Mysterious Relationship between the Cross-Pathogenetic Mechanisms of Neurodegenerative and Oncological Diseases. Int J Mol Sci 2023; 24:14766. [PMID: 37834214 PMCID: PMC10573395 DOI: 10.3390/ijms241914766] [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: 08/10/2023] [Revised: 09/22/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
The relationship between oncological pathologies and neurodegenerative disorders is extremely complex and is a topic of concern among a growing number of researchers around the world. In recent years, convincing scientific evidence has accumulated that indicates the contribution of a number of etiological factors and pathophysiological processes to the pathogenesis of these two fundamentally different diseases, thus demonstrating an intriguing relationship between oncology and neurodegeneration. In this review, we establish the general links between three intersecting aspects of oncological pathologies and neurodegenerative disorders, i.e., oxidative stress, epigenetic dysregulation, and metabolic dysfunction, examining each process in detail to establish an unusual epidemiological relationship. We also focus on reviewing the current trends in the research and the clinical application of the most promising chemical structures and therapeutic platforms that have a modulating effect on the above processes. Thus, our comprehensive analysis of the set of molecular determinants that have obvious cross-functional pathways in the pathogenesis of oncological and neurodegenerative diseases can help in the creation of advanced diagnostic tools and in the development of innovative pharmacological strategies.
Collapse
Affiliation(s)
- Yulia Aleksandrova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
| | - Margarita Neganova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 420088 Kazan, Russia
| |
Collapse
|
37
|
Katanić Stanković JS, Selaković D, Rosić G. Oxidative Damage as a Fundament of Systemic Toxicities Induced by Cisplatin-The Crucial Limitation or Potential Therapeutic Target? Int J Mol Sci 2023; 24:14574. [PMID: 37834021 PMCID: PMC10572959 DOI: 10.3390/ijms241914574] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/11/2023] [Accepted: 09/23/2023] [Indexed: 10/15/2023] Open
Abstract
Cisplatin, an inorganic complex of platinum, is a chemotherapeutic drug that has been used for 45 years. Despite the progress of pharmaceutical sciences and medicine and the successful application of other platinum complexes for the same purpose, cisplatin is still the therapy of choice in many cancers. Treatment for testicular, ovarian, head and neck, urothelial, cervical, esophageal, breast, and pulmonary malignancies is still unthinkable without the use of this drug. However, cisplatin is also known for many side effects, of which the most pronounced are nephrotoxicity leading to acute renal failure, neurotoxicity, and ototoxicity. Mechanistic studies have proven that one of the conditions that plays a major role in the development of cisplatin-induced toxicities is oxidative stress. Knowing the fact that numerous antioxidants can be used to reduce oxidative stress, thereby reducing tissue lesions, organ failure, and apoptosis at the cellular level, many studies have defined antioxidants as a priority for investigation as a cotreatment. To investigate the mechanism of antioxidant action in vivo, many animal models have been employed. In the last few years, studies have mostly used rodents and zebrafish models. In this article, some of the most recent investigations that used animal models are listed, and the advantages and disadvantages of such experimental studies are pointed out.
Collapse
Affiliation(s)
- Jelena S. Katanić Stanković
- Department of Science, Institute for Information Technologies Kragujevac, University of Kragujevac, Jovana Cvijića bb, 34000 Kragujevac, Serbia
| | - Dragica Selaković
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia;
| | - Gvozden Rosić
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia;
| |
Collapse
|
38
|
Mapuskar KA, Pulliam CF, Zepeda-Orozco D, Griffin BR, Furqan M, Spitz DR, Allen BG. Redox Regulation of Nrf2 in Cisplatin-Induced Kidney Injury. Antioxidants (Basel) 2023; 12:1728. [PMID: 37760031 PMCID: PMC10525889 DOI: 10.3390/antiox12091728] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/30/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Cisplatin, a potent chemotherapeutic agent, is marred by severe nephrotoxicity that is governed by mechanisms involving oxidative stress, inflammation, and apoptosis pathways. The transcription factor Nrf2, pivotal in cellular defense against oxidative stress and inflammation, is the master regulator of the antioxidant response, upregulating antioxidants and cytoprotective genes under oxidative stress. This review discusses the mechanisms underlying chemotherapy-induced kidney injury, focusing on the role of Nrf2 in cancer therapy and its redox regulation in cisplatin-induced kidney injury. We also explore Nrf2's signaling pathways, post-translational modifications, and its involvement in autophagy, as well as examine redox-based strategies for modulating Nrf2 in cisplatin-induced kidney injury while considering the limitations and potential off-target effects of Nrf2 modulation. Understanding the redox regulation of Nrf2 in cisplatin-induced kidney injury holds significant promise for developing novel therapeutic interventions. This knowledge could provide valuable insights into potential strategies for mitigating the nephrotoxicity associated with cisplatin, ultimately enhancing the safety and efficacy of cancer treatment.
Collapse
Affiliation(s)
- Kranti A. Mapuskar
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA
| | - Casey F. Pulliam
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA
| | - Diana Zepeda-Orozco
- Pediatric Nephrology and Hypertension at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43210, USA
| | - Benjamin R. Griffin
- Division of Nephrology, The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
- Department of Internal Medicine, The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Muhammad Furqan
- Department of Internal Medicine, The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Douglas R. Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA
| | - Bryan G. Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA
| |
Collapse
|
39
|
Spies J, Covarrubias-Pinto A, Carcamo C, Arancibia Y, Salazar F, Paredes-Martinez C, Otth C, Castro M, Zambrano A. Modulation of Synaptic Plasticity Genes Associated to DNA Damage in a Model of Huntington's Disease. Neurochem Res 2023; 48:2093-2103. [PMID: 36790580 DOI: 10.1007/s11064-023-03889-w] [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: 11/17/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/16/2023]
Abstract
Huntington's disease (HD) is a disease characterized by the progressive degeneration of nerve cells in the brain. DNA damage has been implicated in many neurological disorders; however, the association between this damage and the impaired signaling related to neurodegeneration is still unclear. The transcription factor c-AMP-responsive element binding protein (CREB) has a relevant role in the neuronal plasticity process regulating the expression of several genes, including brain-derived neurotrophic factor (BDNF). Here we analyzed the direct link between DNA damage and the expression of genes involved in neuronal plasticity. The study was performed in model cell lines STHdhQ7 (wild type) and STHdhQ111 (HD model). Treatment with Etoposide (Eto) was used to induce double-strand breaks (DSBs) to evaluate the DNA damage response (DDR) and the expression of synaptic plasticity genes. Eto treatment induced phosphorylation of ATM (p-ATM) and H2AX (γH2AX), markers of DDR, in both cell lines. Interestingly, upon DNA damage, STHdhQ7 cells showed increased expression of activity-regulated cytoskeleton associated protein (Arc) and BDNF when compared to the HD cell line model. Additionally, Eto induced CREB activation with a differential localization of its co-activators in the cell types analyzed. These results suggest that DSBs impact differentially the gene expression patterns of plasticity genes in the normal cell line versus the HD model. This effect is mediated by the impaired localization of CREB-binding protein (CBP) and histone acetylation in the HD model. Our results highlight the role of epigenetics and DNA repair on HD and therefore we suggest that future studies should explore in depth the epigenetic landscape on neuronal pathologies with the goal to further understand molecular mechanisms and pinpoint therapeutic targets.
Collapse
Affiliation(s)
- Johana Spies
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Casilla (P. O. Box) 567, Valdivia, Chile
| | - Adriana Covarrubias-Pinto
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Casilla (P. O. Box) 567, Valdivia, Chile
| | - Constanza Carcamo
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Casilla (P. O. Box) 567, Valdivia, Chile
| | - Yennyfer Arancibia
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Casilla (P. O. Box) 567, Valdivia, Chile
| | - Fernanda Salazar
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Casilla (P. O. Box) 567, Valdivia, Chile
| | - Carolina Paredes-Martinez
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Casilla (P. O. Box) 567, Valdivia, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Carola Otth
- Facultad de Medicina, Instituto de Microbiología Clínica, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Maite Castro
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Casilla (P. O. Box) 567, Valdivia, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Centro Interdisciplinario de Neurociencias de Valparaíso (CINV), Valparaíso, Chile
| | - Angara Zambrano
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Casilla (P. O. Box) 567, Valdivia, Chile.
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile.
| |
Collapse
|
40
|
Hamaya S, Oura K, Morishita A, Masaki T. Cisplatin in Liver Cancer Therapy. Int J Mol Sci 2023; 24:10858. [PMID: 37446035 DOI: 10.3390/ijms241310858] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver tumor and is often diagnosed at an unresectable advanced stage. Systemic chemotherapy as well as transarterial chemoembolization (TACE) and hepatic arterial infusion chemotherapy (HAIC) are used to treat advanced HCC. TACE and HAIC have long been the standard of care for patients with unresectable HCC but are limited to the treatment of intrahepatic lesions. Systemic chemotherapy with doxorubicin or chemohormonal therapy with tamoxifen have also been considered, but neither has demonstrated survival benefits. In the treatment of unresectable advanced HCC, cisplatin is administered transhepatic arterially for local treatment. Subsequently, for cisplatin-refractory cases due to drug resistance, a shift to systemic therapy with a different mechanism of action is expected to produce new antitumor effects. Cisplatin is also used for the treatment of liver tumors other than HCC. This review summarizes the action and resistance mechanism of cisplatin and describes the treatment of the major hepatobiliary cancers for which cisplatin is used as an anticancer agent, with a focus on HCC.
Collapse
Affiliation(s)
- Sae Hamaya
- Department of Gastroenterology and Neurology, Kagawa University Faculty of Medicine, Kita-gun 761-0793, Japan
| | - Kyoko Oura
- Department of Gastroenterology and Neurology, Kagawa University Faculty of Medicine, Kita-gun 761-0793, Japan
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Kagawa University Faculty of Medicine, Kita-gun 761-0793, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Kagawa University Faculty of Medicine, Kita-gun 761-0793, Japan
| |
Collapse
|
41
|
Basirat U, Bin Tariq U, Moeen N, Jawhar ZH, Shoja SJ, Kareem AK, Ramírez-Coronel AA, Romero-Parra RM, Zabibah RS, Gupta J, Mustafa YF, Farhood B. A Systematic Review of the Chemo/Radioprotective Effects of Melatonin against Ototoxic Adverse Effects Induced by Chemotherapy and Radiotherapy. Curr Pharm Des 2023; 29:1218-1229. [PMID: 37138418 DOI: 10.2174/1381612829666230503145707] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 03/05/2023] [Accepted: 03/13/2023] [Indexed: 05/05/2023]
Abstract
BACKGROUND Although chemotherapy and radiotherapy are effective in cancer treatment, different adverse effects induced by these therapeutic modalities (such as ototoxicity) restrict their clinical use. Co-treatment of melatonin may alleviate the chemotherapy/radiotherapy-induced ototoxicity. OBJECTIVE In the present study, the otoprotective potentials of melatonin against the ototoxicity induced by chemotherapy and radiotherapy were reviewed. METHODS According to the PRISMA guideline, a systematic search was carried out to identify all relevant studies on "the role of melatonin against ototoxic damage associated with chemotherapy and radiotherapy" in the different electronic databases up to September 2022. Sixty-seven articles were screened based on a predefined set of inclusion and exclusion criteria. Seven eligible studies were finally included in this review. RESULTS The in vitro findings showed that cisplatin chemotherapy significantly decreased the auditory cell viability compared to the control group; in contrast, the melatonin co-administration increased the cell viability of cisplatin-treated cells. The results obtained from the distortion product otoacoustic emission (DPOAE) and auditory brainstem response (ABR) tests demonstrated a decreased amplitude of DPOAE and increased values of ABR I-IV interval and ABR threshold in mice/rats receiving radiotherapy and cisplatin; nevertheless, melatonin co-treatment indicated an opposite pattern on these evaluated parameters. It was also found that cisplatin and radiotherapy could significantly induce the histological and biochemical changes in the auditory cells/tissue. However, melatonin co-treatment resulted in alleviating the cisplatin/radiotherapy-induced biochemical and histological changes. CONCLUSION According to the findings, it was shown that melatonin co-treatment alleviates the ototoxic damage induced by chemotherapy and radiotherapy. Mechanically, melatonin may exert its otoprotective effects via its anti-oxidant, anti-apoptotic, and anti-inflammatory activities and other mechanisms.
Collapse
Affiliation(s)
| | | | - Nawal Moeen
- Nawaz Sharif Medical College, Gujrat, Pakistan
| | - Zanko Hassan Jawhar
- Department of Medical Laboratory Science, College of Health Sciences, Lebanese French University, Erbil, Kurdistan Region, Iraq
- Clinical Biochemistry Department, College of Health Sciences, Hawler Medical University, Erbil, Kurdistan Region, Iraq
| | - Sarah Jawad Shoja
- College of Health & Medical Technology, Al-Ayen University, Nasiriyah, Iraq
| | - Ali Kamil Kareem
- Biomedical Engineering Department, Al-Mustaqbal University College, Babylon, Iraq
| | | | | | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Pin Code 281406, U.P., India
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
42
|
Khodadadi F, Khorashadizadeh M, Ghasemi F. Thymoquinone enhanced the antitumor activity of cisplatin in human bladder cancer 5637 cells in vitro. Mol Biol Rep 2023:10.1007/s11033-023-08472-8. [PMID: 37219672 DOI: 10.1007/s11033-023-08472-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/18/2023] [Indexed: 05/24/2023]
Abstract
PURPOSE Cisplatin-based chemotherapy is a primary alternative for treating bladder cancer. But drug resistance and various side effects are the main unsightliness challenges. In search of a novel chemotherapeutic approach, this study was conducted to investigate whether thymoquinone (TQ) chemosensitize 5637 bladder cancer cells to cisplatin (CDDP). METHODS The IC50 for each drug was first determined. The cells were then pre-exposed to 40 µM of TQ for 24 h before being treated with 6 µM of cisplatin. The viability and the sub-G1 population of the 5673 cells were respectively evaluated by alamar blue assay and propidium iodide staining. RT-qPCR was also applied to analyze the expression profile of the apoptosis-related genes (Bax, Bcl-2, p53). RESULTS The viability of the cells treated with the combination of TQ and CDDP was significantly decreased compared to CDDP- or TQ-treated cells. TQ at the concentration of 40 µM increased the cytotoxicity of 6 µM CDDP by 35.5%. Moreover, flow cytometry analysis indicated that TQ pre-treatment of the cells resulted in a 55.5% increase in the population of 5637 cells in the sub-G1 phase compared to cells treated with CDDP alone. The results from RT-qPCR exhibited that the exposure of the cells to both TQ and CDDP significantly elevated Bax/Bcl-2 ratio by down-regulating Bcl-2 expression. CONCLUSION TQ significantly increased the cytotoxicity of CDDP in 5637 cells and induced apoptosis by down-regulation of the Bcl-2. Therefore, TQ and CDDP might be an effective therapeutic combination for TCC bladder cancer treatment.
Collapse
Affiliation(s)
- Fatemeh Khodadadi
- Department of Medical Biotechnology, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohsen Khorashadizadeh
- Department of Medical Biotechnology, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Fahimeh Ghasemi
- Department of Medical Biotechnology, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran.
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|
43
|
Jin X, He R, Liu J, Wang Y, Li Z, Jiang B, Lu J, Yang S. An herbal formulation "Shenshuaifu Granule" alleviates cisplatin-induced nephrotoxicity by suppressing inflammation and apoptosis through inhibition of the TLR4/MyD88/NF-κB pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 306:116168. [PMID: 36646160 DOI: 10.1016/j.jep.2023.116168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/23/2022] [Accepted: 01/10/2023] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shenshuaifu Granule (SSF) is an in-hospital preparation approved by the Guangdong Food and Drug Administration of China. It has been clinically used against kidney diseases for more than 20 years with a definite curative effect. AIM OF THE STUDY Cisplatin (CDDP) is a first-line chemotherapeutic drug in clinical practice, primarily excreted by the kidney with nephrotoxicity as a common side effect. Approximately 5-20% of cancer patients develop acute kidney injury (AKI) after chemotherapy; however, prevention and control strategies are currently unavailable. Therefore, it is important to identify safe and effective drugs that can prevent the nephrotoxicity of CDDP. SSF is an herbal formulation with 8 herbs, and has been used to protect the kidney in China. Nonetheless, its mechanism in relieving CDDP nephrotoxicity remains unclear. Therefore, this work attempt to prove that SSF can alleviate CDDP nephrotoxicity. We also explore its mechanism. MATERIALS AND METHODS First, Thin Layer Chromatography (TLC) of a few herbs in SSF were performed for quality control. Several open-access databases were used to identify the active ingredients of SSF, their corresponding targets, and CDDP-induced nephrotoxicity targets. We performed Protein-Protein Interaction (PPI), Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. Next, the results of network pharmacology were validated using CDDP-induced nephrotoxicity mouse models. Renal function in the mice was assessed by analyzing the levels of serum creatinine (Scr) and blood urea nitrogen (BUN). On the other hand, renal damage was assessed by determining the level of tubular injury and apoptotic cells using Periodic acid-Schiff (PAS) staining and Terminal Dutp Nick End-Labeling (TUNEL) staining, respectively. The expression of inflammatory and apoptotic-related targets including IL-1β, IL-6, TNF-α, Cox-2, Bax, Bcl-2, Cleaved-caspase 3, and Cleaved-caspase 9 was determined using Western Blot (WB) and Immunohistochemistry (IHC). Furthermore, WB was used to analyze the expression of proteins associated with the TLR4/MyD88/NF-κB pathway in the kidneys of mice with CDDP-induced nephrotoxicity. Finally, molecular docking simulations were performed to evaluate the binding abilities between major active ingredients of SSF and core targets. RESULT Through network pharmacology, we identified 127 active ingredients of SSF and their corresponding 134 targets. Additional screening identified 14 active ingredients and 17 targets for further analysis. In biological process (BP), the targets were enriched in inflammation and apoptosis, among others. In KEGG terms, they were enriched in apoptosis and NF-κB pathways. Animal experiments revealed that SSF significantly reduced the levels of Scr and BUN and prevented renal tubular damage in mice treated with CDDP. In addition, SSF inhibited inflammation and apoptosis by targeting the TLR4/MyD88/NF-κB pathway. Molecular docking revealed good binding capacities of active ingredients and core targets. CONCLUSION In summary, the experimental findings were consistent with the network pharmacological predictions. SSF can inhibit inflammation and apoptosis by targeting the TLR4/MyD88/NF-κB pathway. Taken together, our data suggest that SSF is an alternative agent for the treatment of CDDP-induced nephrotoxicity.
Collapse
Affiliation(s)
- Xiaoming Jin
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China
| | - Riming He
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China
| | - Jiahui Liu
- Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Shenzhen, Guangdong, 518033, China
| | - Yuzhi Wang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China
| | - Zhongtang Li
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China
| | - Beibei Jiang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China
| | - Jiandong Lu
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Nanjing University of Chinese Medicine, Shenzhen, 518033, China.
| | - Shudong Yang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518033, China.
| |
Collapse
|
44
|
Sanchez VA, Shuey MM, Dinh PC, Monahan PO, Fosså SD, Sesso HD, Dolan ME, Einhorn LH, Vaughn DJ, Martin NE, Feldman DR, Kroenke K, Fung C, Frisina RD, Travis LB. Patient-Reported Functional Impairment Due to Hearing Loss and Tinnitus After Cisplatin-Based Chemotherapy. J Clin Oncol 2023; 41:2211-2226. [PMID: 36626694 PMCID: PMC10489421 DOI: 10.1200/jco.22.01456] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/25/2022] [Accepted: 11/16/2022] [Indexed: 01/12/2023] Open
Abstract
PURPOSE Cisplatin is widely used and highly ototoxic, but patient-reported functional impairment because of cisplatin-related hearing loss (HL) and tinnitus has not been comprehensively evaluated. PATIENTS AND METHODS Testicular cancer survivors (TCS) given first-line cisplatin-based chemotherapy completed validated questionnaires, including the Hearing Handicap Inventory for Adults (HHIA) and Tinnitus Primary Function Questionnaire (TPFQ), each of which quantifies toxicity-specific functional impairment. Spearman correlations evaluated associations between HL and tinnitus severity and level of functional handicap quantified with the HHIA and TPFQ, respectively. Associations between HL or tinnitus and five prespecified adverse health outcomes (cognitive dysfunction, fatigue, depression, anxiety, and overall health) were evaluated. RESULTS HL and tinnitus affected 137 (56.4%) and 147 (60.5%) of 243 TCS, respectively. Hearing aids were used by 10% TCS (14/137). Of TCS with HL, 35.8% reported clinically significant functional impairment. Severe HHIA-assessed functional impairment was associated with cognitive dysfunction (odds ratio [OR], 10.62; P < .001), fatigue (OR, 5.48; P = .003), and worse overall health (OR, 0.19; P = .012). Significant relationships existed between HL severity and HHIA score, and tinnitus severity and TPFQ score (P < .0001 each). TCS with either greater hearing difficulty or more severe tinnitus were more likely to report cognitive dysfunction (OR, 5.52; P = .002; and OR, 2.56; P = .05), fatigue (OR, 6.18; P < .001; and OR, 4.04; P < .001), depression (OR, 3.93; P < .01; and OR, 3.83; P < .01), and lower overall health (OR, 0.39; P = .03; and OR, 0.46; P = .02, respectively). CONCLUSION One in three TCS with HL report clinically significant functional impairment. Follow-up of cisplatin-treated survivors should include routine assessment for HL and tinnitus. Use of the HHIA and TPFQ permit risk stratification and referral to audiologists as needed, since HL adversely affects functional status and is the single largest modifiable risk factor for cognitive decline and dementia in the general population.
Collapse
Affiliation(s)
| | - Megan M. Shuey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Paul C. Dinh
- Department of Medical Oncology, Indiana University, Indianapolis, IN
| | - Patrick O. Monahan
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, IN
| | | | - Howard D. Sesso
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA
| | | | | | - David J. Vaughn
- Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Neil E. Martin
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA
| | | | | | - Chunkit Fung
- J.P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY
| | | | - Lois B. Travis
- Department of Medical Oncology, Indiana University, Indianapolis, IN
| |
Collapse
|
45
|
Guseva Canu I, Bovio N, Arveux P, Bulliard JL, Fournier E, Germann S, Konzelmann I, Maspoli M, Rapiti E, Grzebyk M. Breast cancer and occupation: Non-parametric and parametric net survival analyses among Swiss women (1990-2014). Front Public Health 2023; 11:1129708. [PMID: 37089493 PMCID: PMC10115164 DOI: 10.3389/fpubh.2023.1129708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/14/2023] [Indexed: 04/08/2023] Open
Abstract
Introduction Occupation can contribute to differences in risk and stage at diagnosis of breast cancer. This study aimed at determining whether occupation, along with skill level and the socio-professional category, affect the breast cancer survival (BCS) up to 10 years after diagnosis. Materials and methods We used cancer registry records to identify women diagnosed with primary invasive breast cancer in western Switzerland over the period 1990-2014 and matched them with the Swiss National Cohort. The effect of work-related variables on BCS was assessed using non-parametric and parametric net survival methods. Results Study sample included 8,678 women. In the non-parametric analysis, we observed a statistically significant effect of all work-related variables on BCS. Women in elementary occupations, with low skill level, and in paid employment not classified elsewhere, had the lowest BCS, while professionals, those with the highest skill level and belonging to top management and independent profession category had the highest BCS. The parametric analysis confirmed this pattern. Considering elementary occupations as reference, all occupations but Craft and related trades had a hazard ratio (HR) below 1. Among professionals, technicians and associate professionals, and clerks, the protective effect of occupation was statistically significant and remained unchanged after adjustment for age, calendar period, registry, nationality, and histological type. After adjusting for tumor stage, the HRs increased only slightly, though turned non-significant. The same effect was observed in top management and independent professions and supervisors, low level management and skilled laborers, compared to unskilled employees. Conclusion These results suggest that work-related factors may affect BCS. Yet, this study was conducted using a limited set of covariates and a relatively small study sample. Therefore, further larger studies are needed for more detailed analyses of at risk occupations and working conditions and assessing the potential interaction between work-related variables and tumor stage.
Collapse
Affiliation(s)
- Irina Guseva Canu
- Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Nicolas Bovio
- Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Patrick Arveux
- Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Jean-Luc Bulliard
- Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
- Neuchâtel and Jura Cancer Registry, Neuchâtel, Switzerland
| | - Evelyne Fournier
- Geneva Cancer Registry, University of Geneva, Geneva, Switzerland
| | - Simon Germann
- Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | | | | | | | - Michel Grzebyk
- Department of Occupational Epidemiology, National Research and Safety Institute (INRS), Vandoeuvre lès Nancy, France
| |
Collapse
|
46
|
Li C, Wang X, Qiao X, Fan L, Zhu H, Chen Y, He Y, Zhang Z. 5,7-Dihydroxy-4-methylcoumarin modulates the JNK/FoxO1 signaling pathway to attenuate cisplatin-induced ototoxicity by suppressing oxidative stress and apoptosis in vitro. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119437. [PMID: 36754151 DOI: 10.1016/j.bbamcr.2023.119437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/12/2023] [Accepted: 01/29/2023] [Indexed: 02/08/2023]
Abstract
5,7-Dihydroxy-4-methylcoumarin (D4M) is attributed to free radical scavenging effects, with wide application for anti-oxidation. This work aimed to assess D4M's impact on cisplatin-induced ototoxicity. The cell viability was estimated with CCK-8 assay. Apoptosis was detected by the Annexin V-FITC and PI assay. The reactive oxygen species (ROS) level was determined by MitoSOX-Red and CellROX-Green probes. Mitochondrial membrane potential was analyzed with TMRM staining. Immunofluorescence was utilized for hair cells and spiral ganglion neuron detection. Apoptosis-associated proteins were assessed by cleaved caspase-3 and TUNEL staining. These results showed that D4M pretreatment protected hair cells from cisplatin-induced damage, increased cell viability, and decreased apoptosis in House Ear Institute-Organ of Corti1 (HEI-OC1) cells and neonatal mouse cochlear explants. D4M significantly inhibited cisplatin-induced mitochondrial apoptosis and reduced ROS accumulation. In addition, the protective effect of D4M on cisplatin-induced ototoxicity was also confirmed in cochlear hair cells and spiral ganglion neurons in neonatal mice. Mechanistic studies showed that D4M markedly downregulated p-JNK and elevated the expression ratio of p-FoxO1/FoxO1, thereby reducing cisplatin-induced caspase-dependent apoptosis. Meanwhile, D4M-related protection of HEI-OC1 cells was significantly blunted by JNK signaling induction with anisomycin. This study supports the possibility that D4M may be used as a new compound to prevent cisplatin-related hearing loss.
Collapse
Affiliation(s)
- Cai Li
- Department of Otolaryngology Head and Neck Surgery, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xue Wang
- Department of Otolaryngology Head and Neck Surgery, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xiangyun Qiao
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230031, China
| | - Li Fan
- Department of Otolaryngology Head and Neck Surgery, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Huanhuan Zhu
- Department of Otolaryngology Head and Neck Surgery, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Yutao Chen
- Department of Otolaryngology Head and Neck Surgery, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Yingzi He
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai 200031, China.
| | - Zhiyuan Zhang
- Department of Otolaryngology Head and Neck Surgery, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China.
| |
Collapse
|
47
|
Wang H, Lin H, Kang W, Huang L, Gong S, Zhang T, Huang X, He F, Ye Y, Tang Y, Jia H, Yang H. miR-34a/DRP-1-mediated mitophagy participated in cisplatin-induced ototoxicity via increasing oxidative stress. BMC Pharmacol Toxicol 2023; 24:16. [PMID: 36882858 PMCID: PMC9993635 DOI: 10.1186/s40360-023-00654-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 02/21/2023] [Indexed: 03/09/2023] Open
Abstract
PURPOSE Cisplatin is a widely used and effective chemotherapeutic agent for most solid malignant tumors. However, cisplatin-induced ototoxicity is a common adverse effect that limits the therapeutic efficacy of tumors in the clinic. To date, the specific mechanism of ototoxicity has not been fully elucidated, and the management of cisplatin-induced ototoxicity is also an urgent challenge. Recently, some authors believed that miR34a and mitophagy played a role in age-related and drug-induced hearing loss. Our study aimed to explore the involvement of miR-34a/DRP-1-mediated mitophagy in cisplatin-induced ototoxicity. METHODS In this study, C57BL/6 mice and HEI-OC1 cells were treated with cisplatin. MiR-34a and DRP-1 levels were analyzed by qRT‒PCR and western blotting, and mitochondrial function was assessed via oxidative stress, JC-1 and ATP content. Subsequently, we detected DRP-1 levels and observed mitochondrial function by modulating miR-34a expression in HEI-OC1 cells to determine the effect of miR-34a on DRP-1-mediated mitophagy. RESULTS MiR-34a expression increased and DRP-1 levels decreased in C57BL/6 mice and HEI-OC1 cells treated with cisplatin, and mitochondrial dysfunction was involved in this process. Furthermore, the miR-34a mimic decreased DRP-1 expression, enhanced cisplatin-induced ototoxicity and aggravated mitochondrial dysfunction. We further verified that the miR-34a inhibitor increased DRP-1 expression, partially protected against cisplatin-induced ototoxicity and improved mitochondrial function. CONCLUSION MiR-34a/DRP-1-mediated mitophagy was related to cisplatin-induced ototoxicity and might be a novel target for investigating the treatment and protection of cisplatin-induced ototoxicity.
Collapse
Affiliation(s)
- Haiyan Wang
- Department of Otolaryngology, the First Affiliated Hospital of Jinan University, No.613, West Huangpu Avenue, Guangzhou, 510630, China
| | - Hanqing Lin
- Department of Otolaryngology, the First Affiliated Hospital of Jinan University, No.613, West Huangpu Avenue, Guangzhou, 510630, China
| | - Weibiao Kang
- Department of Otolaryngology, the 2nd hospital, Medical College, Shantou University, Shantou, Guangdong, China
| | - Lingfei Huang
- Department of Otolaryngology, the First Affiliated Hospital of Jinan University, No.613, West Huangpu Avenue, Guangzhou, 510630, China
| | - Sisi Gong
- Department of Otolaryngology, the First Affiliated Hospital of Jinan University, No.613, West Huangpu Avenue, Guangzhou, 510630, China
| | - Tao Zhang
- Department of Otolaryngology, the First Affiliated Hospital of Jinan University, No.613, West Huangpu Avenue, Guangzhou, 510630, China
| | - Xiaotong Huang
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 West Yan Jiang Road, Guangzhou, 510120, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Feinan He
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 West Yan Jiang Road, Guangzhou, 510120, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yongyi Ye
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Department of Hearing and Speech Science, Xinhua College, Sun Yat-Sen University, Guangzhou, China
| | - Yiyang Tang
- Department of Otolaryngology, the First Affiliated Hospital of Jinan University, No.613, West Huangpu Avenue, Guangzhou, 510630, China
| | - Haiying Jia
- Department of Otolaryngology, the First Affiliated Hospital of Jinan University, No.613, West Huangpu Avenue, Guangzhou, 510630, China.
| | - Haidi Yang
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 West Yan Jiang Road, Guangzhou, 510120, China. .,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China. .,Department of Hearing and Speech Science, Xinhua College, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
48
|
Degranulation of Murine Resident Cochlear Mast Cells: A Possible Factor Contributing to Cisplatin-Induced Ototoxicity and Neurotoxicity. Int J Mol Sci 2023; 24:ijms24054620. [PMID: 36902051 PMCID: PMC10003316 DOI: 10.3390/ijms24054620] [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: 01/17/2023] [Revised: 02/17/2023] [Accepted: 02/22/2023] [Indexed: 03/04/2023] Open
Abstract
Permanent hearing loss is one of cisplatin's adverse effects, affecting 30-60% of cancer patients treated with that drug. Our research group recently identified resident mast cells in rodents' cochleae and observed that the number of mast cells changed upon adding cisplatin to cochlear explants. Here, we followed that observation and found that the murine cochlear mast cells degranulate in response to cisplatin and that the mast cell stabilizer cromoglicic acid (cromolyn) inhibits this process. Additionally, cromolyn significantly prevented cisplatin-induced loss of auditory hair cells and spiral ganglion neurons. Our study provides the first evidence for the possible mast cell participation in cisplatin-induced damage to the inner ear.
Collapse
|
49
|
Sanchez VA, Dinh PC, Rooker J, Monahan PO, Althouse SK, Fung C, Sesso HD, Einhorn LH, Dolan ME, Frisina RD, Travis LB. Prevalence and risk factors for ototoxicity after cisplatin-based chemotherapy. J Cancer Surviv 2023; 17:27-39. [PMID: 36637632 DOI: 10.1007/s11764-022-01313-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 12/07/2022] [Indexed: 01/14/2023]
Abstract
PURPOSE Ototoxicity is a prominent side effect of cisplatin-based chemotherapy. There are few reports, however, estimating its prevalence in well-defined cohorts and associated risk factors. METHODS Testicular cancer (TC) survivors given first-line cisplatin-based chemotherapy completed validated questionnaires. Descriptive statistics evaluated the prevalence of ototoxicity, defined as self-reported hearing loss and/or tinnitus. We compared patients with and without tinnitus or hearing loss using Chi-square test, two-sided Fisher's exact test, or two-sided Wilcoxon rank sum test. To evaluate ototoxicity risk factors, a backward selection logistic regression procedure was performed. RESULTS Of 145 TC survivors, 74% reported ototoxicity: 68% tinnitus; 59% hearing loss; and 52% reported both. TC survivors with tinnitus were more likely to indicate hypercholesterolemia (P = 0.008), and difficulty hearing (P < .001). Tinnitus was also significantly related to age at survey completion (OR = 1.79; P = 0.003) and cumulative cisplatin dose (OR = 5.17; P < 0.001). TC survivors with hearing loss were more likely to report diabetes (P = 0.042), hypertension (P = 0.007), hypercholesterolemia (P < 0.001), and family history of hearing loss (P = 0.044). Risk factors for hearing loss included age at survey completion (OR = 1.57; P = 0.036), hypercholesterolemia (OR = 3.45; P = 0.007), cumulative cisplatin dose (OR = 1.94; P = 0.049), and family history of hearing loss (OR = 2.87; P = 0.071). CONCLUSIONS Ototoxicity risk factors included age, cisplatin dose, cardiovascular risk factors, and family history of hearing loss. Three of four TC survivors report some type of ototoxicity; thus, follow-up of cisplatin-treated survivors should include routine assessment for ototoxicity with provision of indicated treatments. IMPLICATIONS FOR CANCER SURVIVORS Survivors should be aware of risk factors associated with ototoxicity. Referrals to audiologists before, during, and after cisplatin treatment is recommended.
Collapse
Affiliation(s)
- Victoria A Sanchez
- Department of Otolaryngology-Head & Neck Surgery, University of South Florida, 12901 Bruce B. Downs Blvd., MDC 73, Tampa, FL, 33612, USA.
| | - Paul C Dinh
- Department of Medical Oncology, Indiana University, Indianapolis, IN, USA
| | - Jennessa Rooker
- College of Nursing, University of South Florida, Tampa, FL, USA
| | - Patrick O Monahan
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, IN, USA
| | - Sandra K Althouse
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, IN, USA
| | - Chunkit Fung
- J.P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Howard D Sesso
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Lawrence H Einhorn
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, IN, USA
| | - M Eileen Dolan
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Robert D Frisina
- Department of Medical Engineering, University of South Florida, Tampa, FL, USA
| | - Lois B Travis
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, IN, USA
| |
Collapse
|
50
|
Ota S, Onimaru H, Izumizaki M. Effect of cisplatin on respiratory activity in neonatal rats. Pflugers Arch 2023; 475:233-248. [PMID: 36289078 DOI: 10.1007/s00424-022-02762-z] [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: 04/12/2022] [Revised: 09/07/2022] [Accepted: 10/10/2022] [Indexed: 02/01/2023]
Abstract
One side effect of cisplatin, a cytotoxic platinum anticancer drug, is peripheral neuropathy; however, its central nervous system effects remain unclear. We monitored respiratory nerve activity from the C4 ventral root in brainstem and spinal cord preparations from neonatal rats (P0-3) to investigate its central effects. Bath application of 10-100 μM cisplatin for 15-20 min dose-dependently decreased the respiratory rate and increased the amplitude of C4 inspiratory activity. These effects were not reversed after washout. In separate perfusion experiments, cisplatin application to the medulla decreased the respiratory rate, and application to the spinal cord increased the C4 burst amplitude without changing the burst rate. Application of other platinum drugs, carboplatin or oxaliplatin, induced no change of respiratory activity. A membrane potential analysis of respiratory-related neurons in the rostral medulla showed that firing frequencies of action potentials in the burst phase tended to decrease during cisplatin application. In contrast, in inspiratory spinal motor neurons, cisplatin application increased the peak firing frequency of action potentials during the inspiratory burst phase. The increased burst amplitude and decreased respiratory frequency were partially antagonized by riluzole and picrotoxin, respectively. Taken together, cisplatin inhibited respiratory rhythm via medullary inhibitory system activation and enhanced inspiratory motor nerve activity by changing the firing property of motor neurons.
Collapse
Affiliation(s)
- Shinichiro Ota
- Department of Physiology, Showa University School of Medicine, Tokyo, 142-8555, Japan
| | - Hiroshi Onimaru
- Department of Physiology, Showa University School of Medicine, Tokyo, 142-8555, Japan.
| | - Masahiko Izumizaki
- Department of Physiology, Showa University School of Medicine, Tokyo, 142-8555, Japan
| |
Collapse
|