1
|
Ding Y, Liu Y, Li D, Hu R, Tian Z, Yang L, Li Y, Lin Y, Qu Y. Melatonin Ameliorates Senescence of Mouse Auditory Cell Line HEI-OC1 Cells by Suppressing NLRP3 Inflammasome-Mediated Pyroptosis. Mol Neurobiol 2025:10.1007/s12035-025-04880-y. [PMID: 40169516 DOI: 10.1007/s12035-025-04880-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 03/21/2025] [Indexed: 04/03/2025]
Abstract
The aim of this study was to determine whether oxidative stress-induced premature senescence in mouse auditory cell line HEI-OC1 cells in vitro is associated with NLRP3 inflammasome activation and pyroptosis, and whether melatonin has a protective effect. HEI-OC1 cells were exposed to different concentrations of hydrogen peroxide (H2O2) to induce oxidative stress and subsequently analyzed by Western blotting to measure pyroptosis-related proteins - NLRP3, caspase-1, and GSDMD-N. Compared with untreated control cells, exposure to different concentrations of hydrogen peroxide (H2O2) promoted premature senescence of HEI-OC1 cells, accompanied by a significant increase in levels of pyroptosis-related proteins - NLRP3, caspase-1, and GSDMD-N. Furthermore, melatonin treatment was shown to decrease the expression of these proteins in HEI-OC1 cells and attenuate the H2O2-induced senescence process. NLRP3 inflammasome activation contributes to oxidative stress-induced premature senescence of HEI-OC1 cells in vitro, leading to pyroptosis. Melatonin attenuates pyroptosis and senescence in HEI-OC1 cells by inhibiting the expression of NLRP3, caspase-1, and GSDMD-N, providing reliable evidence for melatonin as a potential therapeutic agent for age-related hearing loss.
Collapse
Affiliation(s)
- Yongqing Ding
- Department of Otolaryngology, Qiaoxi District, The Third Hospital of Hebei Medical University, No. 13, Ziqiang Road, Shijiazhuang City, Hebei Province, China
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Yachao Liu
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Dong Li
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Ruili Hu
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Zedong Tian
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Lihang Yang
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Yanping Li
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Yantao Lin
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Yan Qu
- Department of Otolaryngology, Qiaoxi District, The Third Hospital of Hebei Medical University, No. 13, Ziqiang Road, Shijiazhuang City, Hebei Province, China.
| |
Collapse
|
2
|
Wang H, Fleishman JS, Wu S, Wang G, Du L, Li J, Du J. cGAS-STING targeting offers novel therapeutic opportunities in neurological diseases. Ageing Res Rev 2025; 105:102691. [PMID: 39954791 DOI: 10.1016/j.arr.2025.102691] [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: 10/08/2024] [Revised: 12/10/2024] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
Cyclic guanosine monophosphate (GMP)-adenosine monophosphate (AMP) (cGAMP) synthase (cGAS) is a cytosolic DNA sensor that produces the secondary messenger cGAMP. cGAMP activates the endoplasmic reticulum-associated adaptor stimulator of interferon genes (STING) and activates the innate immune system to produce a type I interferon response. Besides sensing microbial DNA, cGAS can also be activated by self-DNA or endogenous DNA, including that derived from genotoxic extranuclear chromatin and mitochondrially released DNA, indicating that cGAS-STING is an important mechanism in sterile inflammatory responses, autoimmunity, and cellular senescence. However, aberrant activation of the cGAS-STING pathway results in inflammatory and autoimmune diseases. cGAS-STING has emerged as a vital mechanism driving the pathogenesis of inflammation, implicating cGAS-STING signaling in neurological diseases. In this review, we first outline the principal elements of the cGAS-STING signaling cascade, summarizing recent research highlighting how cGAS-STING activation contributes to the pathogenesis of neurological diseases, including various autoimmune, autoinflammatory, and neurodegenerative diseases. Next, we outline selective small-molecule modulators that function as cGAS-STING inhibitors and summarize their mechanisms for treating multiple neurological diseases. Finally, we discuss key limitations of the current therapeutic paradigm and generate possible strategies to overcome them.
Collapse
Affiliation(s)
- Hongquan Wang
- Department of Geriatrics, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Shuang Wu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Guan Wang
- Aerospace Medical Center, Aerospace Center Hospital, Beijing 100049, China
| | - Lida Du
- Division of Neurobiology, Johns Hopkins University, Baltimore, MD 21205, USA.
| | - Jilai Li
- Aerospace Medical Center, Aerospace Center Hospital, Beijing 100049, China.
| | - Jichen Du
- Aerospace Medical Center, Aerospace Center Hospital, Beijing 100049, China; Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
3
|
Zhang C, Qiu Y, Yuan F. The long non-coding RNA maternally expressed 3-micorRNA-15a-5p axis is modulated by melatonin and prevents nucleus pulposus cell inflammation and apoptosis. Basic Clin Pharmacol Toxicol 2023; 133:603-619. [PMID: 37658573 DOI: 10.1111/bcpt.13939] [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: 02/20/2023] [Revised: 05/05/2023] [Accepted: 08/28/2023] [Indexed: 09/03/2023]
Abstract
Nucleus pulposus (NP) cell apoptosis is regarded as a critical risk factor for intervertebral disc degeneration (IVDD). Melatonin exerts a protective role on NP cells. The study concentrates on the role and mechanism of lncRNA MEG3 in melatonin-mediated effects on NP cells. An in vitro IVDD model was constructed using IL-1β on human NP cells. qRT-PCR investigated MEG3, miR-15a-5p and PGC-1α mRNA levels in tissues and NP cells. IL-1β-treated NP cells subsequent to transfection, followed by melatonin treatment. NP cell proliferation, viability, apoptosis and inflammatory reactions were assayed. Western blot checked the profiles of PGC-1α, SIRT1 and NF-κB p65. Student's t-test or one-way analysis of variance (ANOVA) followed by Tukey's test was used for statistical tests. As indicated by the data, melatonin weakened NP cell inflammation and apoptosis and enhanced MEG3 expression. MEG3 expression was attenuated in IVDD tissues. MEG3 knockdown impaired the function of melatonin, which was, however, strengthened by miR-15a-5p knockdown. MEG3 targeted miR-15a-5p, which targeted PGC-1α and repressed the PGC-1α/SIRT1 pathway. Collectively, this study has disclosed that the MEG3-miR-15a-5p-PGC-1α/SIRT1 pathway modulated by melatonin can hamper NP cell apoptosis and inflammation elicited by IL-1β.
Collapse
Affiliation(s)
- Chengyuan Zhang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongjia Qiu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Yuan
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Dou X, Ma Y, Luo Q, Song C, Liu M, Liu X, Jia D, Li S, Liu X. Therapeutic potential of melatonin in the intervertebral disc degeneration through inhibiting the ferroptosis of nucleus pulpous cells. J Cell Mol Med 2023; 27:2340-2353. [PMID: 37329158 PMCID: PMC10424295 DOI: 10.1111/jcmm.17818] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/28/2023] [Accepted: 06/12/2023] [Indexed: 06/18/2023] Open
Abstract
Ferroptosis, a novel type of cell death mediated by the iron-dependent lipid peroxidation, contributes to the pathogenesis of the intervertebral disc degeneration (IDD). Increasing evidence demonstrated that melatonin (MLT) displayed the therapeutic potential to prevent the development of IDD. Current mechanistic study aims to explore whether the downregulation of ferroptosis contributes to the therapeutic capability of MLT in IDD. Current studies demonstrated that conditioned medium (CM) from the lipopolysaccharide (LPS)-stimulated macrophages caused a series of changes about IDD, including increased intracellular oxidative stress (increased reactive oxygen species and malondialdehyde levels, but decreased glutathione levels), upregulated expression of inflammation-associated factors (IL-1β, COX-2 and iNOS), increased expression of key matrix catabolic molecules (MMP-13, ADAMTS4 and ADAMTS5), reduced the expression of major matrix anabolic molecules (COL2A1 and ACAN), and increased ferroptosis (downregulated GPX4 and SLC7A11 levels, but upregulated ACSL4 and LPCAT3 levels) in nucleus pulposus (NP) cells. MLT could alleviate CM-induced NP cell injury in a dose-dependent manner. Moreover, the data substantiated that intercellular iron overload was involved in CM-induced ferroptosis in NP cells, and MLT treatment alleviated intercellular iron overload and protected NP cells against ferroptosis, and those protective effects of MLT in NP cells further attenuated with erastin and enhanced with ferrostatin-1(Fer-1). This study demonstrated that CM from the LPS-stimulated RAW264.7 macrophages promoted the NP cell injury. MLT alleviated the CM-induced NP cell injury partly through inhibiting ferroptosis. The findings support the role of ferroptosis in the pathogenesis of IDD, and suggest that MLT may serve as a potential therapeutic approach for clinical treatment of IDD.
Collapse
Affiliation(s)
- Xinyu Dou
- Department of OrthopaedicsPeking University Third HospitalBeijingChina
- Beijing Key Laboratory of Spinal Disease ResearchBeijingChina
- Engineering Research Center of Bone and Joint Precision MedicineMinistry of EducationBeijingChina
| | - Yunlong Ma
- Pain Medical CenterPeking University Third HospitalBeijingChina
| | - Qipeng Luo
- Pain Medical CenterPeking University Third HospitalBeijingChina
| | - Chunyu Song
- Department of AnesthesiologyPeking University Third HospitalBeijingChina
| | - Meijuan Liu
- Department of Endocrinology, Genetics and MetabolismBeijing Children's HospitalBeijingChina
| | - Xiao Liu
- Department of OrthopaedicsPeking University Third HospitalBeijingChina
- Beijing Key Laboratory of Spinal Disease ResearchBeijingChina
- Engineering Research Center of Bone and Joint Precision MedicineMinistry of EducationBeijingChina
| | - Donglin Jia
- Pain Medical CenterPeking University Third HospitalBeijingChina
| | - Shuiqing Li
- Pain Medical CenterPeking University Third HospitalBeijingChina
| | - Xiaoguang Liu
- Department of OrthopaedicsPeking University Third HospitalBeijingChina
- Beijing Key Laboratory of Spinal Disease ResearchBeijingChina
- Engineering Research Center of Bone and Joint Precision MedicineMinistry of EducationBeijingChina
| |
Collapse
|
5
|
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
|
6
|
Zou T, Ye B, Chen K, Zhang A, Guo D, Pan Y, Ding R, Hu H, Sun X, Xiang M. Impacts of impaired mitochondrial dynamics in hearing loss: Potential therapeutic targets. Front Neurosci 2022; 16:998507. [PMID: 36278017 PMCID: PMC9579438 DOI: 10.3389/fnins.2022.998507] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Mitochondria are the powerhouse of the cells. Under physiological conditions, mitochondrial fission and fusion maintain a dynamic equilibrium in the cytoplasm, which is referred to as mitochondrial dynamics. As an important approach to regulating mitochondrial function and quantity, the role of mitochondrial dynamics has been demonstrated in the pathogenesis of various disease models, including brain damage, neurodegeneration, and stress. As the vital organ of the peripheral auditory system, the cochlea consumes a significant amount of energy, and the maintenance of mitochondrial homeostasis is essential for the cochlear auditory capacity. OPA1 functions as both a necessary gene regulating mitochondrial fusion and a pathogenic gene responsible for auditory neuropathy, suggesting that an imbalance in mitochondrial dynamics may play a critical role in hearing loss, but relevant studies are few. In this review, we summarize recent evidence regarding the role of mitochondrial dynamics in the pathogenesis of noise-induced hearing loss (NIHL), drug-induced hearing loss, hereditary hearing loss, and age-related hearing loss. The impacts of impaired mitochondrial dynamics on hearing loss are discussed, and the potential of mitochondrial dynamics for the prevention and treatment of hearing loss is considered.
Collapse
Affiliation(s)
- Tianyuan Zou
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Bin Ye
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Kaili Chen
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Andi Zhang
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Dongye Guo
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Yi Pan
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Rui Ding
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Haixia Hu
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Xingmei Sun
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
- *Correspondence: Xingmei Sun,
| | - Mingliang Xiang
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
- Mingliang Xiang,
| |
Collapse
|
7
|
Jing H, Sun X, Li M, Peng J, Gu X, Xiong J. Exogenous Melatonin Activating Nuclear Factor E2-Related Factor 2 (Nrf2) Pathway via Melatonin Receptor to Reduce Oxidative Stress and Apoptosis in Antler Mesenchymal Stem Cells. Molecules 2022; 27:molecules27082515. [PMID: 35458712 PMCID: PMC9029981 DOI: 10.3390/molecules27082515] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/09/2022] [Accepted: 04/11/2022] [Indexed: 11/16/2022] Open
Abstract
Antler growth depends on the proliferation and differentiation of mesenchymal stem cells (MSCs), and this process may be adversely affected by oxidative stress. Melatonin (MLT) has antioxidant functions, but its role in Cervidae remains largely unknown. In this article, flow cytometry, reactive oxygen species (ROS) identification, qPCR, and other methods were used to investigate the protective mechanism of MLT in H2O2-induced oxidative stress of antler MSCs. The results showed that MLT significantly increases cell viability by relieving the oxidative stress of antler MSCs. MLT inhibits cell apoptosis by protecting mitochondrial function. We blocked the melatonin receptor with luzindole (Luz) and found that the receptor blockade significantly increases H2O2-induced hyperoxide levels and causes significant inhibition of mitochondrial function. MLT treatment activates the nuclear factor E2-related factor 2 (Nrf2) antioxidant signaling pathway, up-regulates the expression of NAD(P)H quinone oxidoreductase 1 (NQO1) and other genes and it could inhibit apoptosis. In contrast, the melatonin receptor blockade down-regulates the expression of Nrf2 pathway-related genes, but significantly up-regulates the expression of apoptotic genes. It was indicated that MLT activates the Nrf2 pathway through the melatonin receptor and alleviates H2O2-induced oxidative stress and apoptosis in antler MSCs. This study provides a theoretical basis for further studying the oxidative stress and antioxidant process of antler MSCs and, thereby, increasing antler yields.
Collapse
|