1
|
Wu C, Sun X, Liu L, Cheng L. A Live-Cell Epigenome Manipulation by Photo-Stimuli-Responsive Histone Methyltransferase Inhibitor. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404608. [PMID: 39250325 PMCID: PMC11538670 DOI: 10.1002/advs.202404608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/23/2024] [Indexed: 09/11/2024]
Abstract
Post-translational modifications on the histone H3 tail regulate chromatin structure, impact epigenetics, and hence the gene expressions. Current chemical modulation tools, such as unnatural amino acid incorporation, protein splicing, and sortase-based editing, have allowed for the modification of histones with various PTMs in cellular contexts, but are not applicable for editing native chromatin. The use of small organic molecules to manipulate histone-modifying enzymes alters endogenous histone PTMs but lacks precise temporal and spatial control. To date, there has been no achievement in modulating histone methylation in living cells with spatiotemporal resolution. In this study, a new method is presented for temporally manipulating histone dimethylation H3K9me2 using a photo-responsive inhibitor that specifically targets the methyltransferase G9a on demand. The photo-caged molecule is stable under physiological conditions and cellular environments, but rapidly activated upon exposure to light, releasing the bioactive component that can immediately inhibit the catalytic ability of the G9a in vitro. Besides, this masked compound could also efficiently reactivate the inhibition of methyltransferase activity in living cells, subsequently suppress H3K9me2, a mark that regulates various chromatin functions. Therefore, the chemical system will be a valuable tool for manipulating the epigenome for therapeutic purposes and furthering the understanding of epigenetic mechanisms.
Collapse
Affiliation(s)
- Chuan‐Shuo Wu
- Beijing National Laboratory for Molecular Sciences (BNLMS)CAS Key Laboratory of Molecular Recognition and FunctionCAS Research/Education Center for Excellence in Molecular SciencesInstitute of ChemistryChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Xin Sun
- Beijing National Laboratory for Molecular Sciences (BNLMS)CAS Key Laboratory of Molecular Recognition and FunctionCAS Research/Education Center for Excellence in Molecular SciencesInstitute of ChemistryChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Li Liu
- Beijing National Laboratory for Molecular Sciences (BNLMS)CAS Key Laboratory of Molecular Recognition and FunctionCAS Research/Education Center for Excellence in Molecular SciencesInstitute of ChemistryChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Liang Cheng
- Beijing National Laboratory for Molecular Sciences (BNLMS)CAS Key Laboratory of Molecular Recognition and FunctionCAS Research/Education Center for Excellence in Molecular SciencesInstitute of ChemistryChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
- State Key Laboratory of Elemento‐Organic ChemistryNankai UniversityTianjin300071China
| |
Collapse
|
2
|
Yao Z, Xiao Y, Li W, Kong S, Tu H, Guo S, Liu Z, Ma L, Qiao R, Wang S, Chang M, Zhao X, Zhang Y, Xu L, Sun D, Fu X. FDA-Approved Tedizolid Phosphate Prevents Cisplatin-Induced Hearing Loss Without Decreasing Its Anti-tumor Effect. J Assoc Res Otolaryngol 2024; 25:259-275. [PMID: 38622383 DOI: 10.1007/s10162-024-00945-2] [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: 08/02/2023] [Accepted: 03/04/2024] [Indexed: 04/17/2024] Open
Abstract
PURPOSE Cisplatin is a low-cost clinical anti-tumor drug widely used to treat solid tumors. However, its use could damage cochlear hair cells, leading to irreversible hearing loss. Currently, there appears one drug approved in clinic only used for reducing ototoxicity associated with cisplatin in pediatric patients, which needs to further explore other candidate drugs. METHODS Here, by screening 1967 FDA-approved drugs to protect cochlear hair cell line (HEI-OC1) from cisplatin damage, we found that Tedizolid Phosphate (Ted), a drug indicated for the treatment of acute infections, had the best protective effect. Further, we evaluated the protective effect of Ted against ototoxicity in mouse cochlear explants, zebrafish, and adult mice. The mechanism of action of Ted was further explored using RNA sequencing analysis and verified. Meanwhile, we also observed the effect of Ted on the anti-tumor effect of cisplatin. RESULTS Ted had a strong protective effect on hair cell (HC) loss induced by cisplatin in zebrafish and mouse cochlear explants. In addition, when administered systemically, it protected mice from cisplatin-induced hearing loss. Moreover, antitumor studies showed that Ted had no effect on the antitumor activity of cisplatin both in vitro and in vivo. RNA sequencing analysis showed that the otoprotective effect of Ted was mainly achieved by inhibiting phosphorylation of ERK. Consistently, ERK activator aggravated the damage of cisplatin to HCs. CONCLUSION Collectively, these results showed that FDA-approved Ted protected HCs from cisplatin-induced HC loss by inhibiting ERK phosphorylation, indicating its potential as a candidate for preventing cisplatin ototoxicity in clinical settings.
Collapse
Affiliation(s)
- Zhiwei Yao
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China
| | - Yu Xiao
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China
- School of Life Science, Shandong University, Qingdao, 266237, China
| | - Wen Li
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China.
| | - Shuhui Kong
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250000, China
| | - Hailong Tu
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China
| | - Siwei Guo
- School of Life Science, Shandong University, Qingdao, 266237, China
| | - Ziyi Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China
| | - Lushun Ma
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China
| | - Ruifeng Qiao
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250000, China
| | - Song Wang
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Miao Chang
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China
| | - Xiaoxu Zhao
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yuan Zhang
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Lei Xu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250000, China.
| | - Daqing Sun
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Xiaolong Fu
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 250117, China.
| |
Collapse
|
3
|
Liu H, Kuang H, Wang Y, Bao L, Cao W, Yu L, Qi M, Wang R, Yang X, Ye Q, Ding F, Ren L, Liu S, Ma F, Liu S. MSC-derived exosomes protect auditory hair cells from neomycin-induced damage via autophagy regulation. Biol Res 2024; 57:3. [PMID: 38217055 PMCID: PMC10787390 DOI: 10.1186/s40659-023-00475-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 11/10/2023] [Indexed: 01/14/2024] Open
Abstract
BACKGROUND Sensorineural hearing loss (SNHL) poses a major threat to both physical and mental health; however, there is still a lack of effective drugs to treat the disease. Recently, novel biological therapies, such as mesenchymal stem cells (MSCs) and their products, namely, exosomes, are showing promising therapeutic potential due to their low immunogenicity, few ethical concerns, and easy accessibility. Nevertheless, the precise mechanisms underlying the therapeutic effects of MSC-derived exosomes remain unclear. RESULTS Exosomes derived from MSCs reduced hearing and hair cell loss caused by neomycin-induced damage in models in vivo and in vitro. In addition, MSC-derived exosomes modulated autophagy in hair cells to exert a protective effect. Mechanistically, exogenously administered exosomes were internalized by hair cells and subsequently upregulated endocytic gene expression and endosome formation, ultimately leading to autophagy activation. This increased autophagic activity promoted cell survival, decreased the mitochondrial oxidative stress level and the apoptosis rate in hair cells, and ameliorated neomycin-induced ototoxicity. CONCLUSIONS In summary, our findings reveal the otoprotective capacity of exogenous exosome-mediated autophagy activation in hair cells in an endocytosis-dependent manner, suggesting possibilities for deafness treatment.
Collapse
Affiliation(s)
- Huan Liu
- Department of Otolaryngology Head and Neck Surgery, Peking University Third Hospital, Beijing, China
| | - Huijuan Kuang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology,, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yiru Wang
- Anesthesiology Department, Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Lili Bao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology,, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wanxin Cao
- Department of Otolaryngology Head and Neck Surgery, Peking University Third Hospital, Beijing, China
| | - Lu Yu
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration and Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Meihao Qi
- Department of Otolaryngology-Head and Neck Surgery, Xijing Hospital, Air Force Military, Xi'an, Shaanxi, China
| | - Renfeng Wang
- Department of Otolaryngology-Head and Neck Surgery, Xijing Hospital, Air Force Military, Xi'an, Shaanxi, China
| | - Xiaoshan Yang
- School of Stomatology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Qingyuan Ye
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Digital Dentistry Center, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Feng Ding
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology,, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lili Ren
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology,, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Siying Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology,, The Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Furong Ma
- Department of Otolaryngology Head and Neck Surgery, Peking University Third Hospital, Beijing, China.
| | - Shiyu Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology,, The Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
4
|
Zhou X, Jin L, Li Y, Wang Y, Li W, Shen X. Comprehensive analysis of N6-methyladenosine-related RNA methylation in the mouse hippocampus after acquired hearing loss. BMC Genomics 2023; 24:577. [PMID: 37759187 PMCID: PMC10537436 DOI: 10.1186/s12864-023-09697-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/22/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND The mechanism underlying cognitive impairment after hearing loss (HL) remains unclear. N6-methyladenosine (m6A) is involved in many neurodegenerative diseases; however, its role in cognitive impairment after HL has not yet been investigated. Therefore, we aimed to analyze the m6A modification profile of the mouse hippocampus after HL exposure. A mouse model of neomycin-induced HL was established. An auditory brainstem-response test was utilized for detecting hearing threshold. The passive avoidance test was served as the mean for evaluating cognitive function. The m6A-regulated enzyme expression levels were analyzed by using reverse transcription quantitative real-time polymerase chain reaction and western blot analyses. RNA sequencing (RNA-Seq) and methylated RNA immunoprecipitation sequencing (MeRIP-Seq) were performed with the aim of investigating gene expression differences and m6A modification in the mouse hippocampus. RESULTS Neomycin administration induced severe HL in mice. At four months of age, the mice in the HL group showed poorer cognitive performance than the mice in the control group. METTL14, WTAP, and YTHDF2 mRNA levels were downregulated in the hippocampi of HL mice, whereas ALKBH5 and FTO mRNA levels were significantly upregulated. At the protein level, METTL3 and FTO were significantly upregulated. Methylated RNA immunoprecipitation sequencing analysis revealed 387 and 361 m6A hypermethylation and hypomethylation peaks, respectively. Moreover, combined analysis of mRNA expression levels and m6A peaks revealed eight mRNAs with significantly changed expression levels and methylation. CONCLUSIONS Our findings revealed the m6A transcriptome-wide profile in the hippocampus of HL mice, which may provide a basis for understanding the association between HL and cognitive impairment from the perspective of epigenetic modifications.
Collapse
Affiliation(s)
- Xuehua Zhou
- Department of Anesthesiology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, 200031, Shanghai, China
| | - Lin Jin
- Department of Anesthesiology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, 200031, Shanghai, China
| | - Yufeng Li
- Department of Anesthesiology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, 200031, Shanghai, China
| | - Yiru Wang
- Department of Anesthesiology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, 200031, Shanghai, China
| | - Wen Li
- ENT Institute, Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, 200031, Shanghai, China
| | - Xia Shen
- Department of Anesthesiology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, 200031, Shanghai, China.
| |
Collapse
|
5
|
Xiao Y, Li D. The role of epigenetic modifications in sensory hair cell development, survival, and regulation. Front Cell Neurosci 2023; 17:1210279. [PMID: 37388412 PMCID: PMC10300351 DOI: 10.3389/fncel.2023.1210279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 05/23/2023] [Indexed: 07/01/2023] Open
Abstract
The cochlea is the sensory organ in the periphery, and hair cells are its main sensory cells. The development and survival of hair cells are highly controlled processes. When cells face intracellular and environmental stimuli, epigenetic regulation controls the structure and function of the genome in response to different cell fates. During sensory hair cell development, different histone modifications can induce normal numbers of functional hair cells to generate. When individuals are exposed to environmental-related hair cell damage, epigenetic modification also plays a significant role in the regulation of hair cell fate. Since mammalian hair cells cannot regenerate, their loss can cause permanent sensorineural hearing loss. Many breakthroughs have been achieved in recent years in understanding the signaling pathways that determine hair cell regeneration, and it is fascinating to note that epigenetic regulation plays a significant role in hair cell regeneration. In this review, we discuss the role of epigenetics in inner ear cell development, survival and regeneration and the significant impact on hearing protection.
Collapse
|
6
|
Zheng Z, Nan B, Liu C, Tang D, Li W, Zhao L, Nie G, He Y. Inhibition of histone methyltransferase PRMT5 attenuates cisplatin-induced hearing loss through the PI3K/Akt-mediated mitochondrial apoptotic pathway. J Pharm Anal 2023; 13:590-602. [PMID: 37440906 PMCID: PMC10334280 DOI: 10.1016/j.jpha.2023.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 07/15/2023] Open
Abstract
This study aimed to evaluate the therapeutic potential of inhibiting protein arginine methyltransferase 5 (PRMT5) in cisplatin-induced hearing loss. The effects of PRMT5 inhibition on cisplatin-induced auditory injury were determined using immunohistochemistry, apoptosis assays, and auditory brainstem response. The mechanism of PRMT5 inhibition on hair cell survival was assessed using RNA-seq and Cleavage Under Targets and Tagment-quantitative polymerase chain reaction (CUT&Tag-qPCR) analyses in the HEI-OC1 cell line. Pharmacological inhibition of PRMT5 significantly alleviated cisplatin-induced damage to hair cells and spiral ganglion neurons in the cochlea and decreased apoptosis by protecting mitochondrial function and preventing the accumulation of reactive oxygen species. CUT&Tag-qPCR analysis demonstrated that inhibition of PRMT5 in HEI-OC1 cells reduced the accumulation of H4R3me2s/H3R8me2s marks at the promoter region of the Pik3ca gene, thus activating the expression of Pik3ca. These findings suggest that PRMT5 inhibitors have strong potential as agents against cisplatin-induced ototoxicity and can lay the foundation for further research on treatment strategies of hearing loss.
Collapse
Affiliation(s)
- Zhiwei Zheng
- 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 Medicine (Fudan University), Fudan University, Shanghai, 200031, China
| | - Benyu Nan
- Department of Otorhinolaryngology-Head and Neck Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Chang Liu
- Department of Otolaryngology-Head and Neck Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - 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 Medicine (Fudan University), Fudan University, Shanghai, 200031, China
| | - Wen 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 Medicine (Fudan University), Fudan University, Shanghai, 200031, China
| | - Liping Zhao
- 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 Medicine (Fudan University), Fudan University, Shanghai, 200031, China
| | - Guohui Nie
- Department of Otolaryngology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, Guangdong, 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, NHC Key Laboratory of Hearing Medicine (Fudan University), Fudan University, Shanghai, 200031, China
| |
Collapse
|
7
|
Tang D, Lu Y, Zuo N, Yan R, Wu C, Wu L, Liu S, He Y. The H3K27 demethylase controls the lateral line embryogenesis of zebrafish. Cell Biol Toxicol 2023; 39:1137-1152. [PMID: 34716527 PMCID: PMC10406677 DOI: 10.1007/s10565-021-09669-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 10/11/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Kdm6b, a specific histone 3 lysine 27 (H3K27) demethylase, has been reported to be implicated in a variety of developmental processes including cell differentiation and cell fate determination and multiple organogenesis. Here, we regulated the transcript level of kdm6bb to study the potential role in controlling the hearing organ development of zebrafish. METHODS A morpholino antisense oligonucleotide (MO) strategy was used to induce Kdm6b deficiency; immunohistochemical staining and in situ hybridization analysis were conducted to figure out the morphologic alterations and embryonic mechanisms. RESULTS Kdm6bb is expressed in the primordium and neuromasts at the early stage of zebrafish embryogenesis, suggesting a potential function of Kdm6b in the development of mechanosensory organs. Knockdown of kdm6bb severely influences the cell migration and proliferation in posterior lateral line primordium, abates the number of neuromasts along the trunk, and mRNA-mediated rescue test can partially renew the neuromasts. Loss of kdm6bb might be related to aberrant expressions of chemokine genes encompassing cxcl12a and cxcr4b/cxcr7b in the migrating primordium. Moreover, inhibition of kdm6bb reduces the expression of genes in Fgf signaling pathway, while it increases the axin2 and lef1 expression level of Wnt/β-catenin signaling during the migrating stage. CONCLUSIONS Collectively, our results revealed that Kdm6b plays an essential role in guiding the migration of primordium and in regulating the deposition of zebrafish neuromasts by mediating the gene expression of chemokines and Wnt and Fgf signaling pathway. Since histone methylation and demethylation are reversible, targeting Kdm6b may present as a novel therapeutic regimen for hearing disorders.
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 Medicine, Fudan University, 83 Fenyang Road, Shanghai, 200031, China
| | - Yitong Lu
- Department of Otolaryngology-Head and Neck Surgery, Yijishan Hospital of Wannan Medical College, 2 Zheshanwest Road, Wuhu, 241001, Anhui, China
| | - Na Zuo
- Department of Otolaryngology-Head and Neck Surgery, Yijishan Hospital of Wannan Medical College, 2 Zheshanwest Road, Wuhu, 241001, Anhui, China
| | - Renchun Yan
- Department of Otolaryngology-Head and Neck Surgery, Yijishan Hospital of Wannan Medical College, 2 Zheshanwest Road, Wuhu, 241001, Anhui, China
| | - Cheng Wu
- Department of Otolaryngology-Head and Neck Surgery, Yijishan Hospital of Wannan Medical College, 2 Zheshanwest Road, Wuhu, 241001, Anhui, China
| | - Lijuan Wu
- Department of Otolaryngology-Head and Neck Surgery, Yijishan Hospital of Wannan Medical College, 2 Zheshanwest Road, Wuhu, 241001, Anhui, China
| | - Shaofeng Liu
- Department of Otolaryngology-Head and Neck Surgery, Yijishan Hospital of Wannan Medical College, 2 Zheshanwest Road, Wuhu, 241001, Anhui, 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, NHC Key Laboratory of Hearing Medicine, Fudan University, 83 Fenyang Road, Shanghai, 200031, China.
| |
Collapse
|
8
|
Mu YR, Zou SY, Li M, Ding YY, Huang X, He ZH, Kong WJ. Role and mechanism of FOXG1-related epigenetic modifications in cisplatin-induced hair cell damage. Front Mol Neurosci 2023; 16:1064579. [PMID: 37181652 PMCID: PMC10169754 DOI: 10.3389/fnmol.2023.1064579] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 04/11/2023] [Indexed: 05/16/2023] Open
Abstract
Cisplatin is widely used in clinical tumor chemotherapy but has severe ototoxic side effects, including tinnitus and hearing damage. This study aimed to determine the molecular mechanism underlying cisplatin-induced ototoxicity. In this study, we used CBA/CaJ mice to establish an ototoxicity model of cisplatin-induced hair cell loss, and our results showed that cisplatin treatment could reduce FOXG1 expression and autophagy levels. Additionally, H3K9me2 levels increased in cochlear hair cells after cisplatin administration. Reduced FOXG1 expression caused decreased microRNA (miRNA) expression and autophagy levels, leading to reactive oxygen species (ROS) accumulation and cochlear hair cell death. Inhibiting miRNA expression decreased the autophagy levels of OC-1 cells and significantly increased cellular ROS levels and the apoptosis ratio in vitro. In vitro, overexpression of FOXG1 and its target miRNAs could rescue the cisplatin-induced decrease in autophagy, thereby reducing apoptosis. BIX01294 is an inhibitor of G9a, the enzyme in charge of H3K9me2, and can reduce hair cell damage and rescue the hearing loss caused by cisplatin in vivo. This study demonstrates that FOXG1-related epigenetics plays a role in cisplatin-induced ototoxicity through the autophagy pathway, providing new ideas and intervention targets for treating ototoxicity.
Collapse
Affiliation(s)
- Yu-rong Mu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng-yu Zou
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ming Li
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan-yan Ding
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Huang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zu-hong He
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wei-jia Kong
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Bazard P, Pineros J, Acosta AA, Thivierge M, Paganella LR, Zucker S, Mannering FL, Modukuri S, Zhu X, Frisina RD, Ding B. Post-Translational Modifications and Age-related Hearing Loss. Hear Res 2022; 426:108625. [DOI: 10.1016/j.heares.2022.108625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 08/21/2022] [Accepted: 09/23/2022] [Indexed: 11/04/2022]
|
10
|
Shen J, Jiao Y, Ding N, Xie L, Ma S, Zhang H, Yang A, Zhang H, Jiang Y. Homocysteine facilitates endoplasmic reticulum stress and apoptosis of hepatocytes by suppressing
ERO1α
expression via cooperation between DNMT1 and G9a. Cell Biol Int 2022; 46:1236-1248. [PMID: 35347798 PMCID: PMC9543485 DOI: 10.1002/cbin.11805] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 03/16/2022] [Accepted: 03/27/2022] [Indexed: 11/27/2022]
Abstract
Endoplasmic reticulum (ER) stress and apoptosis play a critical role in liver injury. Endoplasmic reticulum oxidoreductase 1α (ERO1α) is an oxidase that exists in the luminal side of the ER membrane, participating in protein folding and secretion and inhibiting apoptosis, but the underlying mechanism on liver injury induced by homocysteine (Hcy) remains obscure. In this study, hyperhomocysteinemia (HHcy) mice model was established in cbs+/− mice by feeding a high‐methionine diet for 12 weeks; and cbs+/− mice fed with high‐methionine diet exhibited more severe liver injury compared to cbs+/+ mice. Mechanistically, we found that Hcy promoted ER stress and apoptosis of hepatocytes and thereby aggravated liver injury through inhibiting ERO1α expression; accordingly, overexpression of ERO1α remarkably alleviated ER stress and apoptosis of hepatocytes induced by Hcy. Epigenetic modification analysis revealed that Hcy significantly increased levels of DNA methylation and H3 lysine 9 dimethylation (H3K9me2) on ERO1α promoter, which attributed to upregulated DNA methyltransferase 1 (DNMT1) and G9a, respectively. Further study showed that DNMT1 and G9a cooperatively regulated ERO1α expression in hepatocytes exposed to Hcy. Taken together, our work demonstrates that Hcy activates ER stress and apoptosis of hepatocytes by downregulating ERO1α expression via cooperation between DNMT1 and G9a, which provides new insight into the mechanism of Hcy‐induced ER stress and apoptosis of hepatocytes in liver injury.
Collapse
Affiliation(s)
- Jiangyong Shen
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical UniversityYinchuan750004China
- Department of Clinical Medicine, General Hospital of Ningxia Medical UniversityYinchuan750004China
| | - Yun Jiao
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical UniversityYinchuan750004China
- Department of Infectious diseases, General Hospital of Ningxia Medical UniversityYinchuan750004China
| | - Ning Ding
- School of Basic Medical SciencesNingxia Medical UniversityYinchuan750004China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical UniversityYinchuan750004China
| | - Lin Xie
- School of Basic Medical SciencesNingxia Medical UniversityYinchuan750004China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical UniversityYinchuan750004China
| | - Shengchao Ma
- School of Basic Medical SciencesNingxia Medical UniversityYinchuan750004China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical UniversityYinchuan750004China
| | - Hui Zhang
- School of Basic Medical SciencesNingxia Medical UniversityYinchuan750004China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical UniversityYinchuan750004China
| | - Anning Yang
- School of Basic Medical SciencesNingxia Medical UniversityYinchuan750004China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical UniversityYinchuan750004China
| | - Huiping Zhang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical UniversityYinchuan750004China
- Department of Prenatal Diagnosis Center, General Hospital of Ningxia Medical UniversityYinchuan750004China
| | - Yideng Jiang
- School of Basic Medical SciencesNingxia Medical UniversityYinchuan750004China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical UniversityYinchuan750004China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical UniversityYinchuan750004China
| |
Collapse
|
11
|
He Y, Zheng Z, Liu C, Li W, Zhao L, Nie G, Li H. Inhibiting DNA methylation alleviates cisplatin-induced hearing loss by decreasing oxidative stress-induced mitochondria-dependent apoptosis via the LRP1-PI3K/AKT pathway. Acta Pharm Sin B 2022; 12:1305-1321. [PMID: 35530135 PMCID: PMC9069410 DOI: 10.1016/j.apsb.2021.11.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 12/15/2022] Open
Abstract
Cisplatin-related ototoxicity is a critical side effect of chemotherapy and can lead to irreversible hearing loss. This study aimed to assess the potential effect of the DNA methyltransferase (DNMT) inhibitor RG108 on cisplatin-induced ototoxicity. Immunohistochemistry, apoptosis assay, and auditory brainstem response (ABR) were employed to determine the impacts of RG108 on cisplatin-induced injury in murine hair cells (HCs) and spiral ganglion neurons (SGNs). Rhodamine 123 and TMRM were utilized for mitochondrial membrane potential (MMP) assessment. Reactive oxygen species (ROS) amounts were evaluated by Cellrox green and Mitosox-red probes. Mitochondrial respiratory function evaluation was performed by determining oxygen consumption rates (OCRs). The results showed that RG108 can markedly reduce cisplatin induced damage in HCs and SGNs, and alleviate apoptotic rate by protecting mitochondrial function through preventing ROS accumulation. Furthermore, RG108 upregulated BCL-2 and downregulated APAF1, BAX, and BAD in HEI-OC1 cells, and triggered the PI3K/AKT pathway. Decreased expression of low-density lipoprotein receptor-related protein 1 (LRP1) and high methylation of the LRP1 promoter were observed after cisplatin treatment. RG108 treatment can increase LRP1 expression and decrease LRP1 promoter methylation. In conclusion, RG108 might represent a new potential agent for preventing hearing loss induced by cisplatin via activating the LRP1-PI3K/AKT pathway.
Collapse
Key Words
- 5-mC, 5-methylcytosine
- ABR, auditory brainstem response
- Apoptosis
- Cisplatin
- DNMT
- DNMT, DNA methyltransferase
- EdU, 5-ethynyl-2′-deoxyuridine
- HCs, hair cells
- Hair cell
- IHCs, inner hair cells
- LRP1, low-density lipoprotein receptor-related protein 1
- MMP, mitochondrial membrane potential
- Mitochondrial dysfunction
- OCRs, oxygen consumption rates
- OHCs, outer hair cells
- PI, propidium iodide
- RG108
- ROS
- ROS, reactive oxygen species
- SGNs, spiral ganglion neurons
- Spiral ganglion neurons
- TUNEL, terminal deoxynucleotidyl transferase-mediated dUTP nick-end-labeling
Collapse
|
12
|
Li X, Chen X. Inhibition of PRMT6 reduces neomycin-induced inner ear hair cell injury through the restraint of FoxG1 arginine methylation. Inflamm Res 2022; 71:309-320. [PMID: 35190853 DOI: 10.1007/s00011-022-01541-x] [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: 10/11/2021] [Revised: 01/07/2022] [Accepted: 01/14/2022] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVE Hair cells in the inner ear have been demonstrated to be sensitive to the ototoxicity from some beneficial pharmaceutical drugs. This study aimed to explore the role of protein arginine methyltransferase 6 (PRMT6) in the process of neomycin-induced hearing loss and the underlying mechanism. METHODS The neomycin-induced hearing loss mouse model and hair cell injury in vitro model were established. We took advantage of the HEI-OC1 cell line to evaluate PRMT6 expression in neomycin-induced hair cells, and the effect of PRMT6 on mitochondrial function and FoxG1 arginine methylation. Apoptotic cells were assessed and apoptotic marker cleaved caspase-3 level was detected. Reactive oxygen species (ROS) level and mitochondrial membrane potential (MMP) were subsequently measured. RESULT The result showed that PRMT6 was significantly upregulated in neomycin-induced HEI-OC-1 cells, and PRMT6 silencing prevented MMP loss, reduced ROS production, as well as decreased cell apoptosis under neomycin treatment. Further results showed that FoxG1 was downregulated in neomycin-induced HEI-OC-1 cells, and PRMT6 promoted the FoxG1-mediated luciferase activity, while PRMT6 silencing reversed this effect. Mechanistic experiments revealed that PRMT6 silencing reduced the arginine methylation level of FoxG1 protein. In vivo, neomycin-induced upregulation of hearing thresholds and increased cell apoptosis, whereas PRMT6 inhibitor partly reversed these effects. CONCLUSION Our findings suggested that inhibition of PRMT6 reduced neomycin-induced inner ear hair cell injury through the restraint of FoxG1 arginine methylation.
Collapse
Affiliation(s)
- Xingcheng Li
- Department of Otology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Rd., Zhengzhou, 450052, People's Republic of China.
| | - Xi Chen
- Department of Rhinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| |
Collapse
|
13
|
Liu C, Zheng Z, Li W, Tang D, Zhao L, He Y, Li H. Inhibition of KDM5A attenuates cisplatin-induced hearing loss via regulation of the MAPK/AKT pathway. Cell Mol Life Sci 2022; 79:596. [PMID: 36396833 PMCID: PMC9672031 DOI: 10.1007/s00018-022-04565-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 09/17/2022] [Accepted: 09/19/2022] [Indexed: 11/19/2022]
Abstract
The study aimed to investigate the potential role of lysine-specific demethylase 5A (KDM5A) in cisplatin-induced ototoxicity. The effect of the KDM5A inhibitor CPI-455 was assessed by apoptosis assay, immunofluorescence, flow cytometry, seahorse respirometry assay, and auditory brainstem response test. RNA sequencing, qRT-PCR, and CUT&Tag assays were used to explore the mechanism underlying CPI-455-induced protection. Our results demonstrated that the expression of KDM5A was increased in cisplatin-injured cochlear hair cells compared with controls. CPI-455 treatment markedly declined KDM5A and elevated H3K4 trimethylation levels in cisplatin-injured cochlear hair cells. Moreover, CPI-455 effectively prevented the death of hair cells and spiral ganglion neurons and increased the number of ribbon synapses in a cisplatin-induced ototoxicity mouse model both in vitro and in vivo. In HEI-OC1 cells, KDM5A knockdown reduced reactive oxygen species accumulation and improved mitochondrial membrane potential and oxidative phosphorylation under cisplatin-induced stress. Mechanistically, through transcriptomics and epigenomics analyses, a set of apoptosis-related genes, including Sos1, Sos2, and Map3k3, were regulated by CPI-455. Altogether, our findings indicate that inhibition of KDM5A may represent an effective epigenetic therapeutic target for preventing cisplatin-induced hearing loss.
Collapse
Affiliation(s)
- Chang Liu
- Department of ENT Institute and Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 83 Fenyang Road, Shanghai, 200031 China ,NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031 People’s Republic of China
| | - Zhiwei Zheng
- Department of ENT Institute and Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 83 Fenyang Road, Shanghai, 200031 China ,NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031 People’s Republic of China
| | - Wen Li
- Department of ENT Institute and Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 83 Fenyang Road, Shanghai, 200031 China ,NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031 People’s Republic of China
| | - Dongmei Tang
- Department of ENT Institute and Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 83 Fenyang Road, Shanghai, 200031 China ,NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031 People’s Republic of China
| | - Liping Zhao
- Department of ENT Institute and Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 83 Fenyang Road, Shanghai, 200031 China ,NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031 People’s Republic of China
| | - Yingzi He
- Department of ENT Institute and Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 83 Fenyang Road, Shanghai, 200031 China ,NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031 People’s Republic of China
| | - Huawei Li
- Department of ENT Institute and Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 83 Fenyang Road, Shanghai, 200031 China ,NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031 People’s Republic of China ,Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032 People’s Republic of China ,The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032 People’s Republic of China
| |
Collapse
|
14
|
Liu H, Wang W, Weng X, Chen H, Chen Z, Du Y, Liu X, Wang L. The H3K9 histone methyltransferase G9a modulates renal ischemia reperfusion injury by targeting Sirt1. Free Radic Biol Med 2021; 172:123-135. [PMID: 34102281 DOI: 10.1016/j.freeradbiomed.2021.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/27/2021] [Accepted: 06/02/2021] [Indexed: 12/30/2022]
Abstract
Ischemia reperfusion (IR) injury dampens renal function and usually confers a great risk of renal failure. Aberrant expression of G9a, a H3K9 methyltransferase of mammalian histone, has been implicated as a driving event in various kidney diseases. However, the role of G9a plays in renal IR injury is required to be clarified. Herein, our results showed that renal IR injury resulted in a rapid elevation of G9a, accompanying the down-regulation of Sirt1, a deacetylase that has been reported to afford renoprotection. Genetic overexpression or therapeutic activation of Sirt1 efficiently ameliorated renal IR injury by elevating anti-oxidative genes expression and reducing the accumulation of reactive oxygen species, including O2·- and ·OH. In addition, inhibition of G9a activity by BIX01294 (BIX) alleviated IR injury through abolishing O2·- and ·OH levels in a Sirt1-dependent manner. Mechanistically, we observed that demethylated H3K9 was accumulated on the Sirt1 promoter in renal IR injury. Silencing or suppression of G9a activity erased H3K9me2 from Sirt1 promoter and normalized Sirt1 expression. Further exploration revealed that G9a interacted with chromobox homolog 1 (CBX1) to catalyze H3K9 de-methylation and formed a transcription repressor complex on the Sirt1 promoter, ultimately repressing Sirt1 transcription. In this study, we provided strong evidence that G9a modulated renal IR injury through cooperation with CBX1 to form a transcription repressor complex on the Sirt1 promoter and regulate O2·- and ·OH generation, indicating that G9a-Sirt1 axis might be a promising therapeutic target in an epigenetic manner.
Collapse
Affiliation(s)
- Hao Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Wei Wang
- Department of Urology and Institute of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, 230022, Anhui, China
| | - Xiaodong Weng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Hui Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Yang Du
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
15
|
Zheng Z, Zeng S, Liu C, Li W, Zhao L, Cai C, Nie G, He Y. The DNA methylation inhibitor RG108 protects against noise-induced hearing loss. Cell Biol Toxicol 2021; 37:751-771. [PMID: 33723744 PMCID: PMC8490244 DOI: 10.1007/s10565-021-09596-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 02/22/2021] [Indexed: 11/29/2022]
Abstract
Background Noise-induced hearing loss represents a commonly diagnosed type of hearing disability, severely impacting the quality of life of individuals. The current work is aimed at assessing the effects of DNA methylation on noise-induced hearing loss. Methods Blocking DNA methyltransferase 1 (DNMT1) activity with a selective inhibitor RG108 or silencing DNMT1 with siRNA was used in this study. Auditory brainstem responses were measured at baseline and 2 days after trauma in mice to assess auditory functions. Whole-mount immunofluorescent staining and confocal microcopy of mouse inner ear specimens were performed to analyze noise-induced damage in cochleae and the auditory nerve at 2 days after noise exposure. Results The results showed that noise exposure caused threshold elevation of auditory brainstem responses and cochlear hair cell loss. Whole-mount cochlea staining revealed a reduction in the density of auditory ribbon synapses between inner hair cells and spiral ganglion neurons. Inhibition of DNA methyltransferase activity via a non-nucleoside specific pharmacological inhibitor, RG108, or silencing of DNA methyltransferase-1 with siRNA significantly attenuated ABR threshold elevation, hair cell damage, and the loss of auditory synapses. Conclusions This study suggests that inhibition of DNMT1 ameliorates noise-induced hearing loss and indicates that DNMT1 may be a promising therapeutic target. Graphical abstract Graphical Headlights • RG108 protected against noise-induced hearing loss • RG108 administration protected against noise-induced hair cell loss and auditory neural damage. • RG108 administration attenuated oxidative stress-induced DNA damage and subsequent apoptosis-mediated cell loss in the cochlea after noise exposure. ![]()
Supplementary Information The online version contains supplementary material available at 10.1007/s10565-021-09596-y.
Collapse
Affiliation(s)
- Zhiwei Zheng
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, China
| | - Shan Zeng
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, China
| | - Chang Liu
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, China
| | - Wen Li
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, China
| | - Liping Zhao
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, China
| | - Chengfu Cai
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, 361003, People's Republic of China
| | - Guohui Nie
- Department of Otolaryngology and Institute of Translational Medicine, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, China.
| | - Yingzi He
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China.
- NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai, 200031, China.
| |
Collapse
|
16
|
Zheng Z, Wang Y, Yu H, Li W, Wu J, Cai C, He Y. Salvianolic acid B inhibits ototoxic drug-induced ototoxicity by suppression of the mitochondrial apoptosis pathway. J Cell Mol Med 2020; 24:6883-6897. [PMID: 32351026 PMCID: PMC7299715 DOI: 10.1111/jcmm.15345] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 12/04/2019] [Accepted: 03/06/2020] [Indexed: 12/22/2022] Open
Abstract
It has been claimed that salvianolic acid B (Sal B), a natural bioactive antioxidant, exerts protective effects in various types of cells. This study aims to evaluate the antioxidant and anti‐apoptosis effects of Sal B in a cultured HEI‐OC1 cell line and in transgenic zebrafish (Brn3C: EGFP). A CCK‐8 assay, Annexin V Apoptosis Detection Kit, TUNEL and caspase‐3/7 staining, respectively, examined apoptosis and cell viability. The levels of reactive oxygen species (ROS) were evaluated by CellROX and MitoSOX Red staining. JC‐1 staining was employed to detect the mitochondrial membrane potential (ΔΨm). Western blotting was used to assess expressions of Bax and Bcl‐2. The expression pattern of p‐PI3K and p‐Akt was determined by immunofluorescent staining. We found that Sal B protected against neomycin‐ and cisplatin‐induced apoptotic features, enhanced cell viability and accompanied with decreased caspase‐3 activity in the HEI‐OC1 cells. Supplementary experiments determined that Sal B reduced ROS production (increased ΔΨm), promoted Bcl‐2 expression and down‐regulated the expression of Bax, as well as activated PI3K/AKT signalling pathways in neomycin‐ and cisplatin‐injured HEI‐OC1 cells. Moreover, Sal B markedly decreased the TUNEL signal and protected against neomycin‐ and cisplatin‐induced neuromast HC loss in the transgenic zebrafish. These results unravel a novel role for Sal B as an otoprotective agent against ototoxic drug–induced HC apoptosis, offering a potential use in the treatment of hearing loss.
Collapse
Affiliation(s)
- Zhiwei Zheng
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Yunfeng Wang
- Department of ENT institute and Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Huiqian Yu
- Department of ENT institute and Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Wen Li
- Department of ENT institute and Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Jingfang Wu
- Department of ENT institute and Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Chengfu Cai
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, China.,Teaching Hospital of Fujian Medical University, Xiamen, China.,Xiamen Key Laboratory of Otolaryngology Head and Neck Surgery, Xiamen, China
| | - Yingzi He
- Department of ENT institute and Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| |
Collapse
|
17
|
He Y, Li W, Zheng Z, Zhao L, Li W, Wang Y, Li H. Inhibition of Protein arginine methyltransferase 6 reduces reactive oxygen species production and attenuates aminoglycoside- and cisplatin-induced hair cell death. Theranostics 2020; 10:133-150. [PMID: 31903111 PMCID: PMC6929624 DOI: 10.7150/thno.37362] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/12/2019] [Indexed: 02/06/2023] Open
Abstract
Hair cells in the inner ear have been shown to be susceptible to ototoxicity from some beneficial pharmaceutical drugs, such as aminoglycosides and cisplatin. Thus, there is great interest in discovering new targets or compounds that protect hair cells from these ototoxic drugs. Epigenetic regulation is closely related to inner ear development; however, little is known about epigenetic regulation in the process of ototoxic drugs-induced hearing loss. Methods: In this study, we investigated the role of protein arginine methyltransferase 6 (PRMT6) in aminoglycoside- and cisplatin-induced hair cell loss by using EPZ020411, a selective small molecule PRMT6 inhibitor, in vitro in neonatal mouse cochlear explants and in vivo in C57BL/6 mice. We also took advantage of the HEI-OC1 cell line to evaluate the anti-apoptosis effects of PRMT6 knockdown on cisplatin-induced ototoxicity. Apoptotic cells were identified using cleaved caspase-3 staining and TUNEL assay. The levels of reactive oxygen species (ROS) were evaluated by DCFH-DA and cellROX green staining. The mitochondrial membrane potential (ΔΨm) were determined by JC-1, TMRM, and rhodamine 123 staining. Results: We found that EPZ020411 significantly alleviated neomycin- and cisplatin-induced cell apoptosis and increased hair cell survival. Moreover, pretreatment with EPZ020411 could attenuate neomycin- and cisplatin-induced hearing loss in vivo. Mechanistic studies revealed that inhibition of PRMT6 could reverse the increased expression of caspase-3 and cytochrome c translocation, mitochondrial dysfunction, increased accumulation of ROS, and activation of cell apoptosis after cisplatin injury. Conclusions: Our findings suggested that PRMT6 might serve as a new therapeutic target to prevent hearing loss caused by aminoglycoside- and cisplatin-induced ototoxicity by preventing ROS formation and modulating the mitochondria-related damage and apoptosis.
Collapse
|
18
|
Yu C, Zhuang S. Histone Methyltransferases as Therapeutic Targets for Kidney Diseases. Front Pharmacol 2019; 10:1393. [PMID: 31866860 PMCID: PMC6908484 DOI: 10.3389/fphar.2019.01393] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 10/31/2019] [Indexed: 12/31/2022] Open
Abstract
Emerging evidence has demonstrated that epigenetic regulation plays a vital role in gene expression under normal and pathological conditions. Alterations in the expression and activation of histone methyltransferases (HMTs) have been reported in preclinical models of multiple kidney diseases, including acute kidney injury, chronic kidney disease, diabetic nephropathy, polycystic kidney disease, and renal cell carcinoma. Pharmacological inhibition of these enzymes has shown promise in preclinical models of those renal diseases. In this review, we summarize recent knowledge regarding expression and activation of various HMTs and their functional roles in some kidney diseases. The preclinical activity of currently available HMT inhibitors and the mechanisms of their actions are highlighted.
Collapse
Affiliation(s)
- Chao Yu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, United States
| |
Collapse
|
19
|
Guo J, Chai R, Li H, Sun S. Protection of Hair Cells from Ototoxic Drug-Induced Hearing Loss. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1130:17-36. [PMID: 30915699 DOI: 10.1007/978-981-13-6123-4_2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Hair cells are specialized sensory epithelia cells that receive mechanical sound waves and convert them into neural signals for hearing, and these cells can be killed or damaged by ototoxic drugs, including many aminoglycoside antibiotics, platinum-based anticancer agents, and loop diuretics, leading to drug-induced hearing loss. Studies of therapeutic approaches to drug-induced hearing loss have been hampered by the limited understanding of the biological mechanisms that protect and regenerate hair cells. This review briefly discusses some of the most common ototoxic drugs and describes recent research concerning the mechanisms of ototoxic drug-induced hearing loss. It also highlights current developments in potential therapies and explores current clinical treatments for patients with hearing impairments.
Collapse
Affiliation(s)
- Jin Guo
- Key Laboratory of Hearing Medicine of NHFPC, ENT Institute and Otorhinolaryngology Department, Shanghai Engineering Research Centre of Cochlear Implant, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Renjie Chai
- Key Laboratory of Hearing Medicine of NHFPC, ENT Institute and Otorhinolaryngology Department, Shanghai Engineering Research Centre of Cochlear Implant, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,MOE Key Laboratory for Developmental Genes and Human Disease, Institute of Life Sciences, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China
| | - Huawei Li
- Key Laboratory of Hearing Medicine of NHFPC, ENT Institute and Otorhinolaryngology Department, Shanghai Engineering Research Centre of Cochlear Implant, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Shan Sun
- Key Laboratory of Hearing Medicine of NHFPC, ENT Institute and Otorhinolaryngology Department, Shanghai Engineering Research Centre of Cochlear Implant, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.
| |
Collapse
|
20
|
Li A, You D, Li W, Cui Y, He Y, Li W, Chen Y, Feng X, Sun S, Chai R, Li H. Novel compounds protect auditory hair cells against gentamycin-induced apoptosis by maintaining the expression level of H3K4me2. Drug Deliv 2019; 25:1033-1043. [PMID: 30799660 PMCID: PMC6058728 DOI: 10.1080/10717544.2018.1461277] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aminoglycoside-induced hair cell (HC) loss is a major cause of hearing impairment, and the effective prevention of HC loss remains an unmet medical need. Epigenetic mechanisms have been reported to be involved in protecting cochlear cells against ototoxic drug injury, and in this study we developed new bioactive compounds that have similar chemical structures as the epigenetics-related lysine-specific demethylase 1 (LSD1) inhibitors. LSD1 inhibitors have been reported to protect cochlear cells by preventing demethylation of dimethylated histone H3K4 (H3K4me2). To determine whether these new compounds exert similar protective effects on HCs, we treated mouse cochlear explant cultures with the new compounds together with gentamycin. There was a severe loss of HCs in the organ of Corti after gentamycin exposure, while co-treatment with the new compounds significantly protected against gentamycin-induced HC loss. H3K4me2 levels in the nuclei of HCs decreased after exposure to gentamycin, but H3K4me2 levels were maintained in the presence of the new compounds. Apoptosis is also involved in the injury process, and the new compounds protected the inner ear HCs against apoptosis by reducing caspase-3 activation. Together, our findings demonstrate that our new compounds prevent gentamycin-induced HC loss by preventing the demethylation of H3K4me2 and by inhibiting apoptosis, and these results might provide the theoretical basis for novel drug development for hearing protection.
Collapse
Affiliation(s)
- Ao Li
- a ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital , Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University , Shanghai , China.,b Department of Otorhinolaryngology Head and Neck Surgery , Affiliated Drum Tower Hospital of Nanjing University Medical School, Research Institution of Otorhinolaryngology, Jiangsu Provincial Key Medical Discipline (Laboratory) , Nanjing , China
| | - Dan You
- a ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital , Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University , Shanghai , China
| | - Wenyan Li
- a ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital , Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University , Shanghai , China
| | - Yingjie Cui
- c Knowshine (Shanghai) Pharmaceuticals Inc , Shanghai , China
| | - Yingzi He
- a ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital , Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University , Shanghai , China
| | - Wen Li
- a ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital , Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University , Shanghai , China
| | - Yan Chen
- a ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital , Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University , Shanghai , China
| | - Xiao Feng
- c Knowshine (Shanghai) Pharmaceuticals Inc , Shanghai , China
| | - Shan Sun
- a ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital , Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University , Shanghai , China
| | - Renjie Chai
- d Key Laboratory for Developmental Genes and Human Disease , Ministry of Education, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Institute of Life Sciences, Southeast University , Nanjing , China.,e Co-innovation Center of Neuroregeneration, Nantong University , Nantong , China
| | - Huawei Li
- a ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital , Key Laboratory of Hearing Medicine of NHFPC, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University , Shanghai , China.,f Institutes of Biomedical Sciences and The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University , Shanghai , China
| |
Collapse
|
21
|
Abstract
The epigenetic control of gene expression could be affected by addition and/or removal of post-translational modifications such as phosphorylation, acetylation and methylation of histone proteins, as well as methylation of DNA (5-methylation on cytosines). Misregulation of these modifications is associated with altered gene expression, resulting in various disease conditions. G9a belongs to the protein lysine methyltransferases that specifically methylates the K9 residue of histone H3, leading to suppression of several tumor suppressor genes. In this review, G9a functions, role in various diseases, structural biology aspects for inhibitor design, structure-activity relationship among the reported inhibitors are discussed which could aid in the design and development of potent G9a inhibitors for cancer treatment in the future.
Collapse
|
22
|
Walters BJ, Cox BC. Approaches for the study of epigenetic modifications in the inner ear and related tissues. Hear Res 2019; 376:69-85. [PMID: 30679030 PMCID: PMC6456365 DOI: 10.1016/j.heares.2019.01.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/12/2018] [Accepted: 01/11/2019] [Indexed: 12/12/2022]
Abstract
DNA methylation and histone modifications such as methylation, acetylation, and phosphorylation, are two types of epigenetic modifications that alter gene expression. These additions to DNA regulatory elements or to the tails of histones can be inherited or can also occur de novo. Since epigenetic modifications can have significant effects on various processes at both the cellular and organismal level, there has been a rapid increase in research on this topic throughout all fields of biology in recent years. However, epigenetic research is relativity new for the inner ear field, likely due to the limited number of cells present and their quiescent nature. Here, we provide an overview of methods used to detect DNA methylation and histone modifications with a focus on those that have been validated for use with limited cell numbers and a discussion of the strengths and limitations for each. We also provide examples for how these methods have been used to investigate the epigenetic landscape in the inner ear and related tissues.
Collapse
Affiliation(s)
- Bradley J Walters
- Departments of Neurobiology and Anatomical Sciences, and of Otolaryngology and Communicative Sciences, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Brandon C Cox
- Departments of Pharmacology and Surgery, Division of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL 62711, USA.
| |
Collapse
|
23
|
Glucococorticoid receptor activation exacerbates aminoglycoside-induced damage to the zebrafish lateral line. Hear Res 2019; 377:12-23. [PMID: 30878773 DOI: 10.1016/j.heares.2019.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/18/2018] [Accepted: 03/04/2019] [Indexed: 01/14/2023]
Abstract
Aminoglycoside antibiotics have potent antibacterial properties but cause hearing loss in up to 25% of patients. These drugs are commonly administered in patients with high glucocorticoid stress hormone levels and can be combined with exogenous glucocorticoid treatment. However, the interaction of stress and aminoglycoside-induced hearing loss has not been fully explored. In this study, we investigated the effect of the glucocorticoid stress hormone cortisol on hair cells in the zebrafish lateral line as an important step toward understanding how physiological stressors modulate hair cell survival. We found that 24-hr cortisol incubation sensitized hair cells to neomycin damage. Pharmacological and genetic manipulation demonstrates that sensitization depended on the action of the glucocorticoid receptor but not the mineralocorticoid receptor. Blocking endogenous cortisol production reduced hair cell susceptibility to neomycin, further evidence that glucocorticoids modulate aminoglycoside ototoxicity. Glucocorticoid transcriptional activity was apparent in lateral line hair cells, suggesting a direct action of cortisol in these aminoglycoside-sensitive cells. Our work shows that the stress hormone cortisol can increase hair cell sensitivity to aminoglycoside damage, which highlights the importance of recognizing stress and the impacts of glucocorticoid signaling in both ototoxicity research and clinical practice.
Collapse
|
24
|
Inhibition of Histone Methyltransferase G9a Attenuates Noise-Induced Cochlear Synaptopathy and Hearing Loss. J Assoc Res Otolaryngol 2019; 20:217-232. [PMID: 30710318 DOI: 10.1007/s10162-019-00714-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 01/09/2019] [Indexed: 01/06/2023] Open
Abstract
Posttranslational modification of histones alters their interaction with DNA and nuclear proteins, influencing gene expression and cell fate. In this study, we investigated the effect of G9a (KMT1C, EHMT2), a major histone lysine methyltransferase encoded by the human EHMT2 gene and responsible for histone H3 lysine 9 dimethylation (H3K9me2) on noise-induced permanent hearing loss (NIHL) in adult CBA/J mice. The conditions of noise exposure used in this study led to losses of cochlear synapses and outer hair cells (OHCs) and permanent auditory threshold shifts. Inhibition of G9a with its specific inhibitor BIX 01294 or with siRNA significantly attenuated these pathological features. Treatment with BIX 01294 also prevented the noise-induced decrease of KCNQ4 immunolabeling in OHCs. Additionally, G9a was increased in cochlear cells, including both outer and inner sensory hair cells, some spiral ganglion neurons (SGNs), and marginal cells, 1 h after the completion of the noise exposure. Also subsequent to noise exposure, immunoreactivity for H3K9me2 appeared in some nuclei of OHCs following a high-to-low frequency gradient with more labeled OHCs in the 45-kHz than the 32-kHz region, as well as in the marginal cells and in some SGNs of the basal turn. These findings suggest that epigenetic modifications of H3K9me2 are involved in NIHL and that pharmacological targeting of G9a may offer a strategy for protection against cochlear synaptopathy and NIHL.
Collapse
|
25
|
Shen X, Xiao Y, Li W, Chen K, Yu H. Sevoflurane anesthesia during pregnancy in mice induces hearing impairment in the offspring. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:1827-1836. [PMID: 29970957 PMCID: PMC6020999 DOI: 10.2147/dddt.s156040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction Exposure to gamma-aminobutyric acid-mimetics and N-methyl-D-aspartate-receptor antagonists during pregnancy may lead to hearing loss and long-term behavioral abnormalities in the offspring. The purpose of this study was to explore the association between prenatal exposure to sevoflurane (SEV) anesthesia and hearing impairment in mice. Materials and methods On gestational day 15, pregnant Kunming mice were exposed for 2 hours to 2.5% SEV plus 100% oxygen (anesthesia group) or 100% oxygen alone (control group). Results During auditory brainstem response testing on P30, offspring of the anesthesia group mice exhibited higher hearing thresholds at 8, 16, 24, and 32 kHz; longer peak latency of wave II at all four frequencies; and longer interpeak latencies from waves II to V at 16, 24, and 32 kHz, compared to the control offspring. Caspase-3, iNOS, and COX-2 activation occurred in the fetal cochlea of the anesthesia group. Mitochondrial swelling was observed in the anesthesia group offspring at P1 and P15. Conclusion Our results suggest that SEV exposure during pregnancy may cause detrimental effects on the developing auditory system.
Collapse
Affiliation(s)
- Xia Shen
- Department of Anesthesiology, Shanghai Eye, Ear, Nose, and Throat Hospital, Fudan University, Shanghai 200031, People's Republic of China
| | - Yanan Xiao
- Department of Anesthesiology, Shanghai Eye, Ear, Nose, and Throat Hospital, Fudan University, Shanghai 200031, People's Republic of China
| | - Wen Li
- Research Center, Shanghai Eye, Ear, Nose, and Throat Hospital, Fudan University, Shanghai 200031, People's Republic of China
| | - Kaizheng Chen
- Department of Anesthesiology, Shanghai Eye, Ear, Nose, and Throat Hospital, Fudan University, Shanghai 200031, People's Republic of China
| | - Huiqian Yu
- Department of Otorhinolaryngology, Shanghai Eye, Ear, Nose, and Throat Hospital, Fudan University, Shanghai 200031, People's Republic of China
| |
Collapse
|
26
|
He Y, Bao B, Li H. Using zebrafish as a model to study the role of epigenetics in hearing loss. Expert Opin Drug Discov 2017; 12:967-975. [PMID: 28682135 DOI: 10.1080/17460441.2017.1340270] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The rapid progress of bioinformatics and high-throughput screening techniques in recent years has led to the identification of many candidate genes and small-molecule drugs that have the potential to make significant contributions to our understanding of the developmental and pathological processes of hearing, but it remains unclear how these genes and regulatory factors are coordinated. Increasing evidence suggests that epigenetic mechanisms are essential for establishing gene expression profiles and likely play an important role in the development of inner ear and in the pathology of hearing-associated diseases. Zebrafish are a valuable and tractable in vivo model organism for monitoring changes in the epigenome and for identifying new epigenetic processes and drug molecules that can influence vertebrate development. Areas covered: In this review, the authors focus on zebrafish as a model to summarize recent findings concerning the roles of epigenetics in the development, regeneration, and protection of hair cells. Expert opinion: Using the zebrafish model in combination with high-throughput screening and genome-editing technologies to investigate the function of epigenetics in hearing is crucial to help us better understand the molecular and genetic mechanisms of auditory development and function. It will also contribute to the development of new strategies to restore hearing loss.
Collapse
Affiliation(s)
- Yingzi He
- a ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology , Fudan University , Shanghai , China.,c Key Laboratory of Hearing Medicine of NHFPC , Shanghai , China
| | - Beier Bao
- a ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology , Fudan University , Shanghai , China
| | - Huawei Li
- a ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology , Fudan University , Shanghai , China.,b Institutes of Biomedical Sciences , Fudan University , Shanghai , China.,c Key Laboratory of Hearing Medicine of NHFPC , Shanghai , China.,d Shanghai Engineering Research Centre of Cochlear Implant , Shanghai , China.,e The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science , Fudan University , Shanghai , China
| |
Collapse
|
27
|
MiR-182-5p protects inner ear hair cells from cisplatin-induced apoptosis by inhibiting FOXO3a. Cell Death Dis 2016; 7:e2362. [PMID: 27607577 PMCID: PMC5059852 DOI: 10.1038/cddis.2016.246] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 06/14/2016] [Accepted: 07/13/2016] [Indexed: 01/01/2023]
Abstract
Cisplatin is widely used for chemotherapy of a variety of malignancies. However, the clinical application of cisplatin is hampered by the resultant irreversible hearing loss due to hair cell apoptosis. To date, no practical regimen to resolve this has been developed. Meanwhile, the role of microRNA in protecting hair cells from cisplatin-induced apoptosis in the inner ear has not been extensively investigated. In this study, we monitored miR-183, -96, and -182 turnover in the cochlea during cisplatin treatment in vitro. We found that overexpression of miR-182, but not miR-183 and -96, improved hair cell survival after 3 μM cisplatin treatment in vitro. We demonstrated that overexpression of miR-182 repressed the intrinsic apoptotic pathway by inhibiting the translation of FOXO3a. Our study offers a new therapeutic target for alleviating cisplatin-induced hair cell apoptosis in a rapid and tissue-specific manner.
Collapse
|
28
|
Mateo Sánchez S, Freeman SD, Delacroix L, Malgrange B. The role of post-translational modifications in hearing and deafness. Cell Mol Life Sci 2016; 73:3521-33. [PMID: 27147466 PMCID: PMC11108544 DOI: 10.1007/s00018-016-2257-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 04/21/2016] [Accepted: 04/26/2016] [Indexed: 12/20/2022]
Abstract
Post-translational modifications (PTMs) are key molecular events that modify proteins after their synthesis and modulate their ultimate functional properties by affecting their stability, localisation, interaction potential or activity. These chemical changes expand the size of the proteome adding diversity to the molecular pathways governing the biological outcome of cells. PTMs are, thus, crucial in regulating a variety of cellular processes such as apoptosis, proliferation and differentiation and have been shown to be instrumental during embryonic development. In addition, alterations in protein PTMs have been implicated in the pathogenesis of many human diseases, including deafness. In this review, we summarize the recent progress made in understanding the roles of PTMs during cochlear development, with particular emphasis on the enzymes driving protein phosphorylation, acetylation, methylation, glycosylation, ubiquitination and SUMOylation. We also discuss how these enzymes may contribute to hearing impairment and deafness.
Collapse
Affiliation(s)
- Susana Mateo Sánchez
- Developmental Neurobiology Unit, GIGA-Neurosciences, University of Liège, Quartier Hôpital (CHU), Avenue Hippocrate 15, Tour 4, 1er étage, Bât. B36, 4000, Liège, Belgium
| | - Stephen D Freeman
- Developmental Neurobiology Unit, GIGA-Neurosciences, University of Liège, Quartier Hôpital (CHU), Avenue Hippocrate 15, Tour 4, 1er étage, Bât. B36, 4000, Liège, Belgium
| | - Laurence Delacroix
- Developmental Neurobiology Unit, GIGA-Neurosciences, University of Liège, Quartier Hôpital (CHU), Avenue Hippocrate 15, Tour 4, 1er étage, Bât. B36, 4000, Liège, Belgium
| | - Brigitte Malgrange
- Developmental Neurobiology Unit, GIGA-Neurosciences, University of Liège, Quartier Hôpital (CHU), Avenue Hippocrate 15, Tour 4, 1er étage, Bât. B36, 4000, Liège, Belgium.
| |
Collapse
|
29
|
Shimaji K, Konishi T, Yoshida H, Kimura H, Yamaguchi M. Genome-wide genetic screen identified the link between dG9a and epidermal growth factor receptor signaling pathway in vivo. Exp Cell Res 2016; 346:53-64. [PMID: 27343629 DOI: 10.1016/j.yexcr.2016.06.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 05/27/2016] [Accepted: 06/21/2016] [Indexed: 10/21/2022]
Abstract
G9a is one of the histone H3 Lys 9 (H3K9) specific methyltransferases first identified in mammals. Drosophila G9a (dG9a) has been reported to induce H3K9 dimethylation in vivo, and the target genes of dG9a were identified during embryonic and larval stages. Although dG9a is important for a variety of developmental processes, the link between dG9a and signaling pathways are not addressed yet. Here, by genome-wide genetic screen, taking advantage of the rough eye phenotype of flies that over-express dG9a in eye discs, we identified 16 genes that enhanced the rough eye phenotype induced by dG9a over-expression. These 16 genes included Star, anterior open, bereft and F-box and leucine-rich repeat protein 6 which are components of epidermal growth factor receptor (EGFR) signaling pathway. When dG9a over-expression was combined with mutation of Star, differentiation of R7 photoreceptors in eye imaginal discs as well as cone cells and pigment cells in pupal retinae was severely inhibited. Furthermore, the dG9a over-expression reduced the activated ERK signals in eye discs. These data demonstrate a strong genetic link between dG9a and the EGFR signaling pathway.
Collapse
Affiliation(s)
- Kouhei Shimaji
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan; Insect Biomedical Research Center, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Takahiro Konishi
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan; Insect Biomedical Research Center, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Hideki Yoshida
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan; Insect Biomedical Research Center, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Hiroshi Kimura
- Department of Biological Sciences, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Nagatsuta, Midori-ku, Yokohama 226-8501, Japan
| | - Masamitsu Yamaguchi
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan; Insect Biomedical Research Center, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan.
| |
Collapse
|
30
|
Unexpected Distinct Roles of the Related Histone H3 Lysine 9 Methyltransferases G9a and G9a-Like Protein in Myoblasts. J Mol Biol 2016; 428:2329-2343. [DOI: 10.1016/j.jmb.2016.03.029] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 02/26/2016] [Accepted: 03/27/2016] [Indexed: 01/14/2023]
|
31
|
Li M, Liu C, Yang L, Zhang L, Chen C, He M, Lu Y, Feng W, Pi H, Zhang Y, Zhong M, Yu Z, Zhou Z. G9a-mediated histone methylation regulates cadmium-induced male fertility damage in pubertal mice. Toxicol Lett 2016; 252:11-21. [DOI: 10.1016/j.toxlet.2016.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 03/18/2016] [Accepted: 04/05/2016] [Indexed: 10/22/2022]
|
32
|
Tang D, Lin Q, He Y, Chai R, Li H. Inhibition of H3K9me2 Reduces Hair Cell Regeneration after Hair Cell Loss in the Zebrafish Lateral Line by Down-Regulating the Wnt and Fgf Signaling Pathways. Front Mol Neurosci 2016; 9:39. [PMID: 27303264 PMCID: PMC4880589 DOI: 10.3389/fnmol.2016.00039] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 05/12/2016] [Indexed: 11/13/2022] Open
Abstract
The activation of neuromast (NM) supporting cell (SC) proliferation leads to hair cell (HC) regeneration in the zebrafish lateral line. Epigenetic mechanisms have been reported that regulate HC regeneration in the zebrafish lateral line, but the role of H3K9me2 in HC regeneration after HC loss remains poorly understood. In this study, we focused on the role of H3K9me2 in HC regeneration following neomycin-induced HC loss. To investigate the effects of H3K9me2 in HC regeneration, we took advantage of the G9a/GLP-specific inhibitor BIX01294 that significantly reduces the dimethylation of H3K9. We found that BIX01294 significantly reduced HC regeneration after neomycin-induced HC loss in the zebrafish lateral line. BIX01294 also significantly reduced the proliferation of NM cells and led to fewer SCs in the lateral line. In situ hybridization showed that BIX01294 significantly down-regulated the Wnt and Fgf signaling pathways, which resulted in reduced SC proliferation and HC regeneration in the NMs of the lateral line. Altogether, our results suggest that down-regulation of H3K9me2 significantly decreases HC regeneration after neomycin-induced HC loss through inactivation of the Wnt/β-catenin and Fgf signaling pathways. Thus H3K9me2 plays a critical role in HC regeneration.
Collapse
Affiliation(s)
- Dongmei Tang
- Department of Otorhinolaryngology, Affiliated Eye and ENT Hospital of Fudan University Shanghai, China
| | - Qin Lin
- Department of Otolaryngology Head and Neck Surgery, First Affiliated Hospital of Fujian Medical University Fuzhou, China
| | - Yingzi He
- Department of Otorhinolaryngology, Affiliated Eye and ENT Hospital of Fudan University Shanghai, China
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China; Co-innovation Center of Neuroregeneration, Nantong UniversityNantong, China
| | - Huawei Li
- Department of Otorhinolaryngology, Affiliated Eye and ENT Hospital of Fudan UniversityShanghai, China; State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China; Institute of Stem Cell and Regeneration Medicine, Institutions of Biomedical Science, Fudan UniversityShanghai, China; Key Laboratory of Hearing Science, Ministry of Health, EENT Hospital, Fudan UniversityShanghai, China
| |
Collapse
|
33
|
Ebbers L, Runge K, Nothwang HG. Differential patterns of histone methylase EHMT2 and its catalyzed histone modifications H3K9me1 and H3K9me2 during maturation of central auditory system. Cell Tissue Res 2016; 365:247-64. [PMID: 27083448 DOI: 10.1007/s00441-016-2401-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 03/24/2016] [Indexed: 12/31/2022]
Abstract
Histone methylation is an important epigenetic mark leading to changes in DNA accessibility and transcription. Here, we investigate immunoreactivity against the euchromatic histone-lysine N-methyltransferase EHMT2 and its catalyzed mono- and dimethylation marks at histone 3 lysine 9 (H3K9me1 and H3K9me2) during postnatal differentiation of the mouse central auditory system. In the brainstem, expression of EHMT2 was high in the first postnatal week and down-regulated thereafter. In contrast, immunoreactivity in the auditory cortex (AC) remained high during the first year of life. This difference might be related to distinct demands for adult plasticity. Analyses of two deaf mouse models, namely Cldn14 (-/-) and Cacna1d (-/-), demonstrated that sound-driven or spontaneous activity had no influence on EHMT2 immunoreactivity. The methylation marks H3K9me1 and H3K9me2 were high throughout the auditory system up to 1 year. Young auditory neurons showed immunoreactivity against both methylations at similar intensities, whereas many mature neurons showed stronger labeling for either H3K9me1 or H3K9me2. These differences were only poorly correlated with cell types. To identify methyltransferases contributing to the persistent H3K9me1 and H3K9me2 marks in the adult brainstem, EHMT1 and the retinoblastoma-interacting zinc-finger protein RIZ1 were analyzed. Both were down-regulated during brainstem development, similar to EHMT2. Contrary to EHMT2, EHMT1 was also down-regulated in adult cortical areas. Together, our data reveal a marked difference in EHMT2 levels between mature brainstem and cortical areas and a decoupling between EHMT2 abundance and histone 3 lysine 9 methylations during brainstem differentiation. Furthermore, EHMT1 and EHMT2 are differentially expressed in cortical areas.
Collapse
Affiliation(s)
- Lena Ebbers
- Neurogenetics Group, Center of Excellence Hearing4All, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26111, Oldenburg, Germany
| | - Karen Runge
- Neurogenetics Group, Center of Excellence Hearing4All, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26111, Oldenburg, Germany
| | - Hans Gerd Nothwang
- Neurogenetics Group, Center of Excellence Hearing4All, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26111, Oldenburg, Germany. .,Research Center for Neurosensory Science, Carl von Ossietzky University Oldenburg, 26111, Oldenburg, Germany.
| |
Collapse
|
34
|
AHN MJ, JEONG SG, CHO GW. Antisenescence activity of G9a inhibitor BIX01294 on human bone marrow mesenchymal stromal cells. Turk J Biol 2016. [DOI: 10.3906/biy-1507-11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
35
|
He Y, Wang Z, Sun S, Tang D, Li W, Chai R, Li H. HDAC3 Is Required for Posterior Lateral Line Development in Zebrafish. Mol Neurobiol 2015; 53:5103-17. [PMID: 26395281 DOI: 10.1007/s12035-015-9433-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 09/10/2015] [Indexed: 01/03/2023]
Abstract
Histone deacetylases (HDACs) are involved in multiple developmental processes, but their functions in the development of mechanosensory organs are largely unknown. In the present study, we report the presence of HDAC3 in the zebrafish posterior lateral line primordium and newly deposited neuromasts. We used morpholinos to show that HDAC3 knockdown severely disrupts the development of the posterior lateral line and reduces the numbers of neuromasts and sensory hair cells within these organs. In HDAC3 morphants, we also observed decreased cell proliferation and increased apoptosis, which might lead to these defects. Finally, we show that HDAC3 deficiency results in attenuated Fgf signaling in the migrating primordium. In situ hybridizations indicate aberrant expression patterns of Notch signaling pathway genes in HDAC3 morphants. In addition, inhibition of HDAC3 function diminishes cxcr7b and alters cxcl12a expression in the migrating primordium. Our results indicate that HDAC3 plays a crucial role in regulating posterior lateral line (PLL) formation and provide evidence for epigenetic regulation in auditory organ development.
Collapse
Affiliation(s)
- Yingzi He
- Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, People's Republic of China
| | - Zhengmin Wang
- Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, People's Republic of China. .,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission, Shanghai, People's Republic of China.
| | - Shaoyang Sun
- Key Laboratory of Metabolism and Molecular Medicine, the Ministry of Education, Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, People's Republic of China
| | - Dongmei Tang
- Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, People's Republic of China
| | - Wenyan Li
- Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, People's Republic of China
| | - Renjie Chai
- Co-innovation Center of Neuroregeneration, Key Laboratory for Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing, Jiangsu, 210096, People's Republic of China
| | - Huawei Li
- Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, People's Republic of China. .,Key Laboratory of Hearing Medicine of National Health and Family Planning Commission, Shanghai, People's Republic of China. .,State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, People's Republic of China. .,Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
36
|
|
37
|
Sun S, Yu H, Yu H, Honglin M, Ni W, Zhang Y, Guo L, He Y, Xue Z, Ni Y, Li J, Feng Y, Chen Y, Shao R, Chai R, Li H. Inhibition of the activation and recruitment of microglia-like cells protects against neomycin-induced ototoxicity. Mol Neurobiol 2015; 51:252-67. [PMID: 24781382 DOI: 10.1007/s12035-014-8712-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 04/09/2014] [Indexed: 12/18/2022]
Abstract
One of the most unfortunate side effects of aminoglycoside (AG) antibiotics such as neomycin is that they target sensory hair cells (HCs) and can cause permanent hearing impairment. We have observed HC loss and microglia-like cell (MLC) activation in the inner ear (cochlea) following neomycin administration. We focused on CX3CL1, a membrane-bound glycoprotein expressed on neurons and endothelial cells, as a way to understand how the MLCs are activated and the role these cells play in HC loss. CX3CL1 is the exclusive ligand for CX3CR1, which is a chemokine receptor expressed on the surface of macrophages and MLCs. In vitro experiments showed that the expression levels of CX3CL1 and CX3CR1 increased in the cochlea upon neomycin treatment, and CX3CL1 was expressed on HCs, while CX3CR1 was expressed on MLCs. When cultured with 1 μg/mL exogenous CX3CL1, MLCs were activated by CX3CL1, and the cytokine level was increased in the cochleae leading to apoptosis in the HCs. In CX3CR1 knockout mice, a significantly greater number of cochlear HCs survived than in wild-type mice when the cochlear explants were cultured with neomycin in vitro. Furthermore, inhibiting the activation of MLCs with minocycline reduced the neomycin-induced HC loss and improved the hearing function in neomycin-treated mice in vivo. Our results demonstrate that CX3CL1-induced MLC activation plays an important role in the induction of HC death and provide evidence for CX3CL1 and CX3CR1 as promising new therapeutic targets for the prevention of hearing loss.
Collapse
Affiliation(s)
- Shan Sun
- Research Center, Affiliated Eye and ENT Hospital of Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Chen Y, Li L, Ni W, Zhang Y, Sun S, Miao D, Chai R, Li H. Bmi1 regulates auditory hair cell survival by maintaining redox balance. Cell Death Dis 2015; 6:e1605. [PMID: 25611380 PMCID: PMC4669747 DOI: 10.1038/cddis.2014.549] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 11/13/2014] [Accepted: 11/17/2014] [Indexed: 01/06/2023]
Abstract
Reactive oxygen species (ROS) accumulation are involved in noise- and ototoxic drug-induced hair cell loss, which is the major cause of hearing loss. Bmi1 is a member of the Polycomb protein family and has been reported to regulate mitochondrial function and ROS level in thymocytes and neurons. In this study, we reported the expression of Bmi1 in mouse cochlea and investigated the role of Bmi1 in hair cell survival. Bmi1 expressed in hair cells and supporting cells in mouse cochlea. Bmi1−/− mice displayed severe hearing loss and patched outer hair cell loss from postnatal day 22. Ototoxic drug-induced hair cells loss dramatically increased in Bmi1−/− mice compared with that in wild-type controls both in vivo and in vitro, indicating Bmi1−/− hair cells were significantly more sensitive to ototoxic drug-induced damage. Cleaved caspase-3 and TUNEL staining demonstrated that apoptosis was involved in the increased hair cell loss of Bmi1−/− mice. Aminophenyl fluorescein and MitoSOX Red staining showed the level of free radicals and mitochondrial ROS increased in Bmi1−/− hair cells due to the aggravated disequilibrium of antioxidant–prooxidant balance. Furthermore, the antioxidant N-acetylcysteine rescued Bmi1−/− hair cells from neomycin injury both in vitro and in vivo, suggesting that ROS accumulation was mainly responsible for the increased aminoglycosides sensitivity in Bmi1−/− hair cells. Our findings demonstrate that Bmi1 has an important role in hair cell survival by controlling redox balance and ROS level, thus suggesting that Bmi1 may work as a new therapeutic target for the prevention of hair cell death.
Collapse
Affiliation(s)
- Y Chen
- 1] Department of Otorhinolaryngology, Hearing Research Institute, Affiliated Eye and ENT Hospital of Fudan University, Shanghai 200031, China [2] Central Laboratory, Affiliated Eye and ENT Hospital of Fudan University, Shanghai 200031, China
| | - L Li
- Department of Otorhinolaryngology, Hearing Research Institute, Affiliated Eye and ENT Hospital of Fudan University, Shanghai 200031, China
| | - W Ni
- Department of Otorhinolaryngology, Hearing Research Institute, Affiliated Eye and ENT Hospital of Fudan University, Shanghai 200031, China
| | - Y Zhang
- 1] Department of Otorhinolaryngology, Hearing Research Institute, Affiliated Eye and ENT Hospital of Fudan University, Shanghai 200031, China [2] Central Laboratory, Affiliated Eye and ENT Hospital of Fudan University, Shanghai 200031, China [3] Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - S Sun
- 1] Department of Otorhinolaryngology, Hearing Research Institute, Affiliated Eye and ENT Hospital of Fudan University, Shanghai 200031, China [2] Central Laboratory, Affiliated Eye and ENT Hospital of Fudan University, Shanghai 200031, China
| | - D Miao
- State Key Laboratory of Reproductive Medicine, Research Center for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing 210096, China
| | - R Chai
- Co-innovation Center of Neuroregeneration, Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - H Li
- 1] Department of Otorhinolaryngology, Hearing Research Institute, Affiliated Eye and ENT Hospital of Fudan University, Shanghai 200031, China [2] Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China [3] State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| |
Collapse
|
39
|
McGrath J, Trojer P. Targeting histone lysine methylation in cancer. Pharmacol Ther 2015; 150:1-22. [PMID: 25578037 DOI: 10.1016/j.pharmthera.2015.01.002] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 12/03/2014] [Indexed: 02/06/2023]
Abstract
Within the vast landscape of histone modifications lysine methylation has gained increasing attention because of its profound regulatory potential. The methylation of lysine residues on histone proteins modulates chromatin structure and thereby contributes to the regulation of DNA-based nuclear processes such as transcription, replication and repair. Protein families with opposing catalytic activities, lysine methyltransferases (KMTs) and demethylases (KDMs), dynamically control levels of histone lysine methylation and individual enzymes within these families have become candidate oncology targets in recent years. A number of high quality small molecule inhibitors of these enzymes have been identified. Several of these compounds elicit selective cancer cell killing in vitro and robust efficacy in vivo, suggesting that targeting 'histone lysine methylation pathways' may be a relevant, emerging cancer therapeutic strategy. Here, we discuss individual histone lysine methylation pathway targets, the properties of currently available small molecule inhibitors and their application in the context of cancer.
Collapse
Affiliation(s)
- John McGrath
- Constellation Pharmaceuticals, 215 1st Street Suite 200, Cambridge, MA, 02142, USA
| | - Patrick Trojer
- Constellation Pharmaceuticals, 215 1st Street Suite 200, Cambridge, MA, 02142, USA.
| |
Collapse
|
40
|
Inhibition of H3K4me2 Demethylation Protects Auditory Hair Cells from Neomycin-Induced Apoptosis. Mol Neurobiol 2014; 52:196-205. [PMID: 25132091 DOI: 10.1007/s12035-014-8841-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 07/30/2014] [Indexed: 12/26/2022]
Abstract
Aminoglycoside-induced hair cell loss is a major cause of hearing impairment in children and deserves more attention in medical research. Epigenetic mechanisms have been shown to protect hair cells from ototoxic drugs. In this study, we focused on the role of dimethylated histone H3K4 (H3K4me2) in hair cell survival. To investigate the effects of lysine-specific demethylase 1 (LSD1)--the histone demethylase primarily responsible for demethylating H3K4me2--on neomycin-induced hair cell loss, isolated cochleae were pretreated with LSD1 inhibitors followed by neomycin exposure. There was a severe loss of hair cells in the organ of Corti after neomycin exposure, and inhibition of LSD1 significantly protected against neomycin-induced hair cell loss. H3K4me2 expression in the nuclei of hair cells decreased after exposure to neomycin, and blocking the decreased expression of H3K4me2 with LSD1 inhibitors prevented hair cell loss. Local delivery of these inhibitors in vivo also protected hair cells from neomycin-induced ototoxicity and maintained the hearing threshold in mice as determined by auditory brain stem response. This inhibition of neomycin-induced apoptosis occurs via reduced caspase-3 activation. Together, our findings demonstrate the protective role for H3K4me2 against neomycin-induced hair cell loss and hearing loss.
Collapse
|
41
|
Subbanna S, Nagre NN, Shivakumar M, Umapathy NS, Psychoyos D, Basavarajappa BS. Ethanol induced acetylation of histone at G9a exon1 and G9a-mediated histone H3 dimethylation leads to neurodegeneration in neonatal mice. Neuroscience 2013; 258:422-32. [PMID: 24300108 DOI: 10.1016/j.neuroscience.2013.11.043] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 11/19/2013] [Accepted: 11/20/2013] [Indexed: 11/16/2022]
Abstract
The transient exposure of immature rodents to ethanol during postnatal day 7 (P7), comparable to a time point within the third trimester of human pregnancy, induces neurodegeneration. However, the molecular mechanisms underlying the deleterious effects of ethanol on the developing brain are poorly understood. In our previous study, we showed that a high dose administration of ethanol at P7 enhances G9a and leads to caspase-3-mediated degradation of dimethylated H3 on lysine 9 (H3K9me2). In this study, we investigated the potential role of epigenetic changes at G9a exon1, G9a-mediated H3 dimethylation on neurodegeneration and G9a-associated proteins in the P7 brain following exposure to a low dose of ethanol. We found that a low dose of ethanol induces mild neurodegeneration in P7 mice, enhances specific acetylation of H3 on lysine 14 (H3K14ace) at G9a exon1, G9a protein levels, augments the dimethylation of H3K9 and H3 lysine 27 (H3K27me2). However, neither dimethylated H3K9 nor K27 underwent degradation. Pharmacological inhibition of G9a activity prior to ethanol treatment prevented H3 dimethylation and neurodegeneration. Further, our immunoprecipitation data suggest that G9a directly associates with DNA methyltransferase (DNMT3A) and methyl-CpG-binding protein 2 (MeCP2). In addition, DNMT3A and MeCP2 protein levels were enhanced by a low dose of ethanol that was shown to induce mild neurodegeneration. Collectively, these epigenetic alterations lead to association of G9a, DNMT3A and MeCP2 to form a larger repressive complex and have a significant role in low-dose ethanol-induced neurodegeneration in the developing brain.
Collapse
Affiliation(s)
- S Subbanna
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - N N Nagre
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - M Shivakumar
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| | - N S Umapathy
- Vascular Biology Center, Georgia Regents University, Augusta, GA 30912, USA
| | - D Psychoyos
- Institute of Biosciences and Technology, Houston, Texas A&M University Health Science Center, Houston, TX 77030, USA
| | - B S Basavarajappa
- Division of Analytical Psychopharmacology, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA; New York State Psychiatric Institute, College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA; Department of Psychiatry, College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
42
|
Chen Y, Yu H, Zhang Y, Li W, Lu N, Ni W, He Y, Li J, Sun S, Wang Z, Li H. Cotransfection of Pax2 and Math1 promote in situ cochlear hair cell regeneration after neomycin insult. Sci Rep 2013; 3:2996. [PMID: 24141260 PMCID: PMC3801138 DOI: 10.1038/srep02996] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 10/02/2013] [Indexed: 01/08/2023] Open
Abstract
The ideal strategy for hair cell regeneration is promoting residual supporting cell proliferation followed by induction of hair cell differentiation. In this study, cultured neonatal mouse organs of Corti were treated with neomycin to eliminate hair cells followed by incubation with recombined adenovirus expressing Pax2 and/or Math1. Results showed that overexpression of Pax2 significantly promoted proliferation of supporting cells. The number of BrdU+/myosin VIIA+ cells increased significantly in hair cell pre-existing region two weeks after adenovirus infection in Ad-Pax2-IRES-Math1 group compared to Ad-Pax2 and Ad-Math1 groups. This indicated that cotransfection of Pax2 and Math1 induced supporting cells to proliferate and differentiate into hair cells in situ. Most new hair cells were labeled by FM1-43 suggesting they acquired certain function. The results also suggest that inducing proliferating cells rather than quiescent cells to differentiate into hair cells by forced expression of Math1 is feasible for mammalian hair cell regeneration.
Collapse
Affiliation(s)
- Yan Chen
- 1] Otology Skull base Surgery Department, Hearing Research Institute, Eye and ENT Hospital of Shanghai Medical School, Fudan University. Shanghai, 200031, P.R.China [2] Central laboratory, Eye and ENT Hospital of Shanghai Medical School, Fudan University. Shanghai, 200031, P.R.China [3]
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|