1
|
Noda T, Wakizono T, Manabe T, Aoyagi K, Kubota M, Yasui T, Nakagawa T, Nakashima K, Meno C. Sustained Wnt signaling in the mouse inner ear after morphogenesis: In hair cells, supporting cells, and spiral ganglion neurons. Hear Res 2025; 462:109282. [PMID: 40267597 DOI: 10.1016/j.heares.2025.109282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/10/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025]
Abstract
The regenerative capacity of inner ear hair cells in mammals varies between the cochlea and the vestibular system. Hair cells in the cochlea lack regenerative ability, whereas those in the vestibular system exhibit limited regenerative potential. However, supporting cells in the cochlea retain proliferative capacity, making them a key focus in auditory regeneration research. Similarly, spiral ganglion neurons actively proliferate until birth but lose this ability within a week postnatally, sharing the regenerative limitations of hair cells. This study investigated the role of the canonical Wnt signaling pathway as a potential regulator of these cells. Wnt signaling plays a crucial role in otic development and inner ear morphogenesis. Using reporter mice, we analyzed the activity of the Wnt canonical pathway in the inner ear at the cellular stages from embryonic to adult stages, assessing fluorescence intensities as an indicator of signaling activity. Our findings demonstrate that Wnt signaling remains active in the vestibular hair cells and in the supporting cells of both the cochlea and vestibule throughout development and into adulthood. In addition, Wnt activity was observed in spiral ganglion neurons up to 7 days after birth, coinciding with their period of proliferative potential. These findings suggest that Wnt signaling is integral to cell proliferation in the inner ear both before and after birth.
Collapse
Affiliation(s)
- Teppei Noda
- Department of Otolaryngology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Fukuoka, 8128582, Japan.
| | - Takahiro Wakizono
- Department of Otolaryngology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Fukuoka, 8128582, Japan; Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Fukuoka, 8128582, Japan
| | - Takahiro Manabe
- Department of Otolaryngology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Fukuoka, 8128582, Japan; Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Fukuoka, 8128582, Japan
| | - Kei Aoyagi
- Department of Otolaryngology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Fukuoka, 8128582, Japan; Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Fukuoka, 8128582, Japan
| | - Marie Kubota
- Department of Otolaryngology-Head & Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tetsuro Yasui
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Fukuoka, 8128582, Japan
| | - Takashi Nakagawa
- Department of Otolaryngology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Fukuoka, 8128582, Japan
| | - Kinichi Nakashima
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Fukuoka, 8128582, Japan
| | - Chikara Meno
- Department of Developmental Biology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Fukuoka, 8128582, Japan
| |
Collapse
|
2
|
Lin Y, Zhang Q, Tong W, Wang Y, Wu L, Xiao H, Tang X, Dai M, Ye Z, Chai R, Zhang S. Conditional Overexpression of Net1 Enhances the Trans-Differentiation of Lgr5 + Progenitors into Hair Cells in the Neonatal Mouse Cochlea. Cell Prolif 2025; 58:e13787. [PMID: 39675772 PMCID: PMC11969244 DOI: 10.1111/cpr.13787] [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: 07/26/2024] [Revised: 11/05/2024] [Accepted: 11/13/2024] [Indexed: 12/17/2024] Open
Abstract
Sensorineural hearing loss is mainly caused by damage to hair cells (HC), which cannot be regenerated spontaneously in adult mammals once damaged. Cochlear Lgr5+ progenitors are characterised by HC regeneration capacity in neonatal mice, and we previously screened several new genes that might induce HC regeneration from Lgr5+ progenitors. Net1, a guanine nucleotide exchange factor, is one of the screened new genes and is particularly active in cancer cells and is involved in cell proliferation and differentiation. Here, to explore in vivo roles of Net1 in HC regeneration, Net1 loxp/loxp mice were constructed and crossed with Lgr5 CreER/+ mice to conditionally overexpress (cOE) Net1 in cochlear Lgr5+ progenitors. We observed a large number of ectopic HCs in Lgr5 CreER/+ Net1 loxp/loxp mouse cochlea, which showed a dose-dependent effect. Moreover, the EdU assay was unable to detect any EdU+/Sox2+ supporting cells, while lineage tracing showed significantly more regenerated tdTomato+ HCs in Lgr5 CreER/+ Net1 loxp/loxp tdTomato mice, which indicated that Net1 cOE enhanced HC regeneration by inducing the direct trans-differentiation of Lgr5+ progenitors rather than mitotic HC regeneration. Additionally, qPCR results showed that the transcription factors related to HC regeneration, including Atoh1, Gfi1 and Pou4f3, were significantly upregulated and are probably the mechanism behind the HC regeneration induced by Net1. In conclusion, our study provides new evidence for the role of Net1 in enhancing HC regeneration in the neonatal mouse cochlea.
Collapse
Affiliation(s)
- Yanqin Lin
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
- Southeast University Shenzhen Research InstituteShenzhenChina
| | - Qiuyue Zhang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
- Southeast University Shenzhen Research InstituteShenzhenChina
| | - Wei Tong
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
- Southeast University Shenzhen Research InstituteShenzhenChina
| | - Yintao Wang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
- Southeast University Shenzhen Research InstituteShenzhenChina
| | - Leilei Wu
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
- Southeast University Shenzhen Research InstituteShenzhenChina
| | - Hairong Xiao
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
- Southeast University Shenzhen Research InstituteShenzhenChina
| | - Xujun Tang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
- Southeast University Shenzhen Research InstituteShenzhenChina
| | - Mingchen Dai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
- Southeast University Shenzhen Research InstituteShenzhenChina
| | - Zixuan Ye
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
- Southeast University Shenzhen Research InstituteShenzhenChina
| | - Renjie Chai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
- Southeast University Shenzhen Research InstituteShenzhenChina
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
- Institute for Stem Cell and RegenerationChinese Academy of ScienceBeijingChina
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Shasha Zhang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
- Southeast University Shenzhen Research InstituteShenzhenChina
| |
Collapse
|
3
|
Young CA, Burt E, Munnamalai V. A cochlear progenitor pool influences patterning of the mammalian sensory epithelium via MYBL2. Development 2024; 151:dev202635. [PMID: 39254648 PMCID: PMC11423912 DOI: 10.1242/dev.202635] [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: 12/18/2023] [Accepted: 08/01/2024] [Indexed: 09/11/2024]
Abstract
During embryonic development, Wnt signaling influences both proliferation and sensory formation in the cochlea. How this dual nature of Wnt signaling is coordinated is unknown. In this study, we define a novel role for a Wnt-regulated gene, Mybl2, which was already known to be important for proliferation, in determining the size and patterning of the sensory epithelium in the murine cochlea. Using a quantitative spatial analysis approach and analyzing Mybl2 loss-of-function, we show that Mybl2 promoted proliferation in the inner sulcus domain but limited the size of the sensory domain by influencing their adjoining boundary position via Jag1 regulation during development. Mybl2 loss-of-function simultaneously decreased proliferation in the inner sulcus and increased the size of the sensory domain, resulting in a wider sensory epithelium with ectopic inner hair cell formation during late embryonic stages. These data suggest that progenitor cells in the inner sulcus determine boundary formation and pattern the sensory epithelium via MYBL2.
Collapse
Affiliation(s)
- Caryl A. Young
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Emily Burt
- Molecular Biosciences, The University of Kansas, Lawrence, KS 66045, USA
| | - Vidhya Munnamalai
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| |
Collapse
|
4
|
Cai W, Huang Z, Sun B, Lu L, Ding X, Tao F. The differentiation of Lgr5+ progenitor cells on nanostructures of self-assembled silica beads. PLoS One 2024; 19:e0304809. [PMID: 38995923 PMCID: PMC11244819 DOI: 10.1371/journal.pone.0304809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 05/19/2024] [Indexed: 07/14/2024] Open
Abstract
Supporting cells(SCs) have been demonstrated to be a reliable source for regenerating hair cells(HCs). Previous research has reported that Lgr5+ SCs can regenerate HCs both in vitro and in vivo. However, there is limited knowledge about the impact of the material on Lgr5+ cells. In this study, Lgr5+ cells were isolated from neonatal Lgr5-EGFP-CreERT2 transgenic mice by flow cytometry and then plated on self-assembled silica beads (SB). Lgr5+ cell differentiation was observed by immunofluorescence. We found that in the direct differentiation assay, the SB group generated more hair cells than the control group(*p < 0.05). Especially in the SB group, Lgr5+ progenitors generated significantly more Myo7a+ HCs outside of the colony than in the control group(**p < 0.01). In the sphere differentiation assay, we found that the diameter of spheres in the SB group was significantly larger compared to those of the control group(**p < 0.01). However, the difference in the ratio of myo7a+ cell counts was not obvious(P>0.05). The experiment proved that the self-assembled silica beads could promote the differentiation of Lgr5+ progenitors in vitro. Our findings implicate that nanostructures of self-assembled silica beads can be used as vectors for stem cell research in the inner ear.
Collapse
Affiliation(s)
- Wenjun Cai
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhong Da Hospital, Southeast University, Nanjing, China
| | - Zhichun Huang
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhong Da Hospital, Southeast University, Nanjing, China
| | - Baobin Sun
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhong Da Hospital, Southeast University, Nanjing, China
| | - Ling Lu
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhong Da Hospital, Southeast University, Nanjing, China
| | - Xiaoqiong Ding
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhong Da Hospital, Southeast University, Nanjing, China
| | - Feng Tao
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhong Da Hospital, Southeast University, Nanjing, China
| |
Collapse
|
5
|
Qi J, Zhang L, Wang X, Chen X, Li Y, Wang T, Wu P, Chai R. Modeling, applications and challenges of inner ear organoid. SMART MEDICINE 2024; 3:e20230028. [PMID: 39188517 PMCID: PMC11235738 DOI: 10.1002/smmd.20230028] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/28/2023] [Indexed: 08/28/2024]
Abstract
More than 6% of the world's population is suffering from hearing loss and balance disorders. The inner ear is the organ that senses sound and balance. Although inner ear disorders are common, there are limited ways to intervene and restore its sensory and balance functions. The development and establishment of biologically therapeutic interventions for auditory disorders require clarification of the basics of signaling pathways that control inner ear development and the establishment of endogenous or exogenous cell-based therapeutic methods. In vitro models of the inner ear, such as organoid systems, can help identify new protective or regenerative drugs, develop new gene therapies, and be considered as potential tools for future clinical applications. Advances in stem cell technology and organoid culture offer unique opportunities for modeling inner ear diseases and developing personalized therapies for hearing loss. Here, we review and discuss the mechanisms for the establishment and the potential applications of inner ear organoids.
Collapse
Affiliation(s)
- Jieyu Qi
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologyAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
- School of Life ScienceBeijing Institute of TechnologyBeijingChina
| | - Liyan Zhang
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologyAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
| | - Xiaohan Wang
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologyAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
| | - Xin Chen
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologyAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
| | - Yiyuan Li
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologyAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
| | - Tian Wang
- Department of Otolaryngology‐Head and Neck SurgeryStanford University School of MedicineStanfordCaliforniaUSA
- Department of Otolaryngology‐Head and Neck SurgeryThe Second Xiangya HospitalCentral South UniversityChangshaHunan ProvinceChina
| | - Peina Wu
- School of MedicineSouth China University of TechnologyGuangzhouChina
- Department of OtolaryngologyGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Renjie Chai
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologyAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjingChina
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
- School of Life ScienceBeijing Institute of TechnologyBeijingChina
- Department of Otolaryngology Head and Neck SurgerySichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduChina
| |
Collapse
|
6
|
Udagawa T, Takahashi E, Tatsumi N, Mutai H, Saijo H, Kondo Y, Atkinson PJ, Matsunaga T, Yoshikawa M, Kojima H, Okabe M, Cheng AG. Loss of Pax3 causes reduction of melanocytes in the developing mouse cochlea. Sci Rep 2024; 14:2210. [PMID: 38278860 PMCID: PMC10817906 DOI: 10.1038/s41598-024-52629-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 01/22/2024] [Indexed: 01/28/2024] Open
Abstract
Cochlear melanocytes are intermediate cells in the stria vascularis that generate endocochlear potentials required for auditory function. Human PAX3 mutations cause Waardenburg syndrome and abnormalities of skin and retinal melanocytes, manifested as congenital hearing loss (~ 70%) and hypopigmentation of skin, hair and eyes. However, the underlying mechanism of hearing loss remains unclear. Cochlear melanocytes in the stria vascularis originated from Pax3-traced melanoblasts and Plp1-traced Schwann cell precursors, both of which derive from neural crest cells. Here, using a Pax3-Cre knock-in mouse that allows lineage tracing of Pax3-expressing cells and disruption of Pax3, we found that Pax3 deficiency causes foreshortened cochlea, malformed vestibular apparatus, and neural tube defects. Lineage tracing and in situ hybridization show that Pax3+ derivatives contribute to S100+, Kir4.1+ and Dct+ melanocytes (intermediate cells) in the developing stria vascularis, all of which are significantly diminished in Pax3 mutant animals. Taken together, these results suggest that Pax3 is required for the development of neural crest cell-derived cochlear melanocytes, whose absence may contribute to congenital hearing loss of Waardenburg syndrome in humans.
Collapse
Affiliation(s)
- Tomokatsu Udagawa
- Department of Otorhinolaryngology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan.
- Department of Anatomy, The Jikei University School of Medicine, Tokyo, Japan.
- Department of Otorhinolaryngology, Toho University School of Medicine, Tokyo, Japan.
| | - Erisa Takahashi
- Department of Otorhinolaryngology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
- Department of Anatomy, The Jikei University School of Medicine, Tokyo, Japan
| | - Norifumi Tatsumi
- Department of Anatomy, The Jikei University School of Medicine, Tokyo, Japan
| | - Hideki Mutai
- Division Hearing and Balance Research, National Institute of Sensory Organs, NHO Tokyo Medical Center, Tokyo, Japan
| | - Hiroki Saijo
- Department of Anatomy, The Jikei University School of Medicine, Tokyo, Japan
| | - Yuko Kondo
- Department of Otorhinolaryngology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Patrick J Atkinson
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Tatsuo Matsunaga
- Division Hearing and Balance Research, National Institute of Sensory Organs, NHO Tokyo Medical Center, Tokyo, Japan
| | - Mamoru Yoshikawa
- Department of Otorhinolaryngology, Toho University School of Medicine, Tokyo, Japan
| | - Hiromi Kojima
- Department of Otorhinolaryngology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Masataka Okabe
- Department of Anatomy, The Jikei University School of Medicine, Tokyo, Japan
| | - Alan G Cheng
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
7
|
Li L, Shen T, Liu S, Qi J, Zhao Y. Advancements and future prospects of adeno-associated virus-mediated gene therapy for sensorineural hearing loss. Front Neurosci 2024; 18:1272786. [PMID: 38327848 PMCID: PMC10847333 DOI: 10.3389/fnins.2024.1272786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/12/2024] [Indexed: 02/09/2024] Open
Abstract
Sensorineural hearing loss (SNHL), a highly prevalent sensory impairment, results from a multifaceted interaction of genetic and environmental factors. As we continually gain insights into the molecular basis of auditory development and the growing compendium of deafness genes identified, research on gene therapy for SNHL has significantly deepened. Adeno-associated virus (AAV), considered a relatively secure vector for gene therapy in clinical trials, can deliver various transgenes based on gene therapy strategies such as gene replacement, gene silencing, gene editing, or gene addition to alleviate diverse types of SNHL. This review delved into the preclinical advances in AAV-based gene therapy for SNHL, spanning hereditary and acquired types. Particular focus is placed on the dual-AAV construction method and its application, the vector delivery route of mouse inner ear models (local, systemic, fetal, and cerebrospinal fluid administration), and the significant considerations in transforming from AAV-based animal model inner ear gene therapy to clinical implementation.
Collapse
Affiliation(s)
- Linke Li
- Department of Otorhinolaryngology Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Tian Shen
- Department of Otorhinolaryngology Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Shixi Liu
- Department of Otorhinolaryngology Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Jieyu Qi
- State Key Laboratory of Bioelectronics, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China
| | - Yu Zhao
- Department of Otorhinolaryngology Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
8
|
Liu Y, Yang L, Singh S, Beyer LA, Prieskorn DM, Swiderski DL, Groves AK, Raphael Y. Combinatorial Atoh1, Gfi1, Pou4f3, and Six1 gene transfer induces hair cell regeneration in the flat epithelium of mature guinea pigs. Hear Res 2024; 441:108916. [PMID: 38103445 PMCID: PMC11223172 DOI: 10.1016/j.heares.2023.108916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/29/2023] [Accepted: 11/14/2023] [Indexed: 12/19/2023]
Abstract
Flat epithelium (FE) is a condition characterized by the loss of both hair cells (HCs) and supporting cells and the transformation of the organ of Corti into a simple flat or cuboidal epithelium, which can occur after severe cochlear insults. The transcription factors Gfi1, Atoh1, Pou4f3, and Six1 (GAPS) play key roles in HC differentiation and survival in normal ears. Previous work using a single transcription factor, Atoh1, to induce HC regeneration in mature ears in vivo usually produced very few cells and failed to produce HCs in severely damaged organs of Corti, especially those with FE. Studies in vitro suggested combinations of transcription factors may be more effective than any single factor, thus the current study aims to examine the effect of co-overexpressing GAPS genes in deafened mature guinea pig cochleae with FE. Deafening was achieved through the infusion of neomycin into the perilymph, leading to the formation of FE and substantial degeneration of nerve fibers. Seven days post neomycin treatment, adenovirus vectors carrying GAPS were injected into the scala media and successfully expressed in the FE. One or two months following GAPS inoculation, cells expressing Myosin VIIa were observed in regions under the FE (located at the scala tympani side of the basilar membrane), rather than within the FE. The number of cells, which we define as induced HCs (iHCs), was not significantly different between one and two months, but the larger N at two months made it more apparent that there were significantly more iHCs in GAPS treated animals than in controls. Additionally, qualitative observations indicated that ears with GAPS gene expression in the FE had more nerve fibers than FE without the treatment. In summary, our results showed that co-overexpression of GAPS enhances the potential for HC regeneration in a severe lesion model of FE.
Collapse
Affiliation(s)
- Yujie Liu
- Kresge Hearing Research Institute, Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, MI, USA; Department of Otolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Ministry of Education Key Laboratory of Otolaryngology-Head and Neck Surgery, Beijing 100730, China
| | - Lin Yang
- Kresge Hearing Research Institute, Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, MI, USA; Department of Otolaryngology-Head and Neck Surgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Sunita Singh
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
| | - Lisa A Beyer
- Kresge Hearing Research Institute, Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Diane M Prieskorn
- Kresge Hearing Research Institute, Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Donald L Swiderski
- Kresge Hearing Research Institute, Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Andrew K Groves
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Yehoash Raphael
- Kresge Hearing Research Institute, Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
9
|
Qi J, Huang W, Lu Y, Yang X, Zhou Y, Chen T, Wang X, Yu Y, Sun JQ, Chai R. Stem Cell-Based Hair Cell Regeneration and Therapy in the Inner Ear. Neurosci Bull 2024; 40:113-126. [PMID: 37787875 PMCID: PMC10774470 DOI: 10.1007/s12264-023-01130-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 06/01/2023] [Indexed: 10/04/2023] Open
Abstract
Hearing loss has become increasingly prevalent and causes considerable disability, thus gravely burdening the global economy. Irreversible loss of hair cells is a main cause of sensorineural hearing loss, and currently, the only relatively effective clinical treatments are limited to digital hearing equipment like cochlear implants and hearing aids, but these are of limited benefit in patients. It is therefore urgent to understand the mechanisms of damage repair in order to develop new neuroprotective strategies. At present, how to promote the regeneration of functional hair cells is a key scientific question in the field of hearing research. Multiple signaling pathways and transcriptional factors trigger the activation of hair cell progenitors and ensure the maturation of newborn hair cells, and in this article, we first review the principal mechanisms underlying hair cell reproduction. We then further discuss therapeutic strategies involving the co-regulation of multiple signaling pathways in order to induce effective functional hair cell regeneration after degeneration, and we summarize current achievements in hair cell regeneration. Lastly, we discuss potential future approaches, such as small molecule drugs and gene therapy, which might be applied for regenerating functional hair cells in the clinic.
Collapse
Affiliation(s)
- Jieyu Qi
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Wenjuan Huang
- Hospital of Southeast University, Nanjing, 210096, China
| | - Yicheng Lu
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Xuehan Yang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Yinyi Zhou
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Tian Chen
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Xiaohan Wang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Yafeng Yu
- First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Jia-Qiang Sun
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| | - Renjie Chai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, 100101, China.
| |
Collapse
|
10
|
Young CA, Burt E, Munnamalai V. Sensory progenitors influence patterning of the mammalian auditory sensory epithelium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.13.566920. [PMID: 38014307 PMCID: PMC10680690 DOI: 10.1101/2023.11.13.566920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
During embryonic development Wnt signaling has been shown to influence proliferation and sensory formation in the cochlea. How the dual nature of Wnt signaling is coordinated is unknown. In this study, we define a novel role for a Wnt regulated gene, Mybl2, which was already known to be important for proliferation, in influencing patterning and determining the size of the sensory epithelium in the murine cochlea. Using a quantitative spatial analysis approach and analyzing Mybl2 loss-of-function cochleas, we show that Mybl2 simultaneously specifies the progenitor niche and the size of the sensory domain, and influences the positioning of the medial sensory domain boundary via Jag1 regulation during the mid-gestational stages. Mybl2 conditional knockout resulted in a decrease of proliferation within the progenitor niche. During the late embryonic stages, conditional knockout of Mybl2 produced a wider sensory epithelium across the radial axis with an increase in ectopic inner hair cell formation. These data suggest that Mybl2 -positive progenitors play a role in boundary formation and patterning the sensory epithelium. Summary Statement Mybl2 is a Wnt-regulated gene encoding a transcription factor that is expressed in the cochlear progenitor niche and influences the boundary formation between the niche and the sensory domain during mid-cochlear developmental stages, thereby impacting the size of the sensory epithelium.
Collapse
|
11
|
Rose KP, Manilla G, Milon B, Zalzman O, Song Y, Coate TM, Hertzano R. Spatially distinct otic mesenchyme cells show molecular and functional heterogeneity patterns before hearing onset. iScience 2023; 26:107769. [PMID: 37720106 PMCID: PMC10502415 DOI: 10.1016/j.isci.2023.107769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 06/29/2023] [Accepted: 08/25/2023] [Indexed: 09/19/2023] Open
Abstract
The cochlea consists of diverse cellular populations working in harmony to convert mechanical stimuli into electrical signals for the perception of sound. Otic mesenchyme cells (OMCs), often considered a homogeneous cell type, are essential for normal cochlear development and hearing. Despite being the most numerous cell type in the developing cochlea, OMCs are poorly understood. OMCs are known to differentiate into spatially and functionally distinct cell types, including fibrocytes of the lateral wall and spiral limbus, modiolar osteoblasts, and specialized tympanic border cells of the basilar membrane. Here, we show that OMCs are transcriptionally and functionally heterogeneous and can be divided into four distinct populations that spatially correspond to OMC-derived cochlear structures. We also show that this heterogeneity and complexity of OMCs commences during early phases of cochlear development. Finally, we describe the cell-cell communication network of the developing cochlea, inferring a major role for OMC in outgoing signaling.
Collapse
Affiliation(s)
- Kevin P. Rose
- Neurotology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gabriella Manilla
- Neurotology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Beatrice Milon
- Neurotology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ori Zalzman
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yang Song
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Thomas M. Coate
- Department of Biology, Georgetown University, Washington, DC 20007, USA
| | - Ronna Hertzano
- Neurotology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Otorhinolaryngology Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
12
|
Petit C, Bonnet C, Safieddine S. Deafness: from genetic architecture to gene therapy. Nat Rev Genet 2023; 24:665-686. [PMID: 37173518 DOI: 10.1038/s41576-023-00597-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2023] [Indexed: 05/15/2023]
Abstract
Progress in deciphering the genetic architecture of human sensorineural hearing impairment (SNHI) or loss, and multidisciplinary studies of mouse models, have led to the elucidation of the molecular mechanisms underlying auditory system function, primarily in the cochlea, the mammalian hearing organ. These studies have provided unparalleled insights into the pathophysiological processes involved in SNHI, paving the way for the development of inner-ear gene therapy based on gene replacement, gene augmentation or gene editing. The application of these approaches in preclinical studies over the past decade has highlighted key translational opportunities and challenges for achieving effective, safe and sustained inner-ear gene therapy to prevent or cure monogenic forms of SNHI and associated balance disorders.
Collapse
Affiliation(s)
- Christine Petit
- Institut Pasteur, Université Paris Cité, Inserm, Institut de l'Audition, F-75012, Paris, France.
- Collège de France, F-75005, Paris, France.
| | - Crystel Bonnet
- Institut Pasteur, Université Paris Cité, Inserm, Institut de l'Audition, F-75012, Paris, France
| | - Saaïd Safieddine
- Institut Pasteur, Université Paris Cité, Inserm, Institut de l'Audition, F-75012, Paris, France
- Centre National de la Recherche Scientifique, F-75016, Paris, France
| |
Collapse
|
13
|
Liu W, Ming S, Zhao X, Zhu X, Gong Y. Developmental expression of high-mobility group box 1 (HMGB1) in the mouse cochlea. Eur J Histochem 2023; 67:3704. [PMID: 37667832 PMCID: PMC10518653 DOI: 10.4081/ejh.2023.3704] [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: 03/09/2023] [Accepted: 08/18/2023] [Indexed: 09/06/2023] Open
Abstract
The expression changes of high-mobility group box 1 (HMGB1) in the mouse cochlea have recently been implicated in noise-induced hearing loss, suggesting that HMGB1 participates in regulating cochlear function. However, the precise role of HMGB1 in the auditory system remains largely unclear. This study aimed to investigate its function in the developing mouse cochlea by examining the expression pattern of HMGB1 in the mouse cochlea from embryonic day (E) 18.5 to postnatal day (P) 28 using double immunofluorescence on frozen sections. Our findings revealed that HMGB1 was extensively expressed in the cell nucleus across various regions of the mouse cochlea, including the organ of Corti. Furthermore, its expression underwent developmental regulation during mouse cochlear development. Specifically, HMGB1 was found to be localized in the tympanic border cells at each developmental stage, coinciding with the gradual anatomical in this region during development. In addition, HMGB1 was expressed in the greater epithelial ridge (GER) and supporting cells of the organ of Corti, as validated by the supporting cell marker Sox2 at P1 and P8. However, at P14, the expression of HMGB1 disappeared from the GER, coinciding with the degeneration of the GER into the inner sulcus cells. Moreover, we observed that HMGB1 co-localized with Ki-67-positive proliferating cells in several cochlear regions during late embryonic and early postnatal stages, including the GER, the tympanic border cells, cochlear lateral wall, and cochlear nerves. Furthermore, by dual-staining Ki-67 with neuronal marker TUJ1 and glial marker Sox10, we determined the expression of Ki-67 in the neonatal glial cells. Our spatial-temporal analysis demonstrated that HMGB1 exhibited distinct expression patterns during mouse cochlear development. The co-localization of HMGB1 with Ki-67-positive proliferating cells suggested that HMGB1 may play a role in cochlear development.
Collapse
Affiliation(s)
- Wenjing Liu
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing.
| | - Shanshan Ming
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing.
| | - Xiaobing Zhao
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing.
| | - Xin Zhu
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing.
| | - Yuxiang Gong
- Department of Nephrology, Zhongda Hospital, Southeast University, Nanjing.
| |
Collapse
|
14
|
Udagawa T, Takahashi E, Tatsumi N, Mutai H, Kondo Y, Atkinson PJ, Matsunaga T, Yoshikawa M, Kojima H, Okabe M, Cheng AG. Pax3 deficiency diminishes melanocytes in the developing mouse cochlea. RESEARCH SQUARE 2023:rs.3.rs-2990436. [PMID: 37333245 PMCID: PMC10274955 DOI: 10.21203/rs.3.rs-2990436/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Cochlear melanocytes are intermediate cells in the stria vascularis that generate endocochlear potentials required for auditory function. Human PAX3 mutations cause Waardenburg syndrome and abnormalities of melanocytes, manifested as congenital hearing loss and hypopigmentation of skin, hair and eyes. However, the underlying mechanism of hearing loss remains unclear. During development, cochlear melanocytes in the stria vascularis are dually derived from Pax3-Cre+ melanoblasts migrating from neuroepithelial cells including neural crest cells and Plp1+ Schwann cell precursors originated from also neural crest cells, differentiating in a basal-apical manner. Here, using a Pax3-Cre mouse line, we found that Pax3 deficiency causes foreshortened cochlea, malformed vestibular apparatus, and neural tube defects. Lineage tracing and in situ hybridization show that Pax3-Cre derivatives contribute to S100+ , Kir4.1+ and Dct+ melanocytes (intermediate cells) in the developing stria vascularis, all significantly diminished in Pax3 mutant animals. Taken together, these results suggest that Pax3 is required for the development of neural crest cell-derived cochlear melanocytes, whose absence may contribute to congenital hearing loss of Waardenburg syndrome in human.
Collapse
|
15
|
Foster T, Lewkowicz M, Quintas C, Ionescu CM, Jones M, Wagle SR, Kovacevic B, Wong EYM, Mooranian A, Al-Salami H. Novel Nanoencapsulation Technology and its Potential Role in Bile Acid-Based Targeted Gene Delivery to the Inner Ear. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2204986. [PMID: 36538754 DOI: 10.1002/smll.202204986] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 10/05/2022] [Indexed: 06/17/2023]
Abstract
Hearing loss impacts a large proportion of the global population. Damage to the inner ear, in particular the sensitive hair cells, can impact individuals for the rest of their lives. There are very limited options for interventions after damage to these cells has occurred. Targeted gene delivery may provide an effective means to trigger appropriate differentiation of progenitor cells for effective replacement of these sensitive hair cells. There are several hurdles that need to be overcome to effectively deliver these genes. Nanoencapsulation technology has previously been used for the delivery of pharmaceuticals, proteins and nucleic acids, and may provide an effective means of delivering genes to trigger appropriate differentiation. This review investigates the background of hearing loss, current advancements and pitfalls of gene delivery, and how nanoencapsulation may be useful.
Collapse
Affiliation(s)
- Thomas Foster
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia
| | - Michael Lewkowicz
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia
| | - Christina Quintas
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
| | - Corina Mihaela Ionescu
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia
| | - Melissa Jones
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia
| | - Susbin Raj Wagle
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia
| | - Bozica Kovacevic
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia
| | - Elaine Y M Wong
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia
| | - Armin Mooranian
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia
- School of Pharmacy, University of Otago, Dunedin, Otago, 9016, New Zealand
| | - Hani Al-Salami
- The Biotechnology and Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia
- Hearing Therapeutics, Ear Science Institute Australia, Queen Elizabeth II Medical Centre, Nedlands 6009, Perth, Western Australia, Australia
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
16
|
Hou S, Zhang J, Wu Y, Junmin C, Yuyu H, He B, Yang Y, Hong Y, Chen J, Yang J, Li S. FGF22 deletion causes hidden hearing loss by affecting the function of inner hair cell ribbon synapses. Front Mol Neurosci 2022; 15:922665. [PMID: 35966010 PMCID: PMC9366910 DOI: 10.3389/fnmol.2022.922665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/04/2022] [Indexed: 12/04/2022] Open
Abstract
Ribbon synapses are important structures in transmitting auditory signals from the inner hair cells (IHCs) to their corresponding spiral ganglion neurons (SGNs). Over the last few decades, deafness has been primarily attributed to the deterioration of cochlear hair cells rather than ribbon synapses. Hearing dysfunction that cannot be detected by the hearing threshold is defined as hidden hearing loss (HHL). The relationship between ribbon synapses and FGF22 deletion remains unknown. In this study, we used a 6-week-old FGF22 knockout mice model (Fgf22–/–) and mainly focused on alteration in ribbon synapses by applying the auditory brainstem response (ABR) test, the immunofluorescence staining, the patch-clamp recording, and quantitative real-time PCR. In Fgf22–/– mice, we found the decreased amplitude of ABR wave I, the reduced vesicles of ribbon synapses, and the decreased efficiency of exocytosis, which was suggested by a decrease in the capacitance change. Quantitative real-time PCR revealed that Fgf22–/– led to dysfunction in ribbon synapses by downregulating SNAP-25 and Gipc3 and upregulating MEF2D expression, which was important for the maintenance of ribbon synapses’ function. Our research concluded that FGF22 deletion caused HHL by affecting the function of IHC ribbon synapses and may offer a novel therapeutic target to meet an ever-growing demand for deafness treatment.
Collapse
Affiliation(s)
- Shule Hou
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Jifang Zhang
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Yan Wu
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Chen Junmin
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Huang Yuyu
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Baihui He
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
| | - Yan Yang
- Liaoning Medical Device Test Institute, Shenyang, China
| | - Yuren Hong
- Laboratory of Electron Microscope Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiarui Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Shanghai Children’s Hospital, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Jiarui Chen,
| | - Jun Yang
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
- Jun Yang,
| | - Shuna Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Ear Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
- Shuna Li,
| |
Collapse
|
17
|
Lin MJ, Lee CM, Hsu WL, Chen BC, Lee SJ. Macrophages Break Interneuromast Cell Quiescence by Intervening in the Inhibition of Schwann Cells in the Zebrafish Lateral Line. Front Cell Dev Biol 2022; 10:907863. [PMID: 35846366 PMCID: PMC9285731 DOI: 10.3389/fcell.2022.907863] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
In the zebrafish lateral line system, interneuromast cells (INCs) between neuromasts are kept quiescent by underlying Schwann cells (SWCs). Upon severe injuries that cause the complete loss of an entire neuromast, INCs can occasionally differentiate into neuromasts but how they escape from the inhibition by SWCs is still unclear. Using a genetic/chemical method to ablate a neuromast precisely, we found that a small portion of larvae can regenerate a new neuromast. However, the residual regeneration capacity was hindered by inhibiting macrophages. Using in toto imaging, we further discovered heterogeneities in macrophage behavior and distribution along the lateral line. We witnessed the crawling of macrophages between the injured lateral line and SWCs during regeneration and between the second primordium and the first mature lateral line during development. It implies that macrophages may physically alleviate the nerve inhibition to break the dormancy of INCs during regeneration and development in the zebrafish lateral line.
Collapse
Affiliation(s)
- Meng-Ju Lin
- Department of Life Science, National Taiwan University, Taipei, Taiwan, R.O.C.
| | - Chia-Ming Lee
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan, R.O.C.
| | - Wei-Lin Hsu
- Department of Life Science, National Taiwan University, Taipei, Taiwan, R.O.C.
| | - Bi-Chang Chen
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan, R.O.C.
| | - Shyh-Jye Lee
- Department of Life Science, National Taiwan University, Taipei, Taiwan, R.O.C.
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan, R.O.C.
- Center for Biotechnology, National Taiwan University, Taipei, Taiwan, R.O.C.
- *Correspondence: Shyh-Jye Lee,
| |
Collapse
|
18
|
Abstract
It is well established that humans and other mammals are minimally regenerative compared with organisms such as zebrafish, salamander or amphibians. In recent years, however, the identification of regenerative potential in neonatal mouse tissues that normally heal poorly in adults has transformed our understanding of regenerative capacity in mammals. In this Review, we survey the mammalian tissues for which regenerative or improved neonatal healing has been established, including the heart, cochlear hair cells, the brain and spinal cord, and dense connective tissues. We also highlight common and/or tissue-specific mechanisms of neonatal regeneration, which involve cells, signaling pathways, extracellular matrix, immune cells and other factors. The identification of such common features across neonatal tissues may direct therapeutic strategies that will be broadly applicable to multiple adult tissues.
Collapse
Affiliation(s)
| | - Alice H. Huang
- Department of Orthopedic Surgery, Columbia University, New York, NY 10032, USA
| |
Collapse
|
19
|
Rai V, Tu S, Frank JR, Zuo J. Molecular Pathways Modulating Sensory Hair Cell Regeneration in Adult Mammalian Cochleae: Progress and Perspectives. Int J Mol Sci 2021; 23:ijms23010066. [PMID: 35008497 PMCID: PMC8745006 DOI: 10.3390/ijms23010066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/20/2021] [Accepted: 12/20/2021] [Indexed: 12/30/2022] Open
Abstract
Noise-induced, drug-related, and age-related disabling hearing loss is a major public health problem and affect approximately 466 million people worldwide. In non-mammalian vertebrates, the death of sensory hair cells (HCs) induces the proliferation and transdifferentiation of adjacent supporting cells into new HCs; however, this capacity is lost in juvenile and adult mammalian cochleae leading to permanent hearing loss. At present, cochlear implants and hearing devices are the only available treatments and can help patients to a certain extent; however, no biological approach or FDA-approved drug is effective to treat disabling hearing loss and restore hearing. Recently, regeneration of mammalian cochlear HCs by modulating molecular pathways or transcription factors has offered some promising results, although the immaturity of the regenerated HCs remains the biggest concern. Furthermore, most of the research done is in neonates and not in adults. This review focuses on critically summarizing the studies done in adult mammalian cochleae and discusses various strategies to elucidate novel transcription factors for better therapeutics.
Collapse
Affiliation(s)
| | | | | | - Jian Zuo
- Correspondence: ; Tel.: +1-(402)-280-2916
| |
Collapse
|
20
|
Xu S, Yang N. Research Progress on the Mechanism of Cochlear Hair Cell Regeneration. Front Cell Neurosci 2021; 15:732507. [PMID: 34489646 PMCID: PMC8417573 DOI: 10.3389/fncel.2021.732507] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 07/28/2021] [Indexed: 12/26/2022] Open
Abstract
Mammalian inner ear hair cells do not have the ability to spontaneously regenerate, so their irreversible damage is the main cause of sensorineural hearing loss. The damage and loss of hair cells are mainly caused by factors such as aging, infection, genetic factors, hypoxia, autoimmune diseases, ototoxic drugs, or noise exposure. In recent years, research on the regeneration and functional recovery of mammalian auditory hair cells has attracted more and more attention in the field of auditory research. How to regenerate and protect hair cells or auditory neurons through biological methods and rebuild auditory circuits and functions are key scientific issues that need to be resolved in this field. This review mainly summarizes and discusses the recent research progress in gene therapy and molecular mechanisms related to hair cell regeneration in the field of sensorineural hearing loss.
Collapse
Affiliation(s)
- Shan Xu
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, China
| | - Ning Yang
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
21
|
Key Signaling Pathways Regulate the Development and Survival of Auditory Hair Cells. Neural Plast 2021; 2021:5522717. [PMID: 34194486 PMCID: PMC8214483 DOI: 10.1155/2021/5522717] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/01/2021] [Accepted: 05/31/2021] [Indexed: 01/16/2023] Open
Abstract
The loss of auditory sensory hair cells (HCs) is the most common cause of sensorineural hearing loss (SNHL). As the main sound transmission structure in the cochlea, it is necessary to maintain the normal shape and survival of HCs. In this review, we described and summarized the signaling pathways that regulate the development and survival of auditory HCs in SNHL. The role of the mitogen-activated protein kinase (MAPK), phosphoinositide-3 kinase/protein kinase B (PI3K/Akt), Notch/Wnt/Atoh1, calcium channels, and oxidative stress/reactive oxygen species (ROS) signaling pathways are the most relevant. The molecular interactions of these signaling pathways play an important role in the survival of HCs, which may provide a theoretical basis and possible therapeutic interventions for the treatment of hearing loss.
Collapse
|
22
|
Billings SE, Myers NM, Quiruz L, Cheng AG. Opposing effects of Wnt/β-catenin signaling on epithelial and mesenchymal cell fate in the developing cochlea. Development 2021; 148:268974. [PMID: 34061174 PMCID: PMC8217710 DOI: 10.1242/dev.199091] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 05/05/2021] [Indexed: 12/12/2022]
Abstract
During embryonic development, the otic epithelium and surrounding periotic mesenchymal cells originate from distinct lineages and coordinate to form the mammalian cochlea. Epithelial sensory precursors within the cochlear duct first undergo terminal mitosis before differentiating into sensory and non-sensory cells. In parallel, periotic mesenchymal cells differentiate to shape the lateral wall, modiolus and pericochlear spaces. Previously, Wnt activation was shown to promote proliferation and differentiation of both otic epithelial and mesenchymal cells. Here, we fate-mapped Wnt-responsive epithelial and mesenchymal cells in mice and found that Wnt activation resulted in opposing cell fates. In the post-mitotic cochlear epithelium, Wnt activation via β-catenin stabilization induced clusters of proliferative cells that dedifferentiated and lost epithelial characteristics. In contrast, Wnt-activated periotic mesenchyme formed ectopic pericochlear spaces and cell clusters showing a loss of mesenchymal and gain of epithelial features. Finally, clonal analyses via multi-colored fate-mapping showed that Wnt-activated epithelial cells proliferated and formed clonal colonies, whereas Wnt-activated mesenchymal cells assembled as aggregates of mitotically quiescent cells. Together, we show that Wnt activation drives transition between epithelial and mesenchymal states in a cell type-dependent manner. Summary: The developing cochlea comprises spatially and lineally distinct populations of epithelial and mesenchymal cells. This study shows the opposing effects of aberrant Wnt/β-catenin signaling on cell fates of cochlear epithelial and mesenchymal cells.
Collapse
Affiliation(s)
- Sara E Billings
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nina M Myers
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lee Quiruz
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alan G Cheng
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
23
|
Canonical Wnt Signaling Pathway on Polarity Formation of Utricle Hair Cells. Neural Plast 2021; 2021:9950533. [PMID: 34122536 PMCID: PMC8166501 DOI: 10.1155/2021/9950533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/26/2021] [Accepted: 05/11/2021] [Indexed: 02/05/2023] Open
Abstract
As part of the inner ear, the vestibular system is responsible for sense of balance, which consists of three semicircular canals, the utricle, and the saccule. Increasing evidence has indicated that the noncanonical Wnt/PCP signaling pathway plays a significant role in the development of the polarity of the inner ear. However, the role of canonical Wnt signaling in the polarity of the vestibule is still not completely clear. In this study, we found that canonical Wnt pathway-related genes are expressed in the early stage of development of the utricle and change dynamically. We conditionally knocked out β-catenin, a canonical Wnt signaling core protein, and found that the cilia orientation of hair cells was disordered with reduced number of hair cells in the utricle. Moreover, regulating the canonical Wnt pathway (Licl and IWP2) in vitro also affected hair cell polarity and indicated that Axin2 may be important in this process. In conclusion, our results not only confirm that the regulation of canonical Wnt signaling affects the number of hair cells in the utricle but also provide evidence for its role in polarity development.
Collapse
|
24
|
Wang J, Song W, Yang R, Li C, Wu T, Dong XB, Zhou B, Guo X, Chen J, Liu Z, Yu QC, Li W, Fu J, Zeng YA. Endothelial Wnts control mammary epithelial patterning via fibroblast signaling. Cell Rep 2021; 34:108897. [PMID: 33789106 DOI: 10.1016/j.celrep.2021.108897] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/09/2020] [Accepted: 03/02/2021] [Indexed: 12/12/2022] Open
Abstract
Endothelial and fibroblast niches are crucial for epithelial organs. How these heterotypic cells interact is of great interest. In this study, we reveal an axis of signaling in which fibroblasts relay Wnt signals from the endothelial niche to organize epithelial patterning. We generate an Axin2-membrane GFP (mGFP) reporter mouse and observe robust Wnt/β-catenin signaling activities in fibroblasts surrounding the mammary epithelium. To enable cell-type-specific gene manipulation in vitro, we establish an organoid system via coculture of endothelial cells (ECs), fibroblasts, and mammary epithelial cells. Deletion of β-catenin in fibroblasts impedes epithelium branching, and ECs are responsible for the activation of Wnt/β-catenin signaling in fibroblasts. In vivo, EC deletion of Wntless inhibits Wnt/β-catenin signaling activity in fibroblasts, rendering a reduction in epithelial branches. These findings highlight the significance of the endothelial niche in tissue patterning, shedding light on the interactive mechanisms in which distinct niche components orchestrate epithelial organogenesis and tissue homeostasis.
Collapse
Affiliation(s)
- Jingqiang Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou 310052, China
| | - Wenqian Song
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China
| | - Ruikai Yang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Chao Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ting Wu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiao Bing Dong
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Bin Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China
| | - Xizhi Guo
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jianfeng Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China
| | - Zhiyong Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qing Cissy Yu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.
| | - Wen Li
- Center of reproductive medicine, Shanghai Key Laboratory of Embryo Original Diseases, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Junfen Fu
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou 310052, China.
| | - Yi Arial Zeng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China.
| |
Collapse
|
25
|
Azbazdar Y, Karabicici M, Erdal E, Ozhan G. Regulation of Wnt Signaling Pathways at the Plasma Membrane and Their Misregulation in Cancer. Front Cell Dev Biol 2021; 9:631623. [PMID: 33585487 PMCID: PMC7873896 DOI: 10.3389/fcell.2021.631623] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/04/2021] [Indexed: 12/24/2022] Open
Abstract
Wnt signaling is one of the key signaling pathways that govern numerous physiological activities such as growth, differentiation and migration during development and homeostasis. As pathway misregulation has been extensively linked to pathological processes including malignant tumors, a thorough understanding of pathway regulation is essential for development of effective therapeutic approaches. A prominent feature of cancer cells is that they significantly differ from healthy cells with respect to their plasma membrane composition and lipid organization. Here, we review the key role of membrane composition and lipid order in activation of Wnt signaling pathway by tightly regulating formation and interactions of the Wnt-receptor complex. We also discuss in detail how plasma membrane components, in particular the ligands, (co)receptors and extracellular or membrane-bound modulators, of Wnt pathways are affected in lung, colorectal, liver and breast cancers that have been associated with abnormal activation of Wnt signaling. Wnt-receptor complex components and their modulators are frequently misexpressed in these cancers and this appears to correlate with metastasis and cancer progression. Thus, composition and organization of the plasma membrane can be exploited to develop new anticancer drugs that are targeted in a highly specific manner to the Wnt-receptor complex, rendering a more effective therapeutic outcome possible.
Collapse
Affiliation(s)
- Yagmur Azbazdar
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, İzmir, Turkey.,Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, İzmir, Turkey
| | - Mustafa Karabicici
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, İzmir, Turkey.,Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, İzmir, Turkey
| | - Esra Erdal
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, İzmir, Turkey.,Department of Medical Biology and Genetics, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Gunes Ozhan
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, İzmir, Turkey.,Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, İzmir, Turkey
| |
Collapse
|
26
|
An In Vitro Study on Prestin Analog Gene in the Bullfrog Hearing Organs. Neural Plast 2020; 2020:3570732. [PMID: 32714383 PMCID: PMC7352134 DOI: 10.1155/2020/3570732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/22/2020] [Accepted: 05/20/2020] [Indexed: 11/18/2022] Open
Abstract
The prestin-based active process in the mammalian outer hair cells (OHCs) is believed to play a crucial role in auditory signal amplification in the cochlea. Prestin belongs to an anion transporter family (SLC26A). It is densely expressed in the OHC lateral plasma membrane and functions as a voltage-dependent motor protein. Analog genes can be found in the genome of nonmammalian species, but their functions in hearing are poorly understood. In the present study, we used the gerbil prestin sequence as a template and identified an analog gene in the bullfrog genome. We expressed the gene in a stable cell line (HEK293T) and performed patch-clamp recording. We found that these cells exhibited prominent nonlinear capacitance (NLC), a widely accepted assay for prestin functioning as a motor protein. Upon close examination, the key parameters of this NLC are comparable to that conferred by the gerbil prestin, and nontransfected cells failed to display NLC. Lastly, we performed patch-clamp recording in HCs of all three hearing organs in bullfrog. HCs in both the sacculus and the amphibian papilla exhibited a capacitance profile that is similar to NLC while HCs in the basilar papilla showed no sign of NLC. Whether or not this NLC-like capacitance change is involved in auditory signal amplification certainly requires further examination; our results represent the first and necessary step in revealing possible roles of prestin in the active hearing processes found in many nonmammalian species.
Collapse
|
27
|
Durán-Alonso MB. Stem cell-based approaches: Possible route to hearing restoration? World J Stem Cells 2020; 12:422-437. [PMID: 32742560 PMCID: PMC7360988 DOI: 10.4252/wjsc.v12.i6.422] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/08/2020] [Accepted: 05/21/2020] [Indexed: 02/06/2023] Open
Abstract
Disabling hearing loss is the most common sensorineural disability worldwide. It affects around 466 million people and its incidence is expected to rise to around 900 million people by 2050, according to World Health Organization estimates. Most cases of hearing impairment are due to the degeneration of hair cells (HCs) in the cochlea, mechano-receptors that transduce incoming sound information into electrical signals that are sent to the brain. Damage to these cells is mainly caused by exposure to aminoglycoside antibiotics and to some anti-cancer drugs such as cisplatin, loud sounds, age, infections and genetic mutations. Hearing deficits may also result from damage to the spiral ganglion neurons that innervate cochlear HCs. Differently from what is observed in avian and non-mammalian species, there is no regeneration of missing sensory cell types in the adult mammalian cochlea, what makes hearing loss an irreversible process. This review summarizes the research that has been conducted with the aim of developing cell-based strategies that lead to sensory cell replacement in the adult cochlea and, ultimately, to hearing restoration. Two main lines of research are discussed, one directed toward the transplantation of exogenous replacement cells into the damaged tissue, and another that aims at reactivating the regenerative potential of putative progenitor cells in the adult inner ear. Results from some of the studies that have been conducted are presented and the advantages and drawbacks of the various approaches discussed.
Collapse
|
28
|
Zhang S, Qiang R, Dong Y, Zhang Y, Chen Y, Zhou H, Gao X, Chai R. Hair cell regeneration from inner ear progenitors in the mammalian cochlea. AMERICAN JOURNAL OF STEM CELLS 2020; 9:25-35. [PMID: 32699655 PMCID: PMC7364385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/10/2020] [Indexed: 06/11/2023]
Abstract
Cochlear hair cells (HCs) are the mechanoreceptors of the auditory system, and because these cells cannot be spontaneously regenerated in adult mammals, hearing loss due to HC damage is permanent. However, cochleae of neonatal mice harbor some progenitor cells that retain limited ability to give rise to new HCs in vivo. Here we review the regulatory factors, signaling pathways, and epigenetic factors that have been reported to play roles in HC regeneration in the neonatal mammalian cochlea.
Collapse
Affiliation(s)
- Shasha Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing 210096, China
| | - Ruiying Qiang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing 210096, China
| | - Ying Dong
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing 210096, China
| | - Yuan Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing 210096, China
| | - Yin Chen
- Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory)Nanjing 210008, China
| | - Han Zhou
- Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory)Nanjing 210008, China
| | - Xia Gao
- Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory)Nanjing 210008, China
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing 210096, China
- Co-Innovation Center of Neuroregeneration, Nantong UniversityNantong 226001, China
- Institute for Stem Cell and Regeneration, Chinese Academy of ScienceBeijing, China
- Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast UniversityNanjing 211189, China
- Department of Otolaryngology Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiangsu Provincial Key Medical Discipline (Laboratory)Nanjing 210008, China
| |
Collapse
|
29
|
He D, Guo R, Zheng D, Xu M, Li P, Guo L, Gan L. Transcription factor Isl1 is dispensable for the development of the mouse prosensory region. Cytotechnology 2020; 72:407-414. [PMID: 32219582 DOI: 10.1007/s10616-020-00387-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 02/28/2020] [Indexed: 01/17/2023] Open
Abstract
In order to identify genes involved in the development of inner ear hair cells, we investigated the role of the transcription factor Islet-class LIM-homeodomain (LIM-HD) 1 (Isl1) in the development of the mouse prosensory region. Isl1 was deleted using the Pax2-Cre system, and deletion of both alleles was verified using cochlea sections. Changes in the number of prosensory region cells were measured to determine the effect of Isl1 on the development of the mouse prosensory region. In order to test whether Isl1 formed a protein complex with Ldb1 and Gata3, co-immunoprecipitation experiments were performed in HEK293 cells using the Flag-tagged LIM-domain of Isl1, HA-tagged LID of Ldb1 and Myc-tagged C-terminal domain of Gata3. The expression of Gata3, Sox2, Jag1 and P27 proteins in the prosensory region were not affected in Isl1-/- prosensory cells. Thus, Isl1 did not form a protein complex with Gata3 through Ldb1 in the Isl1-/- cells. Our results suggest that Isl1 may be dispensable for the development of the mouse prosensory region.
Collapse
Affiliation(s)
- Daqiang He
- College of Life Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Institute of Life Sciences, Hangzhou Normal University, Hangzhou, 310018, Zhejiang, China
- Department of Laboratory Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Rui Guo
- College of Life Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Dongwang Zheng
- Department of Reproductive Physiology, Zhejiang Academy of Medical Sciences, HangZhou Medical College, Hangzhou, 310000, Zhejiang, China
| | - Mei Xu
- Institute of Life Sciences, Hangzhou Normal University, Hangzhou, 310018, Zhejiang, China
| | - Ping Li
- HangZhou CalyGene Bitechnology Limited Company, Hangzhou, 310013, Zhejiang, China
| | - Luming Guo
- College of Life Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Lin Gan
- College of Life Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
- Institute of Life Sciences, Hangzhou Normal University, Hangzhou, 310018, Zhejiang, China.
| |
Collapse
|
30
|
Zhang S, Liu D, Dong Y, Zhang Z, Zhang Y, Zhou H, Guo L, Qi J, Qiang R, Tang M, Gao X, Zhao C, Chen X, Qian X, Chai R. Frizzled-9+ Supporting Cells Are Progenitors for the Generation of Hair Cells in the Postnatal Mouse Cochlea. Front Mol Neurosci 2019; 12:184. [PMID: 31427926 PMCID: PMC6689982 DOI: 10.3389/fnmol.2019.00184] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/12/2019] [Indexed: 01/27/2023] Open
Abstract
Lgr5+ cochlear supporting cells (SCs) have been reported to be hair cell (HC) progenitor cells that have the ability to regenerate HCs in the neonatal mouse cochlea, and these cells are regulated by Wnt signaling. Frizzled-9 (Fzd9), one of the Wnt receptors, has been reported to be used to mark neuronal stem cells in the brain together with other markers and mesenchymal stem cells from human placenta and bone marrow. Here we used Fzd9-CreER mice to lineage label and trace Fzd9+ cells in the postnatal cochlea in order to investigate the progenitor characteristic of Fzd9+ cells. Lineage labeling showed that inner phalangeal cells (IPhCs), inner border cells (IBCs), and third-row Deiters’ cells (DCs) were Fzd9+ cells, but not inner pillar cells (IPCs) or greater epithelial ridge (GER) cells at postnatal day (P)3, which suggests that Fzd9+ cells are a much smaller cell population than Lgr5+ progenitors. The expression of Fzd9 progressively decreased and was too low to allow lineage tracing after P14. Lineage tracing for 6 days in vivo showed that Fzd9+ cells could also generate similar numbers of new HCs compared to Lgr5+ progenitors. A sphere-forming assay showed that Fzd9+ cells could form spheres after sorting by flow cytometry, and when we compared the isolated Fzd9+ cells and Lgr5+ progenitors there were no significant differences in sphere number or sphere diameter. In a differentiation assay, the same number of Fzd9+ cells could produce similar amounts of Myo7a+ cells compared to Lgr5+ progenitors after 10 days of differentiation. All these data suggest that the Fzd9+ cells have a similar capacity for proliferation, differentiation, and HC generation as Lgr5+ progenitors and that Fzd9 can be used as a more restricted marker of HC progenitors.
Collapse
Affiliation(s)
- Shasha Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Dingding Liu
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology-Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Ying Dong
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Zhong Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Yuan Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Han Zhou
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology-Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Lingna Guo
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Jieyu Qi
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Ruiying Qiang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Mingliang Tang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Xia Gao
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology-Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Chunjie Zhao
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Xiaoyun Chen
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoyun Qian
- Jiangsu Provincial Key Medical Discipline (Laboratory), Department of Otolaryngology-Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Science, Beijing, China.,Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China.,Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| |
Collapse
|
31
|
Lush ME, Diaz DC, Koenecke N, Baek S, Boldt H, St Peter MK, Gaitan-Escudero T, Romero-Carvajal A, Busch-Nentwich EM, Perera AG, Hall KE, Peak A, Haug JS, Piotrowski T. scRNA-Seq reveals distinct stem cell populations that drive hair cell regeneration after loss of Fgf and Notch signaling. eLife 2019; 8:e44431. [PMID: 30681411 PMCID: PMC6363392 DOI: 10.7554/elife.44431] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 01/24/2019] [Indexed: 12/25/2022] Open
Abstract
Loss of sensory hair cells leads to deafness and balance deficiencies. In contrast to mammalian hair cells, zebrafish ear and lateral line hair cells regenerate from poorly characterized support cells. Equally ill-defined is the gene regulatory network underlying the progression of support cells to differentiated hair cells. scRNA-Seq of lateral line organs uncovered five different support cell types, including quiescent and activated stem cells. Ordering of support cells along a developmental trajectory identified self-renewing cells and genes required for hair cell differentiation. scRNA-Seq analyses of fgf3 mutants, in which hair cell regeneration is increased, demonstrates that Fgf and Notch signaling inhibit proliferation of support cells in parallel by inhibiting Wnt signaling. Our scRNA-Seq analyses set the foundation for mechanistic studies of sensory organ regeneration and is crucial for identifying factors to trigger hair cell production in mammals. The data is searchable and publicly accessible via a web-based interface.
Collapse
Affiliation(s)
- Mark E Lush
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Daniel C Diaz
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Nina Koenecke
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Sungmin Baek
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Helena Boldt
- Stowers Institute for Medical ResearchKansas CityUnited States
| | | | | | - Andres Romero-Carvajal
- Stowers Institute for Medical ResearchKansas CityUnited States
- Pontificia Universidad Catolica del EcuadorCiencias BiologicasQuitoEcuador
| | - Elisabeth M Busch-Nentwich
- Wellcome Sanger Institute, Wellcome Genome CampusHinxtonUnited Kingdom
- Department of MedicineUniversity of CambridgeCambridgeUnited Kingdom
| | - Anoja G Perera
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Kathryn E Hall
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Allison Peak
- Stowers Institute for Medical ResearchKansas CityUnited States
| | - Jeffrey S Haug
- Stowers Institute for Medical ResearchKansas CityUnited States
| | | |
Collapse
|
32
|
Ortiz-Matamoros A, Arias C. Differential Changes in the Number and Morphology of the New Neurons after Chronic Infusion of Wnt7a, Wnt5a, and Dkk-1 in the Adult Hippocampus In Vivo. Anat Rec (Hoboken) 2019; 302:1647-1657. [PMID: 30635974 DOI: 10.1002/ar.24069] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 11/15/2018] [Accepted: 11/29/2018] [Indexed: 12/13/2022]
Abstract
In the adult hippocampus of many mammals, a particular microenvironment in the neurogenic niche regulates the proliferation, self-renewal, and differentiation of neuronal stem cells. In this proliferative niche, a variety of molecules provide a finely regulated molecular signaling that controls stem cell properties. During development, Wnt signaling has been implicated in cell fate determination and proliferation, in the establishment of cell polarity, as well as a cue for axonal growth and dendrite orientation. In the adult brain, this pathway also participates in the stem cell self-renewal and neuronal differentiation. However, the effects of the chronic Wnt signaling modulation in the adult hippocampus, through the infusion of Wnt7a, Wnt5a, and Dkk-1, on the rate of neurogenesis and on the induction of neurite arborization have not been studied. In this study, we show that Wnt7a and Wn5a further increased the rate of newly generated neurons. However, Wnt5a exerted additional effects by promoting neurite growth and neurite misorientation in the dentate gyrus of adult rats. The chronic exposure to Dkk-1 also generated aberrant location of growing neurites. These results suggest that the interplay of canonical and non-canonical Wnt ligands participates in neuronal stem cell proliferation and in the establishment of proper neurite maturation. Anat Rec, 302:1647-1657, 2019. © 2019 American Association for Anatomy.
Collapse
Affiliation(s)
- Abril Ortiz-Matamoros
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, AP, Mexico
| | - Clorinda Arias
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, AP, Mexico
| |
Collapse
|
33
|
|
34
|
Zhang J, Sun H, Salvi R, Ding D. Paraquat initially damages cochlear support cells leading to anoikis-like hair cell death. Hear Res 2018; 364:129-141. [PMID: 29563067 PMCID: PMC5984146 DOI: 10.1016/j.heares.2018.03.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/20/2018] [Accepted: 03/09/2018] [Indexed: 12/11/2022]
Abstract
Paraquat (PQ), one of the most widely used herbicides, is extremely dangerous because it generates the highly toxic superoxide radical. When paraquat was applied to cochlear organotypic cultures, it not only damaged the outer hair cells (OHCs) and inner hair cells (IHCs), but also caused dislocation of the hair cell rows. We hypothesized that the dislocation arose from damage to the support cells (SCs) that anchors hair cells within the epithelium. To test this hypothesis, rat postnatal cochlear cultures were treated with PQ. Shortly after PQ treatment, the rows of OHCs separated from one another and migrated radially away from IHCs suggesting loss of cell-cell adhesion that hold the hair cells in proper alignment. Hair cells dislocation was associated with extensive loss of SCs in the organ of Corti, loss of tympanic border cells (TBCs) beneath the basilar membrane, the early appearance of superoxide staining and caspase-8 labeling in SCs below the OHCs and disintegration of E-cadherin and β-catenin in the organ of Corti. Damage to the TBCs and SCs occurred prior to loss of OHC or IHC loss suggesting a form of detachment-induced apoptosis referred to as anoikis.
Collapse
Affiliation(s)
- Jianhui Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, China; Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, 14214, USA
| | - Hong Sun
- Department of Otorhinolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, China; Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, 14214, USA
| | - Richard Salvi
- Department of Otorhinolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, China; Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, 14214, USA; Department of Audiology and Speech-Language Pathology, Asia University, Taichung, Taiwan, ROC
| | - Dalian Ding
- Department of Otorhinolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, China; Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, 14214, USA.
| |
Collapse
|
35
|
Abstract
Programmable nucleases can introduce precise changes to genomic DNA through homology-directed repair (HDR). Unfortunately, HDR is largely restricted to mitotic cells, and is typically accompanied by an excess of stochastic insertions and deletions (indels). Here we present an in vivo base editing strategy that addresses these limitations. We use nuclease-free base editing to install a S33F mutation in β-catenin that blocks β-catenin phosphorylation, impedes β-catenin degradation, and upregulates Wnt signaling. In vitro, base editing installs the S33F mutation with a 200-fold higher editing:indel ratio than HDR. In post-mitotic cells in mouse inner ear, injection of base editor protein:RNA:lipid installs this mutation, resulting in Wnt activation that induces mitosis of cochlear supporting cells and cellular reprogramming. In contrast, injection of HDR agents does not induce Wnt upregulation. These results establish a strategy for modifying posttranslational states in signaling pathways, and an approach to precision editing in post-mitotic tissues. Base editing allows the precise introduction of point mutations into cellular DNA without requiring double-stranded DNA breaks or homology-directed repair, which is inefficient in postmitotic cells. Here the authors demonstrate in vivo base editing of post-mitotic somatic cells in the postnatal mouse inner ear with physiological outcomes.
Collapse
|
36
|
Zhang Y, Guo L, Lu X, Cheng C, Sun S, Li W, Zhao L, Lai C, Zhang S, Yu C, Tang M, Chen Y, Chai R, Li H. Characterization of Lgr6+ Cells as an Enriched Population of Hair Cell Progenitors Compared to Lgr5+ Cells for Hair Cell Generation in the Neonatal Mouse Cochlea. Front Mol Neurosci 2018; 11:147. [PMID: 29867341 PMCID: PMC5961437 DOI: 10.3389/fnmol.2018.00147] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 04/12/2018] [Indexed: 12/20/2022] Open
Abstract
Hair cell (HC) loss is irreversible because only very limited HC regeneration has been observed in the adult mammalian cochlea. Wnt/β-catenin signaling regulates prosensory cell proliferation and differentiation during cochlear development, and Wnt activation promotes the proliferation of Lgr5+ cochlear HC progenitors in newborn mice. Similar to Lgr5, Lgr6 is also a Wnt downstream target gene. Lgr6 is reported to be present in adult stem cells in the skin, nail, tongue, lung, and mammary gland, and this protein is very important for adult stem cell maintenance in rapidly proliferating organs. Our previous studies showed that Lgr6+ cells are a subpopulation of Lgr5+ progenitor cells and that both Lgr6+ and Lgr5+ progenitors can generate Myosin7a+ HCs in vitro. Thus we hypothesized that Lgr6+ cells are an enriched population of cochlear progenitor cells. However, the detailed distinctions between the Lgr5+ and Lgr6+ progenitors are unclear. Here, we systematically compared the proliferation, HC differentiation, and detailed transcriptome expression profiles of these two progenitor populations. We found that the same number of isolated Lgr6+ progenitors generated significantly more Myosin7a+ HCs compared to Lgr5+ progenitors; however, Lgr5+ progenitors formed more epithelial colonies and more spheres than Lgr6+ progenitors in vitro. Using RNA-Seq, we compared the transcriptome differences between Lgr5+ and Lgr6+ progenitors and identified a list of significantly differential expressed genes that might regulate the proliferation and differentiation of these HC progenitors, including 4 cell cycle genes, 9 cell signaling pathway genes, and 54 transcription factors. In conclusion, we demonstrate that Lgr6+ progenitors are an enriched population of inner ear progenitors that generate more HCs compared to Lgr5+ progenitors in the newborn mouse cochlea, and the our research provides a series of genes that might regulate the proliferation of progenitors and HC generation.
Collapse
Affiliation(s)
- Yanping Zhang
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Luo Guo
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Xiaoling Lu
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Cheng Cheng
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Shan Sun
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Wen Li
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Liping Zhao
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Chuijin Lai
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Shasha Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Chenjie Yu
- Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline Laboratory, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Mingliang Tang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Yan Chen
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China.,Jiangsu Province Hi-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Huawei Li
- ENT Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine, National Health and Family Planning Commission (NHFPC), Shanghai, China.,Shanghai Engineering Research Center of Cochlear Implant, Shanghai, China.,The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
37
|
Atkinson PJ, Dong Y, Gu S, Liu W, Najarro EH, Udagawa T, Cheng AG. Sox2 haploinsufficiency primes regeneration and Wnt responsiveness in the mouse cochlea. J Clin Invest 2018; 128:1641-1656. [PMID: 29553487 DOI: 10.1172/jci97248] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 02/01/2018] [Indexed: 12/31/2022] Open
Abstract
During development, Sox2 is indispensable for cell division and differentiation, yet its roles in regenerating tissues are less clear. Here, we used combinations of transgenic mouse models to reveal that Sox2 haploinsufficiency (Sox2haplo) increases rather than impairs cochlear regeneration in vivo. Sox2haplo cochleae had delayed terminal mitosis and ectopic sensory cells, yet normal auditory function. Sox2haplo amplified and expanded domains of damage-induced Atoh1+ transitional cell formation in neonatal cochlea. Wnt activation via β-catenin stabilization (β-cateninGOF) alone failed to induce proliferation or transitional cell formation. By contrast, β-cateninGOF caused proliferation when either Sox2haplo or damage was present, and transitional cell formation when both were present in neonatal, but not mature, cochlea. Mechanistically, Sox2haplo or damaged neonatal cochleae showed lower levels of Sox2 and Hes5, but not of Wnt target genes. Together, our study unveils an interplay between Sox2 and damage in directing tissue regeneration and Wnt responsiveness and thus provides a foundation for potential combinatorial therapies aimed at stimulating mammalian cochlear regeneration to reverse hearing loss in humans.
Collapse
Affiliation(s)
- Patrick J Atkinson
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Yaodong Dong
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, USA.,Department of Otology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shuping Gu
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Wenwen Liu
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Elvis Huarcaya Najarro
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Tomokatsu Udagawa
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Alan G Cheng
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
38
|
Luo WW, Wang XW, Ma R, Chi FL, Chen P, Cong N, Gu YY, Ren DD, Yang JM. Junctional E-cadherin/p120-catenin Is Correlated with the Absence of Supporting Cells to Hair Cells Conversion in Postnatal Mice Cochleae. Front Mol Neurosci 2018. [PMID: 29515364 PMCID: PMC5826362 DOI: 10.3389/fnmol.2018.00020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Notch inhibition is known to generate supernumerary hair cells (HCs) at the expense of supporting cells (SCs) in the mammalian inner ear. However, inhibition of Notch activity becomes progressively less effective at inducing SC-to-HC conversion in the postnatal cochlea and balance organs as the animal ages. It has been suggested that the SC-to-HC conversion capacity is inversely correlated with E-cadherin accumulation in postnatal mammalian utricles. However, whether E-cadherin localization is linked to the SC-to-HC conversion capacity in the mammalian inner ear is poorly understood. In the present study, we treated cochleae from postnatal day 0 (P0) with the Notch signaling inhibitor DAPT and observed apparent SC-to-HC conversion along with E-cadherin/p120ctn disruption in the sensory region. In addition, the SC-to-HC conversion capacity and E-cadherin/p120ctn disorganization were robust in the apex but decreased toward the base. We further demonstrated that the ability to regenerate HCs and the disruption of E-cadherin/p120ctn concomitantly decreased with age and ceased at P7, even after extended DAPT treatments. This timing is consistent with E-cadherin/p120ctn accumulation in the postnatal cochleae. These results suggest that the decreasing capacity of SCs to transdifferentiate into HCs correlates with E-cadherin/p120ctn localization in the postnatal cochleae, which might account for the absence of SC-to-HC conversion in the mammalian cochlea.
Collapse
Affiliation(s)
- Wen-Wei Luo
- Department of Otology and Skull Base Surgery, Eye & ENT Hospital of Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine, Ministry of Health, Shanghai, China
| | - Xin-Wei Wang
- Department of Otology and Skull Base Surgery, Eye & ENT Hospital of Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine, Ministry of Health, Shanghai, China
| | - Rui Ma
- Department of Otology and Skull Base Surgery, Eye & ENT Hospital of Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine, Ministry of Health, Shanghai, China
| | - Fang-Lu Chi
- Department of Otology and Skull Base Surgery, Eye & ENT Hospital of Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine, Ministry of Health, Shanghai, China
| | - Ping Chen
- Department of Cell Biology, Emory University, Atlanta, GA, United States
| | - Ning Cong
- Department of Otology and Skull Base Surgery, Eye & ENT Hospital of Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine, Ministry of Health, Shanghai, China
| | - Yu-Yan Gu
- Department of Otology and Skull Base Surgery, Eye & ENT Hospital of Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine, Ministry of Health, Shanghai, China
| | - Dong-Dong Ren
- Department of Otology and Skull Base Surgery, Eye & ENT Hospital of Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine, Ministry of Health, Shanghai, China
| | - Juan-Mei Yang
- Department of Otology and Skull Base Surgery, Eye & ENT Hospital of Fudan University, Shanghai, China.,Key Laboratory of Hearing Medicine, Ministry of Health, Shanghai, China
| |
Collapse
|
39
|
Guan M, Fang Q, He Z, Li Y, Qian F, Qian X, Lu L, Zhang X, Liu D, Qi J, Zhang S, Tang M, Gao X, Chai R. Inhibition of ARC decreases the survival of HEI-OC-1 cells after neomycin damage in vitro. Oncotarget 2018; 7:66647-66659. [PMID: 27556499 PMCID: PMC5341827 DOI: 10.18632/oncotarget.11336] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 08/10/2016] [Indexed: 12/17/2022] Open
Abstract
Hearing loss is a common sensory disorder mainly caused by the loss of hair cells (HCs). Noise, aging, and ototoxic drugs can all induce apoptosis in HCs. Apoptosis repressor with caspase recruitment domain(ARC) is a key factor in apoptosis that inhibits both intrinsic and extrinsic apoptosis pathways; however, there have been no reports on the role of ARC in HC loss in the inner ear. In this study, we used House Ear Institute Organ of Corti 1 (HEI-OC-1) cells, which is a cochlear hair-cell-like cell line, to investigate the role of ARC in aminoglycoside-induced HC loss. ARC was expressed in the cochlear HCs as well as in the HEI-OC-1 cells, but not in the supporting cells, and the expression level of ARC in HCs was decreased after neomycin injury in both cochlear HCs and HEI-OC-1 cells, suggesting that reduced levels of ARC might correlate with neomycin-induced HC loss. We inhibited ARC expression using siRNA and found that this significantly increased the sensitivity of HEI-OC-1 cells to neomycin toxicity. Finally, we found that ARC inhibition increased the expression of pro-apoptotic factors, decreased the mitochondrial membrane potential, and increased the level of reactive oxygen species (ROS) after neomycin injury, suggesting that ARC inhibits cell death and apoptosis in HEI-OC-1 cells by controlling mitochondrial function and ROS accumulation. Thus the endogenous anti-apoptotic factor ARC might be a new therapeutic target for the prevention of aminoglycoside-induced HC loss.
Collapse
Affiliation(s)
- Ming Guan
- Department of Otolaryngology, The Affiliated Hangzhou Hospital of Nanjing Medical University, Hangzhou 310006, China.,Department of Otolaryngology, Hangzhou First People's Hospital, Hangzhou 310006, China.,Department of Otolaryngology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China
| | - Qiaojun Fang
- MOE Key Laboratory of Developmental Genes and Human Disease, State Key Laboratory of Bioelectronics, Institute of Life Sciences, Southeast University, Nanjing 210096, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Zuhong He
- MOE Key Laboratory of Developmental Genes and Human Disease, State Key Laboratory of Bioelectronics, Institute of Life Sciences, Southeast University, Nanjing 210096, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Yong Li
- Department of Otolaryngology, The Affiliated Hangzhou Hospital of Nanjing Medical University, Hangzhou 310006, China.,Department of Otolaryngology, Hangzhou First People's Hospital, Hangzhou 310006, China
| | - Fuping Qian
- MOE Key Laboratory of Developmental Genes and Human Disease, State Key Laboratory of Bioelectronics, Institute of Life Sciences, Southeast University, Nanjing 210096, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Xiaoyun Qian
- Department of Otolaryngology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China.,Department of Otolaryngology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Ling Lu
- Department of Otolaryngology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China.,Department of Otolaryngology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Xiaoli Zhang
- Department of Otolaryngology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Dingding Liu
- Department of Otolaryngology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jieyu Qi
- MOE Key Laboratory of Developmental Genes and Human Disease, State Key Laboratory of Bioelectronics, Institute of Life Sciences, Southeast University, Nanjing 210096, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Shasha Zhang
- MOE Key Laboratory of Developmental Genes and Human Disease, State Key Laboratory of Bioelectronics, Institute of Life Sciences, Southeast University, Nanjing 210096, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Mingliang Tang
- MOE Key Laboratory of Developmental Genes and Human Disease, State Key Laboratory of Bioelectronics, Institute of Life Sciences, Southeast University, Nanjing 210096, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Xia Gao
- Department of Otolaryngology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China.,Department of Otolaryngology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Renjie Chai
- MOE Key Laboratory of Developmental Genes and Human Disease, State Key Laboratory of Bioelectronics, Institute of Life Sciences, Southeast University, Nanjing 210096, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| |
Collapse
|
40
|
Ni W, Zeng S, Li W, Chen Y, Zhang S, Tang M, Sun S, Chai R, Li H. Wnt activation followed by Notch inhibition promotes mitotic hair cell regeneration in the postnatal mouse cochlea. Oncotarget 2018; 7:66754-66768. [PMID: 27564256 PMCID: PMC5341835 DOI: 10.18632/oncotarget.11479] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 06/29/2016] [Indexed: 12/27/2022] Open
Abstract
Hair cell (HC) loss is the main cause of permanent hearing loss in mammals. Previous studies have reported that in neonatal mice cochleae, Wnt activation promotes supporting cell (SC) proliferation and Notch inhibition promotes the trans-differentiation of SCs into HCs. However, Wnt activation alone fails to regenerate significant amounts of new HCs, Notch inhibition alone regenerates the HCs at the cost of exhausting the SC population, which leads to the death of the newly regenerated HCs. Mitotic HC regeneration might preserve the SC number while regenerating the HCs, which could be a better approach for long-term HC regeneration. We present a two-step gene manipulation, Wnt activation followed by Notch inhibition, to accomplish mitotic regeneration of HCs while partially preserving the SC number. We show that Wnt activation followed by Notch inhibition strongly promotes the mitotic regeneration of new HCs in both normal and neomycin-damaged cochleae while partially preserving the SC number. Lineage tracing shows that the majority of the mitotically regenerated HCs are derived specifically from the Lgr5+ progenitors with or without HC damage. Our findings suggest that the co-regulation of Wnt and Notch signaling might provide a better approach to mitotically regenerate HCs from Lgr5+ progenitor cells.
Collapse
Affiliation(s)
- Wenli Ni
- Otorhinolaryngology Department of The Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China
| | - Shan Zeng
- Otorhinolaryngology Department of The Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China
| | - Wenyan Li
- Otorhinolaryngology Department of The Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China
| | - Yan Chen
- Otorhinolaryngology Department of The Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Central Laboratory, Affiliated Eye and ENT Hospital of Fudan University, Shanghai, PR China.,Key Laboratory of Hearing Medicine of The National Health and Family Planning Commission, Shanghai, PR China
| | - Shasha Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, PR China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Mingliang Tang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, PR China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Shan Sun
- Otorhinolaryngology Department of The Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Central Laboratory, Affiliated Eye and ENT Hospital of Fudan University, Shanghai, PR China.,Key Laboratory of Hearing Medicine of The National Health and Family Planning Commission, Shanghai, PR China
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, PR China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Huawei Li
- Otorhinolaryngology Department of The Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China.,Central Laboratory, Affiliated Eye and ENT Hospital of Fudan University, Shanghai, PR China
| |
Collapse
|
41
|
Characterization of Lgr5+ progenitor cell transcriptomes in the apical and basal turns of the mouse cochlea. Oncotarget 2018; 7:41123-41141. [PMID: 27070092 PMCID: PMC5173047 DOI: 10.18632/oncotarget.8636] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/28/2016] [Indexed: 12/11/2022] Open
Abstract
Lgr5+ supporting cells (SCs) are enriched hair cell (HC) progenitors in the cochlea, and several studies have shown a difference in the proliferation and HC regeneration ability of SCs between the apical and basal turns. However, the detailed differences between the transcriptomes of the apical and basal Lgr5+ SCs have not yet been investigated. We found that when isolated by FACS, Lgr5+ cells from the apex generated significantly more HCs and had significantly higher proliferation and mitotic HC regeneration ability compared to those from the base. Next, we used microarray analysis to determine the transcriptome expression profiles of Lgr5+ progenitors from the apex and the base. We first analyzed the genes that were enriched and differentially expressed in Lgr5+ progenitors from the apex and the base. Then we analyzed the cell cycle genes and the transcription factors that might regulate the proliferation and differentiation of Lgr5+ progenitors. Lastly, to further analyze the role of differentially expressed genes and to gain an overall view of the gene network in cochlear HC regeneration, we created a protein-protein interaction network. Our datasets suggest the possible genes that might regulate the proliferation and HC regeneration ability of Lgr5+ progenitors, and these genes might provide new therapeutic targets for HC regeneration in the future.
Collapse
|
42
|
Tan SH, Barker N. Wnt Signaling in Adult Epithelial Stem Cells and Cancer. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 153:21-79. [PMID: 29389518 DOI: 10.1016/bs.pmbts.2017.11.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Wnt/β-catenin signaling is integral to the homeostasis and regeneration of many epithelial tissues due to its critical role in adult stem cell regulation. It is also implicated in many epithelial cancers, with mutations in core pathway components frequently present in patient tumors. In this chapter, we discuss the roles of Wnt/β-catenin signaling and Wnt-regulated stem cells in homeostatic, regenerative and cancer contexts of the intestines, stomach, skin, and liver. We also examine the sources of Wnt ligands that form part of the stem cell niche. Despite the diversity in characteristics of various tissue stem cells, the role(s) of Wnt/β-catenin signaling is generally coherent in maintaining stem cell fate and/or promoting proliferation. It is also likely to play similar roles in cancer stem cells, making the pathway a salient therapeutic target for cancer. While promising progress is being made in the field, deeper understanding of the functions and signaling mechanisms of the pathway in individual epithelial tissues will expedite efforts to modulate Wnt/β-catenin signaling in cancer treatment and tissue regeneration.
Collapse
Affiliation(s)
- Si Hui Tan
- A*STAR Institute of Medical Biology, Singapore
| | - Nick Barker
- A*STAR Institute of Medical Biology, Singapore; Kanazawa University, Kanazawa, Japan; Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
43
|
Mills KM, Szczerkowski JLA, Habib SJ. Wnt ligand presentation and reception: from the stem cell niche to tissue engineering. Open Biol 2017; 7:rsob.170140. [PMID: 28814649 PMCID: PMC5577451 DOI: 10.1098/rsob.170140] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 07/21/2017] [Indexed: 02/06/2023] Open
Abstract
Stem cells reside in niches where spatially restricted signals maintain a delicate balance between stem cell self-renewal and differentiation. Wnt family proteins are particularly suited for this role as they are modified by lipids, which constrain and spatially regulate their signalling range. In recent years, Wnt/β-catenin signalling has been shown to be essential for the self-renewal of a variety of mammalian stem cells. In this review, we discuss Wnt-responsive stem cells in their niche, and mechanisms by which Wnt ligands are presented to responsive cells. We also highlight recent progress in molecular visualization that has allowed for the monitoring of Wnt signalling within the stem cell compartment and new approaches to recapitulate this niche signalling in vitro Indeed, new technologies that present Wnt in a localized manner and mimic the three-dimensional microenvironment of stem cells will advance our understanding of Wnt signalling in the stem cell niche. These advances will expand current horizons to exploit Wnt ligands in the rapidly evolving fields of tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Kate M Mills
- Centre for Stem Cells and Regenerative Medicine, King's College London, London SE1 9RT, UK
| | - James L A Szczerkowski
- Centre for Stem Cells and Regenerative Medicine, King's College London, London SE1 9RT, UK
| | - Shukry J Habib
- Centre for Stem Cells and Regenerative Medicine, King's College London, London SE1 9RT, UK
| |
Collapse
|
44
|
Kretzschmar K, Clevers H. Wnt/β-catenin signaling in adult mammalian epithelial stem cells. Dev Biol 2017; 428:273-282. [PMID: 28526587 DOI: 10.1016/j.ydbio.2017.05.015] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 05/15/2017] [Accepted: 05/16/2017] [Indexed: 01/06/2023]
Abstract
Adult stem cells self-renew and replenish differentiated cells in various organs and tissues throughout a mammal's life. Over the last 25 years an ever-growing body of knowledge has unraveled the essential regulation of adult mammalian epithelia by the canonical Wnt signaling with its key intracellular effector β-catenin. In this review, we discuss the principles of the signaling pathway and its role in adult epithelial stem cells of the intestine and skin during homeostasis and tumorigenesis. We further highlight the research that led to the identification of new stem cell markers and methods to study adult stem cells ex vivo.
Collapse
Affiliation(s)
- Kai Kretzschmar
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre (UMC) Utrecht, 3584 CT Utrecht, The Netherlands; Cancer Genomics Netherlands, UMC Utrecht, 3584 CG Utrecht, The Netherlands
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre (UMC) Utrecht, 3584 CT Utrecht, The Netherlands; Cancer Genomics Netherlands, UMC Utrecht, 3584 CG Utrecht, The Netherlands; Princess Máxima Centre for Pediatric Oncology, 3584 CT Utrecht, The Netherlands.
| |
Collapse
|
45
|
Xu J, Ueno H, Xu CY, Chen B, Weissman IL, Xu PX. Identification of mouse cochlear progenitors that develop hair and supporting cells in the organ of Corti. Nat Commun 2017; 8:15046. [PMID: 28492243 PMCID: PMC5437288 DOI: 10.1038/ncomms15046] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 02/23/2017] [Indexed: 01/20/2023] Open
Abstract
The adult mammalian cochlear sensory epithelium houses two major types of cells, mechanosensory hair cells and underlying supporting cells, and lacks regenerative capacity. Recent evidence indicates that a subset of supporting cells can spontaneously regenerate hair cells after ablation only within the first week postparturition. Here in vivo clonal analysis of mouse inner ear cells during development demonstrates clonal relationship between hair and supporting cells in sensory organs. We report the identification in mouse of a previously unknown population of multipotent stem/progenitor cells that are capable of not only contributing to the hair and supporting cells but also to other cell types, including glia, in cochlea undergoing development, maturation and repair in response to damage. These multipotent progenitors originate from Eya1-expressing otic progenitors. Our findings also provide evidence for detectable regenerative potential in the postnatal cochlea beyond 1 week of age.
Collapse
Affiliation(s)
- Jinshu Xu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Hiroo Ueno
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305, USA
- Ludwig Center, Stanford University, Stanford, California 94305, USA
- Department of Pathology, Stanford University, Stanford, California 94305, USA
| | - Chelsea Y. Xu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Binglai Chen
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Irving L. Weissman
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California 94305, USA
- Ludwig Center, Stanford University, Stanford, California 94305, USA
- Department of Pathology, Stanford University, Stanford, California 94305, USA
| | - Pin-Xian Xu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| |
Collapse
|
46
|
Hirose K, Rutherford MA, Warchol ME. Two cell populations participate in clearance of damaged hair cells from the sensory epithelia of the inner ear. Hear Res 2017; 352:70-81. [PMID: 28526177 DOI: 10.1016/j.heares.2017.04.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 04/09/2017] [Accepted: 04/10/2017] [Indexed: 12/20/2022]
Abstract
The cochlea and the vestibular organs are populated by resident macrophages, but their role in inner ear maintenance and pathology is not entirely clear. Resident macrophages in other organs are responsible for phagocytosis of injured or infected cells, and it is likely that macrophages in the inner ear serve a similar role. Hair cell injury causes macrophages to accumulate within proximity of damaged regions of the inner ear, either by exiting the vasculature and entering the labyrinth or by the resident macrophages reorganizing themselves through local movement to the areas of injury. Direct evidence for macrophage engulfment of apoptotic hair cells has been observed in several conditions. Here, we review evidence for phagocytosis of damaged hair cells in the sensory epithelium by tissue macrophages in the published literature and in some new experiments that are presented here as original work. Several studies also suggest that macrophages are not the only phaogocytic cells in the inner ear, but that supporting cells of the sensory epithelium also play an important role in debris clearance. We describe the various ways in which the sensory epithelia of the inner ear are adapted to eliminate damaged and dying cells. A collaborative effort between resident and migratory macrophages as well as neighboring supporting cells results in the rapid and efficient clearance of cellular debris, even in cases where hair cell loss is rapid and complete.
Collapse
Affiliation(s)
- Keiko Hirose
- Department of Otolaryngology, Washington University, 660 S. Euclid Avenue, Campus Box 8115, St. Louis, MO 63110, United States.
| | - Mark A Rutherford
- Department of Otolaryngology, Washington University, 660 S. Euclid Avenue, Campus Box 8115, St. Louis, MO 63110, United States.
| | - Mark E Warchol
- Department of Otolaryngology, Washington University, 660 S. Euclid Avenue, Campus Box 8115, St. Louis, MO 63110, United States
| |
Collapse
|
47
|
Cheng C, Guo L, Lu L, Xu X, Zhang S, Gao J, Waqas M, Zhu C, Chen Y, Zhang X, Xuan C, Gao X, Tang M, Chen F, Shi H, Li H, Chai R. Characterization of the Transcriptomes of Lgr5+ Hair Cell Progenitors and Lgr5- Supporting Cells in the Mouse Cochlea. Front Mol Neurosci 2017; 10:122. [PMID: 28491023 PMCID: PMC5405134 DOI: 10.3389/fnmol.2017.00122] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 04/11/2017] [Indexed: 12/27/2022] Open
Abstract
Cochlear supporting cells (SCs) have been shown to be a promising resource for hair cell (HC) regeneration in the neonatal mouse cochlea. Previous studies have reported that Lgr5+ SCs can regenerate HCs both in vitro and in vivo and thus are considered to be inner ear progenitor cells. Lgr5+ progenitors are able to regenerate more HCs than Lgr5- SCs, and it is important to understand the mechanism behind the proliferation and HC regeneration of these progenitors. Here, we isolated Lgr5+ progenitors and Lgr5- SCs from Lgr5-EGFP-CreERT2/Sox2-CreERT2/Rosa26-tdTomato mice via flow cytometry. As expected, we found that Lgr5+ progenitors had significantly higher proliferation and HC regeneration ability than Lgr5- SCs. Next, we performed RNA-Seq to determine the gene expression profiles of Lgr5+ progenitors and Lgr5- SCs. We analyzed the genes that were enriched and differentially expressed in Lgr5+ progenitors and Lgr5- SCs, and we found 8 cell cycle genes, 9 transcription factors, and 24 cell signaling pathway genes that were uniquely expressed in one population but not the other. Last, we made a protein–protein interaction network to further analyze the role of these differentially expressed genes. In conclusion, we present a set of genes that might regulate the proliferation and HC regeneration ability of Lgr5+ progenitors, and these might serve as potential new therapeutic targets for HC regeneration.
Collapse
Affiliation(s)
- Cheng Cheng
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China.,Research Institute of OtolaryngologyNanjing, China.,Co-innovation Center of Neuroregeneration, Nantong UniversityNantong, China
| | - Luo Guo
- Department of Otorhinolaryngology, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China.,Key Laboratory of Hearing Medicine of National Health and Family Planning CommissionShanghai, China
| | - Ling Lu
- Department of Otolaryngology-Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical SchoolNanjing, China.,Department of Otolaryngology-Head and Neck Surgery, Drum Tower Clinical Medical College of Nanjing Medical UniversityNanjing, China
| | - Xiaochen Xu
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - ShaSha Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - Junyan Gao
- Health Management and Policy, College of Public Health, Saint Louis University, St. LouisMO, USA
| | - Muhammad Waqas
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China.,Department of Biotechnology, Federal Urdu University of Arts, Science and TechnologyGulshan-e-Iqbal, Pakistan
| | - Chengwen Zhu
- Department of Otolaryngology-Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical SchoolNanjing, China
| | - Yan Chen
- Department of Otorhinolaryngology, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China.,Key Laboratory of Hearing Medicine of National Health and Family Planning CommissionShanghai, China
| | - Xiaoli Zhang
- Department of Otolaryngology-Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical SchoolNanjing, China
| | - Chuanying Xuan
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - Xia Gao
- Department of Otolaryngology-Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical SchoolNanjing, China
| | - Mingliang Tang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - Fangyi Chen
- Department of Biomedical Engineering, Southern University of Science and TechnologyShenzhen, China
| | - Haibo Shi
- Department of Otorhinolaryngology Head and Neck Surgery, The Sixth People's Hospital, Shanghai Jiao Tong UniversityShanghai, China
| | - Huawei Li
- Department of Otorhinolaryngology, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China.,Key Laboratory of Hearing Medicine of National Health and Family Planning CommissionShanghai, China.,Institutes of Biomedical Sciences, Fudan UniversityShanghai, China.,Shanghai Engineering Research Centre of Cochlear ImplantShanghai, China
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China.,Research Institute of OtolaryngologyNanjing, China.,Co-innovation Center of Neuroregeneration, Nantong UniversityNantong, China
| |
Collapse
|
48
|
A Comparative Perspective on Wnt/β-Catenin Signalling in Cell Fate Determination. Results Probl Cell Differ 2017; 61:323-350. [PMID: 28409312 DOI: 10.1007/978-3-319-53150-2_15] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The Wnt/β-catenin pathway is an ancient and highly conserved signalling pathway that plays fundamental roles in the regulation of embryonic development and adult homeostasis. This pathway has been implicated in numerous cellular processes, including cell proliferation, differentiation, migration, morphological changes and apoptosis. In this chapter, we aim to illustrate with specific examples the involvement of Wnt/β-catenin signalling in cell fate determination. We discuss the roles of the Wnt/β-catenin pathway in specifying cell fate throughout evolution, how its function in patterning during development is often reactivated during regeneration and how perturbation of this pathway has negative consequences for the control of cell fate.The origin of all life was a single cell that had the capacity to respond to cues from the environment. With evolution, multicellular organisms emerged, and as a result, subsets of cells arose to form tissues able to respond to specific instructive signals and perform specialised functions. This complexity and specialisation required two types of messages to direct cell fate: intra- and intercellular. A fundamental question in developmental biology is to understand the underlying mechanisms of cell fate choice. Amongst the numerous external cues involved in the generation of cellular diversity, a prominent pathway is the Wnt signalling pathway in all its forms.
Collapse
|
49
|
Mammalian Cochlear Hair Cell Regeneration and Ribbon Synapse Reformation. Neural Plast 2016; 2016:2523458. [PMID: 28119785 PMCID: PMC5227174 DOI: 10.1155/2016/2523458] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 11/29/2016] [Accepted: 12/01/2016] [Indexed: 01/29/2023] Open
Abstract
Hair cells (HCs) are the sensory preceptor cells in the inner ear, which play an important role in hearing and balance. The HCs of organ of Corti are susceptible to noise, ototoxic drugs, and infections, thus resulting in permanent hearing loss. Recent approaches of HCs regeneration provide new directions for finding the treatment of sensor neural deafness. To have normal hearing function, the regenerated HCs must be reinnervated by nerve fibers and reform ribbon synapse with the dendrite of spiral ganglion neuron through nerve regeneration. In this review, we discuss the research progress in HC regeneration, the synaptic plasticity, and the reinnervation of new regenerated HCs in mammalian inner ear.
Collapse
|
50
|
Zheng F, Zuo J. Cochlear hair cell regeneration after noise-induced hearing loss: Does regeneration follow development? Hear Res 2016; 349:182-196. [PMID: 28034617 DOI: 10.1016/j.heares.2016.12.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 11/22/2016] [Accepted: 12/20/2016] [Indexed: 12/14/2022]
Abstract
Noise-induced hearing loss (NIHL) affects a large number of military personnel and civilians. Regenerating inner-ear cochlear hair cells (HCs) is a promising strategy to restore hearing after NIHL. In this review, we first summarize recent transcriptome profile analysis of zebrafish lateral lines and chick utricles where spontaneous HC regeneration occurs after HC damage. We then discuss recent studies in other mammalian regenerative systems such as pancreas, heart and central nervous system. Both spontaneous and forced HC regeneration occurs in mammalian cochleae in vivo involving proliferation and direct lineage conversion. However, both processes are inefficient and incomplete, and decline with age. For direct lineage conversion in vivo in cochleae and in other systems, further improvement requires multiple factors, including transcription, epigenetic and trophic factors, with appropriate stoichiometry in appropriate architectural niche. Increasing evidence from other systems indicates that the molecular paths of direct lineage conversion may be different from those of normal developmental lineages. We therefore hypothesize that HC regeneration does not have to follow HC development and that epigenetic memory of supporting cells influences the HC regeneration, which may be a key to successful cochlear HC regeneration. Finally, we discuss recent efforts in viral gene therapy and drug discovery for HC regeneration. We hope that combination therapy targeting multiple factors and epigenetic signaling pathways will provide promising avenues for HC regeneration in humans with NIHL and other types of hearing loss.
Collapse
Affiliation(s)
- Fei Zheng
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 322, Memphis, TN 38105, United States.
| | - Jian Zuo
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 322, Memphis, TN 38105, United States.
| |
Collapse
|