1
|
Jiang M, Li H, Zhang Q, Xu T, Huang L, Zhang J, Yu H, Zhang J. The role of RGS12 in tissue repair and human diseases. Genes Dis 2025; 12:101480. [PMID: 40271194 PMCID: PMC12017852 DOI: 10.1016/j.gendis.2024.101480] [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: 02/05/2024] [Revised: 08/05/2024] [Accepted: 11/02/2024] [Indexed: 04/25/2025] Open
Abstract
Regulator of G protein signaling 12 (RGS12) belongs to the superfamily of RGS proteins defined by a conserved RGS domain that canonically binds and deactivates heterotrimeric G-proteins. As the largest family member, RGS12 is widely expressed in many cells and tissues. In the past few decades, it has been found that RGS12 affects the activity of various cells in the human body, participates in many physiological and pathological processes, and plays an important role in the pathogenesis of many diseases. Here, we set out to comprehensively review the role of RGS12 in human diseases and its mechanisms, highlighting the possibility of RGS12 as a therapeutic target for the treatment of human diseases.
Collapse
Affiliation(s)
- Min Jiang
- Department of Geriatric Oncology and Department of Palliative Care, Chongqing University Cancer Hospital, Chongqing 400030, China
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hongmei Li
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qiong Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Tongtong Xu
- General Department of Critical Care Medicine, Zhenjiang Traditional Chinese Medicine Hospital, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Zhenjiang, Jiangsu 212003, China
| | - Le Huang
- Army 72nd Group Military Hospital, Huzhou, Zhejiang 313000, China
| | - Jinghong Zhang
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Huiqing Yu
- Department of Geriatric Oncology and Department of Palliative Care, Chongqing University Cancer Hospital, Chongqing 400030, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Junhui Zhang
- Department of Geriatric Oncology and Department of Palliative Care, Chongqing University Cancer Hospital, Chongqing 400030, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| |
Collapse
|
2
|
McGovern MM, Cox BC. Hearing restoration through hair cell regeneration: A review of recent advancements and current limitations. Hear Res 2025; 461:109256. [PMID: 40157114 PMCID: PMC12052480 DOI: 10.1016/j.heares.2025.109256] [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: 01/07/2025] [Revised: 03/21/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
Hearing loss is extremely common, yet limited treatment options are available to restore hearing, and those that are available provide incomplete recovery of hearing detection. For patients who are born with normal hearing, the most common cause of hearing loss is the loss of the sensory hair cells located in the cochlea of the inner ear. Non-mammals, including birds, fish, and amphibians, naturally regenerate new hair cells after damage and this natural process results in functional recovery. While some limited hair cell regeneration also occurs in the immature cochlea of mice, the mature mammalian cochlea does not naturally produce replacement hair cells, and thus hearing loss is permanent. Since the late 1980s, researchers have been investigating mechanisms to convert supporting cells, the cells that remain once hair cells have been killed, into new replacement hair cells. Here we review the current status of hair cell regeneration in the adult cochlea, highlighting recent achievements, as well as challenges that have yet to be resolved.
Collapse
Affiliation(s)
- Melissa M McGovern
- Departments of Otolaryngology and Neurobiology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Brandon C Cox
- Departments of Pharmacology and Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA.
| |
Collapse
|
3
|
Huisman BD, Zuo C, Mathis D. Thymic Mimetic Cells: Evolutionarily Ancient Mirrors of the Periphery. Immunol Rev 2025; 331:e70028. [PMID: 40247628 PMCID: PMC12067923 DOI: 10.1111/imr.70028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/03/2025] [Accepted: 04/07/2025] [Indexed: 04/19/2025]
Abstract
Thymic mimetic cells are hybrids of medullary thymic epithelial cells and diverse peripheral cell types. They are important for imposition of self-tolerance and perform other functions similar to those of their peripheral counterparts. Following early histological observations of “misplaced” stromal cells in thymi from multiple species, mimetic cells were first molecularly investigated in mice. Recent studies have characterized mimetic cells in humans and zebrafish with high-resolution. Many mimetic cell types are conserved across species, although specialized subtypes as well as variable frequencies and levels of specialization are also apparent. Features of the human mimetic cell repertoire, such as the expanded nature of muscle mimetic cells with potential implications in myasthenia gravis and the similarity of tuft mimetic cells and thymic carcinomas, hint at their relevance in human disease. Here we review what is known about mimetic cells across diverse organisms. We discuss potential pressures shaping the composition of the mimetic cell repertoire within and across species, and highlight potential therapeutic applications in human disease.
Collapse
Affiliation(s)
| | - Chong Zuo
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Xiao H, Wu J, Huang L, Ma Y, Wu L, Lin Y, Ye Z, Tan X, Tang X, Tong W, Dai M, Wang Y, Sheng X, Chai R, Zhang S. Conditional Overexpression of Serpine2 Promotes Hair Cell Regeneration from Lgr5+ Progenitors in the Neonatal Mouse Cochlea. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412653. [PMID: 40091489 PMCID: PMC12079390 DOI: 10.1002/advs.202412653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 02/06/2025] [Indexed: 03/19/2025]
Abstract
Neonatal cochlear Lgr5+ progenitors retain limited hair cells (HCs) regenerative capacity, but the regulatory network remains incompletely defined. Serpin family E member 2 (Serpine2) is shown to participate in regulating proliferation and differentiation of cochlear Lgr5+ progenitors in the previous in vitro study. Here, the expression pattern and in vivo roles of Serpine2 in HC regeneration are explored by transgenic mice. It is found that Serpine2 is expressed in the mouse cochlea after birth with a downward trend as the mice age. In addition, Serpine2 conditional overexpression in vivo in Lgr5+ progenitors of neonatal mice cochlea results in an increased number of ectopic HCs in a dose-dependent manner. Serpine2 knockdown ex vivo and in vivo can inhibit HC regeneration. EdU assay and lineage tracing assay demonstrate these ectopic HCs likely originate from Lgr5+ progenitors through direct transdifferentiation rather than through mitotic regeneration. Moreover, single-nucleus RNA sequencing analysis and mRNA level validation reveal that conditionally overexpressed Serpine2 likely induces HC regeneration via inhibiting sonic hedgehog (SHH) signal pathway and inducing Atoh1 and Pou4f3 transcription factor. In brief, these data indicate that Serpine2 plays a pivotal role in HC regeneration from Lgr5+ progenitors in the neonatal mouse cochlea, and this suggests a new avenue for future research into HC regeneration.
Collapse
Affiliation(s)
- Hairong Xiao
- 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 UniversityNanjing210096China
- Southeast University Shenzhen Research InstituteShenzhen518063China
| | - Jiheng Wu
- 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 UniversityNanjing210096China
| | - Lixuan Huang
- 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 UniversityNanjing210096China
| | - Ying Ma
- 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 UniversityNanjing210096China
| | - Leilei Wu
- 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 UniversityNanjing210096China
| | - Yanqin Lin
- 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 UniversityNanjing210096China
- Southeast University Shenzhen Research InstituteShenzhen518063China
| | - Zixuan Ye
- 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 UniversityNanjing210096China
| | - Xin Tan
- 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 UniversityNanjing210096China
| | - Xujun Tang
- 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 UniversityNanjing210096China
| | - Wei Tong
- 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 UniversityNanjing210096China
| | - Mingchen Dai
- 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 UniversityNanjing210096China
| | - Yintao 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 UniversityNanjing210096China
| | - Xia Sheng
- School of Public HealthTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- Department of Environmental HealthSchool of Environmental Science and EngineeringHainan UniversityHaikou570228China
| | - 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 UniversityNanjing210096China
- Southeast University Shenzhen Research InstituteShenzhen518063China
- Institute for Stem Cells and RegenerationChinese Academy of ScienceBeijing100081China
- Department of Otolaryngology Head and Neck SurgerySichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengdu610000China
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantong226001China
| | - Shasha 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 UniversityNanjing210096China
- Southeast University Shenzhen Research InstituteShenzhen518063China
| |
Collapse
|
5
|
Matern MS, Heller S. A developmental atlas of mouse vestibular-like inner ear organoids. iScience 2025; 28:111817. [PMID: 39967872 PMCID: PMC11834118 DOI: 10.1016/j.isci.2025.111817] [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: 07/12/2024] [Revised: 10/16/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Abstract
Inner ear sensory epithelia can be generated in vitro from embryonic stem cells. The resulting inner ear organoids represent a potentially inexhaustible source of otic tissues, including sensory hair cells and supporting cells, for in vitro manipulation. Here, we present a single-cell atlas of whole mouse embryonic stem cell-derived vestibular-like inner ear organoids at six developmental stages. Our analyses trace the genesis and developmental progression of otic progenitor cells to supporting cells and hair cells. By profiling all organoid cells, we also characterize the development of additional cell groups, such as otic mesenchyme. We further utilize our atlas to describe a proliferative phase where otic progenitors produce hair cells and otic neuroblasts, followed by a transition to a non-proliferative state. The resulting map of otic progression reveals specific time windows that can inform future research on cell cycle regulation and cell lineage specification in inner ear organoids.
Collapse
Affiliation(s)
- Maggie S. Matern
- Department of Otolaryngology — Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94304, USA
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Stefan Heller
- Department of Otolaryngology — Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA 94304, USA
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94304, USA
| |
Collapse
|
6
|
Sabatini C, Lin HJ, Ovik G, Hall R, Lee T. The proneural transcription factor Atoh1 promotes odontogenic differentiation in human dental pulp stem cells (DPSCs). BMC Mol Cell Biol 2025; 26:5. [PMID: 39833721 PMCID: PMC11744864 DOI: 10.1186/s12860-025-00530-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Bioengineering of human teeth for replacement is an appealing regenerative approach in the era of gene therapy. Developmentally regulated transcription factors hold promise in the quest because these transcriptional regulators constitute the gene regulatory networks driving cell fate determination. Atonal homolog 1 (Atoh1) is a transcription factor of the basic helix-loop-helix (bHLH) family essential for neurogenesis in the cerebellum, auditory hair cell differentiation, and intestinal stem cell specification. The functional versatility of Atoh1 prompted us to test the possibility that Atoh1 may intersect the dental pulp stem cell (DPSC) gene regulatory network governing odontogenic differentiation. METHODS We isolated DPSCs from human dental pulps and treated the cells with a replication-deficient adenoviral vector to achieve robust ectopic expression of Atoh1, following which the growth and odontogenic differentiation profiles of DPSCs were characterized. RESULTS DPSCs harboring the Atoh1 expression vector exhibited an approximately 3,000-fold increase in the expression of Atoh1 compared to the negative control, leading to increased DPSC proliferation in the growth medium (P < 0.05). In the odontogenic medium, Atoh1 caused an early induction of BMP2 (P < 0.001) followed by a late induction of BMP7 (P < 0.01) and increased Wnt signaling (P < 0.01). The increased BMP/Wnt signaling led to up to 8-fold increased expression of the master osteogenic transcription factor Osterix (P < 0.005) while exhibiting no significant effect on Runx2 or Dlx5, which are abundantly expressed in DPSCs. Atoh1 stimulated expression of type I collagen (P < 0.005) and small integrin-binding ligand, N-linked glycoproteins (SIBLINGs) such as bone sialoprotein (P < 0.001), dentin matrix protein 1 (P < 0.05), dentin sialophosphoprotein (P < 0.005), and osteopontin (P < 0.001), resulting in increased dentin matrix mineralization (P < 0.05). The odontogenic phenotype is associated with metabolic remodeling marked by enhanced glycolytic flux and attenuated mitochondrial metabolic enzyme activities. CONCLUSIONS Atoh1, despite being a proneural transcription factor in development, possesses a novel odontogenic function upon ectopic expression in DPSCs. This in vitro study demonstrates a novel odontogenic mechanism mediated by ectopic expression of the transcription factor Atoh1 in human DPSCs. The finding may offer an innovative strategy for gene-based regeneration of the pulp-dentin complex.
Collapse
Affiliation(s)
- Camila Sabatini
- Department of Restorative Dentistry, School of Dental Medicine, University at Buffalo, 3435 Main Street, Buffalo, NY, 14214, USA.
| | - Huey-Jiun Lin
- Department of Biochemistry, University at Buffalo, 3435 Main Street, Buffalo, NY, 14214, USA
| | - Galib Ovik
- Department of Biochemistry, University at Buffalo, 3435 Main Street, Buffalo, NY, 14214, USA
| | - Richard Hall
- Department of Oral Surgery, University at Buffalo, 3435 Main Street, Buffalo, NY, 14214, USA
| | - Techung Lee
- Department of Biochemistry, University at Buffalo, 3435 Main Street, Buffalo, NY, 14214, USA
| |
Collapse
|
7
|
Jarvis JE, Miao L, Hallaert P, Martin MR, Collado L, Hill NT, Brownell I. POU4F3 Is Necessary for Normal Merkel Cell Formation, whereas POU4F1 Is Dispensable. J Invest Dermatol 2025; 145:193-196.e5. [PMID: 39009281 DOI: 10.1016/j.jid.2024.06.1284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 05/14/2024] [Accepted: 06/01/2024] [Indexed: 07/17/2024]
Affiliation(s)
- Jordan E Jarvis
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Lingling Miao
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Patrick Hallaert
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Mackenzie R Martin
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Loren Collado
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Natasha T Hill
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Isaac Brownell
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
8
|
Karpinski P, Mendez-Pena JE, Wu CL, Akalin A, Cornejo KM, Hung YP, Hoang MP. POU4F3 Is a Sensitive and Specific Marker of Merkel Cell Carcinoma. Mod Pathol 2025; 38:100627. [PMID: 39341281 DOI: 10.1016/j.modpat.2024.100627] [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: 04/30/2024] [Revised: 09/04/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024]
Abstract
Although of therapeutic importance, a single sensitive and specific immunostain to distinguish Merkel cell carcinoma (MCC) from mimics is not currently available. In addition, single tumor cells are difficult to detect in sentinel lymph node biopsy. Leveraging publicly available data sets of 9264 solid tumors and >600,000 single-cell transcriptomes, we identified POU4F3 to be a specific marker of MCC. Analyses of Pan-Cancer RNA bulk sequencing data of 24 tumor types from Tumor Cancer Genomic Atlas data sets as well as non-Tumor Cancer Genomic Atlas small cell lung carcinoma and MCC data sets confirmed POU4F3 specificity for MCC. Single-cell RNA-sequencing analyses also confirmed the lack of POU4F3 expression in lung small cell carcinoma as well as a variety of normal tissues. Nuclear POU4F3 immunohistochemical expression was noted in 98.7% of 153 MCCs and in only 1.7% of mimics (3 of 180 cases, including 95 small cell carcinomas, of which 55 were from lungs and the remainder from other sites). Three POU4F3-positive non-MCC cases were from lungs (2 cases) and vagina (1 case). All 153 tested MCC cases were negative for ASCL1, a key transcriptional regulator highly expressed in small cell lung carcinoma. NeuroD1 was seen in a subset of MCC cases (20.9%, 32/153). POU4F3 immunostain was performed on 29 sentinel lymph nodes, and strong POU4F3 nuclear expression facilitated the ease of metastasis detection, even single tumor cells. Our study built on prior works shows that POU4F3 is a sensitive and specific clinical marker of MCC.
Collapse
Affiliation(s)
- Paweł Karpinski
- Department of Genetics, Wroclaw Medical University, Wroclaw, Poland.
| | - Javier E Mendez-Pena
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Cheng-Lin Wu
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ali Akalin
- Department of Pathology, UMass Medical Center, Worcester, Massachusetts
| | - Kristine M Cornejo
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Yin P Hung
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Mai P Hoang
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
9
|
Shi T, Kim Y, Llamas J, Wang X, Fabian P, Lozito TP, Segil N, Gnedeva K, Crump JG. Long-range Atoh1 enhancers maintain competency for hair cell regeneration in the inner ear. Proc Natl Acad Sci U S A 2024; 121:e2418098121. [PMID: 39671177 DOI: 10.1073/pnas.2418098121] [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: 09/04/2024] [Accepted: 11/11/2024] [Indexed: 12/14/2024] Open
Abstract
During tissue regeneration, lineage-related cells can switch their fate to replace missing cells. This cell plasticity is particularly prominent in more regenerative vertebrates such as zebrafish, yet the molecular basis by which cells transdifferentiate into another cell type upon injury remains unclear. Here, we investigate the epigenetic basis of regenerative transdifferentiation in the inner ear, where supporting cells (SCs) generate mechanosensory hair cells (HCs) upon damage. By comparing the chromatin landscapes in regenerative zebrafish and green anole lizards versus nonregenerative mice, we identified a class of enhancers that function in progenitors to generate HCs and then are selectively maintained in SCs of regenerative vertebrates to regenerate HCs. In particular, we uncovered a syntenic class of long-range enhancers for Atoh1, a master transcription factor for HC differentiation. In the absence of injury, these enhancers maintain accessibility in SCs through adulthood but are prevented from driving zebrafish atoh1a expression through Notch repression. Deletion of these enhancers not only impaired atoh1a expression and HC formation during development but also blocked the ability of SCs to transdifferentiate into HCs during regeneration. Moreover, defects were specific to the inner ear versus the lateral line, revealing distinct mechanisms of regeneration in these mechanosensory organs. These findings reveal a class of regenerative enhancer that maintains competency of inner ear SCs to upregulate atoh1a and transdifferentiate into HCs upon damage. We propose that the continued accessibility of developmental enhancers for one cell fate in lineage-related cells may be a common theme underlying adult cell plasticity in regenerative vertebrates.
Collapse
Affiliation(s)
- Tuo Shi
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles CA 90033
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Yeeun Kim
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles CA 90033
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Juan Llamas
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles CA 90033
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Xizi Wang
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles CA 90033
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Peter Fabian
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles CA 90033
| | - Thomas P Lozito
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles CA 90033
| | - Neil Segil
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles CA 90033
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Ksenia Gnedeva
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles CA 90033
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - J Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles CA 90033
| |
Collapse
|
10
|
Orlovsky K, Appel E, Hantisteanu S, Olender T, Lotem J, Levanon D, Groner Y. Runx3, Brn3a and Isl1 interplay orchestrates the transcriptional program in the early stages of proprioceptive neuron development. PLoS Genet 2024; 20:e1011401. [PMID: 39715266 PMCID: PMC11729954 DOI: 10.1371/journal.pgen.1011401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 01/13/2025] [Accepted: 12/04/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND The development and diversification of sensory proprioceptive neurons, which reside in the dorsal root ganglia (DRG) and express the tropomyosin receptor kinase C (TrkC), depend on the transcription factor (TF) Runx3. Runx3-deficient mice develop severe limb ataxia due to TrkC neuron cell death. Two additional TFs Pou4f1 (also called Brn3a) and Isl1 also play an important role in sensory neuron development. Thus, we aimed to unravel the chromatin state of early-developing TrkC neurons and decipher the Runx3 high-confidence target genes (HCT) and the possible cooperation between Runx3, Brn3a and Isl1 in the regulation of these genes. METHODS Runx3 expression is driven by the gene proximal P2 promoter. Transcriptome analysis was conducted by RNA-seq on RNA isolated from heterozygous (P2+/-) vs. homozygous (P2-/-) TrkC neurons and differentially expressed genes (DEGs) were determined. Genome-wide occupancy of Runx3, Brn3a, Isl1 and histone H3 acetylated on lysine 27 (H3K27Ac) was determined using CUT&RUN. The landscape of Transposase-accessible chromatin was analyzed via ATAC-seq. FINDINGS The intersection of Runx3 genomic occupancy-associated genes and DEG data discovered 244 Runx3 HCT. Brn3a and Isl1 were found to bind to numerous genomic loci, some of which overlapped with Runx3. Most genomic regions bound by each of these three TFs or co-bound by them resided in distantly located enhancer regions rather than in gene promoters. In activated and suppressed neuronal Runx3 HCT, Runx3 cooperated mainly with Brn3a to regulate expression through distantly located enhancers. Interestingly, suppression of non-neuronal immune genes was mainly managed via Runx3 without Brn3a. The distribution of ATAC and H3K27Ac marked regions in Runx3 peaks containing at least one RUNX binding site (Runx3_RBS) revealed that while most promoter regions were marked by ATAC, a prominent fraction of intron/intergenic regions occupied by Runx3, Brn3a or Isl1 were unmarked by ATAC and/or H3K27Ac. CONCLUSIONS These analyses shed new light on the interplay of Runx3, Brn3a, Isl1, and open chromatin regions in regulating the Runx3 HCT in the early developmental stages of TrkC neurons.
Collapse
Affiliation(s)
- Kira Orlovsky
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Elena Appel
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Shay Hantisteanu
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Tsviya Olender
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Joseph Lotem
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ditsa Levanon
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Yoram Groner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
11
|
Snir A, Cieśla K, Vekslar R, Amedi A. Highly compromised auditory spatial perception in aided congenitally hearing-impaired and rapid improvement with tactile technology. iScience 2024; 27:110808. [PMID: 39290844 PMCID: PMC11407022 DOI: 10.1016/j.isci.2024.110808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/11/2024] [Accepted: 08/21/2024] [Indexed: 09/19/2024] Open
Abstract
Spatial understanding is a multisensory construct while hearing is the only natural sense enabling the simultaneous perception of the entire 3D space. To test whether such spatial understanding is dependent on auditory experience, we study congenitally hearing-impaired users of assistive devices. We apply an in-house technology, which, inspired by the auditory system, performs intensity-weighting to represent external spatial positions and motion on the fingertips. We see highly impaired auditory spatial capabilities for tracking moving sources, which based on the "critical periods" theory emphasizes the role of nature in sensory development. Meanwhile, for tactile and audio-tactile spatial motion perception, the hearing-impaired show performance similar to typically hearing individuals. The immediate availability of 360° external space representation through touch, despite the lack of such experience during the lifetime, points to the significant role of nurture in spatial perception development, and to its amodal character. The findings show promise toward advancing multisensory solutions for rehabilitation.
Collapse
Affiliation(s)
- Adi Snir
- The Baruch Ivcher Institute for Brain, Cognition, and Technology, The Baruch Ivcher School of Psychology, Reichman University, HaUniversita 8 Herzliya 461010, Israel
| | - Katarzyna Cieśla
- The Baruch Ivcher Institute for Brain, Cognition, and Technology, The Baruch Ivcher School of Psychology, Reichman University, HaUniversita 8 Herzliya 461010, Israel
- World Hearing Centre, Institute of Physiology and Pathology of Hearing, Mokra 17, 05-830 Kajetany, Nadarzyn, Poland
| | - Rotem Vekslar
- The Baruch Ivcher Institute for Brain, Cognition, and Technology, The Baruch Ivcher School of Psychology, Reichman University, HaUniversita 8 Herzliya 461010, Israel
| | - Amir Amedi
- The Baruch Ivcher Institute for Brain, Cognition, and Technology, The Baruch Ivcher School of Psychology, Reichman University, HaUniversita 8 Herzliya 461010, Israel
| |
Collapse
|
12
|
Butts JC, Wu SR, Durham MA, Dhindsa RS, Revelli JP, Ljungberg MC, Saulnier O, McLaren ME, Taylor MD, Zoghbi HY. A single-cell transcriptomic map of the developing Atoh1 lineage identifies neural fate decisions and neuronal diversity in the hindbrain. Dev Cell 2024; 59:2171-2188.e7. [PMID: 39106860 DOI: 10.1016/j.devcel.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/03/2024] [Accepted: 07/09/2024] [Indexed: 08/09/2024]
Abstract
Proneural transcription factors establish molecular cascades to orchestrate neuronal diversity. One such transcription factor, Atonal homolog 1 (Atoh1), gives rise to cerebellar excitatory neurons and over 30 distinct nuclei in the brainstem critical for hearing, breathing, and balance. Although Atoh1 lineage neurons have been qualitatively described, the transcriptional programs that drive their fate decisions and the full extent of their diversity remain unknown. Here, we analyzed single-cell RNA sequencing and ATOH1 DNA binding in Atoh1 lineage neurons of the developing mouse hindbrain. This high-resolution dataset identified markers for specific brainstem nuclei and demonstrated that transcriptionally heterogeneous progenitors require ATOH1 for proper migration. Moreover, we identified a sizable population of proliferating unipolar brush cell progenitors in the mouse Atoh1 lineage, previously described in humans as the origin of one medulloblastoma subtype. Collectively, our data provide insights into the developing mouse hindbrain and markers for functional assessment of understudied neuronal populations.
Collapse
Affiliation(s)
- Jessica C Butts
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA.
| | - Sih-Rong Wu
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mark A Durham
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ryan S Dhindsa
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jean-Pierre Revelli
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - M Cecilia Ljungberg
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Olivier Saulnier
- The Arthur and Sonia Labatt Brain Tumor Research Centre, The Hospital for Sick Children, Toronto, ON, Canada; Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada; Genomics and Development of Childhood Cancers, Institut Curie, PSL University, 75005 Paris, France; INSERM U830, Cancer Heterogeneity Instability and Plasticity, Institut Curie, PSL University, 75005 Paris, France; SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, 75005 Paris, France
| | - Madison E McLaren
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Michael D Taylor
- The Arthur and Sonia Labatt Brain Tumor Research Centre, The Hospital for Sick Children, Toronto, ON, Canada; Department of Surgery, Department of Laboratory Medicine and Pathobiology, and Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Department of Pediatrics-Hematology/Oncology and Neurosurgery, Baylor College of Medicine, Houston, TX, USA; Texas Children's Cancer and Hematology Center, Houston, TX 77030, USA; Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Huda Y Zoghbi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
13
|
Singh J, Randle MR, Walters BJ, Cox BC. The transcription factor Pou4f3 is essential for the survival of postnatal and adult mouse cochlear hair cells and normal hearing. Front Cell Neurosci 2024; 18:1369282. [PMID: 38566840 PMCID: PMC10985149 DOI: 10.3389/fncel.2024.1369282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction Hair cells (HCs) of the cochlea are responsible for sound transduction and hearing perception in mammals. Genetic mutations in the transcription factor Pou4f3 cause non-syndromic autosomal dominant hearing loss in humans (DFNA15) which varies in the age of onset depending on the individual mutation. Mouse models with germline deletion or mutations in Pou4f3 have previously demonstrated its critical role in the maturation and survival of cochlear HCs during embryonic development. However, the role of Pou4f3 in auditory function and in the survival or maintenance of cochlear HCs after birth and during adulthood has not been studied. Methods Therefore, using the inducible CreER-loxP system, we deleted Pou4f3 from mouse cochlear HCs at different postnatal ages, relevant to specific stages of HC maturation and hearing function. Results and discussion Elevated auditory brainstem response thresholds and significant HC loss were detected in mice with Pou4f3 deletion compared to their control littermates, regardless of the age when Pou4f3 was deleted. However, HC loss occurred more rapidly when Pou4f3 was deleted from immature HCs. Additionally, HC loss caused by Pou4f3 deletion did not affect the number of cochlear supporting cells, but caused a delayed loss of spiral ganglion neurons at 4 months after the deletion. In conclusion, Pou4f3 is necessary for the survival of cochlear HCs and normal hearing at all postnatal ages regardless of their maturation state. Our data also suggest that Pou4f3 indirectly regulates the survival of spiral ganglion neurons.
Collapse
Affiliation(s)
- Jarnail Singh
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Michelle R. Randle
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Bradley J. Walters
- Department of Otolaryngology-Head and Neck Surgery, University of Mississippi Medical Center, Jackson, MS, United States
| | - Brandon C. Cox
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, United States
| |
Collapse
|
14
|
Wang G, Gu Y, Liu Z. Deciphering the genetic interactions between Pou4f3, Gfi1, and Rbm24 in maintaining mouse cochlear hair cell survival. eLife 2024; 12:RP90025. [PMID: 38483314 PMCID: PMC10939501 DOI: 10.7554/elife.90025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024] Open
Abstract
Mammals harbor a limited number of sound-receptor hair cells (HCs) that cannot be regenerated after damage. Thus, investigating the underlying molecular mechanisms that maintain HC survival is crucial for preventing hearing impairment. Intriguingly, Pou4f3-/- or Gfi1-/- HCs form initially but then rapidly degenerate, whereas Rbm24-/- HCs degenerate considerably later. However, the transcriptional cascades involving Pou4f3, Gfi1, and Rbm24 remain undescribed. Here, we demonstrate that Rbm24 expression is completely repressed in Pou4f3-/- HCs but unaltered in Gfi1-/- HCs, and further that the expression of both POU4F3 and GFI1 is intact in Rbm24-/- HCs. Moreover, by using in vivo mouse transgenic reporter assays, we identify three Rbm24 enhancers to which POU4F3 binds. Lastly, through in vivo genetic testing of whether Rbm24 restoration alleviates the degeneration of Pou4f3-/- HCs, we show that ectopic Rbm24 alone cannot prevent Pou4f3-/- HCs from degenerating. Collectively, our findings provide new molecular and genetic insights into how HC survival is regulated.
Collapse
Affiliation(s)
- Guangqin Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yunpeng Gu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Zhiyong Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
- Shanghai Center for Brain Science and Brain-Inspired Intelligence TechnologyShanghaiChina
| |
Collapse
|
15
|
Anselmi C, Fuller GK, Stolfi A, Groves AK, Manni L. Sensory cells in tunicates: insights into mechanoreceptor evolution. Front Cell Dev Biol 2024; 12:1359207. [PMID: 38550380 PMCID: PMC10973136 DOI: 10.3389/fcell.2024.1359207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Tunicates, the sister group of vertebrates, offer a unique perspective for evolutionary developmental studies (Evo-Devo) due to their simple anatomical organization. Moreover, the separation of tunicates from vertebrates predated the vertebrate-specific genome duplications. As adults, they include both sessile and pelagic species, with very limited mobility requirements related mainly to water filtration. In sessile species, larvae exhibit simple swimming behaviors that are required for the selection of a suitable substrate on which to metamorphose. Despite their apparent simplicity, tunicates display a variety of mechanoreceptor structures involving both primary and secondary sensory cells (i.e., coronal sensory cells). This review encapsulates two decades of research on tunicate mechanoreception focusing on the coronal organ's sensory cells as prime candidates for understanding the evolution of vertebrate hair cells of the inner ear and the lateral line organ. The review spans anatomical, cellular and molecular levels emphasizing both similarity and differences between tunicate and vertebrate mechanoreception strategies. The evolutionary significance of mechanoreception is discussed within the broader context of Evo-Devo studies, shedding light on the intricate pathways that have shaped the sensory system in chordates.
Collapse
Affiliation(s)
- Chiara Anselmi
- Hopkins Marine Station, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Pacific Grove, CA, United States
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, United States
| | - Gwynna K. Fuller
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Alberto Stolfi
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States
| | - Andrew K. Groves
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Lucia Manni
- Dipartimento di Biologia, Università degli Studi di Padova, Padova, Italy
| |
Collapse
|
16
|
Choi SW, Abitbol JM, Cheng AG. Hair Cell Regeneration: From Animals to Humans. Clin Exp Otorhinolaryngol 2024; 17:1-14. [PMID: 38271988 PMCID: PMC10933805 DOI: 10.21053/ceo.2023.01382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 01/07/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
Cochlear hair cells convert sound into electrical signals that are relayed via the spiral ganglion neurons to the central auditory pathway. Hair cells are vulnerable to damage caused by excessive noise, aging, and ototoxic agents. Non-mammals can regenerate lost hair cells by mitotic regeneration and direct transdifferentiation of surrounding supporting cells. However, in mature mammals, damaged hair cells are not replaced, resulting in permanent hearing loss. Recent studies have uncovered mechanisms by which sensory organs in non-mammals and the neonatal mammalian cochlea regenerate hair cells, and outlined possible mechanisms why this ability declines rapidly with age in mammals. Here, we review similarities and differences between avian, zebrafish, and mammalian hair cell regeneration. Moreover, we discuss advances and limitations of hair cell regeneration in the mature cochlea and their potential applications to human hearing loss.
Collapse
Affiliation(s)
- Sung-Won Choi
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Otorhinolaryngology-Head and Neck Surgery and Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Pusan National University School of Medicine, Busan, Korea
| | - Julia M. Abitbol
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Alan G. Cheng
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
17
|
McGovern MM, Hosamani IV, Niu Y, Nguyen KY, Zong C, Groves AK. Expression of Atoh1, Gfi1, and Pou4f3 in the mature cochlea reprograms nonsensory cells into hair cells. Proc Natl Acad Sci U S A 2024; 121:e2304680121. [PMID: 38266052 PMCID: PMC10835112 DOI: 10.1073/pnas.2304680121] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 12/08/2023] [Indexed: 01/26/2024] Open
Abstract
Mechanosensory hair cells of the mature mammalian organ of Corti do not regenerate; consequently, loss of hair cells leads to permanent hearing loss. Although nonmammalian vertebrates can regenerate hair cells from neighboring supporting cells, many humans with severe hearing loss lack both hair cells and supporting cells, with the organ of Corti being replaced by a flat epithelium of nonsensory cells. To determine whether the mature cochlea can produce hair cells in vivo, we reprogrammed nonsensory cells adjacent to the organ of Corti with three hair cell transcription factors: Gfi1, Atoh1, and Pou4f3. We generated numerous hair cell-like cells in nonsensory regions of the cochlea and new hair cells continued to be added over a period of 9 wk. Significantly, cells adjacent to reprogrammed hair cells expressed markers of supporting cells, suggesting that transcription factor reprogramming of nonsensory cochlear cells in adult animals can generate mosaics of sensory cells like those seen in the organ of Corti. Generating such sensory mosaics by reprogramming may represent a potential strategy for hearing restoration in humans.
Collapse
Affiliation(s)
| | - Ishwar V. Hosamani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX77030
| | - Yichi Niu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX77030
| | - Ken Y. Nguyen
- Department of Neuroscience, Baylor College of Medicine, Houston, TX77030
| | - Chenghang Zong
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX77030
| | - Andrew K. Groves
- Department of Neuroscience, Baylor College of Medicine, Houston, TX77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX77030
| |
Collapse
|
18
|
Pan Y, Li S, He S, Wang G, Li C, Liu Z, Xiang M. Fgf8 P2A-3×GFP/+: A New Genetic Mouse Model for Specifically Labeling and Sorting Cochlear Inner Hair Cells. Neurosci Bull 2023; 39:1762-1774. [PMID: 37233921 PMCID: PMC10661496 DOI: 10.1007/s12264-023-01069-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/08/2023] [Indexed: 05/27/2023] Open
Abstract
The cochlear auditory epithelium contains two types of sound receptors, inner hair cells (IHCs) and outer hair cells (OHCs). Mouse models for labelling juvenile and adult IHCs or OHCs exist; however, labelling for embryonic and perinatal IHCs or OHCs are lacking. Here, we generated a new knock-in Fgf8P2A-3×GFP/+ (Fgf8GFP/+) strain, in which the expression of a series of three GFP fragments is controlled by endogenous Fgf8 cis-regulatory elements. After confirming that GFP expression accurately reflects the expression of Fgf8, we successfully obtained both embryonic and neonatal IHCs with high purity, highlighting the power of Fgf8GFP/+. Furthermore, our fate-mapping analysis revealed, unexpectedly, that IHCs are also derived from inner ear progenitors expressing Insm1, which is currently regarded as an OHC marker. Thus, besides serving as a highly favorable tool for sorting early IHCs, Fgf8GFP/+ will facilitate the isolation of pure early OHCs by excluding IHCs from the entire hair cell pool.
Collapse
Affiliation(s)
- Yi Pan
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shuting 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
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shunji He
- 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
| | - Guangqin Wang
- 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
- University of Chinese Academy of Sciences, Beijing, 100049, 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
| | - 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.
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 201210, China.
| | - Mingliang Xiang
- Department of Otolaryngology and Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
19
|
Fang Q, Wei Y, Zhang Y, Cao W, Yan L, Kong M, Zhu Y, Xu Y, Guo L, Zhang L, Wang W, Yu Y, Sun J, Yang J. Stem cells as potential therapeutics for hearing loss. Front Neurosci 2023; 17:1259889. [PMID: 37746148 PMCID: PMC10512725 DOI: 10.3389/fnins.2023.1259889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
Hearing impairment is a global health problem. Stem cell therapy has become a cutting-edge approach to tissue regeneration. In this review, the recent advances in stem cell therapy for hearing loss have been discussed. Nanomaterials can modulate the stem cell microenvironment to augment the therapeutic effects further. The potential of combining nanomaterials with stem cells for repairing and regenerating damaged inner ear hair cells (HCs) and spiral ganglion neurons (SGNs) has also been discussed. Stem cell-derived exosomes can contribute to the repair and regeneration of damaged tissue, and the research progress on exosome-based hearing loss treatment has been summarized as well. Despite stem cell therapy's technical and practical limitations, the findings reported so far are promising and warrant further investigation for eventual clinical translation.
Collapse
Affiliation(s)
- Qiaojun Fang
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- School of Life Sciences and Technology, Southeast University, Nanjing, China
| | - Yongjie Wei
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yuhua Zhang
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Wei Cao
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Lin Yan
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Mengdie Kong
- School of Life Sciences and Technology, Southeast University, Nanjing, China
| | - Yongjun Zhu
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yan Xu
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Lingna Guo
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Lei Zhang
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Weiqing Wang
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yafeng Yu
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jingwu 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, Anhui, China
| | - Jianming Yang
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
20
|
Laureano A, Kim J, Martinez E, Kwan KY. Chromodomain helicase DNA binding protein 4 in cell fate decisions. Hear Res 2023; 436:108813. [PMID: 37329862 PMCID: PMC10463912 DOI: 10.1016/j.heares.2023.108813] [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: 01/06/2022] [Revised: 05/09/2023] [Accepted: 05/24/2023] [Indexed: 06/19/2023]
Abstract
Loss of spiral ganglion neurons (SGNs) in the cochlea causes hearing loss. Understanding the mechanisms of cell fate transition accelerates efforts that employ directed differentiation and lineage conversion to repopulate lost SGNs. Proposed strategies to regenerate SGNs rely on altering cell fate by activating transcriptional regulatory networks, but repressing networks for alternative cell lineages is also essential. Epigenomic changes during cell fate transitions suggest that CHD4 represses gene expression by altering the chromatin status. Despite limited direct investigations, human genetic studies implicate CHD4 function in the inner ear. The possibility of CHD4 in suppressing alternative cell fates to promote inner ear regeneration is discussed.
Collapse
Affiliation(s)
- Alejandra Laureano
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Jihyun Kim
- Department of Cell Biology & Neuroscience, Rutgers University, Nelson Labs D250 604 Allison Rd., Piscataway, NJ 08854, USA; Stem Cell Research Center and Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Edward Martinez
- Department of Cell Biology & Neuroscience, Rutgers University, Nelson Labs D250 604 Allison Rd., Piscataway, NJ 08854, USA; Stem Cell Research Center and Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Kelvin Y Kwan
- Department of Cell Biology & Neuroscience, Rutgers University, Nelson Labs D250 604 Allison Rd., Piscataway, NJ 08854, USA; Stem Cell Research Center and Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
21
|
Wang X, Llamas J, Trecek T, Shi T, Tao L, Makmura W, Crump JG, Segil N, Gnedeva K. SoxC transcription factors shape the epigenetic landscape to establish competence for sensory differentiation in the mammalian organ of Corti. Proc Natl Acad Sci U S A 2023; 120:e2301301120. [PMID: 37585469 PMCID: PMC10450657 DOI: 10.1073/pnas.2301301120] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 06/28/2023] [Indexed: 08/18/2023] Open
Abstract
The auditory organ of Corti is comprised of only two major cell types-the mechanosensory hair cells and their associated supporting cells-both specified from a single pool of prosensory progenitors in the cochlear duct. Here, we show that competence to respond to Atoh1, a transcriptional master regulator necessary and sufficient for induction of mechanosensory hair cells, is established in the prosensory progenitors between E12.0 and 13.5. The transition to the competent state is rapid and is associated with extensive remodeling of the epigenetic landscape controlled by the SoxC group of transcription factors. Conditional loss of Sox4 and Sox11-the two homologous family members transiently expressed in the inner ear at the time of competence establishment-blocks the ability of prosensory progenitors to differentiate as hair cells. Mechanistically, we show that Sox4 binds to and establishes accessibility of early sensory lineage-specific regulatory elements, including ones associated with Atoh1 and its direct downstream targets. Consistent with these observations, overexpression of Sox4 or Sox11 prior to developmental establishment of competence precociously induces hair cell differentiation in the cochlear progenitors. Further, reintroducing Sox4 or Sox11 expression restores the ability of postnatal supporting cells to differentiate as hair cells in vitro and in vivo. Our findings demonstrate the pivotal role of SoxC family members as agents of epigenetic and transcriptional changes necessary for establishing competence for sensory receptor differentiation in the inner ear.
Collapse
Affiliation(s)
- Xizi Wang
- Caruso Department of Otolaryngology–Head and Neck Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
| | - Juan Llamas
- Caruso Department of Otolaryngology–Head and Neck Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
| | - Talon Trecek
- Caruso Department of Otolaryngology–Head and Neck Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
| | - Tuo Shi
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
| | - Litao Tao
- Caruso Department of Otolaryngology–Head and Neck Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
| | - Welly Makmura
- Caruso Department of Otolaryngology–Head and Neck Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
| | - J. Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
| | - Neil Segil
- Caruso Department of Otolaryngology–Head and Neck Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
| | - Ksenia Gnedeva
- Caruso Department of Otolaryngology–Head and Neck Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA90033
| |
Collapse
|
22
|
Nguyen JD, Llamas J, Shi T, Crump JG, Groves AK, Segil N. DNA methylation in the mouse cochlea promotes maturation of supporting cells and contributes to the failure of hair cell regeneration. Proc Natl Acad Sci U S A 2023; 120:e2300839120. [PMID: 37549271 PMCID: PMC10438394 DOI: 10.1073/pnas.2300839120] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 07/11/2023] [Indexed: 08/09/2023] Open
Abstract
Mammalian hair cells do not functionally regenerate in adulthood but can regenerate at embryonic and neonatal stages in mice by direct transdifferentiation of neighboring supporting cells into new hair cells. Previous work showed loss of transdifferentiation potential of supporting cells is in part due to H3K4me1 enhancer decommissioning of the hair cell gene regulatory network during the first postnatal week. However, inhibiting this decommissioning only partially preserves transdifferentiation potential. Therefore, we explored other repressive epigenetic modifications that may be responsible for this loss of plasticity. We find supporting cells progressively accumulate DNA methylation at promoters of developmentally regulated hair cell genes. Specifically, DNA methylation overlaps with binding sites of Atoh1, a key transcription factor for hair cell fate. We further show that DNA hypermethylation replaces H3K27me3-mediated repression of hair cell genes in mature supporting cells, and is accompanied by progressive loss of chromatin accessibility, suggestive of facultative heterochromatin formation. Another subset of hair cell loci is hypermethylated in supporting cells, but not in hair cells. Ten-eleven translocation (TET) enzyme-mediated demethylation of these hypermethylated sites is necessary for neonatal supporting cells to transdifferentiate into hair cells. We also observe changes in chromatin accessibility of supporting cell subtypes at the single-cell level with increasing age: Gene programs promoting sensory epithelium development loses chromatin accessibility, in favor of gene programs that promote physiological maturation and function of the cochlea. We also find chromatin accessibility is partially recovered in a chronically deafened mouse model, which holds promise for future translational efforts in hearing restoration.
Collapse
Affiliation(s)
- John D. Nguyen
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology at the University of Southern California, Los Angeles, CA90033
| | - Juan Llamas
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology at the University of Southern California, Los Angeles, CA90033
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA90033
| | - Tuo Shi
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology at the University of Southern California, Los Angeles, CA90033
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA90033
| | - J. Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology at the University of Southern California, Los Angeles, CA90033
| | - Andrew K. Groves
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX77030
- Department of Neuroscience, Baylor College of Medicine, Houston, TX77030
| | - Neil Segil
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology at the University of Southern California, Los Angeles, CA90033
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA90033
| |
Collapse
|
23
|
Sun Y, Liu Z. Recent advances in molecular studies on cochlear development and regeneration. Curr Opin Neurobiol 2023; 81:102745. [PMID: 37356371 DOI: 10.1016/j.conb.2023.102745] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/06/2023] [Accepted: 05/25/2023] [Indexed: 06/27/2023]
Abstract
The auditory organ cochlea harbors two types of sound receptors, inner hair cells (IHCs) and outer hair cells (OHCs), which are innervated by spiral (auditory) ganglion neurons (SGNs). Recent transcriptomic, epigenetic, and genetic studies have started to reveal various aspects of cochlear development, including how prosensory progenitors are specified and diversified into IHCs or OHCs, as well as the heterogeneity among SGNs and how SGN subtypes are formed. Here, we primarily review advances in this line of research over the past five years and discuss a few key studies (from the past two years) to elucidate (1) how prosensory progenitors are specified; (2) the cis-regulatory control of Atoh1 expression and the synergistic interaction between Atoh1 and Pou4f3; and (3) the essential roles of Insm1 and Ikzf2 in OHC development and Tbx2 in IHC development. Moreover, we highlight the contribution of recent molecular studies on cochlear development toward the goal of regenerating IHCs and OHCs, which holds considerable potential for application in treating human deafness. Lastly, we briefly summarize the most recent progress on uncovering when and how SGN diversity is generated.
Collapse
Affiliation(s)
- Yuwei Sun
- 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
| | - 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; Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 201210, China.
| |
Collapse
|
24
|
Wu SR, Butts JC, Caudill MS, Revelli JP, Dhindsa RS, Durham MA, Zoghbi HY. Atoh1 drives the heterogeneity of the pontine nuclei neurons and promotes their differentiation. SCIENCE ADVANCES 2023; 9:eadg1671. [PMID: 37390208 PMCID: PMC10313176 DOI: 10.1126/sciadv.adg1671] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 05/26/2023] [Indexed: 07/02/2023]
Abstract
Pontine nuclei (PN) neurons mediate the communication between the cerebral cortex andthe cerebellum to refine skilled motor functions. Prior studies showed that PN neurons fall into two subtypes based on their anatomic location and region-specific connectivity, but the extent of their heterogeneity and its molecular drivers remain unknown. Atoh1 encodes a transcription factor that is expressed in the PN precursors. We previously showed that partial loss of Atoh1 function in mice results in delayed PN development and impaired motor learning. In this study, we performed single-cell RNA sequencing to elucidate the cell state-specific functions of Atoh1 during PN development and found that Atoh1 regulates cell cycle exit, differentiation, migration, and survival of PN neurons. Our data revealed six previously not known PN subtypes that are molecularly and spatially distinct. We found that the PN subtypes exhibit differential vulnerability to partial loss of Atoh1 function, providing insights into the prominence of PN phenotypes in patients with ATOH1 missense mutations.
Collapse
Affiliation(s)
- Sih-Rong Wu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, USA
| | - Jessica C. Butts
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
| | - Matthew S. Caudill
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, USA
| | - Jean-Pierre Revelli
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Ryan S. Dhindsa
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Mark A. Durham
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
- Medical Student Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | - Huda Y. Zoghbi
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
25
|
Tian C, Wang J, Ye X, Chen J, Zheng R, Yu H, Li J, Yin G, Liu L, Zhao N, Feng G, Zhu Z, Wang J, Fan G, Liu L. Culture conditions of mouse ESCs impact the tumor appearance in vivo. Cell Rep 2023; 42:112645. [PMID: 37314926 DOI: 10.1016/j.celrep.2023.112645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/22/2023] [Accepted: 05/27/2023] [Indexed: 06/16/2023] Open
Abstract
Various culture conditions by small molecules have been explored to extend pluripotency of stem cells, but their impacts on cell fate in vivo remain elusive. We systematically compared the effects of various culture conditions on the pluripotency and cell fate in vivo of mouse embryonic stem cells (ESCs) by tetraploid embryo complementation assay. Conventional ESC cultures in serum/LIF-based condition produced complete ESC mice and also the survival to adulthood at the highest rates of all other chemical-based cultures. Moreover, long-term examination of the survived ESC mice demonstrated that conventional ESC cultures did not lead to visible abnormality for up to 1.5-2 years, whereas the prolonged chemical-based cultures developed retroperitoneal atypical teratomas or leiomyomas. The chemical-based cultures exhibited transcriptomes and epigenomes that typically differed from those of conventional ESC cultures. Our results warrant further refinement of culture conditions in promoting the pluripotency and safety of ESCs in future applications.
Collapse
Affiliation(s)
- Chenglei Tian
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China; Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jing Wang
- Department of Human Genetics and Broad Stem Cell Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Xiaoying Ye
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China; Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jiyu Chen
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China; Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Rongyan Zheng
- Key Laboratory for Stem Cells and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Hanwen Yu
- Key Laboratory for Stem Cells and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jie Li
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China; Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Guoxing Yin
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China; Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Linlin Liu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China; Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Nannan Zhao
- Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Guofeng Feng
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China; Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Zhengmao Zhu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China; Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jichang Wang
- Key Laboratory for Stem Cells and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.
| | - Guoping Fan
- Department of Human Genetics and Broad Stem Cell Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA; Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China.
| | - Lin Liu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China; Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China; Institute of Translational Medicine, Tianjin Union Medical Center, Nankai University, Tianjin 300071, China.
| |
Collapse
|
26
|
Ueda Y, Nakamura T, Nie J, Solivais AJ, Hoffman JR, Daye BJ, Hashino E. Defining developmental trajectories of prosensory cells in human inner ear organoids at single-cell resolution. Development 2023; 150:dev201071. [PMID: 37381908 PMCID: PMC10323240 DOI: 10.1242/dev.201071] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 05/24/2023] [Indexed: 06/29/2023]
Abstract
The inner ear sensory epithelia contain mechanosensitive hair cells and supporting cells. Both cell types arise from SOX2-expressing prosensory cells, but the mechanisms underlying the diversification of these cell lineages remain unclear. To determine the transcriptional trajectory of prosensory cells, we established a SOX2-2A-ntdTomato human embryonic stem cell line using CRISPR/Cas9, and performed single-cell RNA-sequencing analyses with SOX2-positive cells isolated from inner ear organoids at various time points between differentiation days 20 and 60. Our pseudotime analysis suggests that vestibular type II hair cells arise primarily from supporting cells, rather than bi-fated prosensory cells in organoids. Moreover, ion channel- and ion-transporter-related gene sets were enriched in supporting cells versus prosensory cells, whereas Wnt signaling-related gene sets were enriched in hair cells versus supporting cells. These findings provide valuable insights into how prosensory cells give rise to hair cells and supporting cells during human inner ear development, and may provide a clue to promote hair cell regeneration from resident supporting cells in individuals with hearing loss or balance disorders.
Collapse
Affiliation(s)
- Yoshitomo Ueda
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Takashi Nakamura
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Jing Nie
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alexander J. Solivais
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - John R. Hoffman
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Becca J. Daye
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Eri Hashino
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
27
|
Li S, He S, Lu Y, Jia S, Liu Z. Epistatic genetic interactions between Insm1 and Ikzf2 during cochlear outer hair cell development. Cell Rep 2023; 42:112504. [PMID: 37171961 DOI: 10.1016/j.celrep.2023.112504] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/17/2023] [Accepted: 04/27/2023] [Indexed: 05/14/2023] Open
Abstract
The cochlea harbors two types of sound receptors, outer hair cells (OHCs) and inner hair cells (IHCs). OHCs transdifferentiate into IHCs in Insm1 mutants, and OHCs in Ikzf2-deficient mice are dysfunctional and maintain partial IHC gene expression. Insm1 potentially acts as a positive but indirect regulator of Ikzf2, considering that Insm1 is expressed earlier than Ikzf2 and primarily functions as a transcriptional repressor. However, direct evidence of this possibility is lacking. Here, we report the following results: first, Insm1 overexpression in IHCs leads to ectopic Ikzf2 expression. Second, Ikzf2 expression is repressed in Insm1-deficient OHCs, and forced expression of Ikzf2 mitigates the OHC abnormality in Insm1 mutants. Last, dual ablation of Insm1 and Ikzf2 generates a similar OHC phenotype as does Insm1 ablation alone. Collectively, our findings reveal the transcriptional cascade from Insm1 to Ikzf2, which should facilitate future investigation of the molecular mechanisms underlying OHC development and regeneration.
Collapse
Affiliation(s)
- Shuting 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; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shunji He
- 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
| | - Ying Lu
- 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
| | - Shiqi Jia
- The First Affiliated Hospital, Jinan University, Guangzhou 510630, 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; Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China.
| |
Collapse
|
28
|
Păun O, Tan YX, Patel H, Strohbuecker S, Ghanate A, Cobolli-Gigli C, Llorian Sopena M, Gerontogianni L, Goldstone R, Ang SL, Guillemot F, Dias C. Pioneer factor ASCL1 cooperates with the mSWI/SNF complex at distal regulatory elements to regulate human neural differentiation. Genes Dev 2023; 37:218-242. [PMID: 36931659 PMCID: PMC10111863 DOI: 10.1101/gad.350269.122] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 02/28/2023] [Indexed: 03/19/2023]
Abstract
Pioneer transcription factors are thought to play pivotal roles in developmental processes by binding nucleosomal DNA to activate gene expression, though mechanisms through which pioneer transcription factors remodel chromatin remain unclear. Here, using single-cell transcriptomics, we show that endogenous expression of neurogenic transcription factor ASCL1, considered a classical pioneer factor, defines a transient population of progenitors in human neural differentiation. Testing ASCL1's pioneer function using a knockout model to define the unbound state, we found that endogenous expression of ASCL1 drives progenitor differentiation by cis-regulation both as a classical pioneer factor and as a nonpioneer remodeler, where ASCL1 binds permissive chromatin to induce chromatin conformation changes. ASCL1 interacts with BAF SWI/SNF chromatin remodeling complexes, primarily at targets where it acts as a nonpioneer factor, and we provide evidence for codependent DNA binding and remodeling at a subset of ASCL1 and SWI/SNF cotargets. Our findings provide new insights into ASCL1 function regulating activation of long-range regulatory elements in human neurogenesis and uncover a novel mechanism of its chromatin remodeling function codependent on partner ATPase activity.
Collapse
Affiliation(s)
- Oana Păun
- Neural Stem Cell Biology Laboratory, the Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Yu Xuan Tan
- Neural Stem Cell Biology Laboratory, the Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Harshil Patel
- Bioinformatics and Biostatistics Science and Technology Platform, the Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Stephanie Strohbuecker
- Bioinformatics and Biostatistics Science and Technology Platform, the Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Avinash Ghanate
- Bioinformatics and Biostatistics Science and Technology Platform, the Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Clementina Cobolli-Gigli
- Neural Stem Cell Biology Laboratory, the Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Miriam Llorian Sopena
- Bioinformatics and Biostatistics Science and Technology Platform, the Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Lina Gerontogianni
- Bioinformatics and Biostatistics Science and Technology Platform, the Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Robert Goldstone
- Bioinformatics and Biostatistics Science and Technology Platform, the Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Siew-Lan Ang
- Neural Stem Cell Biology Laboratory, the Francis Crick Institute, London NW1 1AT, United Kingdom
| | - François Guillemot
- Neural Stem Cell Biology Laboratory, the Francis Crick Institute, London NW1 1AT, United Kingdom;
| | - Cristina Dias
- Neural Stem Cell Biology Laboratory, the Francis Crick Institute, London NW1 1AT, United Kingdom;
- Medical and Molecular Genetics, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, King's College London, London SE1 9RT, United Kingdom
| |
Collapse
|
29
|
Future Pharmacotherapy for Sensorineural Hearing Loss by Protection and Regeneration of Auditory Hair Cells. Pharmaceutics 2023; 15:pharmaceutics15030777. [PMID: 36986638 PMCID: PMC10054686 DOI: 10.3390/pharmaceutics15030777] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/14/2023] [Accepted: 02/24/2023] [Indexed: 03/03/2023] Open
Abstract
Sensorineural hearing loss has been a global burden of diseases for decades. However, according to recent progress in experimental studies on hair cell regeneration and protection, clinical trials of pharmacotherapy for sensorineural hearing loss have rapidly progressed. In this review, we focus on recent clinical trials for hair cell protection and regeneration and outline mechanisms based on associated experimental studies. Outcomes of recent clinical trials provided valuable data regarding the safety and tolerability of intra-cochlear and intra-tympanic applications as drug delivery methods. Recent findings in molecular mechanisms of hair cell regeneration suggested the realization of regenerative medicine for sensorineural hearing loss in the near future.
Collapse
|
30
|
Wang J, Zheng J, Wang H, He H, Li S, Zhang Y, Wang Y, Xu X, Wang S. Gene therapy: an emerging therapy for hair cells regeneration in the cochlea. Front Neurosci 2023; 17:1177791. [PMID: 37207182 PMCID: PMC10188948 DOI: 10.3389/fnins.2023.1177791] [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: 03/02/2023] [Accepted: 04/12/2023] [Indexed: 05/21/2023] Open
Abstract
Sensorineural hearing loss is typically caused by damage to the cochlear hair cells (HCs) due to external stimuli or because of one's genetic factors and the inability to convert sound mechanical energy into nerve impulses. Adult mammalian cochlear HCs cannot regenerate spontaneously; therefore, this type of deafness is usually considered irreversible. Studies on the developmental mechanisms of HC differentiation have revealed that nonsensory cells in the cochlea acquire the ability to differentiate into HCs after the overexpression of specific genes, such as Atoh1, which makes HC regeneration possible. Gene therapy, through in vitro selection and editing of target genes, transforms exogenous gene fragments into target cells and alters the expression of genes in target cells to activate the corresponding differentiation developmental program in target cells. This review summarizes the genes that have been associated with the growth and development of cochlear HCs in recent years and provides an overview of gene therapy approaches in the field of HC regeneration. It concludes with a discussion of the limitations of the current therapeutic approaches to facilitate the early implementation of this therapy in a clinical setting.
Collapse
Affiliation(s)
- Jipeng Wang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jianwei Zheng
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haiyan Wang
- Department of Otolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Haoying He
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Shuang Li
- Department of Otolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ya Zhang
- Department of Otolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - You Wang
- Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- *Correspondence: You Wang,
| | - Xiaoxiang Xu
- Department of Otolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Xiaoxiang Xu,
| | - Shuyi Wang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Shuyi Wang,
| |
Collapse
|
31
|
Iyer AA, Hosamani I, Nguyen JD, Cai T, Singh S, McGovern MM, Beyer L, Zhang H, Jen HI, Yousaf R, Birol O, Sun JJ, Ray RS, Raphael Y, Segil N, Groves AK. Cellular reprogramming with ATOH1, GFI1, and POU4F3 implicate epigenetic changes and cell-cell signaling as obstacles to hair cell regeneration in mature mammals. eLife 2022; 11:e79712. [PMID: 36445327 PMCID: PMC9708077 DOI: 10.7554/elife.79712] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 11/16/2022] [Indexed: 11/30/2022] Open
Abstract
Reprogramming of the cochlea with hair-cell-specific transcription factors such as ATOH1 has been proposed as a potential therapeutic strategy for hearing loss. ATOH1 expression in the developing cochlea can efficiently induce hair cell regeneration but the efficiency of hair cell reprogramming declines rapidly as the cochlea matures. We developed Cre-inducible mice to compare hair cell reprogramming with ATOH1 alone or in combination with two other hair cell transcription factors, GFI1 and POU4F3. In newborn mice, all transcription factor combinations tested produced large numbers of cells with the morphology of hair cells and rudimentary mechanotransduction properties. However, 1 week later, only a combination of ATOH1, GFI1 and POU4F3 could reprogram non-sensory cells of the cochlea to a hair cell fate, and these new cells were less mature than cells generated by reprogramming 1 week earlier. We used scRNA-seq and combined scRNA-seq and ATAC-seq to suggest at least two impediments to hair cell reprogramming in older animals. First, hair cell gene loci become less epigenetically accessible in non-sensory cells of the cochlea with increasing age. Second, signaling from hair cells to supporting cells, including Notch signaling, can prevent reprogramming of many supporting cells to hair cells, even with three hair cell transcription factors. Our results shed light on the molecular barriers that must be overcome to promote hair cell regeneration in the adult cochlea.
Collapse
Affiliation(s)
- Amrita A Iyer
- Department of Molecular & Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Ishwar Hosamani
- Department of Molecular & Human Genetics, Baylor College of MedicineHoustonUnited States
| | - John D Nguyen
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology at USCLos AngelesUnited States
| | - Tiantian Cai
- Program in Developmental Biology, Baylor College of MedicineHoustonUnited States
| | - Sunita Singh
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Melissa M McGovern
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Lisa Beyer
- Department of Otolaryngology-Head and Neck Surgery, University of MichiganAnn ArborUnited States
| | - Hongyuan Zhang
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Hsin-I Jen
- Program in Developmental Biology, Baylor College of MedicineHoustonUnited States
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Rizwan Yousaf
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Onur Birol
- Program in Developmental Biology, Baylor College of MedicineHoustonUnited States
| | - Jenny J Sun
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Russell S Ray
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Yehoash Raphael
- Department of Otolaryngology-Head and Neck Surgery, University of MichiganAnn ArborUnited States
| | - Neil Segil
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology at USCLos AngelesUnited States
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine of the University of Southern CaliforniaLos AngelesUnited States
| | - Andrew K Groves
- Department of Molecular & Human Genetics, Baylor College of MedicineHoustonUnited States
- Program in Developmental Biology, Baylor College of MedicineHoustonUnited States
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
32
|
Costa A, Powell LM, Malaguti M, Soufi A, Lowell S, Jarman AP. Repurposing the lineage-determining transcription factor Atoh1 without redistributing its genomic binding sites. Front Cell Dev Biol 2022; 10:1016367. [PMID: 36420143 PMCID: PMC9676683 DOI: 10.3389/fcell.2022.1016367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/17/2022] [Indexed: 11/09/2022] Open
Abstract
Although the lineage-determining ability of transcription factors is often modulated according to cellular context, the mechanisms by which such switching occurs are not well known. Using a transcriptional programming model, we found that Atoh1 is repurposed from a neuronal to an inner ear hair cell (HC) determinant by the combined activities of Gfi1 and Pou4f3. In this process, Atoh1 maintains its regulation of neuronal genes but gains ability to regulate HC genes. Pou4f3 enables Atoh1 access to genomic locations controlling the expression of sensory (including HC) genes, but Atoh1 + Pou4f3 are not sufficient for HC differentiation. Gfi1 is key to the Atoh1-induced lineage switch, but surprisingly does not alter Atoh1's binding profile. Gfi1 acts in two divergent ways. It represses the induction by Atoh1 of genes that antagonise HC differentiation, a function in keeping with its well-known repressor role in haematopoiesis. Remarkably, we find that Gfi1 also acts as a co-activator: it binds directly to Atoh1 at existing target genes to enhance its activity. These findings highlight the diversity of mechanisms by which one TF can redirect the activity of another to enable combinatorial control of cell identity.
Collapse
Affiliation(s)
- Aida Costa
- Centre for Discovery Brain Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Lynn M. Powell
- Centre for Discovery Brain Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
| | - Mattias Malaguti
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Abdenour Soufi
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Sally Lowell
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew P. Jarman
- Centre for Discovery Brain Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
33
|
Gartin AK, Frost TC, Cushman CH, Leeper BA, Gokhale PC, DeCaprio JA. Merkel Cell Carcinoma Sensitivity to EZH2 Inhibition Is Mediated by SIX1 Derepression. J Invest Dermatol 2022; 142:2783-2792.e15. [PMID: 35331717 PMCID: PMC9492898 DOI: 10.1016/j.jid.2022.03.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 02/28/2022] [Accepted: 03/07/2022] [Indexed: 01/08/2023]
Abstract
Polycomb repressive complex 2 has a critical role in the maintenance of bivalent promoters and is often perturbed in cancer, including neuroendocrine tumors. In this study, we investigated the susceptibility of Merkel cell carcinoma (MCC), a neuroendocrine carcinoma of the skin, to inhibitors of the Polycomb repressive complex 2 catalytic subunit EZH2. We show that a subset of MCC cell lines is sensitive to EZH2 inhibitor-induced cell viability loss. We find that inhibitor treatment of susceptible cells derepresses the Polycomb repressive complex 2 target SIX1, a transcription factor in the PAX-SIX-EYA-DACH network normally involved in inner ear hair cell development, and that PAX-SIX-EYA-DACH network transcription factors are critical contributors to EZH2 inhibitor-induced MCC cell viability loss. Furthermore, we show the EZH2 inhibitor tazemetostat slows the growth of MCC xenografts and derepresses SIX1 and its downstream inner ear transcriptional target MYO6 in vivo. We propose that EZH2 inhibition in MCC leads to SIX1 derepression with dysregulation of hearing-related transcriptional programs and growth inhibition. This study provides evidence that MCC tumors may be specifically susceptible to EZH2 inhibitors, while giving mechanistic insight into the transcriptional programs these inhibitors perturb in MCC, and potentially in other neuroendocrine cancers.
Collapse
Affiliation(s)
- Ashley K Gartin
- Program in Virology, The Graduate School of Arts and Sciences, Harvard University, Cambridge, Massachusetts, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Thomas C Frost
- Program in Virology, The Graduate School of Arts and Sciences, Harvard University, Cambridge, Massachusetts, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Camille H Cushman
- Program in Virology, The Graduate School of Arts and Sciences, Harvard University, Cambridge, Massachusetts, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Brittaney A Leeper
- Experimental Therapeutics Core, Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Robert and Renée Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Prafulla C Gokhale
- Experimental Therapeutics Core, Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Robert and Renée Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - James A DeCaprio
- Program in Virology, The Graduate School of Arts and Sciences, Harvard University, Cambridge, Massachusetts, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
34
|
Heuermann ML, Matos S, Hamilton D, Cox BC. Regenerated hair cells in the neonatal cochlea are innervated and the majority co-express markers of both inner and outer hair cells. Front Cell Neurosci 2022; 16:841864. [PMID: 36187289 PMCID: PMC9524252 DOI: 10.3389/fncel.2022.841864] [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: 12/22/2021] [Accepted: 08/29/2022] [Indexed: 11/30/2022] Open
Abstract
After a damaging insult, hair cells can spontaneously regenerate from cochlear supporting cells within the first week of life. While the regenerated cells express several markers of immature hair cells and have stereocilia bundles, their capacity to differentiate into inner or outer hair cells, and ability to form new synaptic connections has not been well-described. In addition, while multiple supporting cell subtypes have been implicated as the source of the regenerated hair cells, it is unclear if certain subtypes have a greater propensity to form one hair cell type over another. To investigate this, we used two CreER mouse models to fate-map either the supporting cells located near the inner hair cells (inner phalangeal and border cells) or outer hair cells (Deiters’, inner pillar, and outer pillar cells) along with immunostaining for markers that specify the two hair cell types. We found that supporting cells fate-mapped by both CreER lines responded early to hair cell damage by expressing Atoh1, and are capable of producing regenerated hair cells that express terminal differentiation markers of both inner and outer hair cells. The majority of regenerated hair cells were innervated by neuronal fibers and contained synapses. Unexpectedly, we also found that the majority of the laterally positioned regenerated hair cells aberrantly expressed both the outer hair cell gene, oncomodulin, and the inner hair cell gene, vesicular glutamate transporter 3 (VGlut3). While this work demonstrates that regenerated cells can express markers of both inner and outer hair cells after damage, VGlut3 expression appears to lack the tight control present during embryogenesis, which leads to its inappropriate expression in regenerated cells.
Collapse
Affiliation(s)
- Mitchell L. Heuermann
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Sophia Matos
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, United States
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Deborah Hamilton
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Brandon C. Cox
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, United States
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
- *Correspondence: Brandon C. Cox,
| |
Collapse
|
35
|
Abstract
Current estimates suggest that nearly half a billion people worldwide are affected by hearing loss. Because of the major psychological, social, economic, and health ramifications, considerable efforts have been invested in identifying the genes and molecular pathways involved in hearing loss, whether genetic or environmental, to promote prevention, improve rehabilitation, and develop therapeutics. Genomic sequencing technologies have led to the discovery of genes associated with hearing loss. Studies of the transcriptome and epigenome of the inner ear have characterized key regulators and pathways involved in the development of the inner ear and have paved the way for their use in regenerative medicine. In parallel, the immense preclinical success of using viral vectors for gene delivery in animal models of hearing loss has motivated the industry to work on translating such approaches into the clinic. Here, we review the recent advances in the genomics of auditory function and dysfunction, from patient diagnostics to epigenetics and gene therapy.
Collapse
Affiliation(s)
- Shahar Taiber
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; ,
| | - Kathleen Gwilliam
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA; ,
| | - Ronna Hertzano
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA; ,
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Karen B Avraham
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; ,
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
36
|
Bi Z, Li X, Ren M, Gu Y, Zhu T, Li S, Wang G, Sun S, Sun Y, Liu Z. Development and transdifferentiation into inner hair cells require Tbx2. Natl Sci Rev 2022; 9:nwac156. [PMID: 36687561 PMCID: PMC9844247 DOI: 10.1093/nsr/nwac156] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 01/25/2023] Open
Abstract
Atoh1 is essential for the development of both outer hair cells (OHCs) and inner hair cells (IHCs) in the mammalian cochlea. Whereas Ikzf2 is necessary for OHC development, the key gene required for IHC development remains unknown. We found that deletion of Tbx2 in neonatal IHCs led to their transdifferentiation into OHCs by repressing 26.7% of IHC genes and inducing 56.3% of OHC genes, including Ikzf2. More importantly, persistent expression of Tbx2 coupled with transient Atoh1 expression effectively reprogrammed non-sensory supporting cells into new IHCs expressing the functional IHC marker vGlut3. The differentiation status of these new IHCs was considerably more advanced than that previously reported. Thus, Tbx2 is essential for IHC development and co-upregulation of Tbx2 with Atoh1 in supporting cells represents a new approach for treating deafness related to IHC degeneration.
Collapse
Affiliation(s)
| | | | | | - Yunpeng Gu
- 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,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tong Zhu
- 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
| | - Shuting 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,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guangqin Wang
- 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,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Suhong Sun
- 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,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuwei Sun
- 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,University of Chinese Academy of Sciences, Beijing 100049, China
| | | |
Collapse
|
37
|
Abstract
Cochlear hair cells (HCs) in the inner ear are responsible for sound detection. For HC fate specification, the master transcription factor Atoh1 is both necessary and sufficient. Atoh1 expression is dynamic and tightly regulated during development, but the cis-regulatory elements mediating this regulation remain unresolved. Unexpectedly, we found that deleting the only recognized Atoh1 enhancer, defined here as Eh1, failed to impair HC development. By using the assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq), we discovered two additional Atoh1 enhancers: Eh2 and Eh3. Notably, Eh2 deletion was sufficient for impairing HC development, and concurrent deletion of Eh1 and Eh2 or all three enhancers resulted in nearly complete absence of HCs. Lastly, we showed that Atoh1 binds to all three enhancers, consistent with its autoregulatory function. Our findings reveal that the cooperative action of three distinct enhancers underpins effective Atoh1 regulation during HC development, indicating potential therapeutic approaches for HC regeneration.
Collapse
|
38
|
GFI1 regulates hair cell differentiation by acting as an off-DNA transcriptional co-activator of ATOH1, and a DNA-binding repressor. Sci Rep 2022; 12:7793. [PMID: 35551236 PMCID: PMC9098437 DOI: 10.1038/s41598-022-11931-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 05/03/2022] [Indexed: 11/08/2022] Open
Abstract
GFI1 is a zinc finger transcription factor that is necessary for the differentiation and survival of hair cells in the cochlea. Deletion of Gfi1 in mice significantly reduces the expression of hundreds of hair cell genes: this is a surprising result, as GFI1 normally acts as a transcriptional repressor by recruiting histone demethylases and methyltransferases to its targets. To understand the mechanisms by which GFI1 promotes hair cell differentiation, we used CUT&RUN to identify the direct targets of GFI1 and ATOH1 in hair cells. We found that GFI1 regulates hair cell differentiation in two distinct ways—first, GFI1 and ATOH1 can bind to the same regulatory elements in hair cell genes, but while ATOH1 directly binds its target DNA motifs in many of these regions, GFI1 does not. Instead, it appears to enhance ATOH1’s transcriptional activity by acting as part of a complex in which it does not directly bind DNA. Second, GFI1 can act in its more typical role as a direct, DNA-binding transcriptional repressor in hair cells; here it represses non-hair cell genes, including many neuronal genes. Together, our results illuminate the function of GFI1 in hair cell development and hair cell reprogramming strategies.
Collapse
|
39
|
Kervarrec T, Pissaloux D, Poilane J, Tirode F, Tallet A, Collin C, Tallegas M, Berthon P, Gaboriaud P, Sohier P, Calonje E, Luzar B, Goto K, Cokelaere K, Lamant L, Balme B, Wild R, Neuville A, Deschamps L, Auberger E, Paumier V, Bonte H, Moulonguet I, Plantier F, Cales V, Pinsolle V, Roblet D, Dupuy F, Dallot A, Seris A, Jouary T, Houben R, Schrama D, Hesbacher S, Macagno N, Battistella M, Cribier B, Vergier B, Fouchardière A, Jullie M. Recurrent
FOXK1
::
GRHL
and
GPS2
::
GRHL
fusions in trichogerminoma. J Pathol 2022; 257:96-108. [DOI: 10.1002/path.5872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 12/14/2021] [Accepted: 01/17/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Thibault Kervarrec
- Department of Pathology Université de Tours, Centre Hospitalier Universitaire de Tours Tours France
- “Biologie des infections à polyomavirus” team, UMR INRA ISP 1282 Université de Tours Tours France
- CARADERM Network
| | - Daniel Pissaloux
- Department of Biopathology Center Léon Bérard Lyon France
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of Lyon, Equipe Labellisée Ligue contre le Cancer Lyon France
| | - Jeremie Poilane
- Department of Pathology Université de Tours, Centre Hospitalier Universitaire de Tours Tours France
- Platform of Somatic Tumor Molecular Genetics Université de Tours, Centre Hospitalier Universitaire de Tours Tours France
- Department of Pathology Centre Hospitalier Universitaire de Angers Angers France
| | - Franck Tirode
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of Lyon, Equipe Labellisée Ligue contre le Cancer Lyon France
| | - Anne Tallet
- Platform of Somatic Tumor Molecular Genetics Université de Tours, Centre Hospitalier Universitaire de Tours Tours France
| | - Christine Collin
- Platform of Somatic Tumor Molecular Genetics Université de Tours, Centre Hospitalier Universitaire de Tours Tours France
| | - Matthias Tallegas
- Department of Pathology Université de Tours, Centre Hospitalier Universitaire de Tours Tours France
- Platform of Somatic Tumor Molecular Genetics Université de Tours, Centre Hospitalier Universitaire de Tours Tours France
| | - Patricia Berthon
- “Biologie des infections à polyomavirus” team, UMR INRA ISP 1282 Université de Tours Tours France
| | - Pauline Gaboriaud
- “Biologie des infections à polyomavirus” team, UMR INRA ISP 1282 Université de Tours Tours France
| | - Pierre Sohier
- CARADERM Network
- Faculté de Médecine Paris Centre Santé University of Paris Paris France
- Department of Pathology Hôpital Cochin, AP‐HP Centre‐Université de Paris Paris France
| | - Eduardo Calonje
- Department of Dermatopathology St John's Institute of Dermatology, St Thomas's Hospital London UK
| | - Boštjan Luzar
- Institute of Pathology Medical Faculty University of Ljubljana Ljubljana Slovenia
| | - Keisuke Goto
- Department of Pathology Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital Tokyo Japan
- Department of Diagnostic Pathology Shizuoka Cancer Center Hospital, Sunto Japan
- Department of Diagnostic Pathology and Cytology Osaka International Cancer Institute Osaka Japan
- Department of Dermatology Hyogo Cancer Center Akashi Japan
| | | | - Laurence Lamant
- CARADERM Network
- Department of Pathology, CHU Toulouse, Institut Universitaire du Cancer Toulouse Oncopole Université Toulouse III Paul Sabatier Toulouse France
| | - Brigitte Balme
- CARADERM Network
- Department of Pathology Centre Hospitalier Universitaire de Lyon Lyon France
| | | | - Agnes Neuville
- Contades Office of Pathological Anatomy and Cytology Strasbourg France
| | - Lydia Deschamps
- CARADERM Network
- Department of Pathology, APHP Bichat Hospital Paris France
| | - Elisabeth Auberger
- Service d'anatomie et cytologie pathologiques Hopital Simone Veil Eaubonne France
| | | | | | | | - Françoise Plantier
- Department of Pathology Hôpital Cochin, AP‐HP Centre‐Université de Paris Paris France
- Cabinet Mathurin Moreau Paris France
| | - Valérie Cales
- Department of Pathology Centre hospitalier de Pau Pau France
| | - Vincent Pinsolle
- Department of plastic and reconstructive surgery Hôpital Haut‐Lévêque, Centre hospitalier universitaire de Bordeaux Pessac France
| | - Denis Roblet
- Department of Pathology Centre hospitalier d'Angouleme Saint Michel France
| | - Frantz Dupuy
- Laboratoire de cytologie et d'anatomie pathologiques», le Bouscat France
| | - Alexiane Dallot
- Centre national de Dermatopathologie‐La Roquette Paris France
| | | | - Thomas Jouary
- CARADERM Network
- Department of Dermatology Centre Hospitalier de Pau Pau France
| | - Roland Houben
- Department of Dermatology, Venereology and Allergology University Hospital Würzburg, Josef‐Schneider‐Straße 2 Würzburg Germany
| | - David Schrama
- Department of Dermatology, Venereology and Allergology University Hospital Würzburg, Josef‐Schneider‐Straße 2 Würzburg Germany
| | - Sonja Hesbacher
- Department of Dermatology, Venereology and Allergology University Hospital Würzburg, Josef‐Schneider‐Straße 2 Würzburg Germany
| | - Nicolas Macagno
- CARADERM Network
- Department of Biopathology Center Léon Bérard Lyon France
- Department of Pathology Timone University Hospital Marseille France
| | - Maxime Battistella
- CARADERM Network
- Department of Pathology, APHP Hôpital Saint Louis Université Paris 7 Paris France
| | - Bernard Cribier
- CARADERM Network
- Dermatology Clinic, Hôpitaux Universitaires & Université de Strasbourg, Hôpital Civil Strasbourg France
| | - Beatrice Vergier
- Department of Pathology, Hôpital Haut‐Lévêque, CHU de Bordeaux Pessac France
| | - Arnaud Fouchardière
- CARADERM Network
- Department of Biopathology Center Léon Bérard Lyon France
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Cancer Research Center of Lyon, Equipe Labellisée Ligue contre le Cancer Lyon France
| | - Marie‐Laure Jullie
- CARADERM Network
- Department of Pathology, Hôpital Haut‐Lévêque, CHU de Bordeaux Pessac France
| |
Collapse
|
40
|
Li S, Fan T, Li C, Wang Y, Li J, Liu Z. Fate-mapping analysis of cochlear cells expressing Atoh1 mRNA via a new Atoh1 3*HA-P2A-Cre knockin mouse strain. Dev Dyn 2022; 251:1156-1174. [PMID: 35038200 DOI: 10.1002/dvdy.453] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Atoh1 is recognized to be essential for cochlear hair cell (HC) development. However, Atoh1 temporal and spatial expression patterns remain widely debated. Here, we aimed to obtain evidence to resolve the controversies regarding Atoh1 expression by generating a new knockin mouse strain: Atoh13*HA-P2A-Cre . RESULTS Fate-mapping analysis of Atoh13*HA-P2A-Cre/+ ; Rosa26-CAG-LSL-tdTomato (Ai9)/+ mice enabled us to concurrently characterize the temporal expression of Atoh1 protein (through HA-tag immunostaining) and visualize the cells expressing Atoh1 mRNA (as tdTomato+ cells). Our findings show that whereas Atoh1 mRNA expression is rapidly turned on in early cochlear progenitors, Atoh1 protein is only detected in differentiating HCs or progenitors just committed to the HC fate. Cre activity is also stronger in Atoh13*HA-P2A-Cre/+ than in previous mouse models, because almost all cochlear HCs and nearby supporting cells here are tdTomato+. Furthermore, tdTomato, but not HA, is expressed in middle and apical spiral ganglion neurons. CONCLUSION Collectively, our findings indicate that Atoh13*HA-P2A-Cre can serve as a powerful genetic model in the developmental biology field. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Shuting Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Ting Fan
- ENT Institute and Otorhinolaryngology Department, 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
| | - 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, China
| | - Yunfeng Wang
- ENT Institute and Otorhinolaryngology Department, 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
| | - Jian Li
- Clinical Laboratory Center, Children's Hospital of Fudan University, Shanghai, 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, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| |
Collapse
|
41
|
Ozment E, Tamvacakis AN, Zhou J, Rosiles-Loeza PY, Escobar-Hernandez EE, Fernandez-Valverde SL, Nakanishi N. Cnidarian hair cell development illuminates an ancient role for the class IV POU transcription factor in defining mechanoreceptor identity. eLife 2021; 10:74336. [PMID: 34939935 PMCID: PMC8846589 DOI: 10.7554/elife.74336] [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: 09/30/2021] [Accepted: 12/22/2021] [Indexed: 12/04/2022] Open
Abstract
Although specialized mechanosensory cells are found across animal phylogeny, early evolutionary histories of mechanoreceptor development remain enigmatic. Cnidaria (e.g. sea anemones and jellyfishes) is the sister group to well-studied Bilateria (e.g. flies and vertebrates), and has two mechanosensory cell types – a lineage-specific sensory effector known as the cnidocyte, and a classical mechanosensory neuron referred to as the hair cell. While developmental genetics of cnidocytes is increasingly understood, genes essential for cnidarian hair cell development are unknown. Here, we show that the class IV POU homeodomain transcription factor (POU-IV) – an indispensable regulator of mechanosensory cell differentiation in Bilateria and cnidocyte differentiation in Cnidaria – controls hair cell development in the sea anemone cnidarian Nematostella vectensis. N. vectensis POU-IV is postmitotically expressed in tentacular hair cells, and is necessary for development of the apical mechanosensory apparatus, but not of neurites, in hair cells. Moreover, it binds to deeply conserved DNA recognition elements, and turns on a unique set of effector genes – including the transmembrane receptor-encoding gene polycystin 1 – specifically in hair cells. Our results suggest that POU-IV directs differentiation of cnidarian hair cells and cnidocytes via distinct gene regulatory mechanisms, and support an evolutionarily ancient role for POU-IV in defining the mature state of mechanosensory neurons.
Collapse
Affiliation(s)
- Ethan Ozment
- Department of Biological Sciences, University of Arkansas, Fayetteville, United States
| | - Arianna N Tamvacakis
- Department of Biological Sciences, University of Arkansas, Fayetteville, United States
| | - Jianhong Zhou
- Department of Biological Sciences, University of Arkansas, Fayetteville, United States
| | - Pablo Yamild Rosiles-Loeza
- Unidad de Genómica Avanzada (Langebio), Centro de Investigación y de Estudios Avanzados del IPN, Irapuato, Mexico
| | | | - Selene L Fernandez-Valverde
- Unidad de Genómica Avanzada (Langebio), Centro de Investigación y de Estudios Avanzados del IPN, Irapuato, Mexico
| | - Nagayasu Nakanishi
- Department of Biological Sciences, University of Arkansas, Fayetteville, United States
| |
Collapse
|
42
|
Sun S, Li S, Luo Z, Ren M, He S, Wang G, Liu Z. Dual expression of Atoh1 and Ikzf2 promotes transformation of adult cochlear supporting cells into outer hair cells. eLife 2021; 10:66547. [PMID: 34477109 PMCID: PMC8439656 DOI: 10.7554/elife.66547] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 09/02/2021] [Indexed: 12/24/2022] Open
Abstract
Mammalian cochlear outer hair cells (OHCs) are essential for hearing. Severe hearing impairment follows OHC degeneration. Previous attempts at regenerating new OHCs from cochlear supporting cells (SCs) have been unsuccessful, notably lacking expression of the key OHC motor protein, Prestin. Thus, regeneration of Prestin+ OHCs represents a barrier to restore auditory function in vivo. Here, we reported the successful in vivo conversion of adult mouse cochlear SCs into Prestin+ OHC-like cells through the concurrent induction of two key transcriptional factors known to be necessary for OHC development: Atoh1 and Ikzf2. Single-cell RNA sequencing revealed the upregulation of 729 OHC genes and downregulation of 331 SC genes in OHC-like cells. The resulting differentiation status of these OHC-like cells was much more advanced than previously achieved. This study thus established an efficient approach to induce the regeneration of Prestin+ OHCs, paving the way for in vivo cochlear repair via SC transdifferentiation.
Collapse
Affiliation(s)
- Suhong Sun
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Shuting Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhengnan Luo
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Minhui Ren
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Shunji He
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Guangqin Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, 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, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| |
Collapse
|