1
|
Catozzi A, Peiris Pagès M, Humphrey S, Revill M, Morgan D, Roebuck J, Chen Y, Davies-Williams B, Brennan K, Mukarram Hossain ASM, Makeev VJ, Satia K, Sfyri PP, Galvin M, Coles D, Lallo A, Pearce SP, Kerr A, Priest L, Foy V, Carter M, Caeser R, Chan JM, Rudin CM, Blackhall F, Frese KK, Dive C, Simpson KL. Functional characterization of the ATOH1 molecular subtype indicates a pro-metastatic role in small cell lung cancer. Cell Rep 2025; 44:115603. [PMID: 40305287 PMCID: PMC12116416 DOI: 10.1016/j.celrep.2025.115603] [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: 05/24/2024] [Revised: 12/09/2024] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Molecular subtypes of small cell lung cancer (SCLC) have been described based on differential expression of the transcription factors (TFs) ASCL1, NEUROD1, and POU2F3 and immune-related genes. We previously reported an additional subtype based on expression of the neurogenic TF ATOH1 within our SCLC circulating tumor cell-derived explant (CDX) model biobank. Here, we show that ATOH1 protein is detected in 7 of 81 preclinical models and 16 of 102 clinical samples of SCLC. In CDX models, ATOH1 directly regulates neurogenesis and differentiation programs, consistent with roles in normal tissues. In ex vivo cultures of ATOH1+ CDXs, ATOH1 is required for cell survival. In vivo, ATOH1 depletion slows tumor growth and suppresses liver metastasis. Our data validate ATOH1 as a bona fide lineage-defining TF of SCLC with cell survival and pro-metastatic functions. Further investigation exploring ATOH1-driven vulnerabilities for targeted treatment with predictive biomarkers is warranted.
Collapse
Affiliation(s)
- Alessia Catozzi
- SCLC Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester M20 4BX, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK
| | - Maria Peiris Pagès
- SCLC Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester M20 4BX, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK
| | - Sam Humphrey
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Mitchell Revill
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Derrick Morgan
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Jordan Roebuck
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Yitao Chen
- SCLC Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester M20 4BX, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK
| | - Bethan Davies-Williams
- SCLC Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester M20 4BX, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK
| | - Kevin Brennan
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - A S Md Mukarram Hossain
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Vsevolod J Makeev
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Karishma Satia
- SCLC Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester M20 4BX, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK
| | - Pagona P Sfyri
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Melanie Galvin
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Darryl Coles
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Alice Lallo
- SCLC Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester M20 4BX, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK
| | - Simon P Pearce
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Alastair Kerr
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Lynsey Priest
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| | - Victoria Foy
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK; Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| | - Mathew Carter
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK; Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| | - Rebecca Caeser
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Joseph M Chan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Fiona Blackhall
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK; Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M20 4BX, UK
| | - Kristopher K Frese
- SCLC Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester M20 4BX, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| | - Caroline Dive
- SCLC Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester M20 4BX, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK.
| | - Kathryn L Simpson
- SCLC Biology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester M20 4BX, UK; Cancer Research UK Lung Cancer Centre of Excellence, Manchester and London, UK; Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, UK
| |
Collapse
|
2
|
Cheng YF, Kempfle JS, Chiang H, Tani K, Wang Q, Chen SH, Lenz D, Chen WY, Wu W, Petrillo M, Edge ASB. Sox2 interacts with Atoh1 and Huwe1 loci to regulate Atoh1 transcription and stability during hair cell differentiation. PLoS Genet 2025; 21:e1011573. [PMID: 39883720 PMCID: PMC11813075 DOI: 10.1371/journal.pgen.1011573] [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: 04/01/2024] [Revised: 02/11/2025] [Accepted: 01/13/2025] [Indexed: 02/01/2025] Open
Abstract
Stem cell pluripotency gene Sox2 stimulates expression of proneural basic-helix-loop-helix transcription factor Atoh1. Sox2 is necessary for the development of cochlear hair cells and binds to the Atoh1 3' enhancer to stimulate Atoh1 expression. We show here that Sox2 deletion in late embryogenesis results in the formation of extra hair cells, in contrast to the absence of hair cell development obtained after Sox2 knockout early in gestation. Sox2 overexpression decreased the level of Atoh1 protein despite an increase in Atoh1 mRNA. Sox2 upregulated E3 ubiquitin ligase, Huwe1, by direct binding to the Huwe1 gene. By upregulating its cognate E3 ligase, Sox2 disrupts the positive feedback loop through which Atoh1 protein increases the expression of Atoh1. We conclude that Sox2 initiates expression, while also limiting continued activity of bHLH transcription factor, Atoh1, and this inhibition represents a new mechanism for regulating the activity of this powerful initiator of hair cell development.
Collapse
Affiliation(s)
- Yen-Fu Cheng
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, United States of America
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
- Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Judith S. Kempfle
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, United States of America
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
| | - Hao Chiang
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, United States of America
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
| | - Kohsuke Tani
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, United States of America
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
| | - Quan Wang
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, United States of America
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
| | - Sheng-Hong Chen
- Lab for Cell Dynamics, Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- National Center for Theoretical Sciences, Physics Division, Taipei, Taiwan
| | - Danielle Lenz
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, United States of America
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
| | - Wei-Yi Chen
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wenjin Wu
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, United States of America
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
| | - Marco Petrillo
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, United States of America
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
| | - Albert S. B. Edge
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, United States of America
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts, United States of America
- Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, Massachusetts, United States of America
- Harvard Stem Cell Institute, Cambridge, Massachusetts, United States of America
| |
Collapse
|
3
|
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
|
4
|
Wang T, Yang T, Kedaigle A, Pregernig G, McCarthy R, Holmes B, Wu X, Becker L, Pan N, So K, Chen L, He J, Mahmoudi A, Negi S, Kowalczyk M, Gibson T, Druckenbrod N, Cheng AG, Burns J. Precise genetic control of ATOH1 enhances maturation of regenerated hair cells in the mature mouse utricle. Nat Commun 2024; 15:9166. [PMID: 39448563 PMCID: PMC11502789 DOI: 10.1038/s41467-024-53153-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Abstract
Vestibular hair cells are mechanoreceptors critical for detecting head position and motion. In mammals, hair cell loss causes vestibular dysfunction as spontaneous regeneration is nearly absent. Constitutive expression of exogenous ATOH1, a hair cell transcription factor, increases hair cell regeneration, however, these cells fail to fully mature. Here, we profiled mouse utricles at 14 time points, and defined transcriptomes of developing and mature vestibular hair cells. To mimic native hair cells which downregulate endogenous ATOH1 as they mature, we engineered viral vectors carrying the supporting cell promoters GFAP and RLBP1. In utricles damaged ex vivo, both CMV-ATOH1 and GFAP-ATOH1 increased regeneration more effectively than RLBP1-ATOH1, while GFAP-ATOH1 and RLBP1-ATOH1 induced hair cells with more mature transcriptomes. In utricles damaged in vivo, GFAP-ATOH1 induced regeneration of hair cells expressing genes indicative of maturing type II hair cells, and more hair cells with bundles and synapses than untreated organs. Together our results demonstrate the efficacy of spatiotemporal control of ATOH1 overexpression in inner ear hair cell regeneration.
Collapse
Affiliation(s)
- Tian Wang
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, 94305, USA
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Tian Yang
- Decibel Therapeutics, Boston, MA, 02215, USA
| | | | - Gabriela Pregernig
- Decibel Therapeutics, Boston, MA, 02215, USA
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Ryan McCarthy
- Decibel Therapeutics, Boston, MA, 02215, USA
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Ben Holmes
- Decibel Therapeutics, Boston, MA, 02215, USA
| | - Xudong Wu
- Decibel Therapeutics, Boston, MA, 02215, USA
| | - Lars Becker
- Decibel Therapeutics, Boston, MA, 02215, USA
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Ning Pan
- Decibel Therapeutics, Boston, MA, 02215, USA
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Kathy So
- Decibel Therapeutics, Boston, MA, 02215, USA
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Leon Chen
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, 94305, USA
| | - Jun He
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, 94305, USA
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Ahmad Mahmoudi
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, 94305, USA
| | - Soumya Negi
- Decibel Therapeutics, Boston, MA, 02215, USA
| | | | | | | | - Alan G Cheng
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Palo Alto, CA, 94305, USA.
| | | |
Collapse
|
5
|
Pedersen EA, Verhaegen ME, Joseph MK, Harms KL, Harms PW. Merkel cell carcinoma: updates in tumor biology, emerging therapies, and preclinical models. Front Oncol 2024; 14:1413793. [PMID: 39136002 PMCID: PMC11317257 DOI: 10.3389/fonc.2024.1413793] [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: 04/07/2024] [Accepted: 07/08/2024] [Indexed: 08/15/2024] Open
Abstract
Merkel cell carcinoma (MCC) is an aggressive cutaneous neuroendocrine carcinoma thought to arise via either viral (Merkel cell polyomavirus) or ultraviolet-associated pathways. Surgery and radiotherapy have historically been mainstays of management, and immunotherapy has improved outcomes for advanced disease. However, there remains a lack of effective therapy for those patients who fail to respond to these established approaches, underscoring a critical need to better understand MCC biology for more effective prognosis and treatment. Here, we review the fundamental aspects of MCC biology and the recent advances which have had profound impact on management. The first genetically-engineered mouse models for MCC tumorigenesis provide opportunities to understand the potential MCC cell of origin and may prove useful for preclinical investigation of novel therapeutics. The MCC cell of origin debate has also been advanced by recent observations of MCC arising in association with a clonally related hair follicle tumor or squamous cell carcinoma in situ. These studies also suggested a role for epigenetics in the origin of MCC, highlighting a potential utility for this therapeutic avenue in MCC. These and other therapeutic targets form the basis for a wealth of ongoing clinical trials to improve MCC management. Here, we review these recent advances in the context of the existing literature and implications for future investigations.
Collapse
Affiliation(s)
| | | | - Mallory K. Joseph
- Department of Dermatology, University of Michigan, Ann Arbor, MI, United States
| | - Kelly L. Harms
- Department of Dermatology, University of Michigan, Ann Arbor, MI, United States
| | - Paul W. Harms
- Department of Dermatology, University of Michigan, Ann Arbor, MI, United States
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
6
|
Ma J, Xia M, Guo J, Li W, Sun S, Chen B. MEK/ERK signaling drives the transdifferentiation of supporting cells into functional hair cells by modulating the Notch pathway. Stem Cells Transl Med 2024; 13:661-677. [PMID: 38709826 PMCID: PMC11227976 DOI: 10.1093/stcltm/szae030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 04/02/2024] [Indexed: 05/08/2024] Open
Abstract
Loss of cochlear hair cells (HCs) leads to permanent hearing loss in mammals, and regenerative medicine is regarded as an ideal strategy for hearing recovery. Limited genetic and pharmaceutical approaches for HC regeneration have been established, and the existing strategies cannot achieve recovery of auditory function. A promising target to promote HC regeneration is MEK/ERK signaling because dynamic shifts in its activity during the critical stages of inner ear development have been observed. Here, we first showed that MEK/ERK signaling is activated specifically in supporting cells (SCs) after aminoglycoside-induced HC injury. We then selected 4 MEK/ERK signaling inhibitors, and PD0325901 (PD03) was found to induce the transdifferentiation of functional supernumerary HCs from SCs in the neonatal mammalian cochlear epithelium. We next found that PD03 facilitated the generation of HCs in inner ear organoids. Through genome-wide high-throughput RNA sequencing and verification, we found that the Notch pathway is the downstream target of MEK/ERK signaling. Importantly, delivery of PD03 into the inner ear induced mild HC regeneration in vivo. Our study thus reveals the importance of MEK/ERK signaling in cell fate determination and suggests that PD03 might serve as a new approach for HC regeneration.
Collapse
Affiliation(s)
- Jiaoyao Ma
- Department of ENT Institute and Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Mingyu Xia
- Department of ENT Institute and Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Jin Guo
- Department of ENT Institute and Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Wen Li
- Department of ENT Institute and Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Shan Sun
- Department of ENT Institute and Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Bing Chen
- Department of ENT Institute and Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai, 200032, People’s Republic of China
| |
Collapse
|
7
|
Huang H, Majumder T, Khot B, Suriyaarachchi H, Yang T, Shao Q, Tirukovalluru S, Liu G. The role of microtubule-associated protein tau in netrin-1 attractive signaling. J Cell Sci 2024; 137:jcs261244. [PMID: 38197773 PMCID: PMC10906489 DOI: 10.1242/jcs.261244] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 11/24/2023] [Indexed: 01/11/2024] Open
Abstract
Direct binding of netrin receptors with dynamic microtubules (MTs) in the neuronal growth cone plays an important role in netrin-mediated axon guidance. However, how netrin-1 (NTN1) regulates MT dynamics in axon turning remains a major unanswered question. Here, we show that the coupling of netrin-1 receptor DCC with tau (MAPT)-regulated MTs is involved in netrin-1-promoted axon attraction. Tau directly interacts with DCC and partially overlaps with DCC in the growth cone of primary neurons. Netrin-1 induces this interaction and the colocalization of DCC and tau in the growth cone. The netrin-1-induced interaction of tau with DCC relies on MT dynamics and TUBB3, a highly dynamic β-tubulin isotype in developing neurons. Netrin-1 increased cosedimentation of DCC with tau and TUBB3 in MTs, and knockdown of either tau or TUBB3 mutually blocked this effect. Downregulation of endogenous tau levels by tau shRNAs inhibited netrin-1-induced axon outgrowth, branching and commissural axon attraction in vitro, and led to defects in spinal commissural axon projection in vivo. These findings suggest that tau is a key MT-associated protein coupling DCC with MT dynamics in netrin-1-promoted axon attraction.
Collapse
Affiliation(s)
- Huai Huang
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Tanushree Majumder
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Bhakti Khot
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Harindi Suriyaarachchi
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Tao Yang
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Qiangqiang Shao
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Shraddha Tirukovalluru
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| | - Guofa Liu
- Department of Biological Sciences, University of Toledo, M. S. 601, 2801 W. Bancroft St., Toledo, OH 43606, USA
| |
Collapse
|
8
|
Sanlav G, Baran B, Kum Özşengezer S, Kizmazoğlu D, Altun Z, Aktaş S, Olgun N. S-100 and MATH-1 Protein Expressions Can Be Useful for the Prediction of Clinical Outcome in Neuroblastoma Patients. J Pediatr Hematol Oncol 2024; 46:21-28. [PMID: 37943051 DOI: 10.1097/mph.0000000000002783] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 10/06/2023] [Indexed: 11/10/2023]
Abstract
Neuroblastoma (NB) is the most frequent extracranial solid tumor of childhood, remarkable for its broad spectrum of clinical behavior. This diversity in behavior correlates closely with defined clinical and biological features and combinations of prognostic variables are used for risk-group assignment. S-100 proteins have roles in differentiation and were shown to be frequently dysregulated in NB. MATH-1 protein plays role in neuronal cell differentiation through development. However, up to date, there are no studies evaluating the relationship between MATH-1 and NB. Grb2-associated binding (Gab) proteins have roles in the regulation of cell growth and differentiation. Gab1 was reported to be related to poor survival of high-risk NB patients. The aim of this study was to investigate the relationship between differentiation-related S-100, MATH-1, and Gab1 proteins and risk group and/or stages of NB. A significant relation was found between S-100 and early stages of NB. This study also revealed a significant association between MATH-1 and low-risk groups. S-100 and MATH-1 were also shown to provide survival advantages among stages and risk groups. The findings of this study support the assumption that S-100 and MATH-1 can be potential prognostic biomarkers for staging and risk-group assignment of NB patients. These proteins can be useful tools for clinicians to guide through treatment options, especially for the evaluation of tumor differentiation.
Collapse
Affiliation(s)
| | | | | | - Deniz Kizmazoğlu
- Pediatric Oncology, Dokuz Eylul University Institute of Oncology, İzmir, Turkey
| | | | | | - Nur Olgun
- Pediatric Oncology, Dokuz Eylul University Institute of Oncology, İzmir, Turkey
| |
Collapse
|
9
|
Veithen M, Huyghe A, Van Den Ackerveken P, Fukada SI, Kokubo H, Breuskin I, Nguyen L, Delacroix L, Malgrange B. Sox9 Inhibits Cochlear Hair Cell Fate by Upregulating Hey1 and HeyL Antagonists of Atoh1. Cells 2023; 12:2148. [PMID: 37681879 PMCID: PMC10486728 DOI: 10.3390/cells12172148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023] Open
Abstract
It is widely accepted that cell fate determination in the cochlea is tightly controlled by different transcription factors (TFs) that remain to be fully defined. Here, we show that Sox9, initially expressed in the entire sensory epithelium of the cochlea, progressively disappears from differentiating hair cells (HCs) and is finally restricted to supporting cells (SCs). By performing ex vivo electroporation of E13.5-E14.5 cochleae, we demonstrate that maintenance of Sox9 expression in the progenitors committed to HC fate blocks their differentiation, even if co-expressed with Atoh1, a transcription factor necessary and sufficient to form HC. Sox9 inhibits Atoh1 transcriptional activity by upregulating Hey1 and HeyL antagonists, and genetic ablation of these genes induces extra HCs along the cochlea. Although Sox9 suppression from sensory progenitors ex vivo leads to a modest increase in the number of HCs, it is not sufficient in vivo to induce supernumerary HC production in an inducible Sox9 knockout model. Taken together, these data show that Sox9 is downregulated from nascent HCs to allow the unfolding of their differentiation program. This may be critical for future strategies to promote fully mature HC formation in regeneration approaches.
Collapse
Affiliation(s)
- Mona Veithen
- Laboratory of Developmental Neurobiology, GIGA-Neurosciences, University of Liege, 4000 Liege, Belgium; (M.V.); (A.H.); (P.V.D.A.); (I.B.); (L.D.)
| | - Aurélia Huyghe
- Laboratory of Developmental Neurobiology, GIGA-Neurosciences, University of Liege, 4000 Liege, Belgium; (M.V.); (A.H.); (P.V.D.A.); (I.B.); (L.D.)
| | - Priscilla Van Den Ackerveken
- Laboratory of Developmental Neurobiology, GIGA-Neurosciences, University of Liege, 4000 Liege, Belgium; (M.V.); (A.H.); (P.V.D.A.); (I.B.); (L.D.)
| | - So-ichiro Fukada
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan;
| | - Hiroki Kokubo
- Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minamiku, Hiroshima 734-8551, Japan;
| | - Ingrid Breuskin
- Laboratory of Developmental Neurobiology, GIGA-Neurosciences, University of Liege, 4000 Liege, Belgium; (M.V.); (A.H.); (P.V.D.A.); (I.B.); (L.D.)
| | - Laurent Nguyen
- Laboratory of Molecular Regulation of Neurogenesis, GIGA-Neurosciences, University of Liege, 4000 Liege, Belgium;
| | - Laurence Delacroix
- Laboratory of Developmental Neurobiology, GIGA-Neurosciences, University of Liege, 4000 Liege, Belgium; (M.V.); (A.H.); (P.V.D.A.); (I.B.); (L.D.)
| | - Brigitte Malgrange
- Laboratory of Developmental Neurobiology, GIGA-Neurosciences, University of Liege, 4000 Liege, Belgium; (M.V.); (A.H.); (P.V.D.A.); (I.B.); (L.D.)
| |
Collapse
|
10
|
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
|
11
|
Wang Q, Wu J, Yang J, Huang S, Yuan Y, Dai P. Two SOX11 variants cause Coffin-Siris syndrome with a new feature of sensorineural hearing loss. Am J Med Genet A 2023; 191:183-189. [PMID: 36369738 PMCID: PMC10100107 DOI: 10.1002/ajmg.a.63011] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 08/24/2022] [Accepted: 09/22/2022] [Indexed: 11/13/2022]
Abstract
Coffin-Siris syndrome (CSS, OMIM#135900) is a rare congenital disorder associated with neurodevelopmental and dysmorphic features. The primary cause of CSS is pathogenic variants in any of 9 BAF chromatin-remodeling complex encoding genes or the genes SOX11 and PHF6. Herein, we performed whole-exome sequencing (WES) and a series of analyses of growth-related, auditory, and radiological findings in two probands with syndromic sensorineural hearing loss and inner ear malformations who exhibited distinctive facial features, intellectual disability, growth retardation, and fifth finger malformation. Two de novo variants in the SOX11 gene (c.148A>C:p.Lys50Asn; c.811_814del:p.Asn271Serfs*10) were detected in these probands and were identified as pathogenic variants as per ACMG guidelines. These probands were diagnosed as having CSS based upon clinical and genetic findings. This is the first report of CSS caused by variants in SOX11 gene in Chinese individuals. Deleterious SOX11 variants can result in sensorineural hearing loss with inner ear malformation, potentially extending the array of phenotypes associated with these pathogenic variants. We suggest that both genetic and clinical findings be considered when diagnosing syndromic hearing loss.
Collapse
Affiliation(s)
- Qiuquan Wang
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, Beijing, China.,State Key Lab of Hearing Science, Ministry of Education, Beijing, China.,Beijing Key Lab of hearing loss Prevention and Treatment, Beijing, China
| | - Jie Wu
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, Beijing, China.,State Key Lab of Hearing Science, Ministry of Education, Beijing, China.,Beijing Key Lab of hearing loss Prevention and Treatment, Beijing, China
| | - Jinyuan Yang
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, Beijing, China.,State Key Lab of Hearing Science, Ministry of Education, Beijing, China.,Beijing Key Lab of hearing loss Prevention and Treatment, Beijing, China
| | - Shasha Huang
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, Beijing, China.,State Key Lab of Hearing Science, Ministry of Education, Beijing, China.,Beijing Key Lab of hearing loss Prevention and Treatment, Beijing, China
| | - Yongyi Yuan
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, Beijing, China.,State Key Lab of Hearing Science, Ministry of Education, Beijing, China.,Beijing Key Lab of hearing loss Prevention and Treatment, Beijing, China
| | - Pu Dai
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, Beijing, China.,State Key Lab of Hearing Science, Ministry of Education, Beijing, China.,Beijing Key Lab of hearing loss Prevention and Treatment, Beijing, China
| |
Collapse
|
12
|
Generation of innervated cochlear organoid recapitulates early development of auditory unit. Stem Cell Reports 2022; 18:319-336. [PMID: 36584686 PMCID: PMC9860115 DOI: 10.1016/j.stemcr.2022.11.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/30/2022] Open
Abstract
Functional cochlear hair cells (HCs) innervated by spiral ganglion neurons (SGNs) are essential for hearing, whereas robust models that recapitulate the peripheral auditory circuity are still lacking. Here, we developed cochlear organoids with functional peripheral auditory circuity in a staging three-dimensional (3D) co-culture system by initially reprogramming cochlear progenitor cells (CPCs) with increased proliferative potency that could be long-term expanded, then stepwise inducing the differentiation of cochlear HCs, as well as the outgrowth of neurites from SGNs. The function of HCs and synapses within organoids was confirmed by a series of morphological and electrophysiological evaluations. Single-cell mRNA sequencing revealed the differentiation trajectories of CPCs toward the major cochlear cell types and the dynamic gene expression during organoid HC development, which resembled the pattern of native HCs. We established the cochlear organoids with functional synapses for the first time, which provides a platform for deciphering the mechanisms of sensorineural hearing loss.
Collapse
|
13
|
Shu M, Hong D, Lin H, Zhang J, Luo Z, Du Y, Sun Z, Yin M, Yin Y, Liu L, Bao S, Liu Z, Lu F, Huang J, Dai J. Single-cell chromatin accessibility identifies enhancer networks driving gene expression during spinal cord development in mouse. Dev Cell 2022; 57:2761-2775.e6. [PMID: 36495874 DOI: 10.1016/j.devcel.2022.11.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 06/22/2022] [Accepted: 11/16/2022] [Indexed: 12/14/2022]
Abstract
Spinal cord development is precisely orchestrated by spatiotemporal gene regulatory programs. However, the underlying epigenetic mechanisms remain largely elusive. Here, we profiled single-cell chromatin accessibility landscapes in mouse neural tubes spanning embryonic days 9.5-13.5. We identified neuronal-cell-cluster-specific cis-regulatory elements in neural progenitors and neurons. Furthermore, we applied a novel computational method, eNet, to build enhancer networks by integrating single-cell chromatin accessibility and gene expression data and identify the hub enhancers within enhancer networks. It was experimentally validated in vivo for Atoh1 that knockout of the hub enhancers, but not the non-hub enhancers, markedly decreased Atoh1 expression and reduced dp1/dI1 cells. Together, our work provides insights into the epigenetic regulation of spinal cord development and a proof-of-concept demonstration of enhancer networks as a general mechanism in transcriptional regulation.
Collapse
Affiliation(s)
- Muya Shu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Danni Hong
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Hongli Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Jixiang Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhengnan Luo
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Yi Du
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Zheng Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Man Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Yanyun Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Lifang Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Shilai Bao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Zhiyong Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Falong Lu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China.
| | - Jialiang Huang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
14
|
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
|
15
|
Avraham KB, Khalaily L, Noy Y, Kamal L, Koffler-Brill T, Taiber S. The noncoding genome and hearing loss. Hum Genet 2022; 141:323-333. [PMID: 34491412 DOI: 10.1007/s00439-021-02359-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 08/29/2021] [Indexed: 12/16/2022]
Abstract
The age of sequencing has provided unprecedented insights into the human genome. The coding region of the genome comprises nearly 20,000 genes, of which approximately 4000 are associated with human disease. Beyond the protein-coding genome, which accounts for only 3% of the genome, lies a vast pool of regulatory elements in the form of promoters, enhancers, RNA species, and other intricate elements. These features undoubtably influence human health and disease, and as a result, a great deal of effort is currently being invested in deciphering their identity and mechanism. While a paucity of material has caused a lag in identifying these elements in the inner ear, the emergence of technologies for dealing with a minimal number of cells now has the field working overtime to catch up. Studies on microRNAs (miRNAs), long non-coding RNAs (lncRNAs), methylation, histone modifications, and more are ongoing. A number of microRNAs and other noncoding elements are known to be associated with hearing impairment and there is promise that regulatory elements will serve as future tools and targets of therapeutics and diagnostics. This review covers the current state of the field and considers future directions for the noncoding genome and implications for hearing loss.
Collapse
Affiliation(s)
- Karen B Avraham
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, 6997801, Tel Aviv, Israel.
| | - Lama Khalaily
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Yael Noy
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Lara Kamal
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Tal Koffler-Brill
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Shahar Taiber
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine and Sagol School of Neuroscience, Tel Aviv University, 6997801, Tel Aviv, Israel
| |
Collapse
|
16
|
Khouri-Farah N, Guo Q, Morgan K, Shin J, Li JYH. Integrated single-cell transcriptomic and epigenetic study of cell state transition and lineage commitment in embryonic mouse cerebellum. SCIENCE ADVANCES 2022; 8:eabl9156. [PMID: 35363520 PMCID: PMC10938588 DOI: 10.1126/sciadv.abl9156] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 02/07/2022] [Indexed: 06/14/2023]
Abstract
Recent studies using single-cell RNA-sequencing have revealed cellular heterogeneity in the developing mammalian cerebellum, yet the regulatory logic underlying this cellular diversity remains to be elucidated. Using integrated single-cell RNA and ATAC analyses, we resolved developmental trajectories of cerebellar progenitors and identified putative trans- and cis-elements that control cell state transition. We reverse engineered gene regulatory networks (GRNs) of each cerebellar cell type. Through in silico simulations and in vivo experiments, we validated the efficacy of GRN analyses and uncovered the molecular control of a posterior transitory zone (PTZ), a distinct progenitor zone residing immediately anterior to the morphologically defined rhombic lip (RL). We showed that perturbing cell fate specification in the PTZ and RL causes posterior cerebellar vermis hypoplasia, the most common cerebellar birth defect in humans. Our study provides a foundation for comprehensive studies of developmental programs of the mammalian cerebellum.
Collapse
Affiliation(s)
- Nagham Khouri-Farah
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06030-6403, USA
| | - Qiuxia Guo
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06030-6403, USA
| | - Kerry Morgan
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06030-6403, USA
| | - Jihye Shin
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06030-6403, USA
| | - James Y. H. Li
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06030-6403, USA
- Institute for Systems Genomics, University of Connecticut, 400 Farmington Avenue, Farmington, CT 06030-6403, USA
| |
Collapse
|
17
|
Graf M, Interlandi M, Moreno N, Holdhof D, Göbel C, Melcher V, Mertins J, Albert TK, Kastrati D, Alfert A, Holsten T, de Faria F, Meisterernst M, Rossig C, Warmuth-Metz M, Nowak J, Meyer Zu Hörste G, Mayère C, Nef S, Johann P, Frühwald MC, Dugas M, Schüller U, Kerl K. Single-cell transcriptomics identifies potential cells of origin of MYC rhabdoid tumors. Nat Commun 2022; 13:1544. [PMID: 35318328 PMCID: PMC8941154 DOI: 10.1038/s41467-022-29152-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 02/26/2022] [Indexed: 11/30/2022] Open
Abstract
Rhabdoid tumors (RT) are rare and highly aggressive pediatric neoplasms. Their epigenetically-driven intertumoral heterogeneity is well described; however, the cellular origin of RT remains an enigma. Here, we establish and characterize different genetically engineered mouse models driven under the control of distinct promoters and being active in early progenitor cell types with diverse embryonic onsets. From all models only Sox2-positive progenitor cells give rise to murine RT. Using single-cell analyses, we identify distinct cells of origin for the SHH and MYC subgroups of RT, rooting in early stages of embryogenesis. Intra- and extracranial MYC tumors harbor common genetic programs and potentially originate from fetal primordial germ cells (PGCs). Using PGC specific Smarcb1 knockout mouse models we validate that MYC RT originate from these progenitor cells. We uncover an epigenetic imbalance in MYC tumors compared to PGCs being sustained by epigenetically-driven subpopulations. Importantly, treatments with the DNA demethylating agent decitabine successfully impair tumor growth in vitro and in vivo. In summary, our work sheds light on the origin of RT and supports the clinical relevance of DNA methyltransferase inhibitors against this disease. Rhabdoid tumors (RT) are aggressive paediatric cancers with yet unknown cells of origin. Here, the authors establish genetically engineered mouse models of RT and, using single-cell RNA-seq and epigenomics, identify potential cells of origin for the SHH and MYC subtypes.
Collapse
Affiliation(s)
- Monika Graf
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149, Münster, Germany
| | - Marta Interlandi
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149, Münster, Germany.,Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
| | - Natalia Moreno
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149, Münster, Germany
| | - Dörthe Holdhof
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.,Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Carolin Göbel
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.,Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Viktoria Melcher
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149, Münster, Germany
| | - Julius Mertins
- Department of Neurology, Schlosspark-Klinik, 14059, Berlin, Germany.,Institute of Molecular Tumor Biology, University of Münster, 48149, Münster, Germany
| | - Thomas K Albert
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149, Münster, Germany
| | - Dennis Kastrati
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149, Münster, Germany
| | - Amelie Alfert
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149, Münster, Germany
| | - Till Holsten
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.,Institute of Molecular Tumor Biology, University of Münster, 48149, Münster, Germany
| | - Flavia de Faria
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149, Münster, Germany.,Department of Pediatric Hematology and Oncology, Children's Hospital of Brasìlia, 70684-831, Brasìlia, Brazil
| | - Michael Meisterernst
- Institute of Molecular Tumor Biology, University of Münster, 48149, Münster, Germany
| | - Claudia Rossig
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149, Münster, Germany
| | - Monika Warmuth-Metz
- Neuroradiological Reference Center, University Hospital Würzburg, Würzburg, Germany
| | - Johannes Nowak
- Neuroradiological Reference Center, University Hospital Würzburg, Würzburg, Germany.,SRH Poliklinik Gera GmbH, Radiological Practice Gotha, Gotha, Germany
| | - Gerd Meyer Zu Hörste
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, 48149, Münster, Germany
| | - Chloe Mayère
- Department of Genetic Medicine and Development, University of Geneva, 1211, Geneva, Switzerland.,iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, 1211, Geneva, Switzerland
| | - Serge Nef
- Department of Genetic Medicine and Development, University of Geneva, 1211, Geneva, Switzerland.,iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, 1211, Geneva, Switzerland
| | - Pascal Johann
- Swabian Children's Cancer Center, Paediatric and Adolescent Medicine, University Medical Center Augsburg, 86156, Augsburg, Germany.,Division of Pediatric Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael C Frühwald
- Swabian Children's Cancer Center, Paediatric and Adolescent Medicine, University Medical Center Augsburg, 86156, Augsburg, Germany
| | - Martin Dugas
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany.,Institute of Medical Informatics, Heidelberg University Hospital, Heidelberg, Germany
| | - Ulrich Schüller
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.,Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.,Research Institute Children's Cancer Center, 20251, Hamburg, Germany
| | - Kornelius Kerl
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149, Münster, Germany.
| |
Collapse
|
18
|
Herrera E, Escalante A. Transcriptional Control of Axon Guidance at Midline Structures. Front Cell Dev Biol 2022; 10:840005. [PMID: 35265625 PMCID: PMC8900194 DOI: 10.3389/fcell.2022.840005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
The development of the nervous system is a time-ordered and multi-stepped process that includes neurogenesis and neuronal specification, axonal navigation, and circuits assembly. During axonal navigation, the growth cone, a dynamic structure located at the tip of the axon, senses environmental signals that guide axons towards their final targets. The expression of a specific repertoire of receptors on the cell surface of the growth cone together with the activation of a set of intracellular transducing molecules, outlines the response of each axon to specific guidance cues. This collection of axon guidance molecules is defined by the transcriptome of the cell which, in turn, depends on transcriptional and epigenetic regulators that modify the structure and DNA accessibility to determine what genes will be expressed to elicit specific axonal behaviors. Studies focused on understanding how axons navigate intermediate targets, such as the floor plate of vertebrates or the mammalian optic chiasm, have largely contributed to our knowledge of how neurons wire together during development. In fact, investigations on axon navigation at these midline structures led to the identification of many of the currently known families of proteins that act as guidance cues and their corresponding receptors. Although the transcription factors and the regulatory mechanisms that control the expression of these molecules are not well understood, important advances have been made in recent years in this regard. Here we provide an updated overview on the current knowledge about the transcriptional control of axon guidance and the selection of trajectories at midline structures.
Collapse
|
19
|
Kwan KY, White PM. Understanding the differentiation and epigenetics of cochlear sensory progenitors in pursuit of regeneration. Curr Opin Otolaryngol Head Neck Surg 2021; 29:366-372. [PMID: 34374667 PMCID: PMC8452321 DOI: 10.1097/moo.0000000000000741] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PURPOSE OF REVIEW Sensory hair cells (HCs) of the inner ear are responsible for our ability to hear and balance. Loss of these cells results in hearing loss. Stem cell replacement and in situ regeneration have the potential to replace lost HCs. Newly discovered contributions of transcription factor regulatory networks and epigenetic mechanisms in regulating HC differentiation and regeneration are placed into context of the literature. RECENT FINDINGS A wealth of new data has helped to define cochlear sensory progenitors in their developmental trajectories. This includes transcription factor networks, epigenetic manipulations, and cochlear HC subtype specification. SUMMARY Understanding how sensory progenitors differ and how HC subtypes arise will substantially inform efforts in hearing restoration.
Collapse
Affiliation(s)
- Kelvin Y. Kwan
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway, New Jersey
| | - Patricia M. White
- Department of Neuroscience, Ernest J. Del Monte Institute of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
20
|
Gómez-Dorado M, Daudet N, Gale JE, Dawson SJ. Differential regulation of mammalian and avian ATOH1 by E2F1 and its implication for hair cell regeneration in the inner ear. Sci Rep 2021; 11:19368. [PMID: 34588543 PMCID: PMC8481459 DOI: 10.1038/s41598-021-98816-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/31/2021] [Indexed: 12/23/2022] Open
Abstract
The mammalian inner ear has a limited capacity to regenerate its mechanosensory hair cells. This lack of regenerative capacity underlies the high incidence of age-related hearing loss in humans. In contrast, non-mammalian vertebrates can form new hair cells when damage occurs, a mechanism that depends on re-activation of expression of the pro-hair cell transcription factor Atoh1. Here, we show that members of the E2F transcription factor family, known to play a key role in cell cycle progression, regulate the expression of Atoh1. E2F1 activates chicken Atoh1 by directly interacting with a cis-regulatory region distal to the avian Atoh1 gene. E2F does not activate mouse Atoh1 gene expression, since this regulatory element is absent in mammals. We also show that E2F1 expression changes dynamically in the chicken auditory epithelium during ototoxic damage and hair cell regeneration. Therefore, we propose a model in which the mitotic regeneration of non-mammalian hair cells is due to E2F1-mediated activation of Atoh1 expression, a mechanism which has been lost in mammals.
Collapse
Affiliation(s)
| | - Nicolas Daudet
- UCL Ear Institute, 332 Gray's Inn Road, London, WC1X 8EE, UK
| | - Jonathan E Gale
- UCL Ear Institute, 332 Gray's Inn Road, London, WC1X 8EE, UK
| | - Sally J Dawson
- UCL Ear Institute, 332 Gray's Inn Road, London, WC1X 8EE, UK.
| |
Collapse
|
21
|
Enhancer decommissioning imposes an epigenetic barrier to sensory hair cell regeneration. Dev Cell 2021; 56:2471-2485.e5. [PMID: 34331868 DOI: 10.1016/j.devcel.2021.07.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/24/2021] [Accepted: 07/08/2021] [Indexed: 01/02/2023]
Abstract
Adult mammalian tissues such as heart, brain, retina, and the sensory structures of the inner ear do not effectively regenerate, although a latent capacity for regeneration exists at embryonic and perinatal times. We explored the epigenetic basis for this latent regenerative potential in the mouse inner ear and its rapid loss during maturation. In perinatal supporting cells, whose fate is maintained by Notch-mediated lateral inhibition, the hair cell enhancer network is epigenetically primed (H3K4me1) but silenced (active H3K27 de-acetylation and trimethylation). Blocking Notch signaling during the perinatal period of plasticity rapidly eliminates epigenetic silencing and allows supporting cells to transdifferentiate into hair cells. Importantly, H3K4me1 priming of the hair cell enhancers in supporting cells is removed during the first post-natal week, coinciding with the loss of transdifferentiation potential. We hypothesize that enhancer decommissioning during cochlear maturation contributes to the failure of hair cell regeneration in the mature organ of Corti.
Collapse
|
22
|
Malawsky DS, Weir SJ, Ocasio JK, Babcock B, Dismuke T, Cleveland AH, Donson AM, Vibhakar R, Wilhelmsen K, Gershon TR. Cryptic developmental events determine medulloblastoma radiosensitivity and cellular heterogeneity without altering transcriptomic profile. Commun Biol 2021; 4:616. [PMID: 34021242 PMCID: PMC8139976 DOI: 10.1038/s42003-021-02099-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 04/12/2021] [Indexed: 12/20/2022] Open
Abstract
It is unclear why medulloblastoma patients receiving similar treatments experience different outcomes. Transcriptomic profiling identified subgroups with different prognoses, but in each subgroup, individuals remain at risk of incurable recurrence. To investigate why similar-appearing tumors produce variable outcomes, we analyzed medulloblastomas triggered in transgenic mice by a common driver mutation expressed at different points in brain development. We genetically engineered mice to express oncogenic SmoM2, starting in multipotent glio-neuronal stem cells, or committed neural progenitors. Both groups developed medulloblastomas with similar transcriptomic profiles. We compared medulloblastoma progression, radiosensitivity, and cellular heterogeneity, determined by single-cell transcriptomic analysis (scRNA-seq). Stem cell-triggered medulloblastomas progressed faster, contained more OLIG2-expressing stem-like cells, and consistently showed radioresistance. In contrast, progenitor-triggered MBs progressed slower, down-regulated stem-like cells and were curable with radiation. Progenitor-triggered medulloblastomas also contained more diverse stromal populations, with more Ccr2+ macrophages and fewer Igf1+ microglia, indicating that developmental events affected the subsequent tumor microenvironment. Reduced mTORC1 activity in M-Smo tumors suggests that differential Igf1 contributed to differences in phenotype. Developmental events in tumorigenesis that were obscure in transcriptomic profiles thus remained cryptic determinants of tumor composition and outcome. Precise understanding of medulloblastoma pathogenesis and prognosis requires supplementing transcriptomic/methylomic studies with analyses that resolve cellular heterogeneity.
Collapse
Affiliation(s)
- Daniel Shiloh Malawsky
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Seth J Weir
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Jennifer Karin Ocasio
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Benjamin Babcock
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Taylor Dismuke
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Abigail H Cleveland
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- UNC Cancer Cell Biology Training Program, University of North Carolina, Chapel Hill, NC, USA
| | - Andrew M Donson
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's, Hospital Colorado, Aurora, CO, USA
| | - Rajeev Vibhakar
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's, Hospital Colorado, Aurora, CO, USA
| | - Kirk Wilhelmsen
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
- Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
- RENCI, Chapel Hill, NC, USA.
| | - Timothy R Gershon
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
- UNC Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
23
|
Abdul-Aziz D, Hathiramani N, Phung L, Sykopetrites V, Edge ASB. HIC1 Represses Atoh1 Transcription and Hair Cell Differentiation in the Cochlea. Stem Cell Reports 2021; 16:797-809. [PMID: 33770497 PMCID: PMC8072069 DOI: 10.1016/j.stemcr.2021.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 02/26/2021] [Accepted: 02/27/2021] [Indexed: 11/23/2022] Open
Abstract
Across species, expression of the basic helix-loop-helix transcription factor ATOH1 promotes differentiation of cochlear supporting cells to sensory hair cells required for hearing. In mammals, this process is limited to development, whereas nonmammalian vertebrates can also regenerate hair cells after injury. The mechanistic basis for this difference is not fully understood. Hypermethylated in cancer 1 (HIC1) is a transcriptional repressor known to inhibit Atoh1 in the cerebellum. We therefore investigated its potential role in cochlear hair cell differentiation. We find that Hic1 is expressed throughout the postnatal murine cochlear sensory epithelium. In cochlear organoids, Hic1 knockdown induces Atoh1 expression and promotes hair cell differentiation, while Hic1 overexpression hinders differentiation. Wild-type HIC1, but not the DNA-binding mutant C521S, suppresses activity of the Atoh1 autoregulatory enhancer and blocks its responsiveness to β-catenin activation. Our findings reveal the importance of HIC1 repression of Atoh1 in the cochlea, which may be targeted to promote hair cell regeneration.
Collapse
Affiliation(s)
- Dunia Abdul-Aziz
- Department of Otolaryngology, Harvard Medical School, Boston, MA, USA; Eaton Peabody Laboratory, Massachusetts Eye and Ear, Boston, MA, USA
| | | | - Lauren Phung
- Eaton Peabody Laboratory, Massachusetts Eye and Ear, Boston, MA, USA
| | - Vittoria Sykopetrites
- Eaton Peabody Laboratory, Massachusetts Eye and Ear, Boston, MA, USA; Università degli Studi di Milano, Milan, Italy
| | - Albert S B Edge
- Department of Otolaryngology, Harvard Medical School, Boston, MA, USA; Eaton Peabody Laboratory, Massachusetts Eye and Ear, Boston, MA, USA; Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
24
|
Behesti H, Kocabas A, Buchholz DE, Carroll TS, Hatten ME. Altered temporal sequence of transcriptional regulators in the generation of human cerebellar granule cells. eLife 2021; 10:67074. [PMID: 34842137 PMCID: PMC8687658 DOI: 10.7554/elife.67074] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 11/27/2021] [Indexed: 02/07/2023] Open
Abstract
Brain development is regulated by conserved transcriptional programs across species, but little is known about the divergent mechanisms that create species-specific characteristics. Among brain regions, human cerebellar histogenesis differs in complexity compared with nonhuman primates and rodents, making it important to develop methods to generate human cerebellar neurons that closely resemble those in the developing human cerebellum. We report a rapid protocol for the derivation of the human ATOH1 lineage, the precursor of excitatory cerebellar neurons, from human pluripotent stem cells (hPSCs). Upon transplantation into juvenile mice, hPSC-derived cerebellar granule cells migrated along glial fibers and integrated into the cerebellar cortex. By Translational Ribosome Affinity Purification-seq, we identified an unexpected temporal shift in the expression of RBFOX3 (NeuN) and NEUROD1, which are classically associated with differentiated neurons, in the human outer external granule layer. This molecular divergence may enable the protracted development of the human cerebellum compared to mice.
Collapse
Affiliation(s)
- Hourinaz Behesti
- Laboratory of Developmental Neurobiology, Rockefeller UniversityNew YorkUnited States
| | - Arif Kocabas
- Laboratory of Developmental Neurobiology, Rockefeller UniversityNew YorkUnited States
| | - David E Buchholz
- Laboratory of Developmental Neurobiology, Rockefeller UniversityNew YorkUnited States
| | - Thomas S Carroll
- Bioinformatics Resource Center, Rockefeller UniversityNew YorkUnited States
| | - Mary E Hatten
- Laboratory of Developmental Neurobiology, Rockefeller UniversityNew YorkUnited States
| |
Collapse
|
25
|
Veleta KA, Cleveland AH, Babcock BR, He YW, Hwang D, Sokolsky-Papkov M, Gershon TR. Antiapoptotic Bcl-2 family proteins BCL-xL and MCL-1 integrate neural progenitor survival and proliferation during postnatal cerebellar neurogenesis. Cell Death Differ 2020; 28:1579-1592. [PMID: 33293647 DOI: 10.1038/s41418-020-00687-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/11/2020] [Accepted: 11/16/2020] [Indexed: 11/09/2022] Open
Abstract
The tendency of brain cells to undergo apoptosis in response to exogenous events varies across neural development, with apoptotic threshold dependent on proliferation state. Proliferative neural progenitors show a low threshold for apoptosis, while terminally differentiated neurons are relatively refractory. To define the mechanisms linking proliferation and apoptotic threshold, we examined the effect of conditionally deleting Bcl2l1, the gene that codes the antiapoptotic protein BCL-xL, in cerebellar granule neuron progenitors (CGNPs), and of co-deleting Bcl2l1 homologs, antiapoptotic Mcl-1, or pro-apoptotic Bax. We found that cerebella in conditional Bcl2l1-deleted (Bcl-xLcKO) mice were severely hypoplastic due to the increased apoptosis of CGNPs and their differentiated progeny, the cerebellar granule neurons (CGNs). Apoptosis was highest as Bcl-xLcKO CGNPs exited the cell cycle to initiate differentiation, with proliferating Bcl-xLcKO CGNPs relatively less affected. Despite the overall reduction in cerebellar growth, SHH-dependent proliferation was prolonged in Bcl-xLcKO mice, as more CGNPs remained proliferative in the second postnatal week. Co-deletion of Bax rescued the Bcl-xLcKO phenotype, while co-deletion of Mcl-1 enhanced the phenotype. These findings show that CGNPs require BCL-xL to regulate BAX-dependent apoptosis, and that this role can be partially compensated by MCL-1. Our data further show that BCL-xL expression regulates MCL-1 abundance in CGNPs, and suggest that excessive MCL-1 in Bcl-xLcKO mice prolongs CGNP proliferation by binding SUFU, resulting in increased SHH pathway activation. Accordingly, we propose that BCL-xL and MCL-1 interact with each other and with developmental mechanisms that regulate proliferation, to adjust the apoptotic threshold as CGNPs progress through postnatal neurogenesis to CGNs.
Collapse
Affiliation(s)
- Katherine A Veleta
- UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Abigail H Cleveland
- UNC Cancer Cell Biology Training Program, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Benjamin R Babcock
- Department of Neurology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - You-Wen He
- Department of Immunology, Duke University, Durham, NC, 27708, USA
| | - Duhyeong Hwang
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Marina Sokolsky-Papkov
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Timothy R Gershon
- UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599, USA. .,Department of Neurology, UNC School of Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA. .,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
26
|
Constitutive Activation of Nrf2 in Mice Expands Enterogenesis in Small Intestine Through Negative Regulation of Math1. Cell Mol Gastroenterol Hepatol 2020; 11:503-524. [PMID: 32896624 PMCID: PMC7797379 DOI: 10.1016/j.jcmgh.2020.08.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Notch signaling coordinates cell differentiation processes in the intestinal epithelium. The transcription factor Nrf2 orchestrates defense mechanisms by regulating cellular redox homeostasis, which, as shown previously in murine liver, can be amplified through signaling crosstalk with the Notch pathway. However, interplay between these 2 signaling pathways in the gut is unknown. METHODS Mice modified genetically to amplify Nrf2 in the intestinal epithelium (Keap1f/f::VilCre) were generated as well as pharmacological activation of Nrf2 and subjected to phenotypic and cell lineage analyses. Cell lines were used for reporter gene assays together with Nrf2 overexpression to study transcriptional regulation of the Notch downstream effector. RESULTS Constitutive activation of Nrf2 signaling caused increased intestinal length along with expanded cell number and thickness of enterocytes without any alterations of secretory lineage, outcomes abrogated by concomitant disruption of Nrf2. The Nrf2 and Notch pathways in epithelium showed inverse spatial profiles, where Nrf2 activity in crypts was lower than villi. In progenitor cells of Keap1f/f::VilCre mice, Notch downstream effector Math1, which regulates a differentiation balance of cell lineage through lateral inhibition, showed suppressed expression. In vitro results demonstrated Nrf2 negatively regulated Math1, where 6 antioxidant response elements located in the regulatory regions contributed to this repression. CONCLUSIONS Activation of Nrf2 perturbed the dialog of the Notch cascade though negative regulation of Math1 in progenitor cells, leading to enhanced enterogenesis. The crosstalk between the Nrf2 and Notch pathways could be critical for fine-tuning intestinal homeostasis and point to new approaches for the pharmacological management of absorptive deficiencies.
Collapse
|
27
|
Wang X, Kohl A, Yu X, Zorio DAR, Klar A, Sela-Donenfeld D, Wang Y. Temporal-specific roles of fragile X mental retardation protein in the development of the hindbrain auditory circuit. Development 2020; 147:dev.188797. [PMID: 32747436 DOI: 10.1242/dev.188797] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 06/29/2020] [Indexed: 01/01/2023]
Abstract
Fragile X mental retardation protein (FMRP) is an RNA-binding protein abundant in the nervous system. Functional loss of FMRP leads to sensory dysfunction and severe intellectual disabilities. In the auditory system, FMRP deficiency alters neuronal function and synaptic connectivity and results in perturbed processing of sound information. Nevertheless, roles of FMRP in embryonic development of the auditory hindbrain have not been identified. Here, we developed high-specificity approaches to genetically track and manipulate throughout development of the Atoh1+ neuronal cell type, which is highly conserved in vertebrates, in the cochlear nucleus of chicken embryos. We identified distinct FMRP-containing granules in the growing axons of Atoh1+ neurons and post-migrating NM cells. FMRP downregulation induced by CRISPR/Cas9 and shRNA techniques resulted in perturbed axonal pathfinding, delay in midline crossing, excess branching of neurites, and axonal targeting errors during the period of circuit development. Together, these results provide the first in vivo identification of FMRP localization and actions in developing axons of auditory neurons, and demonstrate the importance of investigating early embryonic alterations toward understanding the pathogenesis of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Department of Biomedical Sciences, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, FL 32306, USA.,Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou 510632, China
| | - Ayelet Kohl
- Koret School of Veterinary Medicine, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Xiaoyan Yu
- Department of Biomedical Sciences, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Diego A R Zorio
- Department of Biomedical Sciences, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Avihu Klar
- Department of Medical Neurobiology IMRIC, Hebrew University Medical School, Jerusalem 91120, Israel
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Yuan Wang
- Department of Biomedical Sciences, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| |
Collapse
|
28
|
Notch Signalling: The Multitask Manager of Inner Ear Development and Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1218:129-157. [DOI: 10.1007/978-3-030-34436-8_8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
29
|
Ocasio J, Babcock B, Malawsky D, Weir SJ, Loo L, Simon JM, Zylka MJ, Hwang D, Dismuke T, Sokolsky M, Rosen EP, Vibhakar R, Zhang J, Saulnier O, Vladoiu M, El-Hamamy I, Stein LD, Taylor MD, Smith KS, Northcott PA, Colaneri A, Wilhelmsen K, Gershon TR. scRNA-seq in medulloblastoma shows cellular heterogeneity and lineage expansion support resistance to SHH inhibitor therapy. Nat Commun 2019; 10:5829. [PMID: 31863004 PMCID: PMC6925218 DOI: 10.1038/s41467-019-13657-6] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 11/14/2019] [Indexed: 01/23/2023] Open
Abstract
Targeting oncogenic pathways holds promise for brain tumor treatment, but inhibition of Sonic Hedgehog (SHH) signaling has failed in SHH-driven medulloblastoma. Cellular diversity within tumors and reduced lineage commitment can undermine targeted therapy by increasing the probability of treatment-resistant populations. Using single-cell RNA-seq and lineage tracing, we analyzed cellular diversity in medulloblastomas in transgenic, medulloblastoma-prone mice, and responses to the SHH-pathway inhibitor vismodegib. In untreated tumors, we find expected stromal cells and tumor-derived cells showing either a spectrum of neural progenitor-differentiation states or glial and stem cell markers. Vismodegib reduces the proliferative population and increases differentiation. However, specific cell types in vismodegib-treated tumors remain proliferative, showing either persistent SHH-pathway activation or stem cell characteristics. Our data show that even in tumors with a single pathway-activating mutation, diverse mechanisms drive tumor growth. This diversity confers early resistance to targeted inhibitor therapy, demonstrating the need to target multiple pathways simultaneously. Although the hedgehog (HH) pathway is known to be deregulated in medulloblastoma, inhibitors of the pathway have shown disappointing clinical benefit. Using single-cell sequencing in a mouse model of the disease, the authors show that the response to the HH pathway inhibitor vismodegib is cell-type specific.
Collapse
Affiliation(s)
- Jennifer Ocasio
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA.,UNC Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Benjamin Babcock
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA.,Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Daniel Malawsky
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Seth J Weir
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Lipin Loo
- UNC Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA.,Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Jeremy M Simon
- UNC Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA.,Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA.,Carolina Institute for Developmental Disabilities, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Mark J Zylka
- UNC Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA.,Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA.,Carolina Institute for Developmental Disabilities, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Duhyeong Hwang
- UNC Eshelman School of Pharmacy, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Taylor Dismuke
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Marina Sokolsky
- UNC Eshelman School of Pharmacy, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Elias P Rosen
- UNC Eshelman School of Pharmacy, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Rajeev Vibhakar
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, CO, USA
| | - Jiao Zhang
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
| | - Olivier Saulnier
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
| | - Maria Vladoiu
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
| | - Ibrahim El-Hamamy
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5G 0A4, Canada.,Program in Computational Biology, Ontario Institute for Cancer Research, Toronto, ON, M5G 0A3, Canada
| | - Lincoln D Stein
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5G 0A4, Canada.,Program in Computational Biology, Ontario Institute for Cancer Research, Toronto, ON, M5G 0A3, Canada
| | - Michael D Taylor
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada.,Division of Neurosurgery, The Hospital for Sick Children, Toronto, ON, M5S 3E1, Canada
| | - Kyle S Smith
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Paul A Northcott
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Alejandro Colaneri
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA.,Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Kirk Wilhelmsen
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA. .,Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA. .,Renaissance Computing Institute at UNC (RENCI), Chapel Hill, NC, 27517, USA.
| | - Timothy R Gershon
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA. .,UNC Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA. .,Carolina Institute for Developmental Disabilities, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA. .,Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
30
|
Tateya T, Sakamoto S, Ishidate F, Hirashima T, Imayoshi I, Kageyama R. Three-dimensional live imaging of Atoh1 reveals the dynamics of hair cell induction and organization in the developing cochlea. Development 2019; 146:146/21/dev177881. [PMID: 31676552 DOI: 10.1242/dev.177881] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 09/27/2019] [Indexed: 01/06/2023]
Abstract
During cochlear development, hair cells (HCs) and supporting cells differentiate in the prosensory domain to form the organ of Corti, but how one row of inner HCs (IHCs) and three rows of outer HCs (OHCs) are organized is not well understood. Here, we investigated the process of HC induction by monitoring Atoh1 expression in cochlear explants of Atoh1-EGFP knock-in mouse embryos and showed that only the cells that express Atoh1 over a certain threshold are selected for HC fate determination. HC induction initially occurs at the medial edge of the prosensory domain to form IHCs and subsequently at the lateral edge to form OHCs, while Hedgehog signaling maintains a space between IHCs and OHCs, leading to formation of the tunnel of Corti. These results reveal dynamic Atoh1 expression in HC fate control and suggest that multi-directional signals regulate OHC induction, thereby organizing the prototype of the organ of Corti.
Collapse
Affiliation(s)
- Tomoko Tateya
- Department of Otolaryngology - Head and Neck Surgery, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan .,Department of Speech and Hearing Sciences and Disorders, Faculty of Health and Medical Science, Kyoto University of Advanced Science, Kyoto 615-8577, Japan.,Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Susumu Sakamoto
- Department of Otolaryngology - Head and Neck Surgery, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan.,Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Fumiyoshi Ishidate
- Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Tsuyoshi Hirashima
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Itaru Imayoshi
- Research Center for Systemic Life Science, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Ryoichiro Kageyama
- Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan .,Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto 606-8501, Japan.,Department of Growth Regulation, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan.,Department of Molecular and Cellular Biology, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
31
|
Munnamalai V, Fekete DM. The acquisition of positional information across the radial axis of the cochlea. Dev Dyn 2019; 249:281-297. [PMID: 31566832 DOI: 10.1002/dvdy.118] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 09/10/2019] [Accepted: 09/10/2019] [Indexed: 12/11/2022] Open
Affiliation(s)
- Vidhya Munnamalai
- The Jackson Laboratory Bar Harbor Maine
- Graduate Program of Biomedical Sciences and EngineeringUniversity of Maine Orono Maine
- The Neuroscience ProgramSackler School of Biomedical Sciences, Tufts University Boston Massachusetts
| | - Donna M. Fekete
- Department of Biological SciencesPurdue University West Lafayette Indiana
- Purdue Institute for Integrative Neuroscience West Lafayette Indiana
- Purdue Center for Cancer Research West Lafayette Indiana
| |
Collapse
|
32
|
Kaneyama T, Shirasaki R. Post-crossing segment of dI1 commissural axons forms collateral branches to motor neurons in the developing spinal cord. J Comp Neurol 2019; 526:1943-1961. [PMID: 29752714 DOI: 10.1002/cne.24464] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 03/30/2018] [Accepted: 05/03/2018] [Indexed: 11/09/2022]
Abstract
The dI1 commissural axons in the developing spinal cord, upon crossing the midline through the floor plate, make a sharp turn to grow rostrally. These post-crossing axons initially just extend adjacent to the floor plate without entering nearby motor columns. However, it remains poorly characterized how these post-crossing dI1 axons behave subsequently to this process. In the present study, to address this issue, we examined in detail the behavior of post-crossing dI1 axons in mice, using the Atoh1 enhancer-based conditional expression system that enables selective and sparse labeling of individual dI1 axons, together with Hb9 and ChAT immunohistochemistry for precise identification of spinal motor neurons (MNs). We found unexpectedly that the post-crossing segment of dI1 axons later gave off collateral branches that extended laterally to invade motor columns. Interestingly, these collateral branches emerged at around the time when their primary growth cones initiated invasion into motor columns. In addition, although the length of the laterally growing collateral branches increased with age, the majority of them remained within motor columns. Strikingly, these collateral branches further gave rise to multiple secondary branches in the region of MNs that innervate muscles close to the body axis. Moreover, these axonal branches formed presynaptic terminals on MNs. These observations demonstrate that dI1 commissural neurons develop axonal projection to spinal MNs via collateral branches arising later from the post-crossing segment of these axons. Our findings thus reveal a previously unrecognized projection of dI1 commissural axons that may contribute directly to generating proper motor output.
Collapse
Affiliation(s)
- Takeshi Kaneyama
- Cellular and Molecular Neurobiology Laboratory, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Ryuichi Shirasaki
- Cellular and Molecular Neurobiology Laboratory, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
33
|
Open chromatin dynamics in prosensory cells of the embryonic mouse cochlea. Sci Rep 2019; 9:9060. [PMID: 31227770 PMCID: PMC6588700 DOI: 10.1038/s41598-019-45515-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 06/10/2019] [Indexed: 12/13/2022] Open
Abstract
Hearing loss is often due to the absence or the degeneration of hair cells in the cochlea. Understanding the mechanisms regulating the generation of hair cells may therefore lead to better treatments for hearing disorders. To elucidate the transcriptional control mechanisms specifying the progenitor cells (i.e. prosensory cells) that generate the hair cells and support cells critical for hearing function, we compared chromatin accessibility using ATAC-seq in sorted prosensory cells (Sox2-EGFP+) and surrounding cells (Sox2-EGFP−) from E12, E14.5 and E16 cochlear ducts. In Sox2-EGFP+, we find greater accessibility in and near genes restricted in expression to the prosensory region of the cochlear duct including Sox2, Isl1, Eya1 and Pou4f3. Furthermore, we find significant enrichment for the consensus binding sites of Sox2, Six1 and Gata3—transcription factors required for prosensory development—in the open chromatin regions. Over 2,200 regions displayed differential accessibility with developmental time in Sox2-EGFP+ cells, with most changes in the E12-14.5 window. Open chromatin regions detected in Sox2-EGFP+ cells map to over 48,000 orthologous regions in the human genome that include regions in genes linked to deafness. Our results reveal a dynamic landscape of open chromatin in prosensory cells with potential implications for cochlear development and disease.
Collapse
|
34
|
Prajapati-DiNubila M, Benito-Gonzalez A, Golden EJ, Zhang S, Doetzlhofer A. A counter gradient of Activin A and follistatin instructs the timing of hair cell differentiation in the murine cochlea. eLife 2019; 8:47613. [PMID: 31187730 PMCID: PMC6561706 DOI: 10.7554/elife.47613] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 05/27/2019] [Indexed: 12/14/2022] Open
Abstract
The mammalian auditory sensory epithelium has one of the most stereotyped cellular patterns known in vertebrates. Mechano-sensory hair cells are arranged in precise rows, with one row of inner and three rows of outer hair cells spanning the length of the spiral-shaped sensory epithelium. Aiding such precise cellular patterning, differentiation of the auditory sensory epithelium is precisely timed and follows a steep longitudinal gradient. The molecular signals that promote auditory sensory differentiation and instruct its graded pattern are largely unknown. Here, we identify Activin A and its antagonist follistatin as key regulators of hair cell differentiation and show, using mouse genetic approaches, that a local gradient of Activin A signaling within the auditory sensory epithelium times the longitudinal gradient of hair cell differentiation. Furthermore, we provide evidence that Activin-type signaling regulates a radial gradient of terminal mitosis within the auditory sensory epithelium, which constitutes a novel mechanism for limiting the number of inner hair cells being produced.
Collapse
Affiliation(s)
- Meenakshi Prajapati-DiNubila
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States.,Center for Sensory Biology, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Ana Benito-Gonzalez
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States.,Center for Sensory Biology, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Erin Jennifer Golden
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States.,Center for Sensory Biology, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Shuran Zhang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States.,Center for Sensory Biology, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Angelika Doetzlhofer
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States.,Center for Sensory Biology, Johns Hopkins University School of Medicine, Baltimore, United States
| |
Collapse
|
35
|
Hou K, Jiang H, Karim MR, Zhong C, Xu Z, Liu L, Guan M, Shao J, Huang X. A Critical E-box in Barhl1 3' Enhancer Is Essential for Auditory Hair Cell Differentiation. Cells 2019; 8:cells8050458. [PMID: 31096644 PMCID: PMC6562609 DOI: 10.3390/cells8050458] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/06/2019] [Accepted: 05/07/2019] [Indexed: 02/05/2023] Open
Abstract
Barhl1, a mouse homologous gene of Drosophila BarH class homeobox genes, is highly expressed within the inner ear and crucial for the long-term maintenance of auditory hair cells that mediate hearing and balance, yet little is known about the molecular events underlying Barhl1 regulation and function in hair cells. In this study, through data mining and in vitro report assay, we firstly identified Barhl1 as a direct target gene of Atoh1 and one E-box (E3) in Barhl1 3’ enhancer is crucial for Atoh1-mediated Barhl1 activation. Then we generated a mouse embryonic stem cell (mESC) line carrying disruptions on this E3 site E-box (CAGCTG) using CRISPR/Cas9 technology and this E3 mutated mESC line is further subjected to an efficient stepwise hair cell differentiation strategy in vitro. Disruptions on this E3 site caused dramatic loss of Barhl1 expression and significantly reduced the number of induced hair cell-like cells, while no affections on the differentiation toward early primitive ectoderm-like cells and otic progenitors. Finally, through RNA-seq profiling and gene ontology (GO) enrichment analysis, we found that this E3 box was indispensable for Barhl1 expression to maintain hair cell development and normal functions. We also compared the transcriptional profiles of induced cells from CDS mutated and E3 mutated mESCs, respectively, and got very consistent results except the Barhl1 transcript itself. These observations indicated that Atoh1-mediated Barhl1 expression could have important roles during auditory hair cell development. In brief, our findings delineate the detail molecular mechanism of Barhl1 expression regulation in auditory hair cell differentiation.
Collapse
Affiliation(s)
- Kun Hou
- Institute of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Hui Jiang
- Institute of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Md Rezaul Karim
- Institute of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Islamic University, Kushtia 7003, Bangladesh.
| | - Chao Zhong
- Institute of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Zhouwen Xu
- Institute of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Lin Liu
- Institute of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Minxin Guan
- Institute of Genetics, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - Jianzhong Shao
- Institute of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
- Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou 310058, China.
| | - Xiao Huang
- Institute of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
- Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou 310058, China.
| |
Collapse
|
36
|
Zhong C, Fu Y, Pan W, Yu J, Wang J. Atoh1 and other related key regulators in the development of auditory sensory epithelium in the mammalian inner ear: function and interplay. Dev Biol 2019; 446:133-141. [DOI: 10.1016/j.ydbio.2018.12.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/30/2018] [Accepted: 12/30/2018] [Indexed: 01/08/2023]
|
37
|
Lipovsek M, Wingate RJ. Conserved and divergent development of brainstem vestibular and auditory nuclei. eLife 2018; 7:40232. [PMID: 30566077 PMCID: PMC6317910 DOI: 10.7554/elife.40232] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 12/18/2018] [Indexed: 12/20/2022] Open
Abstract
Vestibular function was established early in vertebrates and has remained, for the most part, unchanged. In contrast, each group of tetrapods underwent independent evolutionary processes to solve the problem of hearing on land, resulting in a remarkable mixture of conserved, divergent and convergent features that define extant auditory systems. The vestibuloacoustic nuclei of the hindbrain develop from a highly conserved ground plan and provide an ideal framework on which to address the participation of developmental processes to the evolution of neuronal circuits. We employed an electroporation strategy to unravel the contribution of two dorsoventral and four axial lineages to the development of the chick hindbrain vestibular and auditory nuclei. We compare the chick developmental map with recently established genetic fate-maps of the developing mouse hindbrain. Overall, we find considerable conservation of developmental origin for the vestibular nuclei. In contrast, a comparative analysis of the developmental origin of hindbrain auditory structures echoes the complex evolutionary history of the auditory system. In particular, we find that the developmental origin of the chick auditory interaural time difference circuit supports its emergence from an ancient vestibular network, unrelated to the analogous mammalian counterpart.
Collapse
Affiliation(s)
- Marcela Lipovsek
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Richard Jt Wingate
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
38
|
Zeng X, Kirkpatrick R, Hofmann G, Grillot D, Linhart V, Viviani F, Marino J, Boyer J, Graham TL, Lu Q, Wu Z, Benowitz A, Cousins R. Screen for modulators of atonal homolog 1 gene expression using notch pathway-relevant gene transcription based cellular assays. PLoS One 2018; 13:e0207140. [PMID: 30540745 PMCID: PMC6291236 DOI: 10.1371/journal.pone.0207140] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 10/25/2018] [Indexed: 12/11/2022] Open
Abstract
Atonal homolog 1 (Atoh1) is a basic helix-loop-helix 9 (bHLH) transcription factor acting downstream of Notch and is required for the differentiation of sensory hair cells in the inner ear and the specification of secretory cells during the intestinal crypt cell regeneration. Motivated by the observations that the upregulation of Atoh1 gene expression, through genetic manipulation or pharmacological inhibition of Notch signaling (e.g. γ-secretase inhibitors, GSIs), induces ectopic hair cell growth in the cochlea of the inner ear and partially restores hearing after injuries in experimental models, we decided to identify small molecule modulators of the Notch-Atoh1 pathway, which could potentially regenerate hair cells. However, the lack of cellular models of the inner ear has precluded the screening and characterization of such modulators. Here we report using a colon cancer cell line LS-174T, which displays Notch inhibition-dependent Atoh1 expression as a surrogate cellular model to screen for inducers of Atoh1 expression. We designed an Atoh1 promoter-driven luciferase assay to screen a target-annotated library of ~6000 compounds. We further developed a medium throughput, real-time quantitative RT-PCR assay measuring the endogenous Atoh1 gene expression to confirm the hits and eliminate false positives from the reporter-based screen. This strategy allowed us to successfully recover GSIs of known chemotypes. This LS-174T cell-based assay directly measures Atoh1 gene expression induced through Notch-Hes1 inhibition, and therefore offers an opportunity to identify novel cellular modulators along the Notch-Atoh1 pathway.
Collapse
Affiliation(s)
- Xin Zeng
- R&D Target Sciences, GlaxoSmithKline, Upper Providence, Collegeville, United States of America
- * E-mail: (XZ); (RC)
| | - Robert Kirkpatrick
- R&D Alternative Discovery and Development, GlaxoSmithKline, Upper Providence, Collegeville, United States of America
| | - Glenn Hofmann
- R&D Platform Technology Sciences, Drug Design and Selection, GlaxoSmithKline, Upper Providence, Collegeville, United States of America
| | - Didier Grillot
- R&D Flexible Discovery Unit, Villebon-sur-Yvette, Paris, France
| | - Valerie Linhart
- R&D Flexible Discovery Unit, Villebon-sur-Yvette, Paris, France
| | - Fabrice Viviani
- R&D Flexible Discovery Unit, Villebon-sur-Yvette, Paris, France
| | - Joseph Marino
- R&D Alternative Discovery and Development, GlaxoSmithKline, Upper Providence, Collegeville, United States of America
| | - Joseph Boyer
- R&D Statistical sciences, GlaxoSmithKline, Upper Providence, Collegeville, United States of America
| | - Taylor L. Graham
- R&D Target Sciences, GlaxoSmithKline, Upper Providence, Collegeville, United States of America
| | - Quinn Lu
- R&D Target Sciences, GlaxoSmithKline, Upper Providence, Collegeville, United States of America
| | - Zining Wu
- R&D Platform Technology Sciences, Drug Design and Selection, GlaxoSmithKline, Upper Providence, Collegeville, United States of America
| | - Andrew Benowitz
- R&D Alternative Discovery and Development, GlaxoSmithKline, Upper Providence, Collegeville, United States of America
| | - Rick Cousins
- R&D Alternative Discovery and Development, GlaxoSmithKline, Stevenage, Hertfordshire, United Kingdom
- * E-mail: (XZ); (RC)
| |
Collapse
|
39
|
High-resolution transcriptional dissection of in vivo Atoh1-mediated hair cell conversion in mature cochleae identifies Isl1 as a co-reprogramming factor. PLoS Genet 2018; 14:e1007552. [PMID: 30063705 PMCID: PMC6086484 DOI: 10.1371/journal.pgen.1007552] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 08/10/2018] [Accepted: 07/10/2018] [Indexed: 12/24/2022] Open
Abstract
In vivo direct conversion of differentiated cells holds promise for regenerative medicine; however, improving the conversion efficiency and producing functional target cells remain challenging. Ectopic Atoh1 expression in non-sensory supporting cells (SCs) in mouse cochleae induces their partial conversion to hair cells (HCs) at low efficiency. Here, we performed single-cell RNA sequencing of whole mouse sensory epithelia harvested at multiple time points after conditional overexpression of Atoh1. Pseudotemporal ordering revealed that converted HCs (cHCs) are present along a conversion continuum that correlates with both endogenous and exogenous Atoh1 expression. Bulk sequencing of isolated cell populations and single-cell qPCR confirmed 51 transcription factors, including Isl1, are differentially expressed among cHCs, SCs and HCs. In transgenic mice, co-overexpression of Atoh1 and Isl1 enhanced the HC conversion efficiency. Together, our study shows how high-resolution transcriptional profiling of direct cell conversion can identify co-reprogramming factors required for efficient conversion. The ongoing ATOH1 gene therapy clinical trial offers promise for hearing restoration in humans. However, in animal models, Atoh1-mediated sensory regeneration is inefficient and incomplete. Here we performed high-resolution gene expression profiling of single cochlear cells at multiple time points in a mouse model whereby we discovered a continuous regeneration process that leads to the formation of immature sensory cells. We identified 51 key reprogramming transcription factors that may increase the efficiency and completion of the regeneration process and confirmed that Isl1 in transgenic mice promotes Atoh1-mediated sensory regeneration as a co-reprogramming factor. Our studies identify molecular mechanisms and novel co-reprogramming factors for sensory restoration in humans with irreversible hearing loss.
Collapse
|
40
|
Baker NE, Brown NL. All in the family: proneural bHLH genes and neuronal diversity. Development 2018; 145:145/9/dev159426. [PMID: 29720483 DOI: 10.1242/dev.159426] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Proneural basic Helix-Loop-Helix (bHLH) proteins are required for neuronal determination and the differentiation of most neural precursor cells. These transcription factors are expressed in vastly divergent organisms, ranging from sponges to primates. Here, we review proneural bHLH gene evolution and function in the Drosophila and vertebrate nervous systems, arguing that the Drosophila gene atonal provides a useful platform for understanding proneural gene structure and regulation. We also discuss how functional equivalency experiments using distinct proneural genes can reveal how proneural gene duplication and divergence are interwoven with neuronal complexity.
Collapse
Affiliation(s)
- Nicholas E Baker
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461 USA
| | - Nadean L Brown
- Department of Cell Biology and Human Anatomy, University of California, One Shields Avenue, Davis, CA 95616 USA
| |
Collapse
|
41
|
Ni W, Zeng S, Li W, Chen Y, Zhang S, Tang M, Sun S, Chai R, Li H. Wnt activation followed by Notch inhibition promotes mitotic hair cell regeneration in the postnatal mouse cochlea. Oncotarget 2018; 7:66754-66768. [PMID: 27564256 PMCID: PMC5341835 DOI: 10.18632/oncotarget.11479] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 06/29/2016] [Indexed: 12/27/2022] Open
Abstract
Hair cell (HC) loss is the main cause of permanent hearing loss in mammals. Previous studies have reported that in neonatal mice cochleae, Wnt activation promotes supporting cell (SC) proliferation and Notch inhibition promotes the trans-differentiation of SCs into HCs. However, Wnt activation alone fails to regenerate significant amounts of new HCs, Notch inhibition alone regenerates the HCs at the cost of exhausting the SC population, which leads to the death of the newly regenerated HCs. Mitotic HC regeneration might preserve the SC number while regenerating the HCs, which could be a better approach for long-term HC regeneration. We present a two-step gene manipulation, Wnt activation followed by Notch inhibition, to accomplish mitotic regeneration of HCs while partially preserving the SC number. We show that Wnt activation followed by Notch inhibition strongly promotes the mitotic regeneration of new HCs in both normal and neomycin-damaged cochleae while partially preserving the SC number. Lineage tracing shows that the majority of the mitotically regenerated HCs are derived specifically from the Lgr5+ progenitors with or without HC damage. Our findings suggest that the co-regulation of Wnt and Notch signaling might provide a better approach to mitotically regenerate HCs from Lgr5+ progenitor cells.
Collapse
Affiliation(s)
- Wenli Ni
- Otorhinolaryngology Department of The Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China
| | - Shan Zeng
- Otorhinolaryngology Department of The Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China
| | - Wenyan Li
- Otorhinolaryngology Department of The Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China
| | - Yan Chen
- Otorhinolaryngology Department of The Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Central Laboratory, Affiliated Eye and ENT Hospital of Fudan University, Shanghai, PR China.,Key Laboratory of Hearing Medicine of The National Health and Family Planning Commission, Shanghai, PR China
| | - Shasha Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, PR China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Mingliang Tang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, PR China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Shan Sun
- Otorhinolaryngology Department of The Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Central Laboratory, Affiliated Eye and ENT Hospital of Fudan University, Shanghai, PR China.,Key Laboratory of Hearing Medicine of The National Health and Family Planning Commission, Shanghai, PR China
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, PR China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Huawei Li
- Otorhinolaryngology Department of The Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, PR China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, PR China.,Central Laboratory, Affiliated Eye and ENT Hospital of Fudan University, Shanghai, PR China
| |
Collapse
|
42
|
Hox2 Genes Are Required for Tonotopic Map Precision and Sound Discrimination in the Mouse Auditory Brainstem. Cell Rep 2017; 18:185-197. [PMID: 28052248 DOI: 10.1016/j.celrep.2016.12.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 11/03/2016] [Accepted: 12/06/2016] [Indexed: 11/22/2022] Open
Abstract
Tonotopy is a hallmark of auditory pathways and provides the basis for sound discrimination. Little is known about the involvement of transcription factors in brainstem cochlear neurons orchestrating the tonotopic precision of pre-synaptic input. We found that in the absence of Hoxa2 and Hoxb2 function in Atoh1-derived glutamatergic bushy cells of the anterior ventral cochlear nucleus, broad input topography and sound transmission were largely preserved. However, fine-scale synaptic refinement and sharpening of isofrequency bands of cochlear neuron activation upon pure tone stimulation were impaired in Hox2 mutants, resulting in defective sound-frequency discrimination in behavioral tests. These results establish a role for Hox factors in tonotopic refinement of connectivity and in ensuring the precision of sound transmission in the mammalian auditory circuit.
Collapse
|
43
|
Neumann JE, Swartling FJ, Schüller U. Medulloblastoma: experimental models and reality. Acta Neuropathol 2017; 134:679-689. [PMID: 28725965 DOI: 10.1007/s00401-017-1753-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 06/27/2017] [Accepted: 07/16/2017] [Indexed: 12/11/2022]
Abstract
Medulloblastoma is the most frequent malignant brain tumor in childhood, but it may also affect infants, adolescents, and young adults. Recent advances in the understanding of the disease have shed light on molecular and clinical heterogeneity, which is now reflected in the updated WHO classification of brain tumors. At the same time, it is well accepted that preclinical research and clinical trials have to be subgroup-specific. Hence, valid models have to be generated specifically for every medulloblastoma subgroup to properly mimic molecular fingerprints, clinical features, and responsiveness to targeted therapies. This review summarizes the availability of experimental medulloblastoma models with a particular focus on how well these models reflect the actual disease subgroup. We further describe technical advantages and disadvantages of the models and finally point out how some models have successfully been used to introduce new drugs and why some medulloblastoma subgroups are extraordinary difficult to model.
Collapse
Affiliation(s)
- Julia E Neumann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fredrik J Swartling
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Ulrich Schüller
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Research Institute Children's Cancer Center, Martinistrasse 52, 20251, Hamburg, Germany.
| |
Collapse
|
44
|
Gálvez H, Tena JJ, Giraldez F, Abelló G. The Repression of Atoh1 by Neurogenin1 during Inner Ear Development. Front Mol Neurosci 2017; 10:321. [PMID: 29104531 PMCID: PMC5655970 DOI: 10.3389/fnmol.2017.00321] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 09/21/2017] [Indexed: 01/01/2023] Open
Abstract
Atonal homolog 1 (Atoh1) and Neurogenin1 (Neurog1) are basic Helix-Loop-Helix (bHLH) transcription factors crucial for the generation of hair cells (HCs) and neurons in the inner ear. Both genes are induced early in development, but the expression of Atoh1 is counteracted by Neurog1. As a result, HC development is prevented during neurogenesis. This work aimed at understanding the molecular basis of this interaction. Atoh1 regulation depends on a 3'Atoh1-enhancer that is the site for Atoh1 autoregulation. Reporter assays on chick embryos and P19 cells show that Neurog1 hampers the autoactivation of Atoh1, the effect being cell autonomous and independent on Notch activity. Assay for Transposase-Accessible Chromatin with high throughput sequencing (ATAC-Seq) analysis shows that the region B of the 3'Atoh1-enhancer is accessible during development and sufficient for both activation and repression. Neurog1 requires the regions flanking the class A E-box to show its repressor effect, however, it does not require binding to DNA for Atoh1 repression. This depends on the dimerization domains Helix-1 and Helix-2 and the reduction of Atoh1 protein levels. The results point towards the acceleration of Atoh1 mRNA degradation as the potential mechanism for the reduction of Atoh1 levels. Such a mechanism dissociates the prevention of Atoh1 expression in neurosensory progenitors from the unfolding of the neurogenic program.
Collapse
Affiliation(s)
- Héctor Gálvez
- DCEXS, Universitat Pompeu Fabra (UPF) - Parc de Recerca Biomèdica de Barcelona (PRBB), Barcelona, Spain
| | - Juan J Tena
- Centro Andaluz de Biología del Desarrollo, Consejo Superior de Investigaciones Científicas (CSIC), Sevilla, Spain
| | - Fernando Giraldez
- DCEXS, Universitat Pompeu Fabra (UPF) - Parc de Recerca Biomèdica de Barcelona (PRBB), Barcelona, Spain
| | - Gina Abelló
- DCEXS, Universitat Pompeu Fabra (UPF) - Parc de Recerca Biomèdica de Barcelona (PRBB), Barcelona, Spain
| |
Collapse
|
45
|
Fritzsch B, Elliott KL. Gene, cell, and organ multiplication drives inner ear evolution. Dev Biol 2017; 431:3-15. [PMID: 28866362 DOI: 10.1016/j.ydbio.2017.08.034] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 04/27/2017] [Accepted: 08/25/2017] [Indexed: 12/14/2022]
Abstract
We review the development and evolution of the ear neurosensory cells, the aggregation of neurosensory cells into an otic placode, the evolution of novel neurosensory structures dedicated to hearing and the evolution of novel nuclei in the brain and their input dedicated to processing those novel auditory stimuli. The evolution of the apparently novel auditory system lies in duplication and diversification of cell fate transcription regulation that allows variation at the cellular level [transforming a single neurosensory cell into a sensory cell connected to its targets by a sensory neuron as well as diversifying hair cells], organ level [duplication of organ development followed by diversification and novel stimulus acquisition] and brain nuclear level [multiplication of transcription factors to regulate various neuron and neuron aggregate fate to transform the spinal cord into the unique hindbrain organization]. Tying cell fate changes driven by bHLH and other transcription factors into cell and organ changes is at the moment tentative as not all relevant factors are known and their gene regulatory network is only rudimentary understood. Future research can use the blueprint proposed here to provide both the deeper molecular evolutionary understanding as well as a more detailed appreciation of developmental networks. This understanding can reveal how an auditory system evolved through transformation of existing cell fate determining networks and thus how neurosensory evolution occurred through molecular changes affecting cell fate decision processes. Appreciating the evolutionary cascade of developmental program changes could allow identifying essential steps needed to restore cells and organs in the future.
Collapse
Affiliation(s)
- Bernd Fritzsch
- University of Iowa, Department of Biology, Iowa City, IA 52242, United States.
| | - Karen L Elliott
- University of Iowa, Department of Biology, Iowa City, IA 52242, United States
| |
Collapse
|
46
|
Tech K, Tikunov AP, Farooq H, Morrissy AS, Meidinger J, Fish T, Green SC, Liu H, Li Y, Mungall AJ, Moore RA, Ma Y, Jones SJM, Marra MA, Vander Heiden MG, Taylor MD, Macdonald JM, Gershon TR. Pyruvate Kinase Inhibits Proliferation during Postnatal Cerebellar Neurogenesis and Suppresses Medulloblastoma Formation. Cancer Res 2017; 77:3217-3230. [PMID: 28515149 DOI: 10.1158/0008-5472.can-16-3304] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/20/2017] [Accepted: 04/18/2017] [Indexed: 12/11/2022]
Abstract
Aerobic glycolysis supports proliferation through unresolved mechanisms. We have previously shown that aerobic glycolysis is required for the regulated proliferation of cerebellar granule neuron progenitors (CGNP) and for the growth of CGNP-derived medulloblastoma. Blocking the initiation of glycolysis via deletion of hexokinase-2 (Hk2) disrupts CGNP proliferation and restricts medulloblastoma growth. Here, we assessed whether disrupting pyruvate kinase-M (Pkm), an enzyme that acts in the terminal steps of glycolysis, would alter CGNP metabolism, proliferation, and tumorigenesis. We observed a dichotomous pattern of PKM expression, in which postmitotic neurons throughout the brain expressed the constitutively active PKM1 isoform, while neural progenitors and medulloblastomas exclusively expressed the less active PKM2. Isoform-specific Pkm2 deletion in CGNPs blocked all Pkm expression. Pkm2-deleted CGNPs showed reduced lactate production and increased SHH-driven proliferation. 13C-flux analysis showed that Pkm2 deletion reduced the flow of glucose carbons into lactate and glutamate without markedly increasing glucose-to-ribose flux. Pkm2 deletion accelerated tumor formation in medulloblastoma-prone ND2:SmoA1 mice, indicating the disrupting PKM releases CGNPs from a tumor-suppressive effect. These findings show that distal and proximal disruptions of glycolysis have opposite effects on proliferation, and that efforts to block the oncogenic effect of aerobic glycolysis must target reactions upstream of PKM. Cancer Res; 77(12); 3217-30. ©2017 AACR.
Collapse
Affiliation(s)
- Katherine Tech
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina.,Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Andrey P Tikunov
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina
| | - Hamza Farooq
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - A Sorana Morrissy
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jessica Meidinger
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Taylor Fish
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Sarah C Green
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina
| | - Hedi Liu
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Yisu Li
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Andrew J Mungall
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Richard A Moore
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Yussanne Ma
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Steven J M Jones
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Marco A Marra
- Michael Smith Genome Sciences Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Matthew G Vander Heiden
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Michael D Taylor
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada.,The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Division of Neurosurgery, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jeffrey M Macdonald
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina
| | - Timothy R Gershon
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, North Carolina. .,Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina.,UNC Neuroscience Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
47
|
Verhaegen ME, Mangelberger D, Harms PW, Eberl M, Wilbert DM, Meireles J, Bichakjian CK, Saunders TL, Wong SY, Dlugosz AA. Merkel Cell Polyomavirus Small T Antigen Initiates Merkel Cell Carcinoma-like Tumor Development in Mice. Cancer Res 2017; 77:3151-3157. [PMID: 28512245 DOI: 10.1158/0008-5472.can-17-0035] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 01/17/2017] [Accepted: 04/19/2017] [Indexed: 11/16/2022]
Abstract
Merkel cell carcinoma (MCC) tumor cells express several markers detected in normal Merkel cells, a nonproliferative population of neuroendocrine cells that arise from epidermis. MCCs frequently contain Merkel cell polyomavirus (MCPyV) DNA and express viral transforming antigens, sT and tLT, but the role of these putative oncogenes in MCC development, and this tumor's cell of origin, are unknown. Using a panel of preterm transgenic mice, we show that epidermis-targeted coexpression of sT and the cell fate-determinant atonal bHLH transcription factor 1 (ATOH1) leads to development of widespread cellular aggregates, with histology and marker expression mimicking that of human intraepidermal MCC. The MCC-like tumor phenotype was dependent on the FBXW7-binding domain of sT, but not the sT-PP2A binding domain. Coexpression of MCPyV tLT did not appreciably alter the phenotype driven by either sT or sT combined with ATOH1. MCPyV sT, when coexpressed with ATOH1, is thus sufficient to initiate development of epidermis-derived MCC-like tumors in mice. Cancer Res; 77(12); 3151-7. ©2017 AACR.
Collapse
Affiliation(s)
| | | | - Paul W Harms
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Markus Eberl
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan
| | - Dawn M Wilbert
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan
| | - Julia Meireles
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan
| | | | - Thomas L Saunders
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Sunny Y Wong
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Andrzej A Dlugosz
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan.
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
48
|
Gálvez H, Abelló G, Giraldez F. Signaling and Transcription Factors during Inner Ear Development: The Generation of Hair Cells and Otic Neurons. Front Cell Dev Biol 2017; 5:21. [PMID: 28393066 PMCID: PMC5364141 DOI: 10.3389/fcell.2017.00021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 03/02/2017] [Indexed: 12/21/2022] Open
Abstract
Integration between cell signals and bHLH transcription factors plays a prominent role during the development of hair cells of the inner ear. Hair cells are the sensory receptors of the inner ear, responsible for the mechano-transduction of sound waves into electrical signals. They derive from multipotent progenitors that reside in the otic placode. Progenitor commitment is the result of cell signaling from the surrounding tissues that result in the restricted expression of SoxB1 transcription factors, Sox2 and Sox3. In turn, they induce the expression of Neurog1 and Atoh1, two bHLH factors that specify neuronal and hair cell fates, respectively. Neuronal and hair cell development, however, do not occur simultaneously. Hair cell development is prevented during neurogenesis and prosensory stages, resulting in the delay of hair cell development with respect to neuron production. Negative interactions between Neurog1 and Atoh1, and of Atoh1 with other bHLH factors driven by Notch signaling, like Hey1 and Hes5, account for this delay. In summary, the regulation of Atoh1 and hair cell development relies on interactions between cell signaling and bHLH transcription factors that dictate cell fate and timing decisions during development. Interestingly, these mechanisms operate as well during hair cell regeneration after damage and during stem cell directed differentiation, making developmental studies instrumental for improving therapies for hearing impairment.
Collapse
Affiliation(s)
- Héctor Gálvez
- Developmental Biology, CEXS, Parc de Recerca Biomèdica de Barcelona, Universitat Pompeu Fabra Barcelona, Spain
| | - Gina Abelló
- Developmental Biology, CEXS, Parc de Recerca Biomèdica de Barcelona, Universitat Pompeu Fabra Barcelona, Spain
| | - Fernando Giraldez
- Developmental Biology, CEXS, Parc de Recerca Biomèdica de Barcelona, Universitat Pompeu Fabra Barcelona, Spain
| |
Collapse
|
49
|
Yang X, Qian X, Ma R, Wang X, Yang J, Luo W, Chen P, Chi F, Ren D. Establishment of planar cell polarity is coupled to regional cell cycle exit and cell differentiation in the mouse utricle. Sci Rep 2017; 7:43021. [PMID: 28230212 PMCID: PMC5322371 DOI: 10.1038/srep43021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 01/18/2017] [Indexed: 01/16/2023] Open
Abstract
Sensory hair cells are coordinately oriented within each inner ear sensory organ to exhibit a particular form of planar cell polarity (PCP) necessary for mechanotransduction. However, the developmental events associated with establishing PCP in the vestibule are unclear, hindering data interpretation and employment of the vestibule for PCP studies. Herein, we investigated PCP of the mouse vestibular organs. We further characterised cell cycle exit, cell differentiation, and PCP establishment in the utricle. We found that hair cells formed first in the striolar and medial extrastriolar (MES) regions of the utricle at embryonic day 11.5 (E11.5), while cells in the lateral extrastriolar region (LES) mostly formed at E13.5. Cell differentiation was initiated in the striolar region, which expanded first toward the MES, then to the LES by E15.5. The polarity of hair cells was established at birth along a putative line of polarity reversal (LPR), lateral to the striolar region. Core PCP protein Vangl2 emerged in the cell boundaries since E11.5, while cell intrinsic polarity protein Gαi3 appeared at E12.5, then polarized to the bare zone of individual hair cell at E13.5. These findings provide a blueprint of the developmental events associated with establishing PCP in the utricle.
Collapse
Affiliation(s)
- Xiaoyu Yang
- Department of Otology and Skull Base Surgery, Eye &ENT Hospital of Fudan University, Shanghai 200031, China.,Shanghai Clinical Medical Center of Hearing Medicine, Eye &ENT Hospital of Fudan University, Shanghai 200031, China
| | - Xiaoqing Qian
- Department of Otology and Skull Base Surgery, Eye &ENT Hospital of Fudan University, Shanghai 200031, China.,Shanghai Clinical Medical Center of Hearing Medicine, Eye &ENT Hospital of Fudan University, Shanghai 200031, China
| | - Rui Ma
- Department of Otology and Skull Base Surgery, Eye &ENT Hospital of Fudan University, Shanghai 200031, China.,Shanghai Clinical Medical Center of Hearing Medicine, Eye &ENT Hospital of Fudan University, Shanghai 200031, China.,Department of Research Center, Eye &ENT Hospital of Fudan University, Shanghai 200031, China
| | - Xinwei Wang
- Department of Otology and Skull Base Surgery, Eye &ENT Hospital of Fudan University, Shanghai 200031, China.,Shanghai Clinical Medical Center of Hearing Medicine, Eye &ENT Hospital of Fudan University, Shanghai 200031, China
| | - Juanmei Yang
- Department of Otology and Skull Base Surgery, Eye &ENT Hospital of Fudan University, Shanghai 200031, China.,Shanghai Clinical Medical Center of Hearing Medicine, Eye &ENT Hospital of Fudan University, Shanghai 200031, China
| | - Wenwei Luo
- Department of Otology and Skull Base Surgery, Eye &ENT Hospital of Fudan University, Shanghai 200031, China.,Shanghai Clinical Medical Center of Hearing Medicine, Eye &ENT Hospital of Fudan University, Shanghai 200031, China
| | - Ping Chen
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA
| | - Fanglu Chi
- Department of Otology and Skull Base Surgery, Eye &ENT Hospital of Fudan University, Shanghai 200031, China.,Shanghai Clinical Medical Center of Hearing Medicine, Eye &ENT Hospital of Fudan University, Shanghai 200031, China
| | - Dongdong Ren
- Department of Otology and Skull Base Surgery, Eye &ENT Hospital of Fudan University, Shanghai 200031, China.,Shanghai Clinical Medical Center of Hearing Medicine, Eye &ENT Hospital of Fudan University, Shanghai 200031, China
| |
Collapse
|
50
|
Nie J, Hashino E. Organoid technologies meet genome engineering. EMBO Rep 2017; 18:367-376. [PMID: 28202491 DOI: 10.15252/embr.201643732] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/13/2017] [Accepted: 01/18/2017] [Indexed: 01/08/2023] Open
Abstract
Three-dimensional (3D) stem cell differentiation cultures recently emerged as a novel model system for investigating human embryonic development and disease progression in vitro, complementing existing animal and two-dimensional (2D) cell culture models. Organoids, the 3D self-organizing structures derived from pluripotent or somatic stem cells, can recapitulate many aspects of structural organization and functionality of their in vivo organ counterparts, thus holding great promise for biomedical research and translational applications. Importantly, faithful recapitulation of disease and development processes relies on the ability to modify the genomic contents in organoid cells. The revolutionary genome engineering technologies, CRISPR/Cas9 in particular, enable investigators to generate various reporter cell lines for prompt validation of specific cell lineages as well as to introduce disease-associated mutations for disease modeling. In this review, we provide historical overviews, and discuss technical considerations, and potential future applications of genome engineering in 3D organoid models.
Collapse
Affiliation(s)
- Jing Nie
- Department of Otolaryngology - Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Eri Hashino
- Department of Otolaryngology - Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, USA .,Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|