Chonnam Med J.  2017 Jan;53(1):37-46. 10.4068/cmj.2017.53.1.37.

Atoh1 as a Coordinator of Sensory Hair Cell Development and Regeneration in the Cochlea

Affiliations
  • 1Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Korea. victocho@hanmail.net
  • 2Research Institute of Medical Sciences, Chonnam National University, Gwangju, Korea.
  • 3Department of Physiology, Chonnam National University Medical School, Gwangju, Korea.

Abstract

Cochlear sensory hair cells (HCs) are crucial for hearing as mechanoreceptors of the auditory systems. Clarification of transcriptional regulation for the cochlear sensory HC development is crucial for the improvement of cell replacement therapies for hearing loss. Transcription factor Atoh1 is the key player during HC development and regeneration. In this review, we will focus on Atoh1 and its related signaling pathways (Notch, fibroblast growth factor, and Wnt/β-catenin signaling) involved in the development of cochlear sensory HCs. We will also discuss the potential applicability of these signals for the induction of HC regeneration.

Keyword

Hair Cells, Auditory; Regeneration; Transcription Factors; Hearing Loss

MeSH Terms

Cochlea*
Fibroblast Growth Factors
Hair Cells, Auditory
Hair*
Hearing
Hearing Loss
Mechanoreceptors
Regeneration*
Transcription Factors
Fibroblast Growth Factors
Transcription Factors

Figure

  • FIG. 1 Sensory hair cells in organ of Corti. Inner hair cells are arranged in one row on the medial side of the organ of Corti. Outer hair cells are arranged in three rows on the lateral side. Left side figure is scanning electron microscopy image and right side figure is immunofluorescent image. IHC: inner hair cell, OHC: outer hair cell.

  • FIG. 2 Atoh1 gene expression during inner ear development. Atoh1 is expressed in the proneuronal cells at E14.5 (A, B) and expressed in sensory hair cells in the cochlea at E16.5 and P1 (C-F). In adult mammals, hair cells are located in the organ of Corti (G, H&E staining) and have stereocillia (H, scanning electro-microscopic image). OHC: outer hair cell, IHC: inner hair cell.

  • FIG. 3 Notch signaling during mammalian hair cell development. (A) There are 5 notch ligands (Dll1, Dl12, Dll3, jag 1, jag2) in hair cell and 4 notch receptors in the supporting cells. γ-secretase is an internal protease that cleaves the NICD. (B) Notch signaling is activated upon cell-to-cell contact. Downstream genes (Hes1, 5, Hey, other genes) are produced by Notch signaling. These produced inhibitory basic helix-loop-helix proteins (HES1, HES5) blocking the effects of prosenosry genes (Atoh1) that leads to inhibition of the hair cell fate. These inhibited cells differentiate into supporting cells. Dll: delta-like ligand, NICD: notch intracellular domain, CoRs: co-repressor, CSL: CBF1, Su(H) and LAG-1,CoA: co-activator, MAML: mastermind ligand.

  • FIG. 4 Wnt/β-catenin signaling. Without Wnt ligands, GSK3β complex destructs β-catenin (β-cat) by phosphorylation. This signaling is activated when Wnt ligands bind to Fizzled receptors and their co-receptor lipoprotein receptor-related protein (LRP) 5/6. Subsequently, this lead to sequestration of Axin, recruitment of Disheveled (Dvl), and the release of β-cat. Frizzled receptor is stabilized by attachment of R-spondins (R-spo) to Lgr4/5/6 receptor. Released β-cat is translocated into the nucleus. Ant it will bind to T cell factor/lymphoid enhancer-binding factor (TCF/LEF) family to release Wnt target genes, such as Axin2 and Lgr5.

  • FIG. 5 The proposed Role of Wnt/β-catenin signaling in hair cell regeneration. Wnt/β-catenin signaling can increase hair cell regeneration by stimulating cell mitoic proliferation and up-regulating Atoh1 expression.

  • FIG. 6 Factors that direct sensory hair cell fate in the inner ear. Beginning at otic placode stages, FGF signaling regulates otic placode development as well as early stage of otocyst formation. A gradient of Wnt signaling determines the size of otic placode. Wnt pathway interacts with notch pathway. FGF signals are upstream activators of Atoh1 genes during the development of prosensory cells. Final sensory hair cell development is regulated by the expression of Atoh1. Supporting cell development is activated by the expression of notch pathway.


Reference

1. Richardson RT, Atkinson PJ. Atoh1 gene therapy in the cochlea for hair cell regeneration. Expert Opin Biol Ther. 2015; 15:417–430.
Article
2. Hongmiao R, Wei L, Bing H, Xiong DD, Jihao R. Atoh1: landscape for inner ear cell regeneration. Curr Gene Ther. 2014; 14:101–111.
Article
3. McFadden SL, Ding D, Jiang H, Salvi RJ. Time course of efferent fiber and spiral ganglion cell degeneration following complete hair cell loss in the chinchilla. Brain Res. 2004; 997:40–51.
Article
4. Groves AK. The challenge of hair cell regeneration. Exp Biol Med (Maywood). 2010; 235:434–446.
Article
5. Izumikawa M, Minoda R, Kawamoto K, Abrashkin KA, Swiderski DL, Dolan DF, et al. Auditory hair cell replacement and hearing improvement by Atoh1 gene therapy in deaf mammals. Nat Med. 2005; 11:271–276.
Article
6. Blamey P, Artieres F, Başkent D, Bergeron F, Beynon A, Burke E, et al. Factors affecting auditory performance of postlinguistically deaf adults using cochlear implants: an update with 2251 patients. Audiol Neurootol. 2013; 18:36–47.
Article
7. Blamey PJ, Dooley GJ, Parisi ES, Clark GM. Pitch comparisons of acoustically and electrically evoked auditory sensations. Hear Res. 1996; 99:139–150.
Article
8. Costa A, Sanchez-Guardado L, Juniat S, Gale JE, Daudet N, Henrique D. Generation of sensory hair cells by genetic programming with a combination of transcription factors. Development. 2015; 142:1948–1959.
Article
9. Savary E, Sabourin JC, Santo J, Hugnot JP, Chabbert C, Van De Water T, et al. Cochlear stem/progenitor cells from a postnatal cochlea respond to Jagged1 and demonstrate that notch signaling promotes sphere formation and sensory potential. Mech Dev. 2008; 125:674–686.
Article
10. Pickles JO. An Introduction to the Physiology of Hearing. London: Academic Press;1988.
11. Spoendlin H. Moller AR, editor. Basic Mechanisms in Haring. New York: Academic Press;1988.
12. Kelley MW. Regulation of cell fate in the sensory epithelia of the inner ear. Nat Rev Neurosci. 2006; 7:837–849.
Article
13. Dabdoub A, Puligilla C, Jones JM, Fritzsch B, Cheah KS, Pevny LH, et al. Sox2 signaling in prosensory domain specification and subsequent hair cell differentiation in the developing cochlea. Proc Natl Acad Sci U S A. 2008; 105:18396–18401.
Article
14. Zine A, de Ribaupierre F. Notch/Notch ligands and Math1 expression patterns in the organ of Corti of wild-type and Hes1 and Hes5 mutant mice. Hear Res. 2002; 170:22–31.
Article
15. Pauley S, Kopecky B, Beisel K, Soukup G, Fritzsch B. Stem cells and molecular strategies to restore hearing. Panminerva Med. 2008; 50:41–53.
16. Denoyelle F, Marlin S, Weil D, Moatti L, Chauvin P, Garabédian EN, et al. Clinical features of the prevalent form of childhood deafness, DFNB1, due to a connexin-26 gene defect: implications for genetic counselling. Lancet. 1999; 353:1298–1303.
Article
17. Kelsell DP, Dunlop J, Stevens HP, Lench NJ, Liang JN, Parry G, et al. Connexin 26 mutations in hereditary non-syndromic sensorineural deafness. Nature. 1997; 387:80–83.
Article
18. Chen P, Johnson JE, Zoghbi HY, Segil N. The role of Math1 in inner ear development: Uncoupling the establishment of the sensory primordium from hair cell fate determination. Development. 2002; 129:2495–2505.
Article
19. Lai HC, Klisch TJ, Roberts R, Zoghbi HY, Johnson JE. In vivo neuronal subtype-specific targets of Atoh1 (Math1) in dorsal spinal cord. J Neurosci. 2011; 31:10859–10871.
Article
20. Bermingham NA, Hassan BA, Price SD, Vollrath MA, Ben-Arie N, Eatock RA, et al. Math1: an essential gene for the generation of inner ear hair cells. Science. 1999; 284:1837–1841.
Article
21. Mulvaney J, Dabdoub A. Atoh1, an essential transcription factor in neurogenesis and intestinal and inner ear development: function, regulation, and context dependency. J Assoc Res Otolaryngol. 2012; 13:281–293.
Article
22. Stone JS, Choi YS, Woolley SM, Yamashita H, Rubel EW. Progenitor cell cycling during hair cell regeneration in the vestibular and auditory epithelia of the chick. J Neurocytol. 1999; 28:863–876.
23. Klisch TJ, Xi Y, Flora A, Wang L, Li W, Zoghbi HY. In vivo Atoh1 targetome reveals how a proneural transcription factor regulates cerebellar development. Proc Natl Acad Sci U S A. 2011; 108:3288–3293.
Article
24. Helms AW, Johnson JE. Progenitors of dorsal commissural interneurons are defined by MATH1 expression. Development. 1998; 125:919–928.
Article
25. Rose MF, Ahmad KA, Thaller C, Zoghbi HY. Excitatory neurons of the proprioceptive, interoceptive, and arousal hindbrain networks share a developmental requirement for Math1. Proc Natl Acad Sci U S A. 2009; 106:22462–22467.
Article
26. Gerbe F, van Es JH, Makrini L, Brulin B, Mellitzer G, Robine S, et al. Distinct ATOH1 and Neurog3 requirements define tuft cells as a new secretory cell type in the intestinal epithelium. J Cell Biol. 2011; 192:767–780.
Article
27. Yang Q, Bermingham NA, Finegold MJ, Zoghbi HY. Requirement of Math1 for secretory cell lineage commitment in the mouse intestine. Science. 2001; 294:2155–2158.
Article
28. Driver EC, Sillers L, Coate TM, Rose MF, Kelley MW. The Atoh1-lineage gives rise to hair cells and supporting cells within the mammalian cochlea. Dev Biol. 2013; 376:86–98.
Article
29. Lee YS, Liu F, Segil N. A morphogenetic wave of p27Kip1 transcription directs cell cycle exit during organ of Corti development. Development. 2006; 133:2817–2826.
Article
30. Ruben RJ. Development of the inner ear of the mouse: a radioautographic study of terminal mitoses. Acta Otolaryngol. 1967; Suppl 220. 1–44.
31. Stojanova ZP, Kwan T, Segil N. Epigenetic regulation of Atoh1 guides hair cell development in the mammalian cochlea. Development. 2015; 142:3529–3536.
Article
32. Cai T, Seymour ML, Zhang H, Pereira FA, Groves AK. Conditional deletion of Atoh1 reveals distinct critical periods for survival and function of hair cells in the organ of Corti. J Neurosci. 2013; 33:10110–10122.
Article
33. Chonko KT, Jahan I, Stone J, Wright MC, Fujiyama T, Hoshino M, et al. Atoh1 directs hair cell differentiation and survival in the late embryonic mouse inner ear. Dev Biol. 2013; 381:401–410.
Article
34. Gubbels SP, Woessner DW, Mitchell JC, Ricci AJ, Brigande JV. Functional auditory hair cells produced in the mammalian cochlea by in utero gene transfer. Nature. 2008; 455:537–541.
Article
35. Kelly MC, Chang Q, Pan A, Lin X, Chen P. Atoh1 directs the formation of sensory mosaics and induces cell proliferation in the postnatal mammalian cochlea in vivo. J Neurosci. 2012; 32:6699–6710.
Article
36. Liu Z, Dearman JA, Cox BC, Walters BJ, Zhang L, Ayrault O, et al. Age-dependent in vivo conversion of mouse cochlear pillar and Deiters’ cells to immature hair cells by Atoh1 ectopic expression. J Neurosci. 2012; 32:6600–6610.
Article
37. Liu Z, Fang J, Dearman J, Zhang L, Zuo J. In vivo generation of immature inner hair cells in neonatal mouse cochleae by ectopic Atoh1 expression. PLoS One. 2014; 9:e89377.
Article
38. Shou J, Zheng JL, Gao WQ. Robust generation of new hair cells in the mature mammalian inner ear by adenoviral expression of Hath1. Mol Cell Neurosci. 2003; 23:169–179.
Article
39. Yang J, Cong N, Han Z, Huang Y, Chi F. Ectopic hair cell-like cell induction by Math1 mainly involves direct transdifferentiation in neonatal mammalian cochlea. Neurosci Lett. 2013; 549:7–11.
Article
40. Zheng JL, Gao WQ. Overexpression of Math1 induces robust production of extra hair cells in postnatal rat inner ears. Nat Neurosci. 2000; 3:580–586.
Article
41. Kawamoto K, Ishimoto S, Minoda R, Brough DE, Raphael Y. Math1 gene transfer generates new cochlear hair cells in mature guinea pigs in vivo. J Neurosci. 2003; 23:4395–4400.
Article
42. Atkinson PJ, Wise AK, Flynn BO, Nayagam BA, Richardson RT. Hair cell regeneration after ATOH1 gene therapy in the cochlea of profoundly deaf adult guinea pigs. PLoS One. 2014; 9:e102077.
Article
43. Izumikawa M, Batts SA, Miyazawa T, Swiderski DL, Raphael Y. Response of the flat cochlear epithelium to forced expression of Atoh1. Hear Res. 2008; 240:52–56.
Article
44. Bray SJ. Notch signalling: a simple pathway becomes complex. Nat Rev Mol Cell Biol. 2006; 7:678–689.
Article
45. Kiernan AE. Notch signaling during cell fate determination in the inner ear. Semin Cell Dev Biol. 2013; 24:470–479.
Article
46. Adam J, Myat A, Le Roux I, Eddison M, Henrique D, Ish-Horowicz D, et al. Cell fate choices and the expression of Notch, Delta and Serrate homologues in the chick inner ear: parallels with Drosophila sense-organ development. Development. 1998; 125:4645–4654.
Article
47. Haddon C, Jiang YJ, Smithers L, Lewis J. Delta-Notch signalling and the patterning of sensory cell differentiation in the zebrafish ear: evidence from the mind bomb mutant. Development. 1998; 125:4637–4644.
Article
48. Morrison A, Hodgetts C, Gossler A, Hrabé de, Lewis J. Expression of delta1 and serrate1 (Jagged1) in the mouse inner ear. Mech Dev. 1999; 84:169–172.
Article
49. Lanford PJ, Lan Y, Jiang R, Lindsell C, Weinmaster G, Gridley T, et al. Notch signalling pathway mediates hair cell development in mammalian cochlea. Nat Genet. 1999; 21:289–292.
Article
50. Brooker R, Hozumi K, Lewis J. Notch ligands with contrasting functions: Jagged1 and Delta1 in the mouse inner ear. Development. 2006; 133:1277–1286.
Article
51. Kageyama R, Ohtsuka T, Kobayashi T. The Hes gene family: repressors and oscillators that orchestrate embryogenesis. Development. 2007; 134:1243–1251.
Article
52. Zheng JL, Shou J, Guillemot F, Kageyama R, Gao WQ. Hes1 is a negative regulator of inner ear hair cell differentiation. Development. 2000; 127:4551–4560.
Article
53. Zine A, Aubert A, Qiu J, Therianos S, Guillemot F, Kageyama R, et al. Hes1 and Hes5 activities are required for the normal development of the hair cells in the mammalian inner ear. J Neurosci. 2001; 21:4712–4720.
Article
54. Li S, Mark S, Radde-Gallwitz K, Schlisner R, Chin MT, Chen P. Hey2 functions in parallel with Hes1 and Hes5 for mammalian auditory sensory organ development. BMC Dev Biol. 2008; 8:20.
Article
55. Tateya T, Imayoshi I, Tateya I, Ito J, Kageyama R. Cooperative functions of Hes/Hey genes in auditory hair cell and supporting cell development. Dev Biol. 2011; 352:329–340.
Article
56. Kiernan AE, Cordes R, Kopan R, Gossler A, Gridley T. The Notch ligands DLL1 and JAG2 act synergistically to regulate hair cell development in the mammalian inner ear. Development. 2005; 132:4353–4362.
Article
57. Ma EY, Rubel EW, Raible DW. Notch signaling regulates the extent of hair cell regeneration in the zebrafish lateral line. J Neurosci. 2008; 28:2261–2273.
Article
58. Daudet N, Gibson R, Shang J, Bernard A, Lewis J, Stone J. Notch regulation of progenitor cell behavior in quiescent and regenerating auditory epithelium of mature birds. Dev Biol. 2009; 326:86–100.
Article
59. Li W, Wu J, Yang J, Sun S, Chai R, Chen ZY, et al. Notch inhibition induces mitotically generated hair cells in mammalian cochleae via activating the Wnt pathway. Proc Natl Acad Sci U S A. 2015; 112:166–171.
Article
60. Yamamoto N, Tanigaki K, Tsuji M, Yabe D, Ito J, Honjo T. Inhibition of Notch/RBP-J signaling induces hair cell formation in neonate mouse cochleas. J Mol Med (Berl). 2006; 84:37–45.
Article
61. Burns JC, Cox BC, Thiede BR, Zuo J, Corwin JT. In vivo proliferative regeneration of balance hair cells in newborn mice. J Neurosci. 2012; 32:6570–6577.
Article
62. Jung JY, Avenarius MR, Adamsky S, Alpert E, Feinstein E, Raphael Y. siRNA targeting Hes5 augments hair cell regeneration in aminoglycoside-damaged mouse utricle. Mol Ther. 2013; 21:834–841.
Article
63. Lin V, Golub JS, Nguyen TB, Hume CR, Oesterle EC, Stone JS. Inhibition of Notch activity promotes nonmitotic regeneration of hair cells in the adult mouse utricles. J Neurosci. 2011; 31:15329–15339.
Article
64. Mizutari K, Fujioka M, Hosoya M, Bramhall N, Okano HJ, Okano H, et al. Notch inhibition induces cochlear hair cell regeneration and recovery of hearing after acoustic trauma. Neuron. 2013; 77:58–69.
Article
65. Tona Y, Hamaguchi K, Ishikawa M, Miyoshi T, Yamamoto N, Yamahara K, et al. Therapeutic potential of a gamma-secretase inhibitor for hearing restoration in a guinea pig model with noise-induced hearing loss. BMC Neurosci. 2014; 15:66.
Article
66. Hébert JM. FGFs: neurodevelopment's jack-of-all-trades - how do they do it. Front Neurosci. 2011; 5:133.
Article
67. Pirvola U, Spencer-Dene B, Xing-Qun L, Kettunen P, Thesleff I, Fritzsch B, et al. FGF/FGFR-2(IIIb) signaling is essential for inner ear morphogenesis. J Neurosci. 2000; 20:6125–6134.
Article
68. Schimmang T. Expression and functions of FGF ligands during early otic development. Int J Dev Biol. 2007; 51:473–481.
Article
69. Wright TJ, Mansour SL. Fgf3 and Fgf10 are required for mouse otic placode induction. Development. 2003; 130:3379–3390.
70. Hayashi T, Ray CA, Bermingham-McDonogh O. Fgf20 is required for sensory epithelial specification in the developing cochlea. J Neurosci. 2008; 28:5991–5999.
Article
71. Pirvola U, Ylikoski J, Trokovic R, Hébert JM, McConnell SK, Partanen J. FGFR1 is required for the development of the auditory sensory epithelium. Neuron. 2002; 35:671–680.
Article
72. Colvin JS, Bohne BA, Harding GW, McEwen DG, Ornitz DM. Skeletal overgrowth and deafness in mice lacking fibroblast growth factor receptor 3. Nat Genet. 1996; 12:390–397.
Article
73. Hayashi T, Cunningham D, Bermingham-McDonogh O. Loss of Fgfr3 leads to excess hair cell development in the mouse organ of Corti. Dev Dyn. 2007; 236:525–533.
Article
74. Mueller KL, Jacques BE, Kelley MW. Fibroblast growth factor signaling regulates pillar cell development in the organ of corti. J Neurosci. 2002; 22:9368–9377.
Article
75. Puligilla C, Feng F, Ishikawa K, Bertuzzi S, Dabdoub A, Griffith AJ, et al. Disruption of fibroblast growth factor receptor 3 signaling results in defects in cellular differentiation, neuronal patterning, and hearing impairment. Dev Dyn. 2007; 236:1905–1917.
Article
76. Doetzlhofer A, Basch ML, Ohyama T, Gessler M, Groves AK, Segil N. Hey2 regulation by FGF provides a Notch-independent mechanism for maintaining pillar cell fate in the organ of Corti. Dev Cell. 2009; 16:58–69.
77. Su YX, Hou CC, Yang WX. Control of hair cell development by molecular pathways involving Atoh1, Hes1 and Hes5. Gene. 2015; 558:6–24.
Article
78. Ito H, Nakajima A, Nomoto H, Furukawa S. Neurotrophins facilitate neuronal differentiation of cultured neural stem cells via induction of mRNA expression of basic helix-loop-helix transcription factors Mash1 and Math1. J Neurosci Res. 2003; 71:648–658.
Article
79. Millimaki BB, Sweet EM, Dhason MS, Riley BB. Zebrafish atoh1 genes: classic proneural activity in the inner ear and regulation by Fgf and Notch. Development. 2007; 134:295–305.
Article
80. Miller JR, Hocking AM, Brown JD, Moon RT. Mechanism and function of signal transduction by the Wnt/beta-catenin and Wnt/Ca2+ pathways. Oncogene. 1999; 18:7860–7872.
Article
81. MacDonald BT, Tamai K, He X. Wnt/beta-catenin signaling: components, mechanisms, and diseases. Dev Cell. 2009; 17:9–26.
82. Shi F, Cheng YF, Wang XL, Edge AS. Beta-catenin up-regulates Atoh1 expression in neural progenitor cells by interaction with an Atoh1 3’ enhancer. J Biol Chem. 2010; 285:392–400.
Article
83. Agathocleous M, Iordanova I, Willardsen MI, Xue XY, Vetter ML, Harris WA, et al. A directional Wnt/beta-catenin-Sox2-proneural pathway regulates the transition from proliferation to differentiation in the Xenopus retina. Development. 2009; 136:3289–3299.
Article
84. Jayasena CS, Ohyama T, Segil N, Groves AK. Notch signaling augments the canonical Wnt pathway to specify the size of the otic placode. Development. 2008; 135:2251–2261.
Article
85. Jacques BE, Puligilla C, Weichert RM, Ferrer-Vaquer A, Hadjantonakis AK, Kelley MW, et al. A dual function for canonical Wnt/XMLLink_XYZ-catenin signaling in the developing mammalian cochlea. Development. 2012; 139:4395–4404.
Article
86. Shi F, Hu L, Jacques BE, Mulvaney JF, Dabdoub A, Edge AS. β-Catenin is required for hair-cell differentiation in the cochlea. J Neurosci. 2014; 34:6470–6479.
Article
87. Shi F, Hu L, Edge AS. Generation of hair cells in neonatal mice by β-catenin overexpression in Lgr5-positive cochlear progenitors. Proc Natl Acad Sci U S A. 2013; 110:13851–13856.
Article
88. Chai R, Kuo B, Wang T, Liaw EJ, Xia A, Jan TA, et al. Wnt signaling induces proliferation of sensory precursors in the postnatal mouse cochlea. Proc Natl Acad Sci U S A. 2012; 109:8167–8172.
Article
89. Jansson L, Kim GS, Cheng AG. Making sense of Wnt signaling-linking hair cell regeneration to development. Front Cell Neurosci. 2015; 9:66.
Article
90. Shi F, Kempfle JS, Edge AS. Wnt-responsive Lgr5-expressing stem cells are hair cell progenitors in the cochlea. J Neurosci. 2012; 32:9639–9648.
Article
91. Kuo BR, Baldwin EM, Layman WS, Taketo MM, Zuo J. In vivo cochlear hair cell generation and survival by coactivation of β-catenin and atoh1. J Neurosci. 2015; 35:10786–10798.
Article
92. Atkinson PJ, Huarcaya Najarro E, Sayyid ZN, Cheng AG. Sensory hair cell development and regeneration: similarities and differences. Development. 2015; 142:1561–1571.
Article
93. Cai T, Jen HI, Kang H, Klisch TJ, Zoghbi HY, Groves AK. Characterization of the transcriptome of nascent hair cells and identification of direct targets of the Atoh1 transcription factor. J Neurosci. 2015; 35:5870–5883.
Article
94. Krizhanovsky V, Soreq L, Kliminski V, Ben-Arie N. Math1 target genes are enriched with evolutionarily conserved clustered E-box binding sites. J Mol Neurosci. 2006; 28:211–229.
Article
Full Text Links
  • CMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr