期刊论文详细信息
BMC Developmental Biology
Cooperative and independent functions of FGF and Wnt signaling during early inner ear development
Katherine Shim1  Jian Zhang1  Amanda A. Mahoney Rogers1  Kevin D. Wright1 
[1] Department of Pediatrics, Children’s Research Institute, Medical College of Wisconsin, Milwaukee 53226, WI, USA
关键词: Cross talk;    β-catenin;    FGF;    Wnt8a;    Sprouty2;    Sprouty1;    Otic placode;    Inner ear;   
Others  :  1231016
DOI  :  10.1186/s12861-015-0083-8
 received in 2015-02-05, accepted in 2015-09-18,  发布年份 2015
【 摘 要 】

Background

In multiple vertebrate organisms, including chick, Xenopus, and zebrafish, Fibroblast Growth Factor (FGF) and Wnt signaling cooperate during formation of the otic placode. However, in the mouse, although FGF signaling induces Wnt8a expression during induction of the otic placode, it is unclear whether these two signaling pathways functionally cooperate. Sprouty (Spry) genes encode intracellular antagonists of receptor tyrosine kinase signaling, including FGF signaling. We previously demonstrated that the Sprouty1 (Spry1) and Sprouty2 (Spry2) genes antagonize FGF signaling during induction of the otic placode. Here, we investigate cross talk between FGF/SPRY and Wnt signaling during otic placode induction and assess whether these two signaling pathways functionally cooperate during early inner ear development in the mouse.

Methods

Embryos were generated carrying combinations of a Spry1 null allele, Spry2 null allele, β-catenin null allele, or a Wnt reporter transgene. Otic phenotypes were assessed by in situ hybridization, semi-quantitative reverse transcriptase PCR, immunohistochemistry, and morphometric analysis of sectioned tissue.

Results

Comparison of Spry1, Spry2, and Wnt reporter expression in pre-otic and otic placode cells indicates that FGF signaling precedes and is active in more cells than Wnt signaling. We provide in vivo evidence that FGF signaling activates the Wnt signaling pathway upstream of TCF/Lef transcriptional activation. FGF regulation of Wnt signaling is functional, since early inner ear defects in Spry1 and Spry2 compound mutant embryos can be genetically rescued by reducing the activity of the Wnt signaling pathway. Interestingly, we find that although the entire otic placode increases in size in Spry1 and Spry2 compound mutant embryos, the size of the Wnt-reporter-positive domain does not increase to the same extent as the Wnt-reporter-negative domain.

Conclusions

This study provides genetic evidence that FGF and Wnt signaling cooperate during early inner ear development in the mouse. Furthermore, our data suggest that although specification of the otic placode may be globally regulated by FGF signaling, otic specification of cells in which both FGF and Wnt signaling are active may be more tightly regulated.

【 授权许可】

   
2015 Wright et al.

附件列表
Files Size Format View
Fig. 5. 69KB Image download
Fig. 4. 59KB Image download
Fig. 3. 119KB Image download
Fig. 2. 27KB Image download
Figure 3. 47KB Image download
【 图 表 】

Figure 3.

Fig. 2.

Fig. 3.

Fig. 4.

Fig. 5.

【 参考文献 】
  • [1]Parr BA, Shea MJ, Vassileva G, McMahon AP. Mouse Wnt genes exhibit discrete domains of expression in the early embryonic CNS and limb buds. Development. 1993; 119:247-61.
  • [2]Chen J, Streit A. Induction of the inner ear: Stepwise specification of otic fate from multipotent progenitors. Hear Res. 2013; 297(C):3-12.
  • [3]Yamaguchi TP, Bradley A, McMahon AP, Jones S. A Wnt5a pathway underlies outgrowth of multiple structures in the vertebrate embryo. Development. 1999; 126:1211-23.
  • [4]Groves AK, Fekete DM. Shaping sound in space: the regulation of inner ear patterning. Development. 2012; 139:245-57.
  • [5]Takada S, Stark KL, Shea MJ, Vassileva G, McMahon JA, McMahon AP. Wnt-3a regulates somite and tailbud formation in the mouse embryo. Genes Dev. 1994; 8:174-89.
  • [6]Ladher RK, O’Neill P, Begbie J. From shared lineage to distinct functions: the development of the inner ear and epibranchial placodes. Development. 2010; 137:1777-85.
  • [7]Freyer L, Aggarwal V, Morrow BE. Dual embryonic origin of the mammalian otic vesicle forming the inner ear. Development. 2011; 138:5403-14.
  • [8]Ohyama T, Groves AK, Martin K. The first steps towards hearing: mechanisms of otic placode induction. Int J Dev Biol. 2007; 51:463-72.
  • [9]Groves AK, Labonne C. Setting appropriate boundaries_ Fate, patterning and competence at the neural plate border. Dev Biol. 2014; 389:2-12.
  • [10]Schlosser G. Early embryonic specification of vertebrate cranial placodes. WIREs Dev Biol. 2014; 3:349-63.
  • [11]Borello U, Buffa V, Sonnino C, Melchionna R, Vivarelli E, Cossu G. Differential expression of the Wnt putative receptors Frizzled during mouse somitogenesis. Mech Dev. 1999; 89:173-7.
  • [12]Schneider-Maunoury S, Pujades C. Hindbrain signals in otic regionalization: walk on the wild side. Int J Dev Biol. 2007; 51:495-506.
  • [13]Schimmang T. Expression and functions of FGF ligands during early otic development. Int J Dev Biol. 2007; 51:473-81.
  • [14]Ohyama T. Wnt signals mediate a fate decision between otic placode and epidermis. Development. 2006; 133:865-75.
  • [15]Freter S, Muta Y, Mak S-S, Rinkwitz S, Ladher RK. Progressive restriction of otic fate: the role of FGF and Wnt in resolving inner ear potential. Development. 2008; 135:3415-24.
  • [16]Mccarroll MN, Lewis ZR, Culbertson MD, Martin BL, Kimelman D, Nechiporuk AV. Graded levels of Pax2a and Pax8 regulate cell differentiation during sensory placode formation. Development. 2012; 139:2740-50.
  • [17]Mohamed OA, Clarke HJ, Dufort D. Beta-catenin signaling marks the prospective site of primitive streak formation in the mouse embryo. Dev Dyn. 2004; 231:416-24.
  • [18]Mahoney Rogers AA, Zhang J, Shim K. Sprouty1 and Sprouty2 limit both the size of the otic placode and hindbrain Wnt8a by antagonizing FGF signaling. Dev Biol. 2011; 353:94-104.
  • [19]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; 135:2251-61.
  • [20]Pfeffer PL, Gerster T, Lun K, Brand M, Busslinger M. Characterization of three novel members of the zebrafish Pax2/5/8 family: dependency of Pax5 and Pax8 expression on the Pax2.1 (noi) function. Development. 1998; 125:3063-74.
  • [21]Phillips BT. A direct role for Fgf but not Wnt in otic placode induction. Development. 2004; 131:923-31.
  • [22]Hans S, Westerfield M. Changes in retinoic acid signaling alter otic patterning. Development. 2007; 134:2449-58.
  • [23]Bajoghli B, Aghaallaei N, Jung G, Czerny T. Induction of otic structures by canonical Wnt signalling in medaka. Dev Genes Evol. 2009; 219:391-8.
  • [24]Gutknecht D, Fritzsch B. Lithium can transform ear placodes of Xenopus into multiple otic vesicles connected by tubes. Naturwissenschaften. 1990; 77:235-7.
  • [25]Padanad MS, Bhat N, Guo B, Riley BB. Conditions that influence the response to Fgf during otic placode induction. Dev Biol. 2012; 364:1-10.
  • [26]Domínguez-Frutos E, Vendrell V, Alvarez Y, Zelarayan LC, López-Hernández I, Ros M et al.. Tissue-specific requirements for FGF8 during early inner ear development. Mech Dev. 2009; 126:873-81.
  • [27]Ladher RK. Identification of synergistic signals initiating inner Ear development. Science. 2000; 290:1965-7.
  • [28]Park B-Y, Saint-Jeannet J-P. Hindbrain-derived Wnt and Fgf signals cooperate to specify the otic placode in Xenopus. Dev Biol. 2008; 324:108-21.
  • [29]Urness LD, Paxton CN, Wang X, Schoenwolf GC, Mansour SL. FGF signaling regulates otic placode induction and refinement by controlling both ectodermal target genes and hindbrain Wnt8a. Dev Biol. 2010; 340:595-604.
  • [30]Vendrell V, Vázquez-Echeverría C, López-Hernández I, Alonso BD, Martinez S, Pujades C et al.. Roles of Wnt8a during formation and patterning of the mouse inner ear. Mech Dev. 2013; 130:160-8.
  • [31]Basson M, Akbulut S, Watsonjohnson J, Simon R, Carroll T, Shakya R et al.. Sprouty1 Is a Critical Regulator of GDNF/RET-Mediated Kidney Induction. Dev Cell. 2005; 8:229-39.
  • [32]Shim K, Minowada G, Coling DE, Martin GR. Sprouty2, a mouse deafness gene, regulates cell fate decisions in the auditory sensory epithelium by antagonizing FGF signaling. Dev Cell. 2005; 8:553-64.
  • [33]Brault V, Moore R, Kutsch S, Ishibashi M, Rowitch DH, McMahon AP et al.. Inactivation of the beta-catenin gene by Wnt1-Cre-mediated deletion results in dramatic brain malformation and failure of craniofacial development. Development. 2001; 128:1253-64.
  • [34]Lewandoski M, Meyers EN, Martin GR. Analysis of Fgf8 gene function in vertebrate development. Cold Spring Harb Symp Quant Biol. 1997; 62:159-68.
  • [35]Marone M, Mozzetti S, De Ritis D, Pierelli L. Semiquantitative RT-PCR analysis to assess the expression levels of multiple transcripts from the same sample. Biol Procedures … 2001;3:19-25.
  • [36]Metzger RJ, Klein OD, Martin GR, Krasnow MA. The branching programme of mouse lung development. Nature. 2008;453:745-50.
  • [37]Shim K. Vibratome sectioning for enhanced preservation of the cytoarchitecture of the mammalian organ of Corti. J Visual Exp. 2011;52:e2793.
  • [38]Wright TJ. Fgf3 and Fgf10 are required for mouse otic placode induction. Development. 2003; 130:3379-90.
  • [39]Alvarez Y. Requirements for FGF3 and FGF10 during inner ear formation. Development. 2003; 130:6329-38.
  • [40]Mansour SL, Goddard JM, Capecchi MR. Mice homozygous for a targeted disruption of the proto-oncogene int-2 have developmental defects in the tail and inner ear. Development. 1993; 117:13-28.
  • [41]Min H, Danilenko DM, Scully SA, Bolon B, Ring BD, Tarpley JE et al.. Fgf-10 is required for both limb and lung development and exhibits striking functional similarity to Drosophila branchless. Genes Dev. 1998; 12:3156-61.
  • [42]Zhang J, Wright KD, Mahoney Rogers AA, Barrett MM, Shim K. Compensatory regulation of the size of the inner ear in response to excess induction of otic progenitors by fibroblast growth factor signaling. Dev Dyn. 2014; 243:1317-27.
  • [43]Riccomagno M, Takada S, Epstein D. Wnt-dependent regulation of inner ear morphogenesis is balanced by the opposing and supporting roles of Shh. Genes Dev. 2005; 19:1612-23.
  • [44]Ohyama T, Groves AK. Expression of mouse Foxi class genes in early craniofacial development. Dev Dyn. 2004; 231:640-6.
  • [45]Guy GR, Jackson RA, Yusoff P, Chow SY. Sprouty proteins: modified modulators, matchmakers or missing links? J Endocrinol. 2009; 203:191-202.
  • [46]Cabrita MA, Christofori G. Sprouty proteins, masterminds of receptor tyrosine kinase signaling. Angiogenesis. 2008;11:53–62.
  • [47]Mason J, Morrison D, Albertbasson M, Licht J. Sprouty proteins: multifaceted negative-feedback regulators of receptor tyrosine kinase signaling. Trends Cell Biol. 2006; 16:45-54.
  • [48]Kim HJ, Bar-Sagi D. Modulation of signalling by Sprouty: a developing story. Nat Rev Mol Cell Biol. 2004; 5:441-50.
  • [49]Yang L, O’neill P, Martin K, Maass JC, Vassilev V, Ladher R et al.. Analysis of FGF-dependent and FGF-independent pathways in otic placode induction. PLoS One. 2013; 8: Article ID e55011
  • [50]Li C, Scott DA, Hatch E, Tian X, Mansour SL. Dusp6 (Mkp3) is a negative feedback regulator of FGF-stimulated ERK signaling during mouse development. Development. 2007; 134:167-76.
  • [51]Urness LD, Li C, Wang X, Mansour SL. Expression of ERK signaling inhibitors Dusp6, Dusp7, and Dusp9 during mouse ear development. Dev Dyn. 2008; 237:163-9.
  • [52]Dailey L, Ambrosetti D, Mansukhani A, Basilico C. Mechanisms underlying differential responses to FGF signaling. Cytokine Growth Factor Rev. 2005; 16:233-47.
  • [53]Beurel E, Grieco SF, Jope RS. Glycogen synthase kinase-3 (GSK3): Regulation, actions, and diseases. Pharmacol Ther. 2015;148:114-31.
  • [54]Niehrs C. nrm3470. Nat Rev Mol Cell Biol. 2012; 13:767-79.
  • [55]Lien WH, Fuchs E. Wnt some lose some: transcriptional governance of stem cells by Wnt/ -catenin signaling. Genes Dev. 2014; 28:1517-32.
  • [56]Clevers H, Loh KM, Nusse R. Stem cell signaling. An integral program for tissue renewal and regeneration: Wnt signaling and stem cell control. Science. 2014; 346:1248012.
  • [57]Holland JD, Klaus A, Garratt AN, Birchmeier W. Wnt signaling in stem and cancer stem cells. Curr Opin Cell Biol. 2013; 25:254-64.
  • [58]Jope RS, Johnson GVW. The glamour and gloom of glycogen synthase kinase-3. Trends Biochem Sci. 2004; 29:95-102.
  • [59]Basson M, Watsonjohnson J, Shakya R, Akbulut S, Hyink D, Costantini F et al.. Branching morphogenesis of the ureteric epithelium during kidney development is coordinated by the opposing functions of GDNF and Sprouty1. Dev Biol. 2006; 299:466-77.
  • [60]Kemler R. From cadherins to catenins: cytoplasmic protein interactions and regulation of cell adhesion. Trends Genet. 1993; 9:317-21.
  • [61]Ciruna B, Rossant J. FGF signaling regulates mesoderm cell fate specification and morphogenetic movement at the primitive streak. Dev Cell. 2001; 1:37-49.
  • [62]Rebay I. Keeping the receptor tyrosine kinase signaling pathway in check: lessons from Drosophila. Dev Biol. 2002; 251:1-17.
  • [63]Brunner D, Dücker K, Oellers N, Hafen E, Scholz H, Klämbt C. The ETS domain protein pointed-P2 is a target of MAP kinase in the sevenless signal transduction pathway. Nature. 1994; 370:386-9.
  • [64]Olson ER, Pancratov R, Chatterjee SS, Changkakoty B, Pervaiz Z, DasGupta R. Yan, an ETS-domain transcription factor, negatively modulates the Wingless pathway in the Drosophila eye. EMBO Rep. 2011; 12:1047-54.
  • [65]Webber JL, Zhang J, Cote L, Vivekanand P, Ni X, Zhou J et al.. The relationship between long-range chromatin occupancy and polymerization of the drosophila ETS family transcriptional repressor Yan. Genetics. 2013; 193:633-49.
  • [66]Caviglia S, Luschnig S. The ETS domain transcriptional repressor Anterior open inhibits MAP kinase and Wingless signaling to couple tracheal cell fate with branch identity. Development. 2013; 140:1240-9.
  • [67]Bok J, Raft S, Kong K-A, Koo SK, Drager UC, Wu DK. Transient retinoic acid signaling confers anterior-posterior polarity to the inner ear. Proc Natl Acad Sci. 2011; 108:161-6.
  • [68]Abelló G, Khatri S, Radosevic M, Scotting PJ, Giráldez F, Alsina B. Independent regulation of Sox3 and Lmx1b by FGF and BMP signaling influences the neurogenic and non-neurogenic domains in the chick otic placode. Dev Biol. 2010;339:166–178.
  • [69]Sun X, Mariani FV, Martin GR. Functions of FGF signalling from the apical ectodermal ridge in limb development. Nature. 2002; 418:501-8.
  • [70]Mariani FV, Ahn CP, Martin GR. Genetic evidence that FGFs have an instructive role in limb proximal-distal patterning. Nature. 2008;453:401.
  文献评价指标  
  下载次数:8次 浏览次数:29次