DEVELOPMENTAL DYNAMICS 239:3467–3480, 2010

a PATTERNS & PHENOTYPES

Microarray Identification of Novel Downstream Targets of FoxD4L1/D5, a Critical Component of the Neural Ectodermal Transcriptional Network

Bo Yan, Karen M. Neilson, and Sally A. Moody*

FoxD4L1/D5 is a forkhead that functions as both a transcriptional activator and repressor. FoxD4L1/D5 acts upstream of several other neural transcription factors to maintain neural fate, regulate neural plate patterning, and delay the expression of neural differentiation factors. To iden- tify a more complete list of downstream genes that participate in these earliest steps of neural ectodermal development, we carried out a microarray analysis comparing gene expression in control animal cap ecto- dermal explants (ACs), which will form epidermis, to that in FoxD4L1/D5-expressing ACs. Forty-four genes were tested for validation by RT-PCR of ACs and/or in situ hybridization assays in embryos; 86% of those genes up-regulated and 100% of those genes down-regulated in the microarray were altered accordingly in one of these independent assays. Eleven of these 44 genes are of unknown function, and we provide herein their developmental expression patterns to begin to reveal their roles in ectodermal development. Developmental Dynamics 239:3467–3480, 2010. VC 2010 Wiley-Liss, Inc.

Key words: BMP signaling; FGF signaling; cement gland; olfactory placode; epidermis; Elf-1; FoxD4L1.1; FoxD5

Accepted 12 October 2010

INTRODUCTION gastrulation also represses neural In response to these signaling path- Developmental Dynamics fate (Christian et al., 1991; Heeg- ways, the presumptive neural ecto- During embryonic development, the Truesdell and Labonne, 2006) and derm of Xenopus initially expresses a formation of neural versus non-neu- promotes epidermal fate, in part by large number of transcription factors ral ectoderm is controlled by the dif- ferential activation of three main sig- regulating the duration of the BMP/ that have overlapping expression naling pathways (reviewed in Stern, SMAD1 signal (Fuentealba et al., domains (reviewed in Yan et al., 2009; 2005; Itoh and Sokol, 2007; Rogers 2007). Like BMP, Wnt signaling is Rogers et al., 2009). One of these is et al., 2009). BMP signaling is prevented in the presumptive neural FoxD5, whose gene nomenclature was required to activate an epidermal fate ectoderm by the secretion of Wnt recently changed to foxd4l1.1 (http:// in the embryonic ectoderm, and antagonists from the Organizer. FGF www.xenbase.org/gene/showgene.do? reduction of BMP signaling by secre- signaling promotes a neural fate and method¼display&geneId¼479246&). tion of BMP antagonists from the Or- contributes to the down-regulation of Therefore, we will refer to it as FoxD ganizer/Node converts the embryonic both Bmp gene expression and BMP 4L1/D5 from here on. FoxD4L1/D5 is ectoderm to a neural fate (Hawley signaling (Delaune et al., 2005; Kur- expressed broadly throughout the et al., 1995). Wnt signaling during oda et al., 2005; Marchal et al., 2009). newly induced neural ectoderm

Additional Supporting Information may be found in the online version of this article. Department of Anatomy and Regenerative Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC Grant sponsor: NIH; Grant number: NS23158; Grant sponsor: NSF; Grant number: IOS-0817902; Grant sponsor: George Washington University School of Medicine and Health Sciences. *Correspondence to: Dr. Sally A. Moody, Department of Anatomy and Regenerative Biology, The George Washington University School of Medicine and Health Sciences, 2300 I Street, N.W., Washington, D.C. 20037. E-mail: [email protected] DOI 10.1002/dvdy.22485 Published online 10 November 2010 in Wiley Online Library (wileyonlinelibrary.com).

VC 2010 Wiley-Liss, Inc. 3468 YAN ET AL.

adjacent to the blastopore lip, gradu- tested genes. Several of the genes require the endogenous neuralizing ally becomes confined to the mid- downstream of FoxD4L1/D5 include environment for FoxD4L1/D5 to opti- regions of the neural plate, and at molecules in the BMP and FGF signal- mally affect their transcription. Finally, neural tube stages is expressed only ing pathways, indicating that one it should be noted that although FoxD5 at the midbrain-hindbrain region and function of FoxD4L1/D5 is to maintain is maternally expressed, its transcripts in the tail bud (So¨lter et al., 1999; a signaling environment in the embry- are not detected by RT-PCR in unin- Fetka et al., 2000 Sullivan et al., onic ectoderm that favors a neural jected ACs harvested at the same 2001). We recently showed that fate. Several of the up-regulated genes stages as the microarray analyses (Sul- FoxD4L1/D5 plays an essential role are normally expressed in the neural livan et al., 2001). in maintaining an immature neural ectoderm, and several of the down- fate by regulating a number of other regulated genes are normally ex- FoxD4L1/ neural transcription factors (Yan pressed in the ectoderm bordering the Increased Levels of et al., 2009). Knock-down of endoge- neural plate or in the epidermis. These D5 Alter the Expression of nous FoxD4L1/D5 in the neural ecto- results confirm a role for FoxD4L1/D5 Numerous Genes derm by targeted injection of anti- in maintaining a neural versus non- Increasing the level of FoxD4L1/D5 in sense morpholino oligonucleotides neural ectodermal fate. This study ACs significantly changed by >2-fold (MOs) reduces the size of the neural additionally provides the developmen- the expression of 216 genes repre- plate and causes a loss of 11 other tal expression patterns of genes of sented on the array. Of these, 95 were neural transcription factors. Thus, unknown function that are regulated up-regulated (see Supp. Table S1, FoxD4L1/D5 is necessary for either by FoxD4L1/D5. which is available online) and 121 the induction or the maintenance of were down-regulated (Supp. Table the expression of these genes in RESULTS AND DISCUSSION S2). Of the up-regulated genes, more the neural ectoderm. In contrast, than half are of unknown function. increased expression of FoxD4L1/D5 FoxD4L1/D5 is a member of the fork- The majority of the remainder by mRNA injection into a neural pro- head/winged helix family of transcrip- includes proteins involved in meta- genitor blastomere: (1) increases the tion factors that functions as both an bolic processes and transcription expression of Gem and Zic2; (2) ini- activator and a repressor (Sullivan factors, with smaller percentages tially represses , Sox3,and et al., 2001; Yan et al., 2009). Our pre- involved in signaling pathways and Sox11, and then either increases their vious work demonstrated that more general cellular functions (Fig. endogenous expression level (Sox11) FoxD4L1/D5 acts upstream of 11 other 1A). Of the down-regulated genes, a or expands their expression domains transcription factors (Gem, Sox2, little less than half are of unknown (Sox2, Sox3); and (3) represses the Sox3, Sox11, SoxD, Zic1-3, Xiro1-3) in function, 16% are transcription fac- expression of Zic1, Zic3, SoxD,and the nascent neural ectoderm to main- tors, 12% are involved in metabolic Xiro1-3. Ventral expression of tain an immature neural fate, regulate processes, 13% are involved in signal- FoxD4L1/D5 also promotes ectopic neural plate patterning, and delay the ing pathways, and the remainder rep- expression of some neural genes, onset of expression of bHLH neural resent more general cellular functions Developmental Dynamics decreases epidermal genes (TFAP2, differentiation transcription factors (Fig. 1B). Epi-cytokeratin), and locally reduces (Yan et al., 2009). To identify a more BMP signaling as evidenced by the complete list of downstream genes loss of nuclear phosphoSMAD1/5/8 that may coordinately regulate the Validation of Microarray staining (Yan et al. 2009). Thus, earliest steps of neural ectodermal de- Results by RT-PCR and ISH FoxD4L1/D5 is a key regulator of neu- velopment, we compared the gene Assays ral fate via both transcription and expression profile of control, untreated cell-cell signaling. However, we do not ACs, which would eventually form To predict the level of false-positive yet know the molecular details of how epidermis, to that of FoxD4L1/D5- identification of downstream genes by this is accomplished. expressing ACs. Assays were per- the microarray approach, we assayed To identify a broader cast of mole- formed in ACs to avoid the influence of the expression of a subset of putative cules that may be involved in the gene endogenous signaling centers in whole downstream genes in ACs by RT-PCR network by which FoxD4L1/D5 regu- embryos. We did not treat the ACs and in whole embryos by ISH. Because lates neural ectodermal fate, microar- with neuralizing factors to elicit a neu- FoxD4L1/D5 is known to regulate ray expression analyses were per- ral ectodermal response because previ- other transcription factors and BMP formed on animal cap ectodermal ous work showed that FoxD4L1/D5- signaling (Yan et al., 2009), we explants (ACs) that expressed FoxD expressing ACs express a subset of selected genes for validation if they 4L1/D5. A subset of the candidate neural genes in the absence of early are known to be involved in transcrip- downstream genes identified by micro- mesoderm genes (Sullivan et al., tion or signaling. We also chose genes array was retested by RT-PCR analy- 2001). However, it should be noted of unknown function that displayed sis of FoxD4L1/D5 mRNA-injected that none of the 11 genes listed above the greatest fold-changes (Tables 1 ACs and/or by in situ hybridization that are regulated by FoxD4L1/D5 in and 2). assays of FoxD4L1/D5 mRNA- whole embryo assays (Yan et al., 2009) For the selected FoxD4L1/D5 up- injected embryos. Together these vali- were altered >2-fold in this microar- regulated genes (Table 1), 13/21 were dated the microarray results for 41/44 ray assay, suggesting that these genes significantly increased >1.5-fold in FOXD5 DOWNSTREAM TARGETS 3469

the RT-PCR analysis of FoxD4L1/D5- expressing ACs, consistent with the (18) (17) (58) (34) microarray analysis. For 6/21 genes, : : : : there was no detectable change in expression level (0.9–1.49-fold). Two genes (EST-BJ051730, EST-CB983 184) that scored as up-regulated by the microarray analysis were down- (46)(30) NC (21) (31) NC (25) (18) NC (25) 94.4% (20)(26)(27) NC (24) NC (27) NC (27) (66) 41.2% (21) NC (27) (25) NC (25) (46) 63.8% (39) NC (47) (53) 67.6% (46) NC (45) (35) NC (41) regulated >2-fold in the RT-PCR : : : : : : : : : : : : : : : assay. To analyze whether FoxD4L1/ D5 alters the expression of these ISH, percentage of embryos Dorsal clone Ventral clone genes in whole embryos, we injected FoxD4L1/D5 mRNA into a single dor- sal 16-cell blastomere to cause the a FoxD4L1/D5-expressing clone to over-

PCR fold increase* lap with the endogenous expression domain of each target gene. The ex- pression of Prickle-b, Egr1, and XEvl proved below detection by ISH and, therefore, could not be analyzed by this method. Of the 18 remaining genes, the expression of 15 was either increased or expanded in >50% of the embryos in which FoxD4L1/D5 was expressed in a dorsal clone (Table 1; Fig. 2A). These included 10 that also showed an increase by RT-PCR, 3 that showed no change by RT-PCR, and 2 (EST-BJ051730, EST-CB983184) that showed a decrease by RT-PCR. Fur- ther analysis is required to explain why EST-BJ051730 and EST-CB 983184 are up-regulated by microar- ray and ISH analyses but down-regu- lated by RT-PCR analysis. We also tested whether FoxD4L1/D5 could RT-PCR forward primer RT-PCR reverse primer

Developmental Dynamics ectopically induce these genes by injecting mRNA into a single ventral : 16-cell blastomere; 4/18 genes were induced in the ventral epidermis by FoxD4L1/D5 (Table 1; Fig. 2A). In total, 18/21 genes (85.7%) were con- firmed to be increased in expression by FoxD4L1/D5 by at least one independ- , increased or expanded expression; nd, not detected; NC, no change. Sample size is shown in parentheses.

: ent assay. Only the FoxD4L1/D5 up- regulation of XEvl (Wanner et al., Accession number Array fold TABLE 1. Changes in Expression Levels of Selected FoxD4L1/D5 Up-Regulated Genes 2005), EST-BG161338 and XLef1 (Molenaar et al., 1998) by microarray analyses were not confirmed by either RT-PCR or ISH analyses. For the 23 selected down-regulated genes, 21 were tested by RT-PCR (Table 2). TFAP2 was not tested because we previously demonstrated down-regulation by FoxD4L1/D5 (Yan et al., 2009); Gata3 could not be tested because 3 different primer sets pro- duced inappropriately sized products. Of the remaining 21 genes, 15 were significantly decreased >1.5 fold, con- Array number Gene name Xl.403.1.S1_atXl.20011.1.S1_atXl.637.1.A1_at FoxB1/Fkh-5 FGF-8bXl.3540.1.S1_atXl.1775.1.S1_at AF064810 Egr1 Fullback 4.842 Brat/VegT AF461177 4.591 AF131890 U89707 AF250345 tcctctccatccatgagtcc 3.943 4.501 3.868 gaccaactaagccgacggctcatc tttatgaggttctgtggtgtggtgtc attgtaaggggaacgtgctg gcccacttcatcctcctgtagtag gagatgttagccttgtatctgc 31.67 acccatatagtgcgatgtca tgctcccaactggaaactgtagg 5.75 gtactgttgatagtcttgaggtcc 80.0% 2.45 ctcccaatgcagggttttta 7.33 80.4% 58.1% 8.62 nd 88.9% nd Xl.24509.1.A1_atXl.16539.1.A1_at MGC132176Xl.25938.1.A1_at LOC496224Xl.18191.1.A1_at BC108806 ESTXl.22268.1.S1_at XEvl BC088686Xl.15365.1.A1_at 3.480 EST 3.436 EST BC097640 gaggcaacaaaggaagtcca BI312773 3.301 gtggtacccagaggtggaga BG161338 3.290 BJ051730 ggagttccagggggaaataa 3.050 2.930 cgattggtgtccttgttcct gtttagctggcttggctttg 3.93 tagtggtccaccacctcctc aggctgtggcagaagctaaa tgctgacacctgaacagtcc 0.95 aacaggattgggctgaacac 75.0% ccacaaggacttgagcttcc agccttcttttgagcagcag cacggtgtagctgcgaataa 65.4% 1.82 1.34 1.07 0.28 59.3% nd NC (35) 84.8% NC (24) nd Xl.15990.1.A1_at EST BJ043686 2.872 ttggggatttgctattccag gagcaatgcctaaccccata 0.94 52.4% Xl.9731.1.A1_at EST CB983184 2.842 atactgggctgacctgtgct gcaccggagttcactgtttt 0.49 64.0% Xl.14214.1.A1_atXl.2755.1.S1_at MGC68716 XSpr-2 BC060483 2.820 AY062263 2.226 gggggtcggcaatacttaat caaactgttgcctctcatgag tcagtctggcagccctagtt cacttacacctccggcagcgc 4.02 2.79 NC (30) 89.1% NC (24) Xl.12111.1.S1_atXl.1265.1.S1_at Prickle-b Oct-1 BC073208 2.212 X17190 2.152 gtggatcacgcacaaatgac cttttgcccatccgtacact ctggagggacccctaacatt agtgcgttctggatggaatc 7.06 1.36 nd 82.1% nd Xl.457.1.S1_at Derriere AF065135 2.122 tggcagagttgtggctatca ctatggctgctatggttcctt 1.82 50.9% Xl.410.1.S1_atXl.1182.1.S1_at XLef-1 FGFR2 AF287147 X65943 2.109 2.086 attatccccagcagcaacag cccccttgtgacttggacta gcaccactgggactttgttt ccctgaagaactggcttctg 1.22 1.54 NC (30) 52.2% NC (29) Xl.5454.1.S1_at Xmc AF394111 2.016 ctggtgttacagaccaagggg acctgtgcttttgccactc 2.54 82.9% *, average of three independent experiments;

a sistent with the microarray analysis. 3470 YAN ET AL.

For 6/21 genes, there was no detecta-

b ble change in expression level. The (59) (16) (73) (36) (43) (39) (29) (63) (28) (25) (56) (16) (47) (36) (63) (65) expression of Frb3 and Dlx3-a proved ; ; ; ; ; ; ; : ; ; ; ; ; ; ; ; below detection by ISH and, therefore, could not be analyzed by this method. Of the 21 remaining genes, 20 showed decreased expression in >50% of FoxD4L1/D5 mRNA-injected embryos (17) 39.0% (28) 50.0% (24) NC (21) (33) NC (33) (23) 63.0% (26) 52.8% (23) 52.8% (31) 48.3% (57) 63.5% (39) 35.7% (33) 52.0% (36) NC (18) (15) 58.9% (36) 37.5% (18) 87.2% (41) 47.2% (41) NC (20) (22) NC (59) (33) 58.7% (16) 93.8% (Table 2; Fig. 2B). Expression of Szl ; ; ; ; ; ; ; ; : ; ; ; ; ; ; ; ; ; ; ; was strongly induced both dorsally and ventrally, as previously reported ISH, percentage of embryos Dorsal clone Ventral clone (Yan et al., 2009). Surprisingly, this response is opposite to the microarray and RT-PCR data suggesting that there are additional factors in the

PCR fold decrease* whole embryo that allow for Szl induc- tion by FoxD4L1/D5. It is interesting that although Szl and Frb3 are consid- ered paralogues (Hufton et al., 2006), we did not detect up-regulation of Frb3 by ISH. In total, all 23 genes were confirmed to be decreased in expression by FoxD4L1/D5 by at least one independent method.

Developmental Roles of Validated FoxD4L1/D5 Downstream Genes Previous studies of FoxD4L1/D5 indi- cate that it plays multiple roles in early neural ectodermal development, including: (1) increasing the expres- sion of neural ectoderm genes; (2) reducing the expression of neural dif-

, increased expression; nd, not detected; -, not done; NC, no change; Sample size isferentiation shown in parentheses. genes; (3) reducing the : Developmental Dynamics

RT-PCR forward primer RT-PCR reverse primer expression of epidermis specific genes; and (4) reducing BMP signaling (So¨l-

; ter et al., 1999; Fetka et al., 2000; Sul- livan et al., 2001; Yan et al., 2009). Array fold Analyses of the Gene Ontology func- tion and/or published literature of those genes validated to be down- stream of FoxD4L1/D5 demonstrate , decreased expression; ; concordance with these previous Accession number findings, reveal a newly described

TABLE 2. Changes in Expression Levels of Selected FoxD4L1/D5 Down-Regulated Genes involvement in mesoderm and endo- derm specification and a newly described involvement in FGF signal- ing (Tables 3, 4).

FoxD4L1/D5 up-regulates genes expressed in the neural ectoderm. FoxB1/Fkh5, a forkhead transcrip- tion factor, is expressed in the poste- rior neural ectoderm during gastru- Array number Gene name Xl.5908.1.S1_x_at Xpo X58487 2.611 ctccggttggtaagcacatt ggcaacatatcagggcagtt 1.11 76.5% Xl.14513.1.A1_at Dlx3 BC123268 4.505 atgagtggcccctatgagaagaag ggttctctgtaatggacaaacgg 0.99 64.3% Xl.12881.2.A1_at MGC81002 BC073481 4.810 ccagtggctggagatagagc gctctgaggtctccacgttc 1.45 58.3% Xl.450.1.S1_at Xash1 M98272 4.831 aacttcaatggcttcggcta agcgtctccactttgctcat 5.56 81.8% Xl.2206.1.S1_at Neptune AF353715 2.597 ttgtcatcggaccacctaca atatcctctgtgggcactgg 1.85 78.3% Xl.620.1.S1_s_atXl.7307.1.S1_at Frzb3 LOC443682 BC041544 AF136184 4.149 4.385 aatgtttgctgggggtcata gattgaacatgtgctgctga aggtgcccttatcagcaatg atggtgtctccacctccttg 5.56 2.78 61.5% nd nd Xl.139.1.S1_at FoxI1/XFD-2 X74316 2.451 ccagaactgaaatcttagcaa taacaaagataaagccagaggt 3.85 NC (21) 60.5% Xl.23326.1.S1_at MGC53193 BC045031 3.788 gaaagtgacattgccggact ctgagccctcacatcagtca 3.85 78.3% Xl.3606.1.S1_at Szl AF059570 2.309 gattgaacagtggctgctga atgtctccacctccttgtgg 5.00 86.0% Xl.792.1.S1_at GATA2 M76564 3.745 ggaaggaattcagaccagaaatagga actccagcatggtgacggctatgg 3.57 77.4% Xl.3326.2.S1_a_at BMP7 BC057702 3.175 atctccgcagtgttcgatct atgatccagtcttgccaacc 1.69 71.8% Xl.262.1.S1_at Dlx6 D10259 2.242 gctacatgcctggctactcc ctcctgtgcagccactgtaa 1.61 66.7% Xl.793.1.A1_at GATA3 M76565 2.985 - - - 66.7% Xl.838.1.S1_atXl.1076.1.S1_at Dlx3-a MGC52564 U76752 BC116461 3.003 3.115 aaagaagcaggcaggaacaa atttggctctgcctgagaga tttctgtgccattcgatcag aaggctttgctgctgttgtt 4.76 2.00 83.3% nd nd Xl.15156.1.S1_at MGC52940 BC045214 2.976 tgctgaagacggagtcaatg ttcaaagagccgaggtgagt 1.54 80.6% Xl.563.1.S2_at Xblimp1/Prdm1 AF182280 2.119 gcgagcatgaaagacaaaca agcgggtatgggagagtctt 1.23 77.8% Xl.4619.1.S1_s_at BMP4 BC060340 2.976 gcatgtaaggataagtcgatc gatctcagactcaacggcac 0.93 68.2% Xl.1047.1.S1_at Lim5/Lhx5 L42546 2.088 tgagctctgggccttacagt ttttctgaagggtggtgtcc 1.35 78.0% Xl.21868.1.S1_at Elf-1 BC044973 2.110 ggtgctacaaccccaaagaa agcagaggtgaagtgctggt 2.27 58.6% Xl.1016.1.S1_at FGFR4c AF288453 2.849 gcctttgatatcaccaag agaggatggcactggatc 2.27 69.7% Xl.2143.1.S1_at TFAP-2 M59455 2.681 -lation and in the - diencephalon- - 68.8% *, Average of three independentReported experiments; in Yan et al., 2009.

a b midbrain region in the neural tube FOXD5 DOWNSTREAM TARGETS 3471

(Gamse and Sive, 2001). Fullback,a tion (Wallingford et al., 2002). Oct-1,a FoxD4L1/D5 down-regulates p75-related neurotrophin , is POU homeodomain transcription fac- transcription factors that expressed in the posterior neural ecto- tor, is expressed in the neural ecto- promote neural differentiation. derm during gastrula and neural derm during gastrulation, and then plate stages, and later is expressed in becomes enriched in the anterior neu- Xash1, a bHLH transcription factor, is the diencephalon-midbrain region ral tube (Veenstra et al., 1995). Thus, expressed in differentiating neural (Bromley et al., 2004). Prickle-b, FoxD4L1/D5 up-regulates other neu- progenitors (Ferreiro et al., 1993). It which is involved in the non-canonical ral genes with which it shares com- was significantly down-regulated by Wnt-PCP pathway during convergent mon expression domains (Table 3). FoxD4L1/D5 (Table 4), in accord extension movements (Takeuchi This would be expected of down- with a previous study showing that et al., 2003), is expressed in the poste- stream targets that are positively FoxD4L1/D5 down-regulates other rior neural ectoderm during gastrula- regulated by FoxD4L1/D5. bHLH neural differentiation genes (Ngnr1, NeuroD) (Sullivan et al 2001).

FoxD4L1/D5 down-regulates epidermal-specific genes. Many genes validated to be down- regulated by FoxD4L1/D5 (Bmp4, Bmp7, Dlx3, Dlx3-a, Dlx6, Elf-1, FoxI1, Lim5, Neptune, TFAP-2, Xb limp-1) are known to be involved in non-neural ectodermal development (Table 4). This is consistent with a pre- vious report that FoxD4L1/D5 represses epidermal fate (Yan et al., Fig. 1. Gene ontology analysis of the genes whose expression levels in non-neuralized ACs were significantly altered >2-fold by FoxD4L1/D5 in a microarray analysis. A: Percentages of 2009). However, Elf1 and Lim5 are genes up-regulated by FoxD4L1/D5 in each of several GO functional classes. B: Percentages of also later expressed in the neural genes down-regulated by FoxD4L1/D5 in each of several GO functional classes. The ‘‘Others’’ plate. It will be interesting to deter- category includes metal ion binding proteins, inhibitors, kinases, and undefined nuclear factors. mine whether they are later involved Developmental Dynamics

Fig. 2. Whole mount in situ hybridization assays of embryos injected with FoxD4L1/D5 mRNA. A: Examples of FoxD4L1/D5 causing an increase in downstream gene expression. In neural plate stage embryos, the expression domains of FoxB1 and XSpr2 are broader on the FoxD4L1/D5- injected side (right; indicated by bGal-expressing (red) cells). Lines indicate width of neural plate on injected (right) and control (left) sides of embryos. Dorsal views with anterior to the bottom. The FoxB1 expression domain also extends more anterior (bracket) compared to anterior limit on control side (arrow). At gastrula stages, the endogenous domains of Fgf8b and Oct-1 surround the yolk plug; the anterior extent of the domain is indicated by an arrow on the control side. The cells expressing FoxD4L1/D5 (red nuclei) in the neural plate (bracket) also express increased lev- els of Fgf8b and Oct-1. Dorsal views with anterior to the bottom. Inset in Oct-1 embryo: higher magnification showing FoxD4L1/D5-cells with red nuclei and blue cytoplasm. At gastrula stages, VegT/Brat and BJ051739 are ectopically induced in the animal cap ectoderm. Animal pole views. B: Examples of FoxD4L1/D5 causing a decrease in downstream gene expression. In neural plate stage embryos, MGC52564, MGC52940, Nep- tune, Dlx6, and Blimp1 are expressed in the border zone that separates the anterior neural plate (np) from the epidermis (epi). For each gene, this region shows decreased expression at the site of FoxD4L1/D5-expressing cells (red nuclei, arrows). MGC52564, MGC52940, Dlx6, Blimp1 are an- terior views with dorsal to the top; control side is on the left. Neptune is a side view with anterior to top and dorsal to right. In a gastrula embryo, the normal high level of FoxI1 expression in the animal cap ectoderm (epi) is greatly reduced in the FoxD4L1/D5-expressing clone (*), which is out- lined for clarity. 3472 YAN ET AL.

TABLE 3. Description of Function and Expression Patterns of Validated Up-Regulated Genesa

Gene name Protein description Expression patterns References FoxB1/Fkh5 Transcription factor Posterior NE, DMR Gamse and Sive, 2001 FGF-8b Peptide growth factor Mesoderm, anterior NP, MHR Fletcher et al., 2006 Egr1 DNA binding DBL, mesoderm Panitz et al., 1998 Fullback p75-like receptor Posterior NE, mesoderm, DMR Bromley et al., 2004 Brat/VegT Transcription factor DBL, endomesoderm Horb and Thompsen, 1997; Zhang and King, 1996 MGC132176 Similar to heme-oxygenase AC, NP, BZ, NT, MHR, NC, This study CG, lens, Oto, BA, Neph LOC496224 Similar to uridine-cytidine kinase AC, NP, BZ, NT, retina, lens, Oto, This study BA, somites BC097640 Similar to Solute carrier family AC, NP, BZ, CG, PL, NT, retina, This study 43, member 2 pineal, lens, Oto, BA, heart, somites, Neph, BL BJ051730 Unknown AC, NP, BZ, NT, MHR, NC, This study lens, Oto, BA, somites, Neph BJ043686 Unknown AC, NP, BZ, NT, lens, Oto, BA, This study somites, Neph CB983184 Similar to Rapamycin- AC, NP, BZ, NT, retina, Olf, lens, This study associated protein-1 Oto, BA, somites, heart MGC68716 Similar to Claudin-5 AC, NP, BZ, NT, pineal, Oto, BA, This study heart, somites, notochord XSpr2 Zinc finger protein DBL, Mesoderm, brain Ossipova et al., 2002 Prickle-b LIM domain-PCP pathway Posterior NE, dorsal mesoderm Wallingford et al., 2002 Oct-1 Transcription factor Ectoderm, anterior NT Veenstra et al., 1995 Derriere Peptide growth factor Endoderm, mesoderm Sun et al., 1999 FGFR2 FGF receptor MHR Friesel and Brown, 1992 Xmc Marginal coil protein DBL, mesoderm Frazzetto et al., 2002

aAC, animal cap ectoderm; BA, branchial arches; BL, ventral blood islands; BZ, ectoderm bordering the neural plate; CG, cement gland; DBL, dorsal blastopore lip; DMR, diencephalon-midbrain region; HB, hindbrain; MHR, midbrain-hindbrain region; NC, neu- ral crest; NE, neural ectoderm; Neph, nephric mesoderm; NP, neural plate; NT, neural tube; Olf, olfactory, Oto, otocyst; PL, placodes.

in neural differentiation, as predicted (Yan et al., 2009). The microarray data ectoderm during gastrulation and

Developmental Dynamics by their down-regulation by FoxD4L1/ support the suggestion that FoxD4L1/ neurulation in Xenopus (Sato and D5. This would be in accord with a D5 interferes with the BMP signaling Sargent, 1991). These results are previous study that showed that pathway, but the molecular mecha- consistent with the conclusion that some neural genes (Zic, Xiro)that nism is not yet known. FoxD4L1/D5 down-regulates genes promote bHLH expression of neural that specify tissues other than neural differentiation genes are down-regu- FoxD4L1/D5 down-regulates ectoderm. lated by FoxD4L1/D5 (Yan et al., genes involved in mesoderm and However, there were a few striking 2009). examples of mesoderm- and endo- endoderm development. derm-associated genes that were FoxD4L1/D5 down-regulates Similar to our findings with non-neu- strongly up-regulated by FoxD4L1/D5 ral ectodermal genes, we found that (Table 3). Brat/VegT is a T-box tran- genes involved in BMP signaling. genes involved in mesoderm and endo- scription factor that is essential in Several genes related to BMP signal- derm development (Bmp4, Bmp7, establishing the endoderm in Xenopus ing were down-regulated by FoxD4L1/ Frzb3, GATA2, GATA3, Xpo) were (Horb and Thomsen, 1997; Zhang D5 (Table 4). These include two down-regulated by FoxD4L1/D5 (Ta- et al., 1998), and Derriere is a Nodal ligands (Bmp4, Bmp7) and several ble 4). BMP4 and BMP7 promote ven- factor involved in mesoderm induction genes whose expression is known to tral mesodermal fates (Nishimatsu (Sun et al., 1999) that is directly regu- depend on BMP signaling (Dlx3, Dlx6, and Thomsen, 1998). Frzb3 is a puta- lated by Brat/VegT (White et al., Szl, TFAP-2). Recent studies show tive Wnt inhibitor (AF136184, Gen- 2002). Egr1, a zinc finger protein, is that GATA2 also acts downstream of bank). GATA2 and GATA3 are primar- involved in early mesoderm induction BMP in ectodermal cells (Dalgin et al., ily expressed in blood lineages derived in Xenopus (Panitz et al., 1998), and 2007). These results are in accord with from ventral mesoderm, although Xmc is a novel coil-coiled protein ini- a previous report that FoxD4L1/D5 there may be some neural expression tially expressed in the dorsal marginal reduced nuclear phospho-SMAD 1/5/8 of GATA3 (Zon et al., 1991). Xpo is zone at the onset of gastrulation and expression in ventral epidermal cells expressed in posterior mesoderm and later strongly expressed in the FOXD5 DOWNSTREAM TARGETS 3473

TABLE 4. Description of Function and Expression Patterns of Validated Down-Regulated Genes

Gene name Protein description Expression References Xash1 Transcription factor Brain, retina Ferreiro et al., 1993 MGC81002 Unknown AC, NP, BZ, NT, Epi, retina, Oto, BA This study Frzb3 Secreted protein Ventral mesoderm, ectoderm Hufton et al., 2006 LOC443682 Predicted sulfotransferase AC, Epi, ANR, BZ, Neph This study MGC53193 Predicted cytokeratin AC, Epi, ANR, BZ, NT, This study lens, Oto, BA, Ph GATA2 Transcription factor Mesoderm Bertwistle et al., 1996; Zon et al., 1991 BMP7 Peptide growth factor Ventral mesoderm, ectoderm Hawley et al., 1995 Dlx3-a Transcription factor Ectoderm, CG Papalopulu and Kintner, 1993 MGC52564 XAG-1; Secreted protein Ectoderm, CG Sive et al., 1989 GATA3 Transcription factor Mesoderm, brain Bertwistle et al., 1996; Zon et al., 1991 MGC52940 Predicted Villin1 AC, Epi, BZ, CG, Olf, Ph This study BMP4 Peptide growth factor Ventral mesoderm, ectoderm Fainsod et al., 1994 FGFR4c FGF receptor Ectoderm, NT, brain, eyes Golub et al., 2000 TFAP-2 Transcription factor AC, Epi, NC Luo et al., 2003; Winning et al., 1991 Xpo Transcription factor Posterior mesoderm, Sato and Sargent, 1991 posterior ectoderm Neptune Transcription factor Ectoderm, BZ Takeda et al., 2005 FoxI1/ XFD-2 Transcription factor AC, Epi, BZ Matsuo-Takasaki et al., 2005 Szl Secreted protein Ventral marginal zone Salic et al., 1997 Dlx6 Transcription factor AC, Epi, BZ Luo et al., 2001 Xblimp-1/ Prdm-1 Transcription factor Non-neural ectoderm, de Souza et al., 1999; endomesoderm, BZ Rossi et al., 2008 Elf-1 Transcription factor AC, Epi, NP, BZ, lens, BA This study Lim5/Lhx5 Transcription factor Ectoderm, NP, brain Toyama et al., 1995

aAC, animal cap ectoderm; ANR, anterior neural ridge; BA, branchial arches; BZ, border zone of neural plate; CG, cement gland; Epi, epidermis; NC, neural crest; NP, neural plate; NT, neural tube; Olf, olfactory placode/pit; Oto, otocyst, Ph, anterior pharynx.

mesoderm flanking the neural plate expression by altering FGF signaling, hauer et al., 2000) was down-regulated (Frazzetto et al., 2002). This inconsis- as discussed in the next section. by FoxD4L1/D5 (Table 4). These re- tency requires experimental evalua- sults are consistent with the proposal

Developmental Dynamics tion, but there are two possible explan- that FoxD4L1/D5 promotes neural ations based on previous studies. FoxD4L1/D5 alters genes ectodermal fates and suppresses epi- First, many of these genes are involved in FGF signaling. dermal fates. In addition, three genes expressed early in the dorsal blasto- (Fullback, Xmc, XSpr2) regulated by pore lip region, as is FoxD4L1/D5; FGF signaling has been implicated in the FGF signaling pathway (Frazzetto perhaps FoxD4L1/D5 regulates genes promoting neural induction, regulat- et al., 2002; Ossipova et al., 2002; involved in setting up the dorsal axis, ing patterning of the midbrain, and Sasai et al., 2004; Chung et al., 2005) as previously suggested (Sullivan maintaining neural stem cell prolifer- were up-regulated by FoxD4L1/D5. et al., 2001). Second, while FoxD4L1/ ation (Umemori, 2009), as well as epi- Fullback and Xmc roles in neural and D5 does not induce Xbra expression in dermal and mesodermal formation. mesoderm formation were discussed ACs harvested at neural plate stages, Two members of the FGF pathway above. Although XSpr2 is required for it does induce muscle-specific actin in that are expressed in the neural ecto- gastrulation movements of the meso- ACs harvested at tail bud stages and derm were highly up-regulated by derm (Nutt et al 2001; Zhao et al., in ectopically induced dorsal axes in FoxD4L1/D5 (Table 3). FGF8-b,a 2003), it also is later expressed in the whole embryos (Sullivan et al., 2001). splice form of FGF8, mediates early brain (Ossipova et al., 2002; Fig. 2A); Because FoxD4L1/D5 is normally mesoderm and posterior neural tissue in which of these tissues its expression expressed in the mesoderm of the tail formation (Fletcher et al., 2006), and is influenced by FoxD4L1/D5 requires bud (So¨lter et al., 1999; Sullivan et al., FgfR2 is expressed in the midbrain- further study. One FGF-regulated 2001), these observations suggest that hindbrain region (Blak et al., 2005). gene, Elf-1, which encodes an ETS do- FoxD4L1/D5 may play a later role in Both share expression domains in the main transcription factor, was down- mesoderm formation in the tail bud neural ectoderm with FoxD4L1/D5 regulated by FoxD4L1/D5. This is that is reflected in the up-regulation (So¨lter et al., 1999; Fetka et al., 2000). surprising, because in mouse Elf-1 is of the mesoderm-associated genes One member of the FGF pathway, activated by FGF8 signaling in the reported herein. Perhaps FoxD4L1/D5 FgfR4-c, which causes ectodermal midbrain (Lee et al., 1997). The secondarily influences mesoderm gene cells to develop into epidermis (Umb- expression of Elf-1 has not been 3474 YAN ET AL.

port (Bodoy et al., 2005), one is similar to a Rapamycin-associated protein, which is a serine-threonine kinase involved in cell-cycle progression (Sabatini et al., 1994), and one is simi- lar to Claudin-5, an integral mem- brane protein associated with tight junctions (Sirotkin et al., 1997) (Table 3). Of the 5 down-regulated, validated genes, one remains unknown, one is a cytokeratin, which is an epidermis- enriched cytoskeletal protein, one is the same as XAG-1, which is a secreted protein specific to the cement gland (Sive et al., 1989), one is predicted to be a metabolic enzyme (sulfotransfer- ase), and one is similar to Villin1, which is an actin regulatory protein (Hesterberg and Weber, 1983) (Table 4). Using whole embryo RT-PCR, we determined the temporal pattern of gene expression (Fig. 3), and using whole embryo ISH, we determined the expression patterns of 11 of these vali- dated genes (Figs. 4 and 5); we do not report the expression of MGC52564 because XAG-1 expression is already characterized (Sive et al., 1989). In Supp. Figure S1, we present the expression pattern of the non-vali- dated gene BG161338 because it is a novel gene.

Uncharacterized genes that are up-regulated by FoxD4L1/D5 are Developmental Dynamics all expressed in the neural ectoderm. Transcripts for MGC132176, LOC Fig. 3. RT-PCR assays illustrate the temporal expression patterns of uncharacterized up-regu- lated (A) and down-regulated (B) genes in normal, unmanipulated embryos. RT-, minus RT step; 496224, BC097640, BJ043686, and numbers across the top indicate the developmental stages (st). H4 is an internal control. CB983184 were detected by RT-PCR at the 1-cell and blastula stages (Fig. reported in Xenopus. By RT-PCR, we Expression Patterns of 3A), consistent with the reported observed low levels of Elf-1 expression FoxD4L1/D5-Regulated Genes maternal expression of FoxD4L1/D5 at the 1-cell, blastula, and early gas- of Unknown Function (So¨lter et al., 1999; Fetka et al., 2000; trula stages, high levels at neural plate Sullivan et al., 2001). At gastrula through tail bud stages, and decreased A large number of the genes repre- stages (10.0–12.5), all 7 up-regulated, levels at larval stages (Fig. 3B). By ISH, sented on the Affymetrix Xenopus validated genes are expressed uni- we detected weak expression in the ani- laevis GeneChip (v1.0) have not been formly in the animal cap ectoderm mal cap ectoderm, the neural plate and annotated or fully sequenced, and (Figs. 3A, 4). Each is also detected at border zone, anterior neural tube, lens therefore are of unknown function the neural plate stages (13–16) (Fig. and branchial arch mesoderm (see Fig. (Fig. 1). We performed a BLAST search 3A). By ISH analyses, each is 5B). Only at gastrula and neural plate of the 12 genes of unknown function expressed throughout the neural plate stages does Elf-1 expression overlap that we validated to identify their pre- and the neural plate border zone of with that of FoxD4L1/D5.Intotal, dicted gene functions. Of the seven up- ectoderm that will give rise to cranial these results suggest novel roles for regulated, validated genes, 2 remain placodes, hatching gland, and neural FoxD4L1/D5 in promoting FGF signal- unknown, 2 have high similarity to crest (Fig. 4). Thus, in general, the ing in the neural ectoderm and perhaps metabolic enzymes (heme-oxygenase, expression domains of these genes the mesoderm, and in repressing FGF uridine-cytidine kinase), one is similar overlap with that of FoxD4L1/D5, signaling in the epidermis. to a solute carrier for membrane trans- which is maternally expressed in the FOXD5 DOWNSTREAM TARGETS 3475 Developmental Dynamics

Fig. 4. Whole mount in situ hybridization assays illustrating the expression patterns of 7 uncharacterized genes that are up-regulated by FoxD4L1/D5. Gastrula stages are side views oriented with animal cap (An) to the top and vegetal pole (Veg) to the bottom. Transcripts for all 7 genes are detected in the animal cap ectoderm. At neural plate stages, embryos are oriented in an anterior frontal view, with dorsal to the top. Transcripts for all 7 genes are detected in the neural plate (np) and in the ectoderm bordering the neural plate (arrow) that will give rise to the cra- nial placodes and neural crest. At neural tube stages, MGC132176 is expressed throughout the neural tube (nt), with intense staining in a band in the midbrain-hindbrain (hb) region, in adjacent neural crest (nc), and the dorsal rim of the cement gland (cg). Left embryo is a dorsal view with an- terior at the bottom and right embryo is a ventral view of the head. At larval stages (st32–38), MGC132176 is detected throughout the brain and retina (r), in the lens (L), otocyst (oto), cement gland, branchial arch (BA), and nephric mesoderm (ne). This and all larval stages are side views with anterior to the left. Transverse sections of st38 embryo confirm the labeled tissues identified in whole mount embryos, and identify the profundal ganglion (Pg). At neural tube stages, LOC496224 is expressed throughout the neural tube, retina, and somites (so). Side view with anterior to the left. At larval stages, LOC496224 is additionally expressed in the lens, otocyst, and branchial arches. BC097640 is expressed throughout the neu- ral tube, with strong expression in the pineal primordium (pin), cement gland, several cranial placodes (dlp, dorso-lateral placode; olf, olfactory pla- code; Vp, trigeminal placode), and scattered cells on the ventral midline (bracket) caudal to the cement gland. Left embryo is an anterior frontal view with dorsal to the top; right embryo is a ventral view with anterior to the top. At larval stages, BC097640 is additionally expressed in the lens, otocyst, branchial arches, head mesoderm (hm), heart (ht), ventral blood islands (bl), somites, and nephric mesoderm. At neural tube stages, BJ051730 is weakly expressed throughout the neural tube, with strong expression in the midbrain-hindbrain region and adjacent neural crest. Dor- sal anterior-view from the top. At larval stages, BJ051730 is additionally expressed in the lens, otocyst, branchial arches, somites, and nephric mesoderm. At neural tube stages, BJ043686 is expressed in the neural tube and the ectoderm bordering it (bz). Side view with anterior to the left. At larval stages, BJ043686 is additionally expressed at low levels in the lens, otocyst, somites, branchial arch, head, and nephric mesoderm. At neural tube stages, CB983184 is expressed in the neural tube and the ectoderm bordering it. At larval stages, CB983184 is additionally expressed in the olfactory pit, lens, retina, otocyst, somites, branchial arch mesoderm, and heart. At neural tube stages, MGC68716 is expressed in the neu- ral tube, the ectoderm bordering it, and pineal primordium. At larval stages, MGC68716 is additionally expressed in the otocyst, somites, branchial arch mesoderm, and heart. fb, forebrain; mb, midbrain. 3476 YAN ET AL. Developmental Dynamics

Fig. 5. Whole mount in situ hybridization assays illustrating the expression patterns of 5 genes that are down-regulated by FoxD4L1/D5. Abbrevi- ations are the same as in Figure 4. A: Down-regulated, validated genes of unknown function. Gastrula stages are side views oriented with animal pole to the top and vegetal pole to the bottom. Transcripts for all 4 genes are detected in the animal cap ectoderm. At neural plate stages, embryos are oriented in an anterior frontal view, with dorsal to the top. MGC81002 is weakly detected in the neural plate, the ectoderm bordering it, and epidermis (epi). At neural tube stages, it is detected diffusely in the neural tube, retina and epidermis. Side view. At larval stages, MGC81002 is additionally detected in the otocyst and branchial arch mesoderm. Side view and transverse sections. At neural plate stages, LOC443682 is strongly expressed in the epidermis, the ectoderm bordering the neural plate, and the anterior neural ridge (anr). At neural tube stages, LOC443682 is detected throughout the epidermis, with enhanced expression bordering the retina, hatching gland (hg), and cement gland. Left embryo is a dorso-anterior view with anterior to the bottom; top right embryo is a dorsal view of the head with anterior to the left, and bottom right embryo is a ventral view of the head with anterior to the top. At larval stages, LOC443682 is detected throughout the epidermis, most strongly on the dorsal side, and in the nephric mesoderm. Side view and transverse sections. At neural plate stages, MGC53193 is detected in the epidermis, border of the neural plate, and anterior neural ridge. At neural tube stages, MGC53193 is strongly expressed in the head epidermis and tissue surrounding the cloaca (cl). Left embryo is a frontal view with dorsal to the top and right embryo is a ventral view with posterior to the left. At larval stages, MGC53193 is weakly detected in the epidermis of whole mount embryos (side view), but transverse sections reveal additional weak expression in brain (b), lens, otocyst, and anterior pharynx (ph). At neural plate stages, MGC52940 is strongly expressed in the ectoderm bordering the neural plate and cement gland, and weakly in the epidermis. At neural tube stages, MGC52940 expression is pronounced in a dorsal midline stripe in the epidermis, surrounding the retinas, in a patch over the forebrain (between the retinas), and in the cement gland. Anterior-fron- tal view with dorsal to the top. At larval stages, MGC52940 is highly expressed in the olfactory placode and in cells dotted throughout the epider- mis; it is weakly expressed in the cement gland and anterior pharynx. Side view and transverse sections. B: Expression pattern of Xenopus Elf-1. Elf-1 is weakly expressed in the animal cap ectoderm at gastrulation, throughout the neural plate and border zone, and then is barely detectable in the anterior neural tube (*). At larval stages it is weakly detected in the brain, lens, and branchial arch mesoderm (side views and transverse sections). FOXD5 DOWNSTREAM TARGETS 3477

animal pole at cleavage and blastula mordium (Fig. 4). At late tail bud chial arch mesoderm, and the anterior stages and zygotically expressed in the through larval stages, MGC68716 is pharynx (Fig. 5A). At neural plate neural ectoderm. However, FoxD4L1/ additionally expressed in some placode stages, MGC52940 is strongly ex- D5 expression is not detected in the derivatives, somites, notochord, bran- pressed in the border zone surrounding neural plate border zone or the lateral chial arch, and cardiac mesoderm (Fig. the neural plate and cement gland, and neural plate, indicating that other fac- 4). In summary, while the maternal, is weakly expressed throughout the ep- tors also regulate these genes. blastula, gastrula, and neural plate idermis (Fig. 5A). At later stages, FoxD4L1/D5 expression wanes at expression patterns of these 7 genes MGC52940 is strongly expressed in the neural tube (20–28) and larval (st32– overlap with that of FoxD4L1/D5, olfactory placode and scattered cells 38) stages except at the midbrain- starting at neural plate stages their throughout the epidermis and is hindbrain boundary and in the tail expression domains are far more exten- weakly expressed in the anterior phar- bud (So¨lter et al., 1999; Fetka et al., sive than FoxD4L1/D5, which is only ynx (Fig. 5A). 2000 Sullivan et al., 2001). In contrast, maintained in the midbrain-hindbrain all 7 up-regulated, validated genes are region. This indicates that these genes Summary strongly expressed at most of these may be initially regulated by Fox stages (Fig. 3A), and spatially their D4L1/D5, but their later expression is The expression patterns of these expression domains are more exten- likely regulated by other factors. uncharacterized genes are consistent sive than those of FoxD4L1/D5. with previous studies showing that MGC132176 is expressed throughout FoxD4L1/D5 promotes neural plate the neural tube, with strong expres- Unknown genes that are down- genes and represses non-neural ecto- dermal genes (Yan et al., 2009). The sion in the midbrain-hindbrain region, regulated by FoxD4L1/D5 are expression domains of all of the up- in the associated neural crest, the dor- all expressed in non-neural sal edge of the cement gland, several regulated genes overlap with FoxD4L1/ ectoderm. placode derivatives, branchial arch, D5 expression at maternal, blastula, and nephric mesoderm (Fig. 4). Only LOC443682 expression was gastrula, and neural plate stages, but LOC496224 is expressed throughout detected by RT-PCR at the 1-cell and they are more extensive than that of the neural tube, some placode deriva- blastula stages (Fig. 3B). All 4 down- FoxD4L1/D5 at neural tube to larval tives, somites, and branchial arch regulated, validated genes are ex- stages. This indicates that only their mesoderm (Fig. 4). BC097640 is pressed at gastrula stages uniformly early phases of expression might be expressed in the neural tube with through the animal cap ectoderm regulated by FoxD4L1/D5. The down- enhanced staining in the pineal, (Figs. 3B, 5A; Table 4), overlapping regulated genes are highly expressed cement gland, several cranial placo- with FoxD4L1/D5 expression. RT- in the epidermis and non-neural ecto- des, heart, somites, branchial arch PCR analysis shows strong expression derm that surrounds the neural plate and nephric mesoderm, and ventral continuing through neural plate and border and gives rise to placodes and blood islands (Fig. 4). BJ051730 is neural tube stages (Fig. 3B), but at neural crest, suggesting that FoxD4L1/ expressed diffusely throughout the larval stages the expression of LOC D5 may restrict their expression from encroaching into the neural ectodermal Developmental Dynamics neural tube with enhanced expression 443682 wanes. in the midbrain-hindbrain region, in While temporally FoxD4L1/D5 domain. It is now important to func- the adjacent neural crest, several pla- expression overlaps initially with these tionally characterize these novel genes code derivatives, somites, branchial four genes, spatially it only overlaps to further elucidate the gene regulatory arch, and nephric mesoderm (Fig. 4). significantly at gastrula stages. At neu- network that controls early ectodermal At early stages, BJ043686 is weakly ral plate/tube stages, MGC81002 is fate decisions. In particular, we need to expressed throughout the neural tube weakly expressed in the dorsal epider- test which of these genes are directly and the border zone containing the mis, neural plate/tube, and border zone regulated by FoxD4L1/D5, and which cranial placodes and neural crest (Fig. (Fig. 5A). By late tail bud and larval ones are regulated by transcriptional 4). At late tail bud through larval stages, MGC81002 is additionally activation or repression. These new stages, BJ043686 expression is expressed in the otocyst and branchial FoxD4L1/D5 targets should be valuable detected in some placode derivatives, arch mesoderm (Fig. 5A). At neural in understanding the molecular me- somites, branchial arch, and nephric plate/tube stages, LOC443682 is chanisms of embryonic neural mesoderm (Fig. 4). At early stages, strongly expressed in the epidermis, development. CB983184 is expressed in the anterior border zone and anterior neural ridge neural tube and border zone containing (Fig. 5A). At later stages, LOC443682 EXPERIMENTAL the cranial placodes and neural crest is additionally expressed in the nephric PROCEDURES (Fig. 4). At late tail bud through larval mesoderm (Fig. 5A). At neural plate/ Embryos and Microinjections stages, CB983184 is expressed dif- tube stages, MGC53193 is expressed in fusely in the brain, retina, some pla- the epidermis, border zone, and ante- Fertilized Xenopus laevis eggs were code derivatives, somites, branchial rior neural ridge (Fig. 5A). At tail bud obtained by either in vitro fertilization arch, and cardiac mesoderm (Fig. 4). At stages, MGC53193 expression is nota- (for ACs) or gonadotropin-induced early stages, MGC68716 is weakly ble in tissue surrounding the cloaca; at natural mating of adult frogs (for tar- expressed in the anterior neural tube, larval stages, it is weakly expressed in geted blastomere microinjections) as with intense staining in the pineal pri- brain, some placode derivatives, bran- described elsewhere (Moody, 2000). 3478 YAN ET AL.

FoxD4L1/D5 and nuclear b-galactosi- These chips have been successfully trol gene (H4). To determine the devel- dase (nbgal) mRNAs were synthesized used for analyzing gene expression in opmental expression of unknown by in vitro transcription (Ambion, Aus- the Organizer, endoderm, neural FoxD4L1/D5 downstream genes, wild tin, TX; mMessage mMachine kit). For crest, and regenerating hind limb type embryos were snap-frozen at a se- ACs, FoxD4L1/D5 mRNA (150 pg) was (Grow et al., 2006; Hufton et al., 2006; ries of developmental stages. Total injected into the animal poles of both Sinner et al., 2006; Pearl et al., 2008; RNAs were extracted and reverse-tran- cells at the 2-cell stage. ACs were dis- Zhao et al., 2008). The chips were scribed as above. Standard PCR ampli- sected at stages 8.5– 9, cultured in 1 washed and scanned as recommended fications (primers in Tables 1, 2) were MAB, and collected at stages 11.5–12.5. by the Ovation Biotin RNA Amplifica- performed to show the expression lev- For whole embryo in situ hybridization tion and Labeling System User Guide els of each gene, with H4 as a loading (ISH) assays, one blastomere of the 16- (version 1.0). GCOS software was used control. These analyses were repeated cell embryo having a defined neural or to determine signal intensities and three times with independent samples. epidermal lineage (Moody, 1987) was detection calls for each gene. The repli- injected with a mixture of FoxD4L1/ cate correlation value for the four con- Whole Mount In Situ D5 (150 pg) and nbgal (100 pg) mRNAs. trol samples was 0.94 and for the Hybridization Embryos were cultured in 50% Stein- FoxD4L1/D5-injected samples it was berg’s solution and collected at stages 0.95, indicating consistency between Plasmids encoding a subset of the pu- 12–14. replicates despite random genetic var- tative downstream genes identified in iance between parental frogs. Using the microarray assay were either GeneSpring software, all experimen- obtained from the laboratory in which Microarray Analyses tal arrays were normalized to their they were originally reported (Tables Four independent samples of matched control array. Genes were 3, 4), or purchased from Open Biosys- FoxD4L1/D5 mRNA-injected ACs then filtered for at least 2 present calls tems (Thermo Scientific, Waltham, were collected. Each sample was out of 8 calls. The remaining list was MA). Anti-sense digoxigenin-labeled derived from one different male and tested for significant changes by RNA probes were synthesized by two different females and contained ANOVA (P < 0.05). The raw and proc- in vitro transcription (Ambion, Mega- 100 ACs. For each FoxD4L1/D5- essed data are available (GEO acces- script kit). FoxD4L1/D5 mRNA-in- expressing sample, a control, unin- sion number GSE11143). jected embryos were fixed, stained for jected sample from sibling embryos expression of the nbGal lineage tracer, also was collected. All samples were and processed for whole mount ISH RT-PCR processed in parallel for cDNA label- according to standard protocols (Sive ing and chip hybridization to reduce Animal poles of 2-cell embryos were et al., 2000). Embryos were analyzed for inter-sample variations. Total RNAs injected with FoxD4L1/D5 mRNA and whether the expression domain of the were extracted from ACs with the ACs collected as described for the putative downstream gene was RNeasy mini kit (Qiagen, Chatsworth, microarray analyses. Total RNA was expanded or decreased in size or stain- CA). Integrity of RNAs was assessed extracted the with RNeasy mini kit ing intensity within the clone of Fox

Developmental Dynamics using the Agilent 2100 bioanalyzer (Qiagen). Semi-quantitative RT-PCR D4L1/D5-expressing cells. The unin- (Agilent Technologies, Santa Clara, within linear ranges was performed as jectedsideofthesameembryowasused CA) and only samples with an integ- previously described (Yan et al., 2009). as an internal control. To analyze the rity number >8.0 were used. Total Primers are listed in Tables 1 and 2; gene expression patterns of putative RNAs were labeled and fragmented some primer sequences were obtained downstream genes with unknown func- with the Ovation Biotin RNA Amplifi- from published studies, whereas the tion, uninjected wild type embryos were cation and Labeling System (NuGEN others were designed with the Primer3 fixed at gastrulation (stages 10.0–12.5), Technologies, Inc., San Carlos, CA). software (Rozen and Skaletsky, 2000). neural plate (stages 13–16), early neural Briefly, 50 ng total RNA was used for Reverse transcription was performed tube (stages 20–24), tail bud (stages 25– first- and second-strand cDNA synthe- with 1.0 mg of RNA using the Super- 28), and larval (stages 32–38) stages ses. The synthesized cDNAs were Script first-strand synthesis system and processed for ISH as above. amplified, purified with a PCR purifi- (Invitrogen, Carlsbad, CA). Standard cation kit (Qiagen), then labeled and PCR amplifications were performed fragmented following the Ovation kit with PCR Supermix (Invitrogen) ACKNOWLEDGMENTS instructions. The labeled cDNAs were including 1.0 mCi a-[32P]dATP (Amer- We thank our many colleagues in the purified with the Dye Ex kit (Qiagen). sham, Pittsburgh, PA) in the reaction Xenopus community who supplied Chip hybridization and statistical mixture. All PCR assays were ISH clones for several of the genes analyses were performed by the repeated at least 3 times. Bands were investigated herein. NINDS-NIMH Microarray Consor- visualized with a Storm 860 phos- tium at The Translational Genomics phorimager (Molecular Dynamics, REFERENCES Research Institute (T-GEN, Tempe, Sunnyvale, CA), the band intensities AZ). The Affymetrix (Santa Clara, CA) were measured with the ImageQuant Bertwistle D, Walmsley ME, Read EM, Pizzey JA, Patient RK. 1996. GATA fac- VR GeneChip Xenopus laevis Genome analysis program and the relative tors and the origins of adult and embry- Array (v1.0) bears 15,503 probe sets expression levels of the genes were cal- onic blood in Xenopus: responses to representing about 14,400 transcripts. culated as compared to an internal con- retinoic acid. Mech Dev 57:199–214. FOXD5 DOWNSTREAM TARGETS 3479

Blak AA, Naserke T, Weisenhorn DM, Fuentealba LC, Eivers E, Ikeda A, Hur- Zic genes. Proc Natl Acad Sci USA 106: Prakash N, Partanen J, Wurst W. 2005. tado C, Kuroda H, Pera EM, De Rob- 17437–17442. Expression of Fgf receptors 1, 2, and 3 ertis EM. 2007. Integrating patterning Matsuo-Takasaki M, Matsumura M, Sasai in the developing mid- and hindbrain of signals: Wnt/GSK3 regulates the dura- Y. 2005. An essential role of Xenopus the mouse. Dev Dyn 233:1023–1030. tion of the BMP/Smad1 signal. Cell 131: Foxi1a for ventral specification of the Bodoy S, Martin L, Zorzano A, Palacin M, 980–993. cephalic ectoderm during gastrulation. Estevez R, Bertran J. 2005. Identifica- Gamse JT, Sive H. 2001. Early anteropos- Development 132:3885–3894. tion of LAT4, a novel amino acid trans- terior division of the presumptive neu- Molenaar M, Roose J, Peterson J, Venanzi porter with system L activity. J Biol rectoderm in Xenopus. Mech Dev 104: S, Clevers H, Destre´e O. 1998. Differen- Chem 280:12002–12011. 21–36. tial expression of the HMG box tran- Bromley E, Knapp D, Wardle FC, Sun BI, Golub R, Adelman Z, Clementi J, Weiss R, scription factors XTcf-3 and XLef-1 du- Collins-Racie L, LaVallie E, Smith JC, Bonasera J, Servetnick M. 2000. Evolu- ring early Xenopus development. Mech Sive HL. 2004. Identification and char- tionarily conserved and divergent ex- Dev 75:151–154. acterisation of the posteriorly-expressed pression of members of the FGF receptor Moody SA. 1987. Fates of the blastomeres Xenopus neurotrophin receptor homolog family among vertebrate embryos, as of the 16-cell stage Xenopus embryo. genes fullback and fullback-like. Gene revealed by FGFR expression patterns in Dev Biol 119:560–578. Expr Patterns 5:135–140. Xenopus. Dev Genes Evol 210:345–357. Moody SA. 2000. Cell lineage analysis in Christian JL, McMahon JA, McMahon Grow M, Neff AW, Mescher AL, King MW. Xenopus embryos. Methods Mol Biol 135: AP, Moon RT. 1991. Xwnt-8, a Xenopus 2006. Global analysis of gene expression 331–347. Wnt-1/int-1-related gene responsive to in Xenopus hind limbs during stage-de- Nishimatsu S, Thomsen GH. 1998. Ven- mesoderm-inducing growth factors, may pendent complete and incomplete regen- tral mesoderm induction and patterning play a role in ventral mesodermal pat- eration. Dev Dyn 235:2667–2685. by bone morphogenetic protein hetero- terning during embryogenesis. Develop- Hawley SH, Wu¨ nnenberg-Stapleton K, dimers in Xenopus embryos. Mech Dev ment 111:1045–1055. Hashimoto C, Laurent MN, Watabe T, 74:75–88. Chung HA, Hyodo-Miura J, Nagamune T, Blumberg BW, Cho KW. 1995. Disrup- Nutt SL, Dingwell KS, Holt CE, Amaya E. Ueno N. 2005. FGF signal regulates tion of BMP signals in embryonic Xeno- 2001. Xenopus Sprouty2 inhibits FGF- gastrulation cell movements and mor- pus ectoderm leads to direct neural mediated gastrulation movements but phology through its target NRH. Dev induction. Genes Dev 9:2923–2935. does not affect mesoderm induction and Biol 282:95–110. Heeg-Truesdell E, LaBonne C. 2006. Neu- patterning. Genes Dev 15:1152–1166. Dalgin G, Goldman DC, Donley N, Ahmed ral induction in Xenopus requires inhi- Ossipova O, Stick R, Pieler T. 2002. R, Eide CA, Christian JL. 2007. GATA- bition of Wnt-beta-catenin signaling. XSPR-1 and XSPR-2, novel Sp1 related 2 functions downstream of BMPs and Dev Biol 298:71–86. zinc finger containing genes, are CaM KIV in ectodermal cells during Hesterberg LK, Weber K. 1983. Demon- dynamically expressed during Xenopus primitive hematopoiesis. Dev Biol 310: stration of three distinct calcium-bind- embryogenesis. Mech Dev 115:117–122. 454–469. ing sites in villin, a modulator of actin Panitz F, Krain B, Hollemann T, Nord- Delaune E, Lemaire P, Kodjabachian L. assembly. J Biol Chem 258:365–369. heim A, Pieler T. 1998. The Spemann 2005. Neural induction in Xenopus Horb ME, Thomsen GH. 1997. A vegetally organizer-expressed zinc finger gene requires early FGF signalling in addi- localized T-box transcription factor in Xegr-1 responds to the MAP kinase/Ets- tion to BMP inhibition. Development Xenopus eggs specifies mesoderm and SRF signal transduction pathway. 132:299–310. endoderm and is essential for embry- EMBO J 17:4414–4425. de Souza FS, Gawantka V, Go´mez AP, onic mesoderm formation. Development Papalopulu N, Kintner C. 1993. Xenopus Delius H, Ang SL, Niehrs C. 1999. The 124:1689–1698. Distal-less related genes are zinc finger gene Xblimp1 controls ante- Hufton AL, Vinayagam A, Suhai S, Baker expressed in the developing forebrain rior endomesodermal cell fate in Spe- JC. 2006. Genomic analysis of Xenopus and are induced by planar signals. De-

Developmental Dynamics mann’s organizer. EMBO J 18:6062– organizer function. BMC Dev Biol 6:27. velopment 117:961–975. 6072. Itoh K, Sokol SY. 2007. Early develop- Pearl EJ, Barker D, Day RC, Beck CW. Fainsod A, Steinbeisser H, De Robertis ment of epidermis and neural tissue. 2008. Identification of genes associated EM. 1994. On the function of BMP-4 in In: Moody SA, editor. Principles of de- with regenerative success of Xenopus patterning the marginal zone of the velopmental genetics. San Diego: Aca- laevis hindlimbs. BMC Dev Biol 8:66. Xenopus embryo. EMBO J. 13:5015– demic Press. p 241–258. Rogers CD, Moody SA, Casey ES. 2009. 5025. Kuroda H, Fuentealba L, Ikeda A, Rever- Neural induction and factors that stabi- Ferreiro B, Skoglund P, Bailey A, Dorsky sade B, De Robertis EM. 2005. Default lize a neural fate. Birth Defects Res C R, Harris WA. 1993. XASH1, a Xenopus neural induction: neuralization of disso- Embryo Today 87:249–262. homolog of achaete-scute: a proneural ciated Xenopus cells is mediated by Rossi CC, Hernandez-Lagunas L, Zhang gene in anterior regions of the verte- Ras/MAPK activation. Genes Dev 19: C, Choi IF, Kwok L, Klymkowsky M, brate CNS. Mech Dev 40:25–36. 1022–1027. Artinger KB. 2008. Rohon-Beard sen- Fetka I, Doederlein G, Bouwmeester T. Lee SM, Danielian PS, Fritzsch B, McMa- sory neurons are induced by BMP4 2000. Neuroectodermal specification and hon AP. 1997. Evidence that FGF8 sig- expressing non-neural ectoderm in Xen- regionalization of the Spemann orga- nalling from the midbrain-hindbrain opus laevis. Dev Biol 314:351–361. nizer in Xenopus. Mech Dev 93:49–58. junction regulates growth and polarity Rozen S, Skaletsky H. 2000. Primer3 on Fletcher RB, Baker JC, Harland RM. in the developing midbrain. Develop- the WWW for general users and for bi- 2006. FGF8 spliceforms mediate early ment 124:959–969. ologist programmers. Methods Mol Biol mesoderm and posterior neural tissue Luo T, Matsuo-Takasaki M, Lim JH, Sar- 132:365–386. formation in Xenopus. Development gent TD. 2001. Differential regulation Sabatini DM, Erdjument-Bromage H, Lui 133:1703–1714. of Dlx gene expression by a BMP mor- M, Tempst P, Snyder SH. 1994. RAFT1: Frazzetto G, Klingbeil P, Bouwmeester T. phogenetic gradient. Int J Dev Biol 45: a mammalian protein that binds to 2002. Xenopus marginal coil (Xmc), a 681–684. FKBP12 in a rapamycin-dependent novel FGF inducible cytosolic coiled-coil Luo T, Lee YH, Saint-Jeannet JP, Sargent fashion and is homologous to yeast protein regulating gastrulation move- TD. 2003. Induction of neural crest in Xen- TORs. Cell 78:35–43. ments. Mech Dev 113:3–14. opus by transcription factor AP2alpha. Salic AN, Kroll KL, Evans LM, Kirschner Friesel R, Brown SA. 1992. Spatially re- Proc Natl Acad Sci USA 100:532–537. MW. 1997. Sizzled: a secreted Xwnt8 stricted expression of fibroblast growth Marchal L, Luxardi G, Thome´ V, Kodjaba- antagonist expressed in the ventral factor receptor-2 during Xenopus devel- chian L. 2009. BMP inhibition initiates marginal zone of Xenopus embryos. De- opment. Development 116:1051–1058. neural induction via FGF signaling and velopment 124:4739–4748. 3480 YAN ET AL.

Sasai N, Nakazawa Y, Haraguchi T, Sasai derm via transcriptional repression Wanner SJ, Danos MC, Lohr JL, Miller Y. 2004. The neurotrophin-receptor- that is dependent on the C-terminal do- JR. 2005. Molecular cloning and expres- related protein NRH1 is essential for main. Dev Biol 232:439–457. sion of Ena/Vasp-like (Evl) during Xeno- convergent extension movements. Na- Sun BI, Bush SM, Collins-Racie LA, LaV- pus development. Gene Expr Patterns ture Cell Biol 6:741–748. allie ER, DiBlasio-Smith EA, Wolfman 5:423–428. Sato SM, Sargent TD. 1991. Localized NM, McCoy JM, Sive HL. 1999. der- White RJ, Sun BI, Sive HL, Smith JC. 2002. and inducible expression of Xenopus- rie`re: a TGF-beta family member Direct and indirect regulation of derrie`re, posterior (Xpo), a novel gene active in required for posterior development in a Xenopus mesoderm-inducing factor, by early frog embryos, encoding a protein Xenopus. Development 126:1467–1482. VegT. Development 129:4867–4876. with a ‘CCHC’ finger domain. Develop- Takeda M, Kurauchi T, Yamazaki T, Winning RS, Shea LJ, Marcus SJ, Sar- ment 112:747–753. Izutsu Y, Mae´no M. 2005. Neptune is gent TD. 1991. Developmental regula- Sinner D, Kirilenko P, Rankin S, Wei E, involved in posterior axis and tail for- tion of transcription factor AP-2 during Howard L, Kofron M, Heasman J, mation in Xenopus embryogenesis. Dev Xenopus laevis embryogenesis. Nucleic Woodland HR, Zorn AM. 2006. Global Dyn 234:63–73. Acids Res 19:3709–3714. analysis of the transcriptional network Takeuchi M, Nakabayashi J, Sakaguchi T, Yan B, Neilson KM, Moody SA. 2009. controlling Xenopus endoderm forma- Yamamoto TS, Takahashi H, Takeda H, FoxD5 plays a critical upstream role in tion. Development 133:1955–1966. Ueno N. 2003. The prickle-related gene regulating neural ectodermal fate and Sirotkin H, Morrow B, Saint-Jore B, in vertebrates is essential for gastrula- the onset of neural differentiation. Dev Puech A, Das Gupta R, Patanjali SR, tion cell movements. Curr Biol 13: Biol 329:80–95. Skoultchi A, Weissman SM, Kucherla- 674–679. Zhang J, King ML. 1996. Xenopus VegT pati R. 1997. Identification, characteri- Toyama R, Curtiss PE, Otani H, Kimura RNA is localized to the vegetal cortex zation, and precise mapping of a M, Dawid IB, Taira M. 1995. The LIM during oogenesis and encodes a novel T- human gene encoding a novel mem- class homeobox gene lim5: implied role box transcription factor involved in brane-spanning protein from the 22q11 in CNS patterning in Xenopus and mesodermal patterning. Development region deleted in velo-cardio-facial syn- zebrafish. Dev Biol 170:583–593. 122:4119–4129. drome. Genomics 42:245–251. Umbhauer M, Penzo-Me´ndez A, Clavilier Zhang J, Houston DW, King ML, Payne Sive HL, Hattori K, Weintraub H. 1989. L, Boucaut J, Riou J. 2000. Signaling C, Wylie C, Heasman J. 1998. The role Progressive determination during for- specificities of fibroblast growth factor of maternal VegT in establishing the mation of the anteroposterior axis in receptors in early Xenopus embryo. J primary germ layers in Xenopus Xenopus laevis. Cell 58:171–180. Cell Sci 113:2865–2875. embryos. Cell 94:515–524. Sive HL, Grainger RM, Harland RM. 2000. Umemori H. 2009. Weaving the neuronal Zhao H, Tanegashima K, Ro H, Dawid IB. Early development of Xenopus laevis,a net with target-derived fibroblast 2008. Lrig3 regulates neural crest for- laboratory manual. Cold Spring Harbor, growth factors. Dev Growth Differ 51: mation in Xenopus by modulating Fgf NY: Cold Spring Harbor Laboratory Press. 263–270. and Wnt signaling pathways. Develop- So¨lter M, Ko¨ster M, Hollemann T, Brey Veenstra GJ, Beumer TL, Peterson-Maduro ment 135:1283–1293. A, Pieler T, Kno¨chel W. 1999. Charac- J, Stegeman BI, Karg HA, van der Vliet Zhao J, Cao Y, Zhao C, Postlethwait J, terization of a subfamily of related PC, Destre´e OH. 1995. Dynamic and dif- Meng A. 2003. An SP1-like transcrip- winged helix genes, XFD-12/120/1200 ferential Oct-1 expression during early tion factor Spr2 acts downstream of Fgf (XFLIP), during Xenopus embryogene- Xenopus embryogenesis: persistence of signaling to mediate mesoderm induc- sis. Mech Dev 89:161–165. Oct-1 protein following down-regulation tion. EMBO J. 22:6078–6088. Stern CD. 2005. Neural induction: old of the RNA. Mech Dev 50:103–117. Zon LI, Mather C, Burgess S, Bolce ME, problem, new findings, yet more ques- Wallingford JB, Goto T, Keller R, Harland Harland RM, Orkin SH. 1991. Expres- tions. Development 132:2007–2021. RM. 2002. Cloning and expression of sion of GATA-binding proteins during Sullivan SA, Akers L, Moody SA. 2001. Xenopus Prickle, an orthologue of a embryonic development in Xenopus lae-

Developmental Dynamics foxD5a, a Xenopus winged helix gene, Drosophila planar cell polarity gene. vis. Proc Natl Acad Sci USA 88: maintains an immature neural ecto- Mech Dev 116:183–186. 10642–10646.