Abstract

Intrinsic regulatory factors play critical roles in early cortical patterning, including the development of the anteroposterior (A-P) axis. To identify genes that are differentially expressed along the A-P axis of the developing cerebral cortex, we analyzed gene expression in presumptive frontal, parietal, and occipital cerebral walls of E12.5 mouse using complementary DNA microarrays. We identified 106 genes, including expressed sequence tags (ESTs), expressed in an A-P gradient in the embryonic brain and screened 88 by in situ hybridization for confirmation. Central nervous system (CNS) expression patterns of many of these genes were previously unknown. Others, such as Sfrp1, CoupTF1, and FABP7, were expressed in a manner consistent with previous studies, providing independent confirmation. Two related transcription factors, previously not implicated in CNS development, Fhl1 and Fhl2, were observed to be enriched in posterior and anterior telencephalon, respectively. We studied patterning gradients in Fhl1 knockout mice but observed no changes in gene expression related to A-P regionalization in the Fhl1 knockout mice. These data provide an important set of new candidates for studies of cortical patterning and maturation.

Introduction

There has been significant progress in our understanding of the processes of regional patterning in the cerebral cortex during embryogenesis. The now classic and enduring model for cortical regionalization is the “protomap” model (Rakic 1988) in which the cells derived from neural progenitors have distinct intrinsic identity that drives regionalization. A growing body of data demonstrates that intrinsic gradients of homeobox transcription factors, such as Emx2 and Pax6, and other signaling molecules play a critical role in cortical regionalization (Miyashita-Lin et al. 1999; Bishop et al. 2000, 2002; Mallamaci et al. 2000; Muzio et al. 2002a,b; Muzio and Mallamaci 2003; Sansom et al. 2005). Otx1 and Otx2 provide further examples of homeobox transcription factors with restricted expression patterns that are involved in proper cortical organization and development (Acampora, Avantaggiato, et al. 1999; Acampora, Barone, et al. 1999). The identification of genes, many of which are transcription factors, with distinct temporal and spatial gradients in the developing telencephalon show that there are intrinsic mechanisms controlling early cortical regionalization and that the pattern of gene expression is functionally relevant (Levitt et al. 1997; Miyashita-Lin et al. 1999).

To further define other factors that show spatial gradients during the period of cortical regionalization, we used a genomic screening approach based on custom complementary DNA (cDNA) microarrays to assess gene expression gradients during corticogenesis. We chose to study gene expression at E12.5, a time during which time intrinsic signals are known to direct early telencephalic patterning (Barbe and Levitt 1991; Arimatsu et al. 1992; De Carlos and O'Leary 1992; Cohen-Tannoudji et al. 1994). We identified 106 genes with consistent and significant (P < 0.01) anteroposterior (A-P) gene gradients. We analyzed the majority of identified gene expression patterns (n = 88) using in situ hybridization and reverse transcription–polymerase chain reaction (RT-PCR) and confirmed a gradient of telencephalic expression for a subset of genes. We also investigated the role of one of these genes, Fhl1, a transcription factor without a previous known role in cortical development, in vivo (Morgan et al. 1995; Morgan and Madgwick 1996). We were unable to demonstrate a clear anterior–posterior patterning phenotype in Fhl1 knockouts, suggesting either redundancy with other close family members or a role in later neuronal maturation. This gene and other transcription factors, or signaling molecules identified here clearly provide an important group of genes for future study of cortical development.

Materials and Methods

Dissections and RNA Preparation

Anterior, middle, and posterior regions of the dorsal pallium, the forerunners of frontal parietal and occipital cortical areas, were dissected (A, M, P) from E12.5 C57Bl6J mice from Jackson Laboratory. Anatomical limits were chosen to increase reproducible dissection of samples. The cut between the anterior and the middle regions was made about 1–2 mm behind the middle of the ganglionic eminence because there are no surface landmarks. The posterior section is a tissue slice of approximately 2 mm above the rhinal sulcus identifiable at E12.5. Four litters were dissected resulting in 4 sets of pooled, independent replicate samples. The microdissected regions were flash frozen on dry ice and stored at −80 °C. Total RNA was extracted from each of the pooled samples using TRIzol reagent (GibcoBRL, Carlsbad, CA) according to the manufacturer's recommendations. RNA 6000 Nano Assay (Agilent Technologies, Palo Alto, CA) and Agilent 2100 Bioanalyzer software were used to analyze and quantify the RNA samples. Only high-quality RNA samples were used, for example 28S:18S ratio > 1.6 and A260/280 ∼2.0 (Ultrospec 2000, Pharmacia, Piscataway, NJ).

Fhl1 Knockout Animals

Fhl1 knockouts had been generated on a black Swiss background by inserting a LacZ sequence into the Fhl1 locus (Chu, Ruiz-Lozano, et al. 2000). Because Fhl1 is located on the X chromosome, heterozygous females were mated to wild-type males to generate Fhl1 knockout males and wild-type littermates. Genotyping of the animals was performed by using 2 sets of primers for wild-type and the knockout, for which the primers incorporate a portion of the LacZ gene.

Microarrays

Transcripts were analyzed using a mouse 10 000-element custom cDNA microarray printed at University of California at Los Angeles (UCLA) in collaboration with the microarray core (http://www.genetics.ucla.edu/microarray/) containing 7604 known genes and 1383 ESTs. This array is based on an Incyte cDNA library, which has highly reproducible and confirmable gene expression (Karsten et al. 2003). Four independent experiments were carried out using the pooled samples (A, M, and P) to compare A versus M and A versus P (Fig. 1). For each comparison 2 hybridizations were made with the dyes reversed in order to account for dye incorporation effects. A total of 16 array hybridizations were performed. Labeling and detection of cDNA were performed according to the tyramide signal amplification protocol (NEN, PerkinElmer) with minor modifications (Karsten et al. 2002). cDNA probes synthesized from 2 μg of total RNA of a pooled sample were independently labeled with either Cy3 or Cy5 fluorophore for each slide.

Figure 1.

Flowchart schema of the microarray experiment. The schematic represents the steps for an experiment. Dorsal dissections of A, M, and P from each litter were combined to obtain pooled RNA samples, which were consequently used to for comparative hybridizations of A versus M and A versus P on cDNA microarrays. Each comparison was run in duplicate with a dye flip, resulting in a total of 4 slides per litter. The microarray experiment presented used 4 litters for a total of 16 slides.

Images were analyzed using ImaGene 4.2 (Biodiscovery, Marina del Rey). Quantified signal intensity data were downloaded into Genesight 3.0 (Biodiscovery) and analyzed as described below. Poor quality spots flagged in ImaGene were omitted, and local background correction was performed. Signals with values below 20 were given a low positive value (=20). The ratios of signal intensities were log transformed for each hybridization, and normalized ratios were combined for each dye-flip experimental pair following global normalization. Genes differentially regulated in 2 conditions throughout were identified based on passing a statistical threshold of the 99.5% confidence interval, a stringent criteria (Genesight 3.0). The resulting gene list was narrowed (Karsten et al. 2003) by thresholding those genes with a ratio of at least 1.5 in at least 3 out of the 4 replicate experiments for either A versus M or A versus P. For genes showing similar trends in both comparisons, the criterion was such that at least 4 out of the total of 8 experiments had a ratio of 1.5 or above.

Tissue and Slide Preparation

Tissue samples were flash frozen upon dissection and stored at −80 °C until use. Human tissue was obtained from the National Institutes of Health (NIH) Maryland Brain and Tissue Bank under a protocol approved by the UCLA Institutional Review Board. Cryostat sections of 20 μm were cut and thaw-mounted onto Fisher Superfrost Plus slides. For in situ hybridization, sections were air dried, fixed in 4% paraformaldehyde (PFA) in 0.1 M phosphate buffer (PB) for 20 min, washed in PB for 5 min., rinsed in H2O, dried at room temperature (RT), then stored at −80 °C until use. Slides for immunohistochemistry and for X-gal staining were air dried and stored at −80 °C until use without treatment.

In Situ Hybridization

Tissues fixed on slides were washed/incubated in the following series of solutions at RT: 1) 0.1 M glycine in PB twice for 3 min each, 2) PB for 15 min, 3) 0.25% acetic anhydride, 1.3% tris-acetate buffer in PB for 10 min, 4) 2× sodium chloride-sodium citrate (SSC) twice for 15 min each, 5) 50, 70, 95, and 100% ethanol for 1 min each, 6) chloroform for 5 min, 7) 100 and 95% ethanol for 1 min each. Probes were diluted in hybridization solution to 106 counts/mL. Hybridized slides were coverslipped, sealed with DPX mountant (Gallard Schlesinger, Plainview, NY), and incubated overnight at 60 °C. Following hybridization, slides were washed in the following series of solutions: 1) 4× SSC twice for 20 min each at 60 °C, 2) 30 μg/mL RNase A in RNase buffer for 30 min at 45 °C, 3) 2× SSC 4 times for 20 min at RT, 4), 0.5× SSC twice for 20 min at 60 °C, 5) 0.1× SSC for 20 min at 60 °C, 6) 0.1× SSC for 20 min at RT. Slides were briefly rinsed in H2O, and then air dried. Hybridized slides were opposed to film prior to dipping in Kodak NTB2 emulsion. Following exposure to emulsion at 4 °C the slides were developed, stained with Cresyl violet or hematoxylin and eosin, coverslipped with Permount (Fisher, Houston, TX), and photographed in bright and dark field optics (SPOT).

Probe Synthesis

Inserts from clones spotted on the microarrays were used as templates to generate strand-specific 35S-labeled cRNA probes for in situ hybridization. Bacteria from glycerol stocks were grown in LB + ampicillin media, and the vectors containing the inserts were purified using Miniprep (Qiagen, Valencia, CA), which were subsequently digested with appropriate enzymes to produce linearized plasmids.

Fhl1Bm-specific probe template was generated by PCR, using primers up- and downstream of the alternately spliced region that contained fewer shared sequences with the other isoforms. The primer pair used for Fhl1Bm was 5′-TGTGGCCAAGAAGTGTGCT-3′ and 5′-AGTAGTCGTGCCAGGATTGTC-3′. The PCR product shares about 50 bp on either end of its sequence with AA047966, with about 200 bp specific to Fhl1Bm isoform, which should minimize binding to the other isoforms. The PCR product was gel purified and cloned into a vector (Invitrogen, Carlsbad, CA), and was subsequently used as a template to generate strand-specific 35S- and 32P-labeled cRNA probes for in situ hybridization and Northern blots, respectively.

Northern Blots

Total RNA was extracted from E14 embryos, E17 head, and brains from P0, P7, P14 and P21, using TRIzol reagent (GibcoBRL) according to the manufacturers recommendations. From each sample 10 μg of total RNA was run on a 1.5% agarose/formaldehyde gel in N-(2-hydroxyethyl)piperazine-N′-(2-ethanesulfonic acid)/ethylenediaminetetraacetic acid buffer. Transfer of electrophoresed RNA was carried out onto Nytran SPC (Schleicher & Schuell, Florham Park, NJ) overnight via capillary transfer. Single-stranded DNA probe for the Fhl1 clones was obtained by restriction digestion from the plasmid vector and purified through columns (Qiagen). The probes were labeled with 32P-deoxy-cytidine triphosphate using strand-specific StripEZ RNA Kit (Ambion). The membrane was hybridized with Ultrahyb (Ambion) containing 106 cpm/mL of probe overnight and washed as previously described (Geschwind et al. 2001). The blots were opposed to Kodak AR film against an intensifying screen.

Semiquantitative RT-PCR

Primers for Fhl1 (Taniguchi et al. 1998) were used to test the differential expression by semiquantitative RT-PCR from the pooled RNA samples from A, M, and P. cDNA was reverse transcribed from pooled RNA samples (Promega, Madison, WI), and semiquantitative expression analysis was performed by incorporating SYBR Green I dye into the RT-PCR product. Primers for mouse ribosomal RNA 18S (5′-GTAACCCGTTGAACCCCATT-3′ and 5′-CCATCCAATCGGTAGTAGCG-3′) were used in conjunction for normalization. Typhoon 9410 (Amersham Biosciences, Piscataway, NJ) was used to scan and quantitate the RT-PCR products. For each pooled region of A, M, and P, 500 ng of RNA was used with random primers for cDNA synthesis (Promega). A total of 18 PCR reactions were performed with 3 different pools of RNA. Reactions of 20 μL containing 2 μL of cDNA, 0.2 mM dNTP, 0.2 μM Fhl1 primers (each), 0.0375 μM control primers for ribosomal RNA (each), and 0.5 U Taq polymerase, were run from 21 to 26 cycles. The program used was 1) 2 min at 94 °C, 2) 40 s at 94 °C, 3) 30 s at 61 °C, 4) 1 min at 72 °C, 5) repeat (2)–(4) 20–25 times, (4) 5 min at 72 °C. The products (10–16 μL) were run on agarose gels, and incubated in 1:10 000 dilution of SYBR Green in H2O for 30 min. The gel was subsequently scanned and quantified using the Typhoon 9410 (Amersham).

LacZ Staining

Staining for β-Gal activity was performed as described in Schmidt et al. (1998). Briefly, tissue on slides were fixed in 0.2% glutaraldehyde for 2 min then washed twice for 5 min. each in phosphate-buffered saline (PBS) at RT. Slides were incubated in freshly made staining solution overnight at RT. Subsequent to a short rinse in H2O, slides were counterstained with eosin and mounted with Permount (Fisher).

Immunohistochemistry

Sections on slides were washed and incubated in the following series of solutions at RT unless otherwise stated: 1) 4% PFA for 20 min, 2) PBS 3 times for 5 min each, 3) 0.5% TritonX-100 in PBS for 1 h, 4) PBS 3 times for 5 min each, 5) 50 mM NH4Cl in PBS for 20 min, 6) PBS 3 times for 5 min each, 7) 5% goat serum in 0.01% TritonX-100 in PBS for 1 h, 8) primary antibody incubation in the blocking solution at dilutions ranging from 1:5000 to 1:1000 overnight in 4 °C, 9) PBS 4 times for 20 min each, 10) secondary antibody incubation in blocking solution 1:5000 dilution for 1 h, 11) PBS 4 times for 20 min each. Slides were mounted with Aqua Poly Mount (Polysciences, Warrington, PA). Images were scanned using Leica confocal microscope.

Results

Microarray Expression Results

E12.5 mice were chosen for the microarray analysis because it is relatively early cortical neurogenesis and corresponds to the stage of early intrinsic regionalization and specification of cortical regions (Barbe and Levitt 1991, 1992, 1995; McConnell and Kaznowski 1991; Muzio et al. 2002b; Muzio and Mallamaci 2003). The dissections were performed by removing the dorsal portions of the isolated cerebral hemispheres (Fig. 1). Each litter, consisting of an average of 10 unborn pups of both sexes, was dissected and the resulting samples pooled by region. RNA quality was checked, and in total, 4 pools were used for 2 microarray comparisons of A versus M and A versus P, for a total of 16 slides, including dye swapping. Eighty-eight percent of arrayed clones were identified as present, as they generated average signal intensity greater than local background (Mean Negative Control was subtracted from Mean Signal, and those below 0 are omitted.) when hybridized by A, M, or P samples.

The analysis of the array data revealed 106 known genes and ESTs that are differentially expressed along the anterior–posterior axis (Table 1). Of the 80 genes with higher expression levels in anterior cortical anlage (at least in one of the 2 comparisons), 74 were known or annotated and 6 were ESTs. For the 26 genes identified to be posteriorly enriched (in at least one of the 2 comparisons), there were 25 known or annotated genes and 1 EST. CoupTF1 was found to be differentially expressed at the 99.5% confidence limit, but did not pass the more stringent a second screen (see Methods). Other genes known to be involved in cortical regionalization, such as Emx1 and Pax6 were not included on the array, and thus were not identified.

Table 1

Anterior- and posterior-enriched genes identified by microarray

Accession no.SymbolNameIn situ hybridization
Genes with anterior-enriched expression
AA073913AmnAmnionlessUI
W75669Anapc2Anaphase-promoting complex subunit 2
AA120679Arl7ADP-ribosylation factor-like 7+
W96830Brp16Brain protein 16UI
W36837Cacnb1Calcium channel, voltage-dependent, β1 subunit+
W65230Cldn13Claudin 13
AA120697Clec1bC-type lectin-like receptor 2+
AA108438Cml4Camello-like 4UI
W63987Cnp1Cyclic nucleotide phosphodiesterase 1UI
AI594926DcppDemilune cell and parotid protein
AA008734Ddx27DEAD (Asp–Glu–Ala–Asp) box polypeptide 27
W45825Dyrk2dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 2+CTX
AA008687Ecgf1Endothelial cell growth factor 1 (platelet-derived)
AA032559Eef2kEukaryotic elongation factor-2 kinaseUI
AA289606Elk3ELK3, member of ETS oncogene family+
AA060723EtfaElectron transferring flavoprotein, alpha polypeptide
AI385754Fabp7Fatty acid binding protein 7, brain
AA031219Fbxl17F-box and leucine-rich repeat protein 17+
AA023645Fhl2FHL domains 2+CTX
AA002314Flt3lFMS-like tyrosine kinase 3 ligand
AA060844Fxyd6FXYD domain-containing ion transport regulator 6
AA175441Ggta1Glycoprotein galactosyltransferase alpha 1, 3UI
AA466087H2afzH2A histone family, member Z
W10072Igf1Insulin-like growth factor 1
AA175538Il7rInterleukin 7 receptor
AA403846KitlKit ligand+
W97446Klf1Kruppel-like factor 1 (erythroid)
W66749Letmd1LETM1 domain containing 1
AA003791Loxl2Lysyl oxidase–like 2
AA231293Meox2Mesenchyme homeobox 2
AA098361Mfhas1Malignant fibrous histiocytoma amplified sequence 1UI
AA033466MpoMyeloperoxidase
AA109944Myo7bMyosin VIIb+
AA473269Ndrg1N-myc downstream regulated 1+
W85646Nek9NIMA (never in mitosis gene a)–related expressed kinase 9+
AI425728Nupl1Nucleoporin like 1+
AA119236NvlNuclear VCP-like
W14332Pcbd6-Pyruvoyl-tetrahydropterin synthase dimerization cofactor of hepatocyte nuclear factor 1 alpha+
W98241Pcsk5Proprotein convertase subtilisin/kexin type 5+
AA062140Pitx3Paired-like homeodomain transcription factor 3
AA031057Ppp2r2bProtein phosphatase 2 (formerly 2A), regulatory subunit B (PR 52), β isoform
AI429829Rab13RAB13, member RAS oncogene family
AA275118Rap2cRAP2C, member of RAS oncogene familyUI
CA748280Rbp1Retinol binding protein 1, cellular
W80210RbpmsRNA binding protein gene with multiple splicing
AA184614RhofRas homolog gene family, member f (in filopodia)+
AA000347Sec24cSEC24 related gene family, member C (Saccharomyces cerevisiae)UI
AA268953Sfrp1Secreted frizzled-related sequence protein 1+CTX
AA175259Sh3kbp1SH3-domain kinase binding protein 1+
AA413194Slc25a13Solute carrier family 25 (mitochondrial carrier; adenine nucleotide translocator), member 13+CTX
AI467559Slit3Slit homolog 3 (Drosophila)UI
AA000223Smoc1SPARC related modular calcium binding 1+CTX
AA200737Snap25bpSynaptosomal-associated protein 25 binding protein+
W34018Spnb1Spectrin β1
AA062142Sprrl7Small proline rich-like 7UI
AA038788StcStanniocalcinUI
AA270232Tal1T-cell acute lymphocytic leukemia 1UI
AA547208Tcl1T-cell lymphoma breakpoint 1
W59265TrioTriple functional domain (PTPRF interacting)+
AA522127Uqcrc2Ubiquinol cytochrome c reductase core protein 2
W49208Usp29Ubiquitin specific protease 29
AA061458Usp52Ubiquitin specific protease 52
AA0026362310030G09RikSimilar to zinc finger protein 560+
AA003367Wdr33WD repeat domain 33UI
W972441810012P15RikRIKEN cDNA 1810012P15 geneUI
AA1639963110048L19RikRIKEN cDNA 3110048L19 geneUI
AA1451374930420O11RikRIKEN cDNA 4930420O11 gene+
AA1440464930432B04RikRIKEN cDNA 4930432B04 geneUI
AI4138385430413K10RikSimilar to RIKEN cDNA 5430413K10UI
AA1703355830403E09RikRIKEN cDNA 5830403E09 geneUI
AA204360AA960558Expressed sequence AA960558UI
BX527908B230369L08RikRIKEN cDNA B230369L08 geneUI
AA002786BC023488cDNA sequence BC023488UI
W59215BC037527cDNA sequence BC037527
AK053778C80587Expressed sequence C80587
AA217113Similar to very large inducible GTPase-1UI
AA050124EST, unknownUI
AA155387EST, unknownUI
W70958EST, unknownUI
W77508EST, unknownUI
Genes with posterior-enriched expression
AA245324AipAryl-hydrocarbon receptor-interacting protein
AA064247Coro7Coronin 7
AA008001Ctsb/Fdft1Cathepsin BUI
AA073904Dkk3Dickkopf homolog 3 (Xenopus laevis)+CTX
AA153223Dusp11Dual-specificity phosphatase 11 (RNA/RNP complex 1-interacting)
AA544895Farp1FERM, RhoGEF (Arhgef) and pleckstrin domain protein 1 (chondrocyte-derived)+CTX
AA047966Fhl1FHL domains 1+CTX
AA030294Fzd1Frizzled homolog 1 (Drosophila)+CTX
AA255078Galk1Galactokinase 1UI
AA048560GartPhosphoribosylglycinamide formyltransferase+CTX
AA289992Ifitm2Interferon-induced transmembrane protein 3-like+CTX
AA261160McamMelanoma cell adhesion molecule+CTX
W55827NfixNuclear factor I/X+CTX
W98998Notch1Notch gene homolog 1 (Drosophila)
BX513775PfasPhosphoribosylformylglycinamidine synthase (FGAR amidotransferase)
AA015088Prpf3PRP3 pre-mRNA processing factor 3 homolog (yeast)
AA253750Rbm9RNA binding motif protein 9+
AA518455SEF2/Tcf4Transcription factor 4+CTX
W17647TtrTransthyretin+
W37018Zfp261Zinc finger protein 261
AA041916E130016E03RikFibrinogen silencer binding proteinUI
AA051406D5Wsu46eDNA segment, Chr 5, Wayne State University 46, expressedUI
BX5229372900001G08RikRIKEN cDNA 2900001G08 geneUI
AA2733865033406L14RikRIKEN cDNA B230110O18 geneUI
AA2705271110019L22RikRIKEN cDNA 1110019L22 gene+CTX
AA231557EST, unknown+
Accession no.SymbolNameIn situ hybridization
Genes with anterior-enriched expression
AA073913AmnAmnionlessUI
W75669Anapc2Anaphase-promoting complex subunit 2
AA120679Arl7ADP-ribosylation factor-like 7+
W96830Brp16Brain protein 16UI
W36837Cacnb1Calcium channel, voltage-dependent, β1 subunit+
W65230Cldn13Claudin 13
AA120697Clec1bC-type lectin-like receptor 2+
AA108438Cml4Camello-like 4UI
W63987Cnp1Cyclic nucleotide phosphodiesterase 1UI
AI594926DcppDemilune cell and parotid protein
AA008734Ddx27DEAD (Asp–Glu–Ala–Asp) box polypeptide 27
W45825Dyrk2dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 2+CTX
AA008687Ecgf1Endothelial cell growth factor 1 (platelet-derived)
AA032559Eef2kEukaryotic elongation factor-2 kinaseUI
AA289606Elk3ELK3, member of ETS oncogene family+
AA060723EtfaElectron transferring flavoprotein, alpha polypeptide
AI385754Fabp7Fatty acid binding protein 7, brain
AA031219Fbxl17F-box and leucine-rich repeat protein 17+
AA023645Fhl2FHL domains 2+CTX
AA002314Flt3lFMS-like tyrosine kinase 3 ligand
AA060844Fxyd6FXYD domain-containing ion transport regulator 6
AA175441Ggta1Glycoprotein galactosyltransferase alpha 1, 3UI
AA466087H2afzH2A histone family, member Z
W10072Igf1Insulin-like growth factor 1
AA175538Il7rInterleukin 7 receptor
AA403846KitlKit ligand+
W97446Klf1Kruppel-like factor 1 (erythroid)
W66749Letmd1LETM1 domain containing 1
AA003791Loxl2Lysyl oxidase–like 2
AA231293Meox2Mesenchyme homeobox 2
AA098361Mfhas1Malignant fibrous histiocytoma amplified sequence 1UI
AA033466MpoMyeloperoxidase
AA109944Myo7bMyosin VIIb+
AA473269Ndrg1N-myc downstream regulated 1+
W85646Nek9NIMA (never in mitosis gene a)–related expressed kinase 9+
AI425728Nupl1Nucleoporin like 1+
AA119236NvlNuclear VCP-like
W14332Pcbd6-Pyruvoyl-tetrahydropterin synthase dimerization cofactor of hepatocyte nuclear factor 1 alpha+
W98241Pcsk5Proprotein convertase subtilisin/kexin type 5+
AA062140Pitx3Paired-like homeodomain transcription factor 3
AA031057Ppp2r2bProtein phosphatase 2 (formerly 2A), regulatory subunit B (PR 52), β isoform
AI429829Rab13RAB13, member RAS oncogene family
AA275118Rap2cRAP2C, member of RAS oncogene familyUI
CA748280Rbp1Retinol binding protein 1, cellular
W80210RbpmsRNA binding protein gene with multiple splicing
AA184614RhofRas homolog gene family, member f (in filopodia)+
AA000347Sec24cSEC24 related gene family, member C (Saccharomyces cerevisiae)UI
AA268953Sfrp1Secreted frizzled-related sequence protein 1+CTX
AA175259Sh3kbp1SH3-domain kinase binding protein 1+
AA413194Slc25a13Solute carrier family 25 (mitochondrial carrier; adenine nucleotide translocator), member 13+CTX
AI467559Slit3Slit homolog 3 (Drosophila)UI
AA000223Smoc1SPARC related modular calcium binding 1+CTX
AA200737Snap25bpSynaptosomal-associated protein 25 binding protein+
W34018Spnb1Spectrin β1
AA062142Sprrl7Small proline rich-like 7UI
AA038788StcStanniocalcinUI
AA270232Tal1T-cell acute lymphocytic leukemia 1UI
AA547208Tcl1T-cell lymphoma breakpoint 1
W59265TrioTriple functional domain (PTPRF interacting)+
AA522127Uqcrc2Ubiquinol cytochrome c reductase core protein 2
W49208Usp29Ubiquitin specific protease 29
AA061458Usp52Ubiquitin specific protease 52
AA0026362310030G09RikSimilar to zinc finger protein 560+
AA003367Wdr33WD repeat domain 33UI
W972441810012P15RikRIKEN cDNA 1810012P15 geneUI
AA1639963110048L19RikRIKEN cDNA 3110048L19 geneUI
AA1451374930420O11RikRIKEN cDNA 4930420O11 gene+
AA1440464930432B04RikRIKEN cDNA 4930432B04 geneUI
AI4138385430413K10RikSimilar to RIKEN cDNA 5430413K10UI
AA1703355830403E09RikRIKEN cDNA 5830403E09 geneUI
AA204360AA960558Expressed sequence AA960558UI
BX527908B230369L08RikRIKEN cDNA B230369L08 geneUI
AA002786BC023488cDNA sequence BC023488UI
W59215BC037527cDNA sequence BC037527
AK053778C80587Expressed sequence C80587
AA217113Similar to very large inducible GTPase-1UI
AA050124EST, unknownUI
AA155387EST, unknownUI
W70958EST, unknownUI
W77508EST, unknownUI
Genes with posterior-enriched expression
AA245324AipAryl-hydrocarbon receptor-interacting protein
AA064247Coro7Coronin 7
AA008001Ctsb/Fdft1Cathepsin BUI
AA073904Dkk3Dickkopf homolog 3 (Xenopus laevis)+CTX
AA153223Dusp11Dual-specificity phosphatase 11 (RNA/RNP complex 1-interacting)
AA544895Farp1FERM, RhoGEF (Arhgef) and pleckstrin domain protein 1 (chondrocyte-derived)+CTX
AA047966Fhl1FHL domains 1+CTX
AA030294Fzd1Frizzled homolog 1 (Drosophila)+CTX
AA255078Galk1Galactokinase 1UI
AA048560GartPhosphoribosylglycinamide formyltransferase+CTX
AA289992Ifitm2Interferon-induced transmembrane protein 3-like+CTX
AA261160McamMelanoma cell adhesion molecule+CTX
W55827NfixNuclear factor I/X+CTX
W98998Notch1Notch gene homolog 1 (Drosophila)
BX513775PfasPhosphoribosylformylglycinamidine synthase (FGAR amidotransferase)
AA015088Prpf3PRP3 pre-mRNA processing factor 3 homolog (yeast)
AA253750Rbm9RNA binding motif protein 9+
AA518455SEF2/Tcf4Transcription factor 4+CTX
W17647TtrTransthyretin+
W37018Zfp261Zinc finger protein 261
AA041916E130016E03RikFibrinogen silencer binding proteinUI
AA051406D5Wsu46eDNA segment, Chr 5, Wayne State University 46, expressedUI
BX5229372900001G08RikRIKEN cDNA 2900001G08 geneUI
AA2733865033406L14RikRIKEN cDNA B230110O18 geneUI
AA2705271110019L22RikRIKEN cDNA 1110019L22 gene+CTX
AA231557EST, unknown+

Note: Data analysis resulted in the identification of 80 genes anteriorly enriched and 26 genes with posterior telencephalic enrichment. Expression patterns of 88 genes were examined with in situ hybridization, confirming regionalized patterns for 52 genes in the head or within the telencephalon. Among these genes, 15 possessed a clearly visible anterior–posterior gradient within the cortical plate and dorsal telencephalon based upon in situ hybridization. The symbols represent the following. +CTX, region-specific expression in the dorsal telencephalon was confirmed by in situ hybridization. +, region-specific expression in the head/telencephalon was confirmed by in situ hybridization. —, in situ hybridization did not show any anterior–posterior gradient in the head. UI, in situ hybridization gave uninterpretable results. X, in situ hybridization disagreed with the predicted gradient in the telencephalon.

Table 1

Anterior- and posterior-enriched genes identified by microarray

Accession no.SymbolNameIn situ hybridization
Genes with anterior-enriched expression
AA073913AmnAmnionlessUI
W75669Anapc2Anaphase-promoting complex subunit 2
AA120679Arl7ADP-ribosylation factor-like 7+
W96830Brp16Brain protein 16UI
W36837Cacnb1Calcium channel, voltage-dependent, β1 subunit+
W65230Cldn13Claudin 13
AA120697Clec1bC-type lectin-like receptor 2+
AA108438Cml4Camello-like 4UI
W63987Cnp1Cyclic nucleotide phosphodiesterase 1UI
AI594926DcppDemilune cell and parotid protein
AA008734Ddx27DEAD (Asp–Glu–Ala–Asp) box polypeptide 27
W45825Dyrk2dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 2+CTX
AA008687Ecgf1Endothelial cell growth factor 1 (platelet-derived)
AA032559Eef2kEukaryotic elongation factor-2 kinaseUI
AA289606Elk3ELK3, member of ETS oncogene family+
AA060723EtfaElectron transferring flavoprotein, alpha polypeptide
AI385754Fabp7Fatty acid binding protein 7, brain
AA031219Fbxl17F-box and leucine-rich repeat protein 17+
AA023645Fhl2FHL domains 2+CTX
AA002314Flt3lFMS-like tyrosine kinase 3 ligand
AA060844Fxyd6FXYD domain-containing ion transport regulator 6
AA175441Ggta1Glycoprotein galactosyltransferase alpha 1, 3UI
AA466087H2afzH2A histone family, member Z
W10072Igf1Insulin-like growth factor 1
AA175538Il7rInterleukin 7 receptor
AA403846KitlKit ligand+
W97446Klf1Kruppel-like factor 1 (erythroid)
W66749Letmd1LETM1 domain containing 1
AA003791Loxl2Lysyl oxidase–like 2
AA231293Meox2Mesenchyme homeobox 2
AA098361Mfhas1Malignant fibrous histiocytoma amplified sequence 1UI
AA033466MpoMyeloperoxidase
AA109944Myo7bMyosin VIIb+
AA473269Ndrg1N-myc downstream regulated 1+
W85646Nek9NIMA (never in mitosis gene a)–related expressed kinase 9+
AI425728Nupl1Nucleoporin like 1+
AA119236NvlNuclear VCP-like
W14332Pcbd6-Pyruvoyl-tetrahydropterin synthase dimerization cofactor of hepatocyte nuclear factor 1 alpha+
W98241Pcsk5Proprotein convertase subtilisin/kexin type 5+
AA062140Pitx3Paired-like homeodomain transcription factor 3
AA031057Ppp2r2bProtein phosphatase 2 (formerly 2A), regulatory subunit B (PR 52), β isoform
AI429829Rab13RAB13, member RAS oncogene family
AA275118Rap2cRAP2C, member of RAS oncogene familyUI
CA748280Rbp1Retinol binding protein 1, cellular
W80210RbpmsRNA binding protein gene with multiple splicing
AA184614RhofRas homolog gene family, member f (in filopodia)+
AA000347Sec24cSEC24 related gene family, member C (Saccharomyces cerevisiae)UI
AA268953Sfrp1Secreted frizzled-related sequence protein 1+CTX
AA175259Sh3kbp1SH3-domain kinase binding protein 1+
AA413194Slc25a13Solute carrier family 25 (mitochondrial carrier; adenine nucleotide translocator), member 13+CTX
AI467559Slit3Slit homolog 3 (Drosophila)UI
AA000223Smoc1SPARC related modular calcium binding 1+CTX
AA200737Snap25bpSynaptosomal-associated protein 25 binding protein+
W34018Spnb1Spectrin β1
AA062142Sprrl7Small proline rich-like 7UI
AA038788StcStanniocalcinUI
AA270232Tal1T-cell acute lymphocytic leukemia 1UI
AA547208Tcl1T-cell lymphoma breakpoint 1
W59265TrioTriple functional domain (PTPRF interacting)+
AA522127Uqcrc2Ubiquinol cytochrome c reductase core protein 2
W49208Usp29Ubiquitin specific protease 29
AA061458Usp52Ubiquitin specific protease 52
AA0026362310030G09RikSimilar to zinc finger protein 560+
AA003367Wdr33WD repeat domain 33UI
W972441810012P15RikRIKEN cDNA 1810012P15 geneUI
AA1639963110048L19RikRIKEN cDNA 3110048L19 geneUI
AA1451374930420O11RikRIKEN cDNA 4930420O11 gene+
AA1440464930432B04RikRIKEN cDNA 4930432B04 geneUI
AI4138385430413K10RikSimilar to RIKEN cDNA 5430413K10UI
AA1703355830403E09RikRIKEN cDNA 5830403E09 geneUI
AA204360AA960558Expressed sequence AA960558UI
BX527908B230369L08RikRIKEN cDNA B230369L08 geneUI
AA002786BC023488cDNA sequence BC023488UI
W59215BC037527cDNA sequence BC037527
AK053778C80587Expressed sequence C80587
AA217113Similar to very large inducible GTPase-1UI
AA050124EST, unknownUI
AA155387EST, unknownUI
W70958EST, unknownUI
W77508EST, unknownUI
Genes with posterior-enriched expression
AA245324AipAryl-hydrocarbon receptor-interacting protein
AA064247Coro7Coronin 7
AA008001Ctsb/Fdft1Cathepsin BUI
AA073904Dkk3Dickkopf homolog 3 (Xenopus laevis)+CTX
AA153223Dusp11Dual-specificity phosphatase 11 (RNA/RNP complex 1-interacting)
AA544895Farp1FERM, RhoGEF (Arhgef) and pleckstrin domain protein 1 (chondrocyte-derived)+CTX
AA047966Fhl1FHL domains 1+CTX
AA030294Fzd1Frizzled homolog 1 (Drosophila)+CTX
AA255078Galk1Galactokinase 1UI
AA048560GartPhosphoribosylglycinamide formyltransferase+CTX
AA289992Ifitm2Interferon-induced transmembrane protein 3-like+CTX
AA261160McamMelanoma cell adhesion molecule+CTX
W55827NfixNuclear factor I/X+CTX
W98998Notch1Notch gene homolog 1 (Drosophila)
BX513775PfasPhosphoribosylformylglycinamidine synthase (FGAR amidotransferase)
AA015088Prpf3PRP3 pre-mRNA processing factor 3 homolog (yeast)
AA253750Rbm9RNA binding motif protein 9+
AA518455SEF2/Tcf4Transcription factor 4+CTX
W17647TtrTransthyretin+
W37018Zfp261Zinc finger protein 261
AA041916E130016E03RikFibrinogen silencer binding proteinUI
AA051406D5Wsu46eDNA segment, Chr 5, Wayne State University 46, expressedUI
BX5229372900001G08RikRIKEN cDNA 2900001G08 geneUI
AA2733865033406L14RikRIKEN cDNA B230110O18 geneUI
AA2705271110019L22RikRIKEN cDNA 1110019L22 gene+CTX
AA231557EST, unknown+
Accession no.SymbolNameIn situ hybridization
Genes with anterior-enriched expression
AA073913AmnAmnionlessUI
W75669Anapc2Anaphase-promoting complex subunit 2
AA120679Arl7ADP-ribosylation factor-like 7+
W96830Brp16Brain protein 16UI
W36837Cacnb1Calcium channel, voltage-dependent, β1 subunit+
W65230Cldn13Claudin 13
AA120697Clec1bC-type lectin-like receptor 2+
AA108438Cml4Camello-like 4UI
W63987Cnp1Cyclic nucleotide phosphodiesterase 1UI
AI594926DcppDemilune cell and parotid protein
AA008734Ddx27DEAD (Asp–Glu–Ala–Asp) box polypeptide 27
W45825Dyrk2dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 2+CTX
AA008687Ecgf1Endothelial cell growth factor 1 (platelet-derived)
AA032559Eef2kEukaryotic elongation factor-2 kinaseUI
AA289606Elk3ELK3, member of ETS oncogene family+
AA060723EtfaElectron transferring flavoprotein, alpha polypeptide
AI385754Fabp7Fatty acid binding protein 7, brain
AA031219Fbxl17F-box and leucine-rich repeat protein 17+
AA023645Fhl2FHL domains 2+CTX
AA002314Flt3lFMS-like tyrosine kinase 3 ligand
AA060844Fxyd6FXYD domain-containing ion transport regulator 6
AA175441Ggta1Glycoprotein galactosyltransferase alpha 1, 3UI
AA466087H2afzH2A histone family, member Z
W10072Igf1Insulin-like growth factor 1
AA175538Il7rInterleukin 7 receptor
AA403846KitlKit ligand+
W97446Klf1Kruppel-like factor 1 (erythroid)
W66749Letmd1LETM1 domain containing 1
AA003791Loxl2Lysyl oxidase–like 2
AA231293Meox2Mesenchyme homeobox 2
AA098361Mfhas1Malignant fibrous histiocytoma amplified sequence 1UI
AA033466MpoMyeloperoxidase
AA109944Myo7bMyosin VIIb+
AA473269Ndrg1N-myc downstream regulated 1+
W85646Nek9NIMA (never in mitosis gene a)–related expressed kinase 9+
AI425728Nupl1Nucleoporin like 1+
AA119236NvlNuclear VCP-like
W14332Pcbd6-Pyruvoyl-tetrahydropterin synthase dimerization cofactor of hepatocyte nuclear factor 1 alpha+
W98241Pcsk5Proprotein convertase subtilisin/kexin type 5+
AA062140Pitx3Paired-like homeodomain transcription factor 3
AA031057Ppp2r2bProtein phosphatase 2 (formerly 2A), regulatory subunit B (PR 52), β isoform
AI429829Rab13RAB13, member RAS oncogene family
AA275118Rap2cRAP2C, member of RAS oncogene familyUI
CA748280Rbp1Retinol binding protein 1, cellular
W80210RbpmsRNA binding protein gene with multiple splicing
AA184614RhofRas homolog gene family, member f (in filopodia)+
AA000347Sec24cSEC24 related gene family, member C (Saccharomyces cerevisiae)UI
AA268953Sfrp1Secreted frizzled-related sequence protein 1+CTX
AA175259Sh3kbp1SH3-domain kinase binding protein 1+
AA413194Slc25a13Solute carrier family 25 (mitochondrial carrier; adenine nucleotide translocator), member 13+CTX
AI467559Slit3Slit homolog 3 (Drosophila)UI
AA000223Smoc1SPARC related modular calcium binding 1+CTX
AA200737Snap25bpSynaptosomal-associated protein 25 binding protein+
W34018Spnb1Spectrin β1
AA062142Sprrl7Small proline rich-like 7UI
AA038788StcStanniocalcinUI
AA270232Tal1T-cell acute lymphocytic leukemia 1UI
AA547208Tcl1T-cell lymphoma breakpoint 1
W59265TrioTriple functional domain (PTPRF interacting)+
AA522127Uqcrc2Ubiquinol cytochrome c reductase core protein 2
W49208Usp29Ubiquitin specific protease 29
AA061458Usp52Ubiquitin specific protease 52
AA0026362310030G09RikSimilar to zinc finger protein 560+
AA003367Wdr33WD repeat domain 33UI
W972441810012P15RikRIKEN cDNA 1810012P15 geneUI
AA1639963110048L19RikRIKEN cDNA 3110048L19 geneUI
AA1451374930420O11RikRIKEN cDNA 4930420O11 gene+
AA1440464930432B04RikRIKEN cDNA 4930432B04 geneUI
AI4138385430413K10RikSimilar to RIKEN cDNA 5430413K10UI
AA1703355830403E09RikRIKEN cDNA 5830403E09 geneUI
AA204360AA960558Expressed sequence AA960558UI
BX527908B230369L08RikRIKEN cDNA B230369L08 geneUI
AA002786BC023488cDNA sequence BC023488UI
W59215BC037527cDNA sequence BC037527
AK053778C80587Expressed sequence C80587
AA217113Similar to very large inducible GTPase-1UI
AA050124EST, unknownUI
AA155387EST, unknownUI
W70958EST, unknownUI
W77508EST, unknownUI
Genes with posterior-enriched expression
AA245324AipAryl-hydrocarbon receptor-interacting protein
AA064247Coro7Coronin 7
AA008001Ctsb/Fdft1Cathepsin BUI
AA073904Dkk3Dickkopf homolog 3 (Xenopus laevis)+CTX
AA153223Dusp11Dual-specificity phosphatase 11 (RNA/RNP complex 1-interacting)
AA544895Farp1FERM, RhoGEF (Arhgef) and pleckstrin domain protein 1 (chondrocyte-derived)+CTX
AA047966Fhl1FHL domains 1+CTX
AA030294Fzd1Frizzled homolog 1 (Drosophila)+CTX
AA255078Galk1Galactokinase 1UI
AA048560GartPhosphoribosylglycinamide formyltransferase+CTX
AA289992Ifitm2Interferon-induced transmembrane protein 3-like+CTX
AA261160McamMelanoma cell adhesion molecule+CTX
W55827NfixNuclear factor I/X+CTX
W98998Notch1Notch gene homolog 1 (Drosophila)
BX513775PfasPhosphoribosylformylglycinamidine synthase (FGAR amidotransferase)
AA015088Prpf3PRP3 pre-mRNA processing factor 3 homolog (yeast)
AA253750Rbm9RNA binding motif protein 9+
AA518455SEF2/Tcf4Transcription factor 4+CTX
W17647TtrTransthyretin+
W37018Zfp261Zinc finger protein 261
AA041916E130016E03RikFibrinogen silencer binding proteinUI
AA051406D5Wsu46eDNA segment, Chr 5, Wayne State University 46, expressedUI
BX5229372900001G08RikRIKEN cDNA 2900001G08 geneUI
AA2733865033406L14RikRIKEN cDNA B230110O18 geneUI
AA2705271110019L22RikRIKEN cDNA 1110019L22 gene+CTX
AA231557EST, unknown+

Note: Data analysis resulted in the identification of 80 genes anteriorly enriched and 26 genes with posterior telencephalic enrichment. Expression patterns of 88 genes were examined with in situ hybridization, confirming regionalized patterns for 52 genes in the head or within the telencephalon. Among these genes, 15 possessed a clearly visible anterior–posterior gradient within the cortical plate and dorsal telencephalon based upon in situ hybridization. The symbols represent the following. +CTX, region-specific expression in the dorsal telencephalon was confirmed by in situ hybridization. +, region-specific expression in the head/telencephalon was confirmed by in situ hybridization. —, in situ hybridization did not show any anterior–posterior gradient in the head. UI, in situ hybridization gave uninterpretable results. X, in situ hybridization disagreed with the predicted gradient in the telencephalon.

To functionally categorize the identified genes, an analysis was performed using EASE (http://apps1.niaid.nih.gov/david/). This revealed the following overrepresented categories of genes with P values below 0.05 in the ontology category of Biological Processes: “development,” “cell differentiation,” and “extracellular.” Within these categories were genes of the Wnt pathway, several of these being factors known to play a role in cortical patterning, and other family members not known to be expressed in an A-P gradient. Strikingly, 8 of 41 (19.5%) classified genes were transcription factors, a larger proportion than usually identified in studies involving similar stages of mammalian brain development using the same microarray slides as those in this study (χ2 = 7.38, P < 0.007). For example, in a previous study of stem cell proliferation and neurogenesis using the same microarray, only 12 out of 110 (11%) genes were transcription factors (Karsten et al. 2003). This finding, using an unbiased genetic screening approach, lends additional support to the notion of a key role for transcriptional effects on cortical identity, and provides a set of genes likely to regulate the expression of other genes in an A-P gradient as well.

Database Confirmation of Known Genes

The Mouse Genome Informatics and National Center for Biotechnology Information PubMed databases were searched for previously published expression data or reports on the genes identified. Of the set of A-P differential genes, 47 had been previously studied. Published expression data in the form of in situ hybridization in the embryonic mouse central nervous system (CNS) were available for 20 of the identified genes but many contained insufficient data to determine whether there were A-P gradients of expression. There were, however, 8 genes that displayed graded telencephalic expression patterns by in situ hybridization at embryonic ages, supporting the present array analysis. Dkk3 (Diep et al. 2004), Rbpms (Gerber et al. 1999), Tcf4 (Dorflinger et al. 1999), Eef2k (Sakagami et al. 2002), Sfrp1 (Augustine et al. 2001; Kim et al. 2001), Elk3 (Ayadi et al. 2001), and FABP7 (Sansom et al. 2005) showed regionalized expression in the telencephalon in a pattern predicted by the array analysis. Gradients in the neuroepithelium were evident for both Sfrp1 (Kim et al. 2001), and FABP7 (Sansom et al. 2005). Kitl (Matsui et al. 1990) and Ttr (Grove et al. 1998) were expressed in the olfactory bulb (OB) and the choroid plexus, respectively. Highly expressed genes unique to the choroid plexus often exhibit differential expression in microarray experiments due to the difficulty of removing choroid plexus tissue completely in the dissection. A number of genes expressed in the OB, including Kit1, were identified as anteriorly expressed, consistent with the array analysis because fragments of the developing OB were likely present in the frontal samples. Further, because a low level of expression or a subtle gradient may not be detectable using in situ hybridization, we cannot rule out the possibility that the other genes also have graded expression.

Thirty-two of the identified genes had been knocked out in mice. Reports of CNS phenotypes were available for 7 genes (Table 2), but others had either no mention of a CNS phenotype, or resulted in embryonic lethality. This does not exclude the possibility that these animals may have subtle morphological or behavioral phenotypes that were not readily detected. To provide direct confirmation, we performed a large number of in situ hybridization experiments, summarized below.

Table 2

Abnormal CNS phenotypes observed when genes identified in this study have been inactivated in mouse

GeneCNS phenotype of knockout or transgenic animals
KitlDeficient baseline synaptic transmission between the dentate gyrus and CA3 (Motro et al. 1996)
TrioAltered organization of the dentate gyrus and the mitral cells (O'Brien et al. 2000)
Cnp1Hydrocephalus and neurodegeneration (Lappe-Siefke et al. 2003)
Sfrp1Decreased brain size (Bodine et al. 2004)
Pitx3/Elovl3Loss of neurons in the substantia nigra (Nunes et al. 2003; van den Munckhof et al. 2003)
Ctsb/Fdft1Cerebral and cerebellar atrophy (Felbor et al. 2002)
Igf1Decreased number of axons and oligodendrocytes; overexpression leads to increase in volume of primary somatosensory and primary motor cortices (Beck et al. 1995; Hodge et al. 2004)
GeneCNS phenotype of knockout or transgenic animals
KitlDeficient baseline synaptic transmission between the dentate gyrus and CA3 (Motro et al. 1996)
TrioAltered organization of the dentate gyrus and the mitral cells (O'Brien et al. 2000)
Cnp1Hydrocephalus and neurodegeneration (Lappe-Siefke et al. 2003)
Sfrp1Decreased brain size (Bodine et al. 2004)
Pitx3/Elovl3Loss of neurons in the substantia nigra (Nunes et al. 2003; van den Munckhof et al. 2003)
Ctsb/Fdft1Cerebral and cerebellar atrophy (Felbor et al. 2002)
Igf1Decreased number of axons and oligodendrocytes; overexpression leads to increase in volume of primary somatosensory and primary motor cortices (Beck et al. 1995; Hodge et al. 2004)

Note: The resulting phenotypes of knockouts and transgenics for these regionalized genes indicate that they are involved in telencephalic development. The published data do not indicate deficits or abnormalities in A-P patterning.

Table 2

Abnormal CNS phenotypes observed when genes identified in this study have been inactivated in mouse

GeneCNS phenotype of knockout or transgenic animals
KitlDeficient baseline synaptic transmission between the dentate gyrus and CA3 (Motro et al. 1996)
TrioAltered organization of the dentate gyrus and the mitral cells (O'Brien et al. 2000)
Cnp1Hydrocephalus and neurodegeneration (Lappe-Siefke et al. 2003)
Sfrp1Decreased brain size (Bodine et al. 2004)
Pitx3/Elovl3Loss of neurons in the substantia nigra (Nunes et al. 2003; van den Munckhof et al. 2003)
Ctsb/Fdft1Cerebral and cerebellar atrophy (Felbor et al. 2002)
Igf1Decreased number of axons and oligodendrocytes; overexpression leads to increase in volume of primary somatosensory and primary motor cortices (Beck et al. 1995; Hodge et al. 2004)
GeneCNS phenotype of knockout or transgenic animals
KitlDeficient baseline synaptic transmission between the dentate gyrus and CA3 (Motro et al. 1996)
TrioAltered organization of the dentate gyrus and the mitral cells (O'Brien et al. 2000)
Cnp1Hydrocephalus and neurodegeneration (Lappe-Siefke et al. 2003)
Sfrp1Decreased brain size (Bodine et al. 2004)
Pitx3/Elovl3Loss of neurons in the substantia nigra (Nunes et al. 2003; van den Munckhof et al. 2003)
Ctsb/Fdft1Cerebral and cerebellar atrophy (Felbor et al. 2002)
Igf1Decreased number of axons and oligodendrocytes; overexpression leads to increase in volume of primary somatosensory and primary motor cortices (Beck et al. 1995; Hodge et al. 2004)

Note: The resulting phenotypes of knockouts and transgenics for these regionalized genes indicate that they are involved in telencephalic development. The published data do not indicate deficits or abnormalities in A-P patterning.

In Situ Hybridization Confirmation of Micorarray Results

The expression patterns of 88 clones were assayed by in situ hybridization at ages E12.5 to E14.5, E17.5, and P0 (Tables 1, 3). The data clearly confirmed the differential A-P expression of 15 genes resulting from the array data analysis. Because the in situ hybridizations were run as a large-scale screening, they could not be quantified at this throughput. Rather, we took a very conservative approach, such that only the genes with the graded A-P expression patterns that were most evident are reported (Figs 2, 3). A few genes were strongly expressed outside of the CNS, often in mesenchymal structures surrounding and/or rostral to the telencephalon, often corresponding to anterior-enriched genes in the microarray screen, such as Meox (data not shown). Others that had been identified by the array for enriched anterior expression showed strong signals in the OB and rostral migratory stream and rostral ventral telencephalon, as in the cases of Arl7, Fbxl17, and Kitl, (as well as other facial structures in the anterior head). This is expected, because the dissections of numerous embryos likely included some tissues surrounding the neocortex. These genes are not included in our list of confirmed genes, even though they are consistent with the array data. The in situ hybridization results of the genes identified by the microarrays are presented in Table 1.

Table 3

Summary of experimental results by mircoarray and in situ hybridization

Microarray resultNumber of in situ hybridizations runConfirmed regionalized expression in the headRegionalized in dorsal telencephalon
<F8066 (40)235
>F2622 (16)1310
Total10688 (56)3715
Microarray resultNumber of in situ hybridizations runConfirmed regionalized expression in the headRegionalized in dorsal telencephalon
<F8066 (40)235
>F2622 (16)1310
Total10688 (56)3715

Note: The numbers in parentheses are numbers of in situ hybridization with interpretable results as characterized in the Methods and Results. A clear, qualitatively visible gradient was necessary for the gene to be considered confirmed, a stringent criterion. A subset of those genes that were expressed regionally in an anterior–posterior gradient, which could include extra-neural tissues or the OB, was also found to be regionalized in the dorsal telencephalon.

Table 3

Summary of experimental results by mircoarray and in situ hybridization

Microarray resultNumber of in situ hybridizations runConfirmed regionalized expression in the headRegionalized in dorsal telencephalon
<F8066 (40)235
>F2622 (16)1310
Total10688 (56)3715
Microarray resultNumber of in situ hybridizations runConfirmed regionalized expression in the headRegionalized in dorsal telencephalon
<F8066 (40)235
>F2622 (16)1310
Total10688 (56)3715

Note: The numbers in parentheses are numbers of in situ hybridization with interpretable results as characterized in the Methods and Results. A clear, qualitatively visible gradient was necessary for the gene to be considered confirmed, a stringent criterion. A subset of those genes that were expressed regionally in an anterior–posterior gradient, which could include extra-neural tissues or the OB, was also found to be regionalized in the dorsal telencephalon.

Figure 2.

Genes with enriched anterior expression. (A) Autoradiographic images of in situ hybridization with probes representing anterior-enriched genes showing a clear gradient along the A-P axis of the developing telencephalon. (B) Images from emulsion dipped slides representing mRNA expression (silver grains) of anterior-enriched genes. Slc25a13 is expressed strongly in the anterior telencephalon at E13.5. Fhl2 is expressed anteriorly most prominently at E17.5. Note that the expression is strong in all the cortical layers but is low in the ventricular zone, suggesting that Fhl2 is expressed by postmitotic neural cells. It is also expressed at high levels in the choroid plexus, the differentiating hippocampus, and the thalamus. Dyrk2 is expressed strongly in the cortical plate and the hippocampus but expressed weakly in the VZ/SVZ. chp: choroid plexus, CP: cortical plate, Ctx: cortex, H: hippocampus, IZ: intermediate zone, SP: subplate. Arrows point to the regions of enriched expression. Asterisks indicate expression boundaries. Anterior is to the left.

Figure 3.

Posterior-enriched genes. (A) Autoradiographic film images of posterior-enriched genes showing a clear gradient along the A-P axis of the developing telencephalon. (B) Dark field images representing mRNA expression of posterior-enriched genes after developing slides dipped in photographic emulsion. Fhl1 and AI463886 show a strong A-P gradient at E14.5 (see Methods). Anterior is to the left. Arrows point to the regions of enriched expression. Asterisks indicate relative expression boundaries, which in most cases were gradual.

Anterior-Enriched Genes

In situ hybridization confirmed the A-P gradient of enriched expression for Fhl2, Sfrp1, Slc25a13, Smoc1, and Dyrk2. Three of these genes, Sfrp1, Slc25a13, and Smoc1 were all expressed in the ventricular and the subventricular zones (VZ/SVZ) with a rostral-to-caudal gradient, consistent with a potential intrinsic patterning role, whereas Dyrk2 and Fhl2 showed broader radial expression, including the cortical plate (Fig. 2). Slc25a13, which encodes a calcium-mitochondrial solute carrier protein with EF-hand domains (Kobayashi et al. 1999), showed intense anterior expression at E13.5, largely restricted to the neuroepithelium. Smoc1 encodes a secreted modular protein with EF-hand calcium-binding domain known to be expressed in the brain (Vannahme et al. 2002). Fhl2 encodes a LIM domain protein (Genini et al. 1997; Chan et al. 1998) that interacts with β-catenin of the Wnt pathway and act as a Wnt agonist (Chen et al. 2003; Wei et al. 2003; Labalette et al. 2004). At E17.5, Fhl2 shows strong signal in the intermediate zone and cortical plate, as well as the hippocampus and the choroid plexus (Fig. 2). Dyrk2, a dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 2, is expressed in the anterior cortical plate at E12.5, E14.5, and E17.5 (Fig. 2; Becker et al. 1998; Matsuo et al. 2001; Woods et al. 2001; Campbell and Proud 2002).

Posterior-Enriched Genes

Ten posterior-enriched genes, including one EST, identified by the microarrays show distinct posterior enrichment gradients by in situ hybridization, 6 of which are shown in Figure 3. During early corticogenesis, 5 of these genes, Farp1, Fhl1, Gart, Mcam, and the previously uncharacterized gene, A1463886, were restricted to the germinal neuroepithelium of the VZ and SVZ. Fhl1, a LIM domain gene, is expressed with a ventricular zone-to-cortical plate gradient at E14.5 (Fig. 3). Farp, involved in the adhesion, proliferation, and differentiation of chondrocytes (Koyano et al. 1997, 2001), is expressed in VZ/SVZ at E14.5, and mostly in the posterior cortical plate at E17.5 (data not shown).

Two genes involved in the Wnt pathway, Dkk3, a Wnt antagonist, and Fzd1, a Wnt receptor, are expressed in a somewhat complementary pattern in the neocortical anlage, as Fzd1 is expressed strongly in posterior IZ, where Dkk3 expression is weak (Fig. 3). At E17.5 Dkk3 is expressed in the ventricular zone, the cortical plate, the marginal zone, and the hippocampus with a caudal-to-rostral gradient in the cortical plate. Fzd1 is expressed more strongly in the posterior intermediate zone at E14.5 and E17.5, as well as in the hippocampus and less prominently in the choroid plexus and VZ/SVZ.

Two transcription factors, Nfix and Tcf4 (SEF2), are strongly expressed in the developing cortex and the hippocampus (Fig. 3). Nfix is expressed in all the zones of the cerebral wall at E14.5 with a cortical plate-to-ventricular zone gradient and a caudal-to-rostral gradient. It is expressed at E17.5 in all the embryonic zones dorsal to the perirhinal sulcus but its expression in the subplate extends into the rhinencephalon. Tcf4 is weakly expressed in the marginal zone at E14.5. At E17.5, it is strongly expressed in the cortical plate and the VZ, but not the subplate, which is complementary to the expression of Nfix. In contrast to Nfix, Tcf4 expression in all the zones extends into the rhinencephalon, with the exception of the subplate.

One gene involved in biosynthesis and 2 others encoding membrane-associated proteins were also expressed in a caudal-to-rostral gradient. Gart, a glycinamide ribonucleotide formyltransferase, is a developmentally regulated protein that is implicated in Down syndrome (Brodsky et al. 1997). Mcam is a cell adhesion molecule involved in neurite extension (Taira, Kohama, et al. 2004; Taira, Tsukamoto, et al. 2004; Taira et al. 2005; Kohama et al. 2005). Gart and Mcam probes showed stronger signals in the caudoventral neuroepithelium at E14.5 (data not shown). Ifitm2 encodes an evolutionarily conserved interferon-induced transmembrane protein associated with germ cell specification in mice with known expression throughout the epiblast (Tanaka and Matsui 2002; Lange et al. 2003). It is expressed in the VZ and the outer cortical plate at E17.5, as well as in the choroid plexus and the hippocampus (data not shown).

AI463886, an EST that encodes for a previously uncharacterized protein (AAH55405, NP_081032), is expressed in the posterior marginal zone and the neuroepithelium, as well as the hippocampus and the choroid plexus of the embryonic brain (Fig. 3). It is enriched in the posterior telencephalon during periods of neurogenesis, and is expressed at high levels throughout the developing cortical plate and the ventricular zone at E17. By P0, the expression becomes weak in the brain. Structural analysis of the protein product of this gene using PROSITE (ExPASY) and PredictProtein (Rost and Sander 1993; Rost 1996; Rost et al. 1996; Gasteiger et al. 2003) predicts a transmembrane domain at its C-terminus with the rest of the sequence in the form of cytoplasmic loops. Homology searching revealed no other homologies or clear functional domains. Based on these data, we hypothesize that this gene may encode a novel membrane-bound protein.

Expression of Multiple Fhl1 Isoforms in the Developing Brain

The identification of 2 four-and-a-half LIM (FHL) domain family genes, Fhl1 and Fhl2, piqued our interest, as neither had previously been implicated in brain development. Both proteins contain FHL domains, a cysteine rich motif that coordinately binds 2 zinc atoms and mediates protein–protein interactions (Arber and Caroni 1996). Some LIM domain–containing proteins are nuclear proteins involved in cell lineage determination and pattern formation during development (Lumsden 1995; Beckerle 1997; Dawid et al. 1998; Zhou, Ruiz-Lozano, et al. 1999) and both of these FHL genes are enriched in muscle (Chan et al. 1998; Lee et al. 1998).

However, Fhl1 is subject to complex alternative splicing, which has been incompletely characterized. The murine homolog of Fhl1, KyoT1, produces a shorter splice variant, KyoT2, with a frame shift resulting in a protein domain that interacts with RBP-J (recombination signal binding protein-Jκ) and negatively regulates its transcriptional activity (Taniguchi et al. 1998). In the human, another variant, FHL1B has been identified as a brain-specific isoform of FHL1 with a putative nuclear localization signal and a RBP-J binding region (Lee et al. 1999) identical to KyoT2. This corresponds to the longer splice variant reported but not characterized, by Taniguchi et al. (1998), which will be referred to here as Fhl1Bm (for Fhl1B, mouse).

The expression of Fhl1 messenger RNA (mRNA) and its splice variants was examined using several methods. In the E12.5 mouse brain, semiquantitative RT-PCR confirmed that all isoforms of Fhl1 were consistently expressed at higher levels in the posterior telencephalon. The A, M, and P tissue samples from the microarray studies were used to assess the differential expression of Fhl1 along the A-P axis. PCR products from A, M, and P samples all expressed the 3 isoforms. Ratios were obtained by dividing the amount of PCR product in O by the amount of product in F. All 3 isoforms were significantly enriched in the posterior telencephalon (P values = 0.0005, 0.002, and 0.006 for product sizes 700, 500, and 300 bp, respectively). Northern blot hybridization analyses for E14 embryos, E17 head, and brains from P0, P7, P14, and P21 revealed the expression of Fhl1 and Fhl1Bm in embryonic and postnatal brains, with a lower expression level for Fhl1Bm during the embryonic stages (Fig. 4). The posterior-enriched expression of Fhl1 in the developing neocortex was confirmed by in situ hybridization but the gradient was absent by E17 (Fig. 5). In situ hybridization with a probe recognizing all Fhl1 isoforms showed posterior expression at E12.5, which became more ubiquitous at E17.5.

Figure 4.

Northern blot analysis of Fhl1 and Fhl1Bm expression in total RNA. Fhl1 (lanes a) was expressed at high levels in the embryonic tissue and decreased postnatally. Note that the bands are narrower for the Fhl1Bm (lanes b) hybridization, indicating the specificity of the probe to this particular isoform. Most difference is seen at E14 and E17, showing that Fhl1Bm is not high during embryogenesis, and does not seem to be the predominant isoform of Fhl1 in embryonic or fetal brain. Arrows point to the bands for Fhl1Bm that is larger than that for the wider bands seen with Fhl1 probe.

Figure 5.

Fhl1 expression patterns in the telencephalon. (A) P0 sagittal section showed strong expression of Fhl1 in most areas of the cerebral cortex and other telencephalic structures, as well as the cerebellum. (B) Coronal sections of the germinal layer (medial to the left) showed Fhl1 expression through adulthood. (C) Fhl1 was expressed in the hippocampus throughout development, and in the adult the expression became more limited to the CA1 and CA3 regions, with much less robust expression in the dentate gyrus. (D) In situ hybridization of Fhl1 on adult human brain shows that Fhl1 signal is strong in pyramidal neurons in the gray matter in all cortical layers. The expression of Fhl1 appears to be expressed by different cell types in the cerebellum but most prominently in the Purkinje cells. CA1 and CA3: cornu ammonis 1 and 3, Cbl: cerebellum, Cbr: cerebral cortex; Ctx: cortex, DG: dentate gyrus, G: granular layer, GM: gray matter; GZ: germinal zone, lv: lateral ventricle, M: molecular layer of the cerebellum, ML: molecular layer, P: Purkinje cell layer, WM: white matter.

We also analyzed X-gal staining in heterozygote Fhl1 knockouts, in which the β-galactosidase gene was inserted to inactivate Fhl1 (see below). This revealed a comparable expression pattern to that of the mRNA pattern for Fhl1 in the wild-type mice, except that dentate gyrus X-gal staining was not observed. Close inspection of the stained cells revealed that those cells with LacZ staining (Fig. 6) were mostly pyramidal neurons, including hippocampal neurons of CA1 and CA3, motor neurons, and neurons of the thalamic nuclei. The staining was conspicuously weak in the CA2 region of the hippocampus and also absent from the marginal zone, where Cajal Retzius cells reside.

Figure 6.

LacZ staining on Fhl1 +/− adult brain. (A) Sagittal section of an E14 Fhl1 +/− mouse facing left. (B and C) Coronal sections (rostral to caudal in left to right) of brains from an E17 Fhl1 +/− mouse and an adult Fhl1 −/y mouse, respectively. The staining pattern shows highest levels in the ventral-most regions of the VZ at E17.5 and pyramidal neurons of the adult hippocampus, identical to that of Fhl1 expression in the wild-type brains. CA1 and CA3: cornu ammonis 1 and 3, GZ: germinal zone, H: hippocampus, Str: striatum.

Finally, we examined the expression of FHL1 mRNA in the human cortex and cerebellum on adult brain. Similar to the mouse, Fhl1 expression is prominent in pyramidal neurons of the gray matter of the cerebral cortex and throughout the cerebellum, especially in Purkinje cells (Fig. 5).

Fhl1 Knockout Mice

We decided to assess FHL1's function as a potential regulator of A-P patterning in mice carrying a null allele of Fhl11 in which Fhl1 has been inactivated by insertion of β-gal into the first exon (Chu, Ruiz-Lozano, et al. 2000). We recognized that although its expression within postmitotic neurons suggested a role in neuronal maturation rather than A-P patterning, this avenue would be worth more detailed study, as Fhl1 previously had no known function in brain development. Animals deficient in Fhl1 do not show any gross anatomical or behavioral abnormalities. The brains of homozygous Fhl1 null mice have normal morphology and are comparable in size and appearance with those of their wild-type littermates (Table 4).

Table 4

Measurements and weight comparison of adult wild-type and Fhl1 knockout littermates

Body weight (g)Brain weight (g)a (cm)b (cm)c (cm)Brain/body ratio (%)A-P cortical length/width (%)
WT44.9(7.7)0.49(0.02)0.93(0.06)1.04(0.04)0.72(0.07)1.1(0.3)89
Fhl1 KO37.9(5.7)0.43(0.03)0.93(0.05)0.97(0.01)0.68(0.08)1.1(0.2)96
Body weight (g)Brain weight (g)a (cm)b (cm)c (cm)Brain/body ratio (%)A-P cortical length/width (%)
WT44.9(7.7)0.49(0.02)0.93(0.06)1.04(0.04)0.72(0.07)1.1(0.3)89
Fhl1 KO37.9(5.7)0.43(0.03)0.93(0.05)0.97(0.01)0.68(0.08)1.1(0.2)96

Note: Average body weights and weights of the entire brain were recorded from adult male mice. The measurements of the cortex were made as depicted in the diagram. Values in parentheses are standard deviations.

graphic

Table 4

Measurements and weight comparison of adult wild-type and Fhl1 knockout littermates

Body weight (g)Brain weight (g)a (cm)b (cm)c (cm)Brain/body ratio (%)A-P cortical length/width (%)
WT44.9(7.7)0.49(0.02)0.93(0.06)1.04(0.04)0.72(0.07)1.1(0.3)89
Fhl1 KO37.9(5.7)0.43(0.03)0.93(0.05)0.97(0.01)0.68(0.08)1.1(0.2)96
Body weight (g)Brain weight (g)a (cm)b (cm)c (cm)Brain/body ratio (%)A-P cortical length/width (%)
WT44.9(7.7)0.49(0.02)0.93(0.06)1.04(0.04)0.72(0.07)1.1(0.3)89
Fhl1 KO37.9(5.7)0.43(0.03)0.93(0.05)0.97(0.01)0.68(0.08)1.1(0.2)96

Note: Average body weights and weights of the entire brain were recorded from adult male mice. The measurements of the cortex were made as depicted in the diagram. Values in parentheses are standard deviations.

graphic

To examine the possibility of more subtle changes in telenephalic development, anterior–posterior regionalization was tested by assessing the expression of known molecular gradients, focusing on those genes defined in this study (Fhl2, Slc25a13, Smoc1, Dkk3, Farp1, Fzd1, Ifitm2, Nfix, Tcf4, and AI463886), or other genes known to mark regional boundaries, that is, Cdh6, CoupTF1, EphA5, Emx1, Emx2, and Otx2 (Simeone et al. 1993; Gulisano et al. 1996; Mallamaci et al. 1996; Suzuki et al. 1997; Mackarehtschian et al. 1999; Rubenstein et al. 1999; Liu et al. 2000) in the knockout animals. In situ hybridization was performed on sagittal sections of E14.5 and E17.5 embryos of Fhl1 knockouts and their wild-type littermates (Fig. 7). No changes in the expression patterns of these genes were detected in the Fhl1 heterozygote or homozygote null mice, suggesting that this gene is not likely an upstream regulator of these factors. Importantly, we detected no alterations in Hes-1 and Hes-5 in Fhl1 knockout mice, suggesting that Notch signaling is compensated and not disrupted with Fhl1 loss of function (Fig. 8). Furthermore, because Fhl2 expression is normal in the Fhl1 null mice, it is unlikely that the basis for normal telencephalic patterning in the Fhl1 null mouse is due to compensation by Fhl2.

Figure 7.

Expression of A-P gradient genes in Fhl1 knockout mice. The images are low-power autoradiographs of sagittal sections. The expression patterns of both known and newly identified genes regionalized along the A-P axis show comparable gradients in the Fhl1 knockout animals compared with their wild-type littermates. Arrows point to the regions of enriched expression in the cortex.

Figure 8.

Expression of genes downstream of Notch1 in Fhl1 knockout mice. Low-power autoradiographs of sagittal sections show that the expression patterns and levels of Hes1, Hes5, and Dll1 appear unaltered in the E14 and E17 mice that are deficient in Fhl1. Dashed lines outline the cerebral cortex.

Discussion

We used microarray analysis followed by in situ hybridization on a large scale to identify a number of genes previously not known to be expressed in anterior–posterior gradients in the developing cortex. These data add to a growing number of known genes with such patterns, and provide an additional foundation for future work on the molecular basis of mammalian forebrain patterning. The enrichment in transcription factors among those with A-P gradients at this developmental stage is particularly interesting in this regard, as it provides additional molecular support for the protomap concept, in which factors located in the dorsal pallium are hypothesized to direct initial regionalization of the cerebral cortex, independent of afferent input (Rakic 1988). The subset of genes with strong A-P gradients that are highly expressed or restricted to the VZ and SVZ, such as Sfrp1, Smoc1, Slc25a13, Gart, and the previously uncharacterized A1463886 are most likely to play direct roles in the process of intrinsic patterning within the germinal neuroepithelia.

It is also noteworthy to mention that of the 15 genes that were identified and confirmed in this study, 4 are known to be involved in the Wnt pathway, Fhl2, Fzd1, Dkk3, and Sfrp1 and 4 encode transcription factors/regulators, Fhl1, Fhl2, Tcf4, and Nfix. The use of genetic screening methods here provides a group of genes that were identified using unbiased sampling methods, without a predetermined focus on putative functions or homologies. These data will be useful for assessing putative roles in A-P patterning, as we did in the Fhl1 knockout mouse and have begun in a preliminary manner in another patterning mutant, the foxg1 heterozygous mouse (Levitt, unpublished data).

Wnt Pathway and Cortical Regionalization

Wnt signaling is well known to be critical in differentiation and patterning of the developing embryo (Cadigan and Nusse 1997). Our study identified novel expression patterns of 2 genes involved in the Wnt pathway, Dkk3 (Glinka et al. 1998; Krupnik et al. 1999; Monaghan et al. 1999) and Fzd1 (Vinson et al. 1989; Bhanot et al. 1996), which were expressed in a caudal-to-rostral gradient, as well as Sfrp1 (Finch et al. 1997; Leyns et al. 1997; Rattner et al. 1997) and Fhl2 (Peifer and Polakis 2000; Wei et al. 2003; Labalette et al. 2004) with a rostral-to-caudal gradient. Because Sfrp1 and Dkk3 are Wnt antagonists (Finch et al. 1997; Leyns et al. 1997; Rattner et al. 1997; Caricasole et al. 2003; Suwa et al. 2003), and Fzd1 and Fhl2 are Wnt agonists (Vinson et al. 1989; Bhanot et al. 1996; Peifer and Polakis 2000; Wei et al. 2003; Labalette et al. 2004), the expression data does not support a simple model for Wnt signaling (e.g., Soley proneurogenic). The biological significance of these Wnt antagonists (Zorn 2001; Jones and Jomary 2002; Diep et al. 2004; Roman-Roman et al. 2004), expressed in specific zones of the developing cerebral wall with strong gradients along the rostrocaudal axis, requires further investigation.

Known and Novel Transcription Factors Including Fhl1 and Fhl2

A growing number of transcription factors with identified roles in CNS development have been identified (Miyashita-Lin et al. 1999; Zhou, Qiu, et al. 1999; Zhou et al. 2001; Bishop et al. 2000, 2002; Mallamaci et al. 2000; Muzio and Mallamaci 2003). The present study revealed several of these that have previously characterized roles, including Tcf4 (Pscherer et al. 1996; Dorflinger et al. 1999) and Nfix (Chaudhry et al. 1997). Along with other basic-helix-loop-helix proteins, (Johnson et al. 1992; Kageyama et al. 1997; Nieto et al. 2001), Tcf4 may also regulate lineage fates in regionalizing the developing brain (Liu et al. 1998).

The novel finding of graded expression patterns of Fhl1 and Fhl2 piqued our interest for further study because they were not previously identified in embryonic brain. We examined whether Fhl2 expression might be impacted by Fhl1 loss because Fhl and Fhl2 appear to compensate for each other in the heart (Chu, Bardwell, et al. 2000). No change in Fhl2 expression was observed in Fhl1 knockouts nor was any change detected in the expression of other genes expressed in known developmental gradients in the dorsal telencephalon. It is notable that Fhl1 is conserved from Caenorhabditis elegans to humans (Hobert et al. 1999; Lee et al. 1999; Ng et al. 2001). Aside from the fact that its sequence contains an RBP-J binding site and a nuclear localization signal (Lee et al. 1999) not much is known about its function in the brain. Here we show that Fhl1 is concentrated mainly in postmitotic pyramidal neurons, suggesting its possible role in the maintenance of a mature neuronal phenotype, rather than influencing patterning gradients per se. Until this study, Fhl2 was also not known to be involved in CNS development. Based on its expression pattern in embryonic and perinatal telencephalon and its association with the Wnt pathway, as well as its known transcriptional regulatory activities outside the CNS (Du et al. 2002; McLoughlin et al. 2002), Fhl2 will be an attractive candidate for further investigation.

Methodological Considerations and Limitations

Although several previously reported and novel genes showing expression gradients in the developing dorsal telencephalon were identified, there are methodological limitations. A number of genes that were identified in the array data analysis were expressed in non-CNS tissue or in neighboring regions of the CNS that were likely the result of imprecise tissue dissection, emphasizing the value of anatomical validation, as performed here. The recent emergence of new techniques for preparing distinct neuronal types from developing brain permits future studies to be able to examine different cell types separately (Arlotta et al. 2005; Lobo et al. 2006). As technologies become available for single cell studies and isolating specific cell types from tissue samples, more definitive array studies can be performed (Dougherty and Geschwind 2005). Still the combination of genomic screening followed by anatomical confirmation (Geschwind et al. 2001; Zirlinger et al. 2001; Karsten et al. 2003) as performed here is valuable, as evidenced by the genes identified in this study.

Probe templates were kind gifts provided by Dr John Rubenstein for Otx2, Emx1, and Emx2; Dr Elizabeth Grove for Cdh6; and Dr Harley Kornblum for GFAP. Authors would like to thank Farah Sheik for providing us with Fhl1 knockout mice, Zugen Chen for providing the clones, and Matt Schibler for assistance with confocal microscopy. We also thank Sam Levitt for his technical assistance with the dissections. This work was supported by grants NIH 5T32 MH19384-08 (L.C.K.), MH60233 (D.H.G.), and NICHD P30 HD15052 to the Vanderbilt Kennedy center (PL). Conflict of Interest: None declared.

References

Acampora
D
Avantaggiato
V
Tuorto
F
Barone
P
Perera
M
Choo
D
Wu
D
Corte
G
Simeone
A
Differential transcriptional control as the major molecular event in generating Otx1-/- and Otx2-/- divergent phenotypes
Development
1999
, vol. 
126
 (pg. 
1417
-
1426
)
Acampora
D
Barone
P
Simeone
A
Otx genes in corticogenesis and brain development
Cereb Cortex
1999
, vol. 
9
 (pg. 
533
-
542
)
Arber
S
Caroni
P
Specificity of single LIM motifs in targeting and LIM/LIM interactions in situ
Genes Dev
1996
, vol. 
10
 (pg. 
289
-
300
)
Arimatsu
Y
Miyamoto
M
Nihonmatsu
I
Hirata
K
Uratani
Y
Hatanaka
Y
Takiguchi-Hayashi
K
Early regional specification for a molecular neuronal phenotype in the rat neocortex
Proc Natl Acad Sci USA
1992
, vol. 
89
 (pg. 
8879
-
8883
)
Arlotta
P
Molyneaux
BJ
Chen
J
Inoue
J
Kominami
R
Macklis
JD
Neuronal subtype-specific genes that control corticospinal motor neuron development in vivo
Neuron
2005
, vol. 
45
 (pg. 
207
-
221
)
Augustine
C
Gunnersen
J
Spirkoska
V
Tan
SS
Place- and time-dependent expression of mouse sFRP-1 during development of the cerebral neocortex
Mech Dev
2001
, vol. 
109
 (pg. 
395
-
397
)
Ayadi
A
Suelves
M
Dolle
P
Wasylyk
B
Net, an Ets ternary complex transcription factor, is expressed in sites of vasculogenesis, angiogenesis, and chondrogenesis during mouse development
Mech Dev
2001
, vol. 
102
 (pg. 
205
-
208
)
Barbe
MF
Levitt
P
The early commitment of fetal neurons to the limbic cortex
J Neurosci
1991
, vol. 
11
 (pg. 
519
-
533
)
Barbe
MF
Levitt
P
Attraction of specific thalamic input by cerebral grafts depends on the molecular identity of the implant
Proc Natl Acad Sci USA
1992
, vol. 
89
 (pg. 
3706
-
3710
)
Barbe
MF
Levitt
P
Age-dependent specification of the corticocortical connections of cerebral grafts
J Neurosci
1995
, vol. 
15
 (pg. 
1819
-
1834
)
Beck
KD
Powell-Braxton
L
Widmer
HR
Valverde
J
Hefti
F
Igf1 gene disruption results in reduced brain size, CNS hypomyelination, and loss of hippocampal granule and striatal parvalbumin-containing neurons
Neuron
1995
, vol. 
14
 (pg. 
717
-
730
)
Becker
W
Weber
Y
Wetzel
K
Eirmbter
K
Tejedor
FJ
Joost
HG
Sequence characteristics, subcellular localization, and substrate specificity of DYRK-related kinases, a novel family of dual specificity protein kinases
J Biol Chem
1998
, vol. 
273
 (pg. 
25893
-
25902
)
Beckerle
MC
Zyxin: zinc fingers at sites of cell adhesion
Bioessays
1997
, vol. 
19
 (pg. 
949
-
957
)
Bhanot
P
Brink
M
Samos
CH
Hsieh
JC
Wang
Y
Macke
JP
Andrew
D
Nathans
J
Nusse
R
A new member of the frizzled family from Drosophila functions as a Wingless receptor
Nature
1996
, vol. 
382
 (pg. 
225
-
230
)
Bishop
KM
Goudreau
G
O'Leary
DD
Regulation of area identity in the mammalian neocortex by Emx2 and Pax6
Science
2000
, vol. 
288
 (pg. 
344
-
349
)
Bishop
KM
Rubenstein
JL
O'Leary
DD
Distinct actions of Emx1, Emx2, and Pax6 in regulating the specification of areas in the developing neocortex
J Neurosci
2002
, vol. 
22
 (pg. 
7627
-
7638
)
Bodine
PV
Zhao
W
Kharode
YP
Bex
FJ
Lambert
AJ
Goad
MB
Gaur
T
Stein
GS
Lian
JB
Komm
BS
The Wnt antagonist secreted frizzled-related protein-1 is a negative regulator of trabecular bone formation in adult mice
Mol Endocrinol
2004
, vol. 
18
 (pg. 
1222
-
1237
)
Brodsky
G
Barnes
T
Bleskan
J
Becker
L
Cox
M
Patterson
D
The human GARS-AIRS-GART gene encodes two proteins which are differentially expressed during human brain development and temporally overexpressed in cerebellum of individuals with Down syndrome
Hum Mol Genet
1997
, vol. 
6
 (pg. 
2043
-
2050
)
Cadigan
KM
Nusse
R
Wnt signaling: a common theme in animal development
Genes Dev
1997
, vol. 
11
 (pg. 
3286
-
3305
)
Campbell
LE
Proud
CG
Differing substrate specificities of members of the DYRK family of arginine-directed protein kinases
FEBS Lett
2002
, vol. 
510
 (pg. 
31
-
36
)
Caricasole
A
Ferraro
T
Iacovelli
L
Barletta
E
Caruso
A
Melchiorri
D
Terstappen
GC
Nicoletti
F
Functional characterization of WNT7A signaling in PC12 cells: interaction with A FZD5 × LRP6 receptor complex and modulation by Dickkopf proteins
J Biol Chem
2003
, vol. 
278
 (pg. 
37024
-
37031
)
Chan
KK
Tsui
SK
Lee
SM
Luk
SC
Liew
CC
Fung
KP
Waye
MM
Lee
CY
Molecular cloning and characterization of FHL2, a novel LIM domain protein preferentially expressed in human heart
Gene
1998
, vol. 
210
 (pg. 
345
-
350
)
Chaudhry
AZ
Lyons
GE
Gronostajski
RM
Expression patterns of the four nuclear factor I genes during mouse embryogenesis indicate a potential role in development
Dev Dyn
1997
, vol. 
208
 (pg. 
313
-
325
)
Chen
D
Xu
W
Bales
E
Colmenares
C
Conacci-Sorrell
M
Ishii
S
Stavnezer
E
Campisi
J
Fisher
DE
Ben-Ze'ev
A
, et al. 
SKI activates Wnt/beta-catenin signaling in human melanoma
Cancer Res
2003
, vol. 
63
 (pg. 
6626
-
6634
)
Chu
PH
Bardwell
WM
Gu
Y
Ross
J
Jr
Chen
J
FHL2 (SLIM3) is not essential for cardiac development and function
Mol Cell Biol
2000
, vol. 
20
 (pg. 
7460
-
7462
)
Chu
PH
Ruiz-Lozano
P
Zhou
Q
Cai
C
Chen
J
Expression patterns of FHL/SLIM family members suggest important functional roles in skeletal muscle and cardiovascular system
Mech Dev
2000
, vol. 
95
 (pg. 
259
-
265
)
Cohen-Tannoudji
M
Babinet
C
Wassef
M
Early determination of a mouse somatosensory cortex marker
Nature
1994
, vol. 
368
 (pg. 
460
-
463
)
Dawid
IB
Breen
JJ
Toyama
R
LIM domains: multiple roles as adapters and functional modifiers in protein interactions
Trends Genet
1998
, vol. 
14
 (pg. 
156
-
162
)
De Carlos
JA
O'Leary
DD
Growth and targeting of subplate axons and establishment of major cortical pathways
J Neurosci
1992
, vol. 
12
 (pg. 
1194
-
1211
)
Diep
DB
Hoen
N
Backman
M
Machon
O
Krauss
S
Characterisation of the Wnt antagonists and their response to conditionally activated Wnt signalling in the developing mouse forebrain
Brain Res Dev Brain Res
2004
, vol. 
153
 (pg. 
261
-
270
)
Dorflinger
U
Pscherer
A
Moser
M
Rummele
P
Schule
R
Buettner
R
Activation of somatostatin receptor II expression by transcription factors MIBP1 and SEF-2 in the murine brain
Mol Cell Biol
1999
, vol. 
19
 (pg. 
3736
-
3747
)
Dougherty
JD
Geschwind
DH
Progress in realizing the promise of microarrays in systems neurobiology
Neuron
2005
, vol. 
45
 (pg. 
183
-
185
)
Du
X
Hublitz
P
Gunther
T
Wilhelm
D
Englert
C
Schule
R
The LIM-only coactivator FHL2 modulates WT1 transcriptional activity during gonadal differentiation
Biochim Biophys Acta
2002
, vol. 
1577
 (pg. 
93
-
101
)
Felbor
U
Kessler
B
Mothes
W
Goebel
HH
Ploegh
HL
Bronson
RT
Olsen
BR
Neuronal loss and brain atrophy in mice lacking cathepsins B and L
Proc Natl Acad Sci USA
2002
, vol. 
99
 (pg. 
7883
-
7888
)
Finch
PW
He
X
Kelley
MJ
Uren
A
Schaudies
RP
Popescu
NC
Rudikoff
S
Aaronson
SA
Varmus
HE
Rubin
JS
Purification and molecular cloning of a secreted, Frizzled-related antagonist of Wnt action
Proc Natl Acad Sci USA
1997
, vol. 
94
 (pg. 
6770
-
6775
)
Gasteiger
E
Gattiker
A
Hoogland
C
Ivanyi
I
Appel
RD
Bairoch
A
ExPASy: the proteomics server for in-depth protein knowledge and analysis
Nucleic Acids Res
2003
, vol. 
31
 (pg. 
3784
-
3788
)
Genini
M
Schwalbe
P
Scholl
FA
Remppis
A
Mattei
MG
Schafer
BW
Subtractive cloning and characterization of DRAL, a novel LIM-domain protein down-regulated in rhabdomyosarcoma
DNA Cell Biol
1997
, vol. 
16
 (pg. 
433
-
442
)
Gerber
WV
Yatskievych
TA
Antin
PB
Correia
KM
Conlon
RA
Krieg
PA
The RNA-binding protein gene, hermes, is expressed at high levels in the developing heart
Mech Dev
1999
, vol. 
80
 (pg. 
77
-
86
)
Geschwind
DH
Ou
J
Easterday
MC
Dougherty
JD
Jackson
RL
Chen
Z
Antoine
H
Terskikh
A
Weissman
IL
Nelson
SF
, et al. 
A genetic analysis of neural progenitor differentiation
Neuron
2001
, vol. 
29
 (pg. 
325
-
339
)
Glinka
A
Wu
W
Delius
H
Monaghan
AP
Blumenstock
C
Niehrs
C
Dickkopf-1 is a member of a new family of secreted proteins and functions in head induction
Nature
1998
, vol. 
391
 (pg. 
357
-
362
)
Grove
EA
Tole
S
Limon
J
Yip
L
Ragsdale
CW
The hem of the embryonic cerebral cortex is defined by the expression of multiple Wnt genes and is compromised in Gli3-deficient mice
Development
1998
, vol. 
125
 (pg. 
2315
-
2325
)
Gulisano
M
Broccoli
V
Pardini
C
Boncinelli
E
Emx1 and Emx2 show different patterns of expression during proliferation and differentiation of the developing cerebral cortex in the mouse
Eur J Neurosci
1996
, vol. 
8
 (pg. 
1037
-
1050
)
Hobert
O
Moerman
DG
Clark
KA
Beckerle
MC
Ruvkun
G
A conserved LIM protein that affects muscular adherens junction integrity and mechanosensory function in Caenorhabditis elegans
J Cell Biol
1999
, vol. 
144
 (pg. 
45
-
57
)
Hodge
RD
D'Ercole
AJ
O'Kusky
JR
Insulin-like growth factor-I accelerates the cell cycle by decreasing G1 phase length and increases cell cycle reentry in the embryonic cerebral cortex
J Neurosci
2004
, vol. 
24
 (pg. 
10201
-
10210
)
Johnson
JE
Zimmerman
K
Saito
T
Anderson
DJ
Induction and repression of mammalian achaete-scute homologue (MASH) gene expression during neuronal differentiation of P19 embryonal carcinoma cells
Development
1992
, vol. 
114
 (pg. 
75
-
87
)
Jones
SE
Jomary
C
Secreted frizzled-related proteins: searching for relationships and patterns
Bioessays
2002
, vol. 
24
 (pg. 
811
-
820
)
Kageyama
R
Ishibashi
M
Takebayashi
K
Tomita
K
bHLH transcription factors and mammalian neuronal differentiation
Int J Biochem Cell Biol
1997
, vol. 
29
 (pg. 
1389
-
1399
)
Karsten
SL
Kudo
LC
Jackson
R
Sabatti
C
Kornblum
HI
Geschwind
DH
Global analysis of gene expression in neural progenitors reveals specific cell-cycle, signaling, and metabolic networks
Dev Biol
2003
, vol. 
261
 (pg. 
165
-
182
)
Karsten
SL
Van Deerlin
VM
Sabatti
C
Gill
LH
Geschwind
DH
An evaluation of tyramide signal amplification and archived fixed and frozen tissue in microarray gene expression analysis
Nucleic Acids Res
2002
, vol. 
30
 pg. 
E4
 
Kim
AS
Lowenstein
DH
Pleasure
SJ
Wnt receptors and Wnt inhibitors are expressed in gradients in the developing telencephalon
Mech Dev
2001
, vol. 
103
 (pg. 
167
-
172
)
Kobayashi
K
Sinasac
DS
Iijima
M
Boright
AP
Begum
L
Lee
JR
Yasuda
T
Ikeda
S
Hirano
R
Terazono
H
, et al. 
The gene mutated in adult-onset type II citrullinaemia encodes a putative mitochondrial carrier protein
Nat Genet
1999
, vol. 
22
 (pg. 
159
-
163
)
Kohama
K
Tsukamoto
Y
Furuya
M
Okamura
K
Tanaka
H
Miki
N
Taira
E
Molecular cloning and analysis of the mouse gicerin gene
Neurochem Int
2005
, vol. 
46
 (pg. 
465
-
470
)
Koyano
Y
Kawamoto
T
Kikuchi
A
Shen
M
Kuruta
Y
Tsutsumi
S
Fujimoto
K
Noshiro
M
Fujii
K
Kato
Y
Chondrocyte-derived ezrin-like domain containing protein (CDEP), a rho guanine nucleotide exchange factor, is inducible in chondrocytes by parathyroid hormone and cyclic AMP and has transforming activity in NIH3T3 cells
Osteoarthritis Cartilage
2001
, vol. 
9
 
Suppl A
(pg. 
S64
-
S68
)
Koyano
Y
Kawamoto
T
Shen
M
Yan
W
Noshiro
M
Fujii
K
Kato
Y
Molecular cloning and characterization of CDEP, a novel human protein containing the ezrin-like domain of the band 4.1 superfamily and the Dbl homology domain of Rho guanine nucleotide exchange factors
Biochem Biophys Res Commun
1997
, vol. 
241
 (pg. 
369
-
375
)
Krupnik
VE
Sharp
JD
Jiang
C
Robison
K
Chickering
TW
Amaravadi
L
Brown
DE
Guyot
D
Mays
G
Leiby
K
, et al. 
Functional and structural diversity of the human Dickkopf gene family
Gene
1999
, vol. 
238
 (pg. 
301
-
313
)
Labalette
C
Renard
CA
Neuveut
C
Buendia
MA
Wei
Y
Interaction and functional cooperation between the LIM protein FHL2, CBP/p300, and beta-catenin
Mol Cell Biol
2004
, vol. 
24
 (pg. 
10689
-
10702
)
Lange
UC
Saitou
M
Western
PS
Barton
SC
Surani
MA
The fragilis interferon-inducible gene family of transmembrane proteins is associated with germ cell specification in mice
BMC Dev Biol
2003
, vol. 
3
 pg. 
1
 
Lappe-Siefke
C
Goebbels
S
Gravel
M
Nicksch
E
Lee
J
Braun
PE
Griffiths
IR
Nave
KA
Disruption of Cnp1 uncouples oligodendroglial functions in axonal support and myelination
Nat Genet
2003
, vol. 
33
 (pg. 
366
-
374
)
Lee
SM
Li
HY
Ng
EK
Or
SM
Chan
KK
Kotaka
M
Chim
SS
Tsui
SK
Waye
MM
Fung
KP
, et al. 
Characterization of a brain-specific nuclear LIM domain protein (FHL1B) which is an alternatively spliced variant of FHL1
Gene
1999
, vol. 
237
 (pg. 
253
-
263
)
Lee
SM
Tsui
SK
Chan
KK
Garcia-Barcelo
M
Waye
MM
Fung
KP
Liew
CC
Lee
CY
Chromosomal mapping, tissue distribution and cDNA sequence of four-and-a-half LIM domain protein 1 (FHL1)
Gene
1998
, vol. 
216
 (pg. 
163
-
170
)
Levitt
P
Barbe
MF
Eagleson
KL
Patterning and specification of the cerebral cortex
Annu Rev Neurosci
1997
, vol. 
20
 (pg. 
1
-
24
)
Leyns
L
Bouwmeester
T
Kim
SH
Piccolo
S
De Robertis
EM
Frzb-1 is a secreted antagonist of Wnt signaling expressed in the Spemann organizer
Cell
1997
, vol. 
88
 (pg. 
747
-
756
)
Liu
Q
Dwyer
ND
O'Leary
DD
Differential expression of COUP-TFI, CHL1, and two novel genes in developing neocortex identified by differential display PCR
J Neurosci
2000
, vol. 
20
 (pg. 
7682
-
7690
)
Liu
Y
Ray
SK
Yang
XQ
Luntz-Leybman
V
Chiu
IM
A splice variant of E2-2 basic helix-loop-helix protein represses the brain-specific fibroblast growth factor 1 promoter through the binding to an imperfect E-box
J Biol Chem
1998
, vol. 
273
 (pg. 
19269
-
19276
)
Lobo
MK
Karsten
SL
Gray
M
Geschwind
DH
Yang
XW
FACS-array profiling of striatal projection neuron subtypes in juvenile and adult mouse brains
Nat Neurosci
2006
, vol. 
9
 (pg. 
443
-
452
)
Lumsden
A
Neural development. A ‘LIM code’ for motor neurons?
Curr Biol
1995
, vol. 
5
 (pg. 
491
-
495
)
Mackarehtschian
K
Lau
CK
Caras
I
McConnell
SK
Regional differences in the developing cerebral cortex revealed by ephrin-A5 expression
Cereb Cortex
1999
, vol. 
9
 (pg. 
601
-
610
)
Mallamaci
A
Di Blas
E
Briata
P
Boncinelli
E
Corte
G
OTX2 homeoprotein in the developing central nervous system and migratory cells of the olfactory area
Mech Dev
1996
, vol. 
58
 (pg. 
165
-
178
)
Mallamaci
A
Muzio
L
Chan
CH
Parnavelas
J
Boncinelli
E
Area identity shifts in the early cerebral cortex of Emx2-/- mutant mice
Nat Neurosci
2000
, vol. 
3
 (pg. 
679
-
686
)
Matsui
Y
Zsebo
KM
Hogan
BL
Embryonic expression of a haematopoietic growth factor encoded by the Sl locus and the ligand for c-kit
Nature
1990
, vol. 
347
 (pg. 
667
-
669
)
Matsuo
R
Ochiai
W
Nakashima
K
Taga
T
A new expression cloning strategy for isolation of substrate-specific kinases by using phosphorylation site-specific antibody
J Immunol Methods
2001
, vol. 
247
 (pg. 
141
-
151
)
McConnell
SK
Kaznowski
CE
Cell cycle dependence of laminar determination in developing neocortex
Science
1991
, vol. 
254
 (pg. 
282
-
285
)
McLoughlin
P
Ehler
E
Carlile
G
Licht
JD
Schafer
BW
The LIM-only protein DRAL/FHL2 interacts with and is a corepressor for the promyelocytic leukemia zinc finger protein
J Biol Chem
2002
, vol. 
277
 (pg. 
37045
-
37053
)
Miyashita-Lin
EM
Hevner
R
Wassarman
KM
Martinez
S
Rubenstein
JL
Early neocortical regionalization in the absence of thalamic innervation
Science
1999
, vol. 
285
 (pg. 
906
-
909
)
Monaghan
AP
Kioschis
P
Wu
W
Zuniga
A
Bock
D
Poustka
A
Delius
H
Niehrs
C
Dickkopf genes are co-ordinately expressed in mesodermal lineages
Mech Dev
1999
, vol. 
87
 (pg. 
45
-
56
)
Morgan
MJ
Madgwick
AJ
Slim defines a novel family of LIM-proteins expressed in skeletal muscle
Biochem Biophys Res Commun
1996
, vol. 
225
 (pg. 
632
-
638
)
Morgan
MJ
Madgwick
AJ
Charleston
B
Pell
JM
Loughna
PT
The developmental regulation of a novel muscle LIM-protein
Biochem Biophys Res Commun
1995
, vol. 
212
 (pg. 
840
-
846
)
Motro
B
Wojtowicz
JM
Bernstein
A
van der Kooy
D
Steel mutant mice are deficient in hippocampal learning but not long-term potentiation
Proc Natl Acad Sci USA
1996
, vol. 
93
 (pg. 
1808
-
1813
)
Muzio
L
DiBenedetto
B
Stoykova
A
Boncinelli
E
Gruss
P
Mallamaci
A
Conversion of cerebral cortex into basal ganglia in Emx2(-/-) Pax6(Sey/Sey) double-mutant mice
Nat Neurosci
2002
, vol. 
5
 (pg. 
737
-
745
)
Muzio
L
DiBenedetto
B
Stoykova
A
Boncinelli
E
Gruss
P
Mallamaci
A
Emx2 and Pax6 control regionalization of the pre-neuronogenic cortical primordium
Cereb Cortex
2002
, vol. 
12
 (pg. 
129
-
139
)
Muzio
L
Mallamaci
A
Emx1, emx2 and pax6 in specification, regionalization and arealization of the cerebral cortex
Cereb Cortex
2003
, vol. 
13
 (pg. 
641
-
647
)
Ng
EK
Lee
SM
Li
HY
Ngai
SM
Tsui
SK
Waye
MM
Lee
CY
Fung
KP
Characterization of tissue-specific LIM domain protein (FHL1C) which is an alternatively spliced isoform of a human LIM-only protein (FHL1)
J Cell Biochem
2001
, vol. 
82
 (pg. 
1
-
10
)
Nieto
M
Schuurmans
C
Britz
O
Guillemot
F
Neural bHLH genes control the neuronal versus glial fate decision in cortical progenitors
Neuron
2001
, vol. 
29
 (pg. 
401
-
413
)
Nunes
I
Tovmasian
LT
Silva
RM
Burke
RE
Goff
SP
Pitx3 is required for development of substantia nigra dopaminergic neurons
Proc Natl Acad Sci USA
2003
, vol. 
100
 (pg. 
4245
-
4250
)
O'Brien
SP
Seipel
K
Medley
QG
Bronson
R
Segal
R
Streuli
M
Skeletal muscle deformity and neuronal disorder in Trio exchange factor-deficient mouse embryos
Proc Natl Acad Sci USA
2000
, vol. 
97
 (pg. 
12074
-
12078
)
Peifer
M
Polakis
P
Wnt signaling in oncogenesis and embryogenesis—a look outside the nucleus
Science
2000
, vol. 
287
 (pg. 
1606
-
1609
)
Pscherer
A
Dorflinger
U
Kirfel
J
Gawlas
K
Ruschoff
J
Buettner
R
Schule
R
The helix-loop-helix transcription factor SEF-2 regulates the activity of a novel initiator element in the promoter of the human somatostatin receptor II gene
EMBO J
1996
, vol. 
15
 (pg. 
6680
-
6690
)
Rakic
P
Specification of cerebral cortical areas
Science
1988
, vol. 
241
 (pg. 
170
-
176
)
Rattner
A
Hsieh
JC
Smallwood
PM
Gilbert
DJ
Copeland
NG
Jenkins
NA
Nathans
J
A family of secreted proteins contains homology to the cysteine-rich ligand-binding domain of frizzled receptors
Proc Natl Acad Sci USA
1997
, vol. 
94
 (pg. 
2859
-
2863
)
Roman-Roman
S
Shi
DL
Stiot
V
Hay
E
Vayssiere
B
Garcia
T
Baron
R
Rawadi
G
Murine Frizzled-1 behaves as an antagonist of the canonical Wnt/beta-catenin signaling
J Biol Chem
2004
, vol. 
279
 (pg. 
5725
-
5733
)
Rost
B
PHD: predicting one-dimensional protein structure by profile-based neural networks
Methods Enzymol
1996
, vol. 
266
 (pg. 
525
-
539
)
Rost
B
Fariselli
P
Casadio
R
Topology prediction for helical transmembrane proteins at 86% accuracy
Protein Sci
1996
, vol. 
5
 (pg. 
1704
-
1718
)
Rost
B
Sander
C
Prediction of protein secondary structure at better than 70% accuracy
J Mol Biol
1993
, vol. 
232
 (pg. 
584
-
599
)
Rubenstein
JL
Anderson
S
Shi
L
Miyashita-Lin
E
Bulfone
A
Hevner
R
Genetic control of cortical regionalization and connectivity
Cereb Cortex
1999
, vol. 
9
 (pg. 
524
-
532
)
Sakagami
H
Nishimura
H
Saito
R
Kondo
H
Transient up-regulation of elongation factor-2 kinase (Ca2+/calmodulin-dependent protein kinase III) messenger RNA in developing mouse brain
Neurosci Lett
2002
, vol. 
330
 (pg. 
41
-
44
)
Sansom
SN
Hebert
JM
Thammongkol
U
Smith
J
Nisbet
G
Surani
MA
McConnell
SK
Livesey
FJ
Genomic characterisation of a Fgf-regulated gradient-based neocortical protomap
Development
2005
, vol. 
132
 (pg. 
3947
-
3961
)
Schmidt
A
Tief
K
Foletti
A
Hunziker
A
Penna
D
Hummler
E
Beermann
F
lacZ transgenic mice to monitor gene expression in embryo and adult
Brain Res Brain Res Protoc
1998
, vol. 
3
 (pg. 
54
-
60
)
Simeone
A
Acampora
D
Mallamaci
A
Stornaiuolo
A
D'Apice
MR
Nigro
V
Boncinelli
E
A vertebrate gene related to orthodenticle contains a homeodomain of the bicoid class and demarcates anterior neuroectoderm in the gastrulating mouse embryo
EMBO J
1993
, vol. 
12
 (pg. 
2735
-
2747
)
Suwa
T
Chen
M
Hawks
CL
Hornsby
PJ
Zonal expression of dickkopf-3 and components of the Wnt signalling pathways in the human adrenal cortex
J Endocrinol
2003
, vol. 
178
 (pg. 
149
-
158
)
Suzuki
SC
Inoue
T
Kimura
Y
Tanaka
T
Takeichi
M
Neuronal circuits are subdivided by differential expression of type-II classic cadherins in postnatal mouse brains
Mol Cell Neurosci
1997
, vol. 
9
 (pg. 
433
-
447
)
Taira
E
Kohama
K
Tsukamoto
Y
Okumura
S
Miki
N
Characterization of Gicerin/MUC18/CD146 in the rat nervous system
J Cell Physiol
2004
, vol. 
198
 (pg. 
377
-
387
)
Taira
E
Kohama
K
Tsukamoto
Y
Okumura
S
Miki
N
Gicerin/CD146 is involved in neurite extension of NGF-treated PC12 cells
J Cell Physiol
2005
, vol. 
204
 (pg. 
632
-
637
)
Taira
E
Tsukamoto
Y
Kohama
K
Maeda
M
Kiyama
H
Miki
N
Expression and involvement of gicerin, a cell adhesion molecule, in the development of chick optic tectum
J Neurochem
2004
, vol. 
88
 (pg. 
891
-
899
)
Tanaka
SS
Matsui
Y
Developmentally regulated expression of mil-1 and mil-2, mouse interferon-induced transmembrane protein like genes, during formation and differentiation of primordial germ cells
Mech Dev
2002
, vol. 
119
 
Suppl 1
(pg. 
S261
-
S267
)
Taniguchi
Y
Furukawa
T
Tun
T
Han
H
Honjo
T
LIM protein KyoT2 negatively regulates transcription by association with the RBP-J DNA-binding protein
Mol Cell Biol
1998
, vol. 
18
 (pg. 
644
-
654
)
van den Munckhof
P
Luk
KC
Ste-Marie
L
Montgomery
J
Blanchet
PJ
Sadikot
AF
Drouin
J
Pitx3 is required for motor activity and for survival of a subset of midbrain dopaminergic neurons
Development
2003
, vol. 
130
 (pg. 
2535
-
2542
)
Vannahme
C
Smyth
N
Miosge
N
Gosling
S
Frie
C
Paulsson
M
Maurer
P
Hartmann
U
Characterization of SMOC-1, a novel modular calcium-binding protein in basement membranes
J Biol Chem
2002
, vol. 
277
 (pg. 
37977
-
37986
)
Vinson
CR
Conover
S
Adler
PN
A Drosophila tissue polarity locus encodes a protein containing seven potential transmembrane domains
Nature
1989
, vol. 
338
 (pg. 
263
-
264
)
Wei
Y
Renard
CA
Labalette
C
Wu
Y
Levy
L
Neuveut
C
Prieur
X
Flajolet
M
Prigent
S
Buendia
MA
Identification of the LIM protein FHL2 as a coactivator of beta-catenin
J Biol Chem
2003
, vol. 
278
 (pg. 
5188
-
5194
)
Woods
YL
Cohen
P
Becker
W
Jakes
R
Goedert
M
Wang
X
Proud
CG
The kinase DYRK phosphorylates protein-synthesis initiation factor eIF2Bepsilon at Ser539 and the microtubule-associated protein tau at Thr212: potential role for DYRK as a glycogen synthase kinase 3-priming kinase
Biochem J
2001
, vol. 
355
 (pg. 
609
-
615
)
Zhou
C
Qiu
Y
Pereira
FA
Crair
MC
Tsai
SY
Tsai
MJ
The nuclear orphan receptor COUP-TFI is required for differentiation of subplate neurons and guidance of thalamocortical axons
Neuron
1999
, vol. 
24
 (pg. 
847
-
859
)
Zhou
C
Tsai
SY
Tsai
MJ
COUP-TFI: an intrinsic factor for early regionalization of the neocortex
Genes Dev
2001
, vol. 
15
 (pg. 
2054
-
2059
)
Zhou
Q
Ruiz-Lozano
P
Martone
ME
Chen
J
Cypher, a striated muscle-restricted PDZ and LIM domain-containing protein, binds to alpha-actinin-2 and protein kinase C
J Biol Chem
1999
, vol. 
274
 (pg. 
19807
-
19813
)
Zirlinger
M
Kreiman
G
Anderson
DJ
Amygdala-enriched genes identified by microarray technology are restricted to specific amygdaloid subnuclei
Proc Natl Acad Sci USA
2001
, vol. 
98
 (pg. 
5270
-
5275
)
Zorn
AM
Wnt signalling: antagonistic Dickkopfs
Curr Biol
2001
, vol. 
11
 (pg. 
R592
-
R595
)