Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2010 Nov 19.
Published in final edited form as: Biochem Pharmacol. 2007 Nov 22;75(5):1129–1160. doi: 10.1016/j.bcp.2007.11.006

Gene Expression Profiling Elucidates a Specific Role for RARγ in the Retinoic Acid Induced Differentiation of F9 Teratocarcinoma Stem Cells

Dan Su 1, Lorraine J Gudas 1,*
PMCID: PMC2988767  NIHMSID: NIHMS242556  PMID: 18164278

Abstract

The biological effects of all-trans-retinoic acid (RA), a major active metabolite of retinol, are mainly mediated through its interactions with retinoic acid receptor (RARs α, β, γ) and retinoid X receptor (RXRs α, β, γ) heterodimers. RAR/RXR heterodimers activate transcription by binding to RA-response elements (RAREs or RXREs) in the promoters of primary target genes. Murine F9 teratocarcinoma stem cells have been widely used as a model for cellular differentiation and RA signaling during embryonic development. We identified and characterized genes that are differentially expressed in F9 wild type (Wt) and F9 RAR γ−/− cells, with and without RA treatment, through the use of oligonucleotide based microarrays. Our data indicate that RARγ, in the absence of exogenous RA, modulates gene expression. Genes such as Sfrp2, Tie1, Fbp2, Emp1, and Emp3 exhibited higher transcript levels in RA treated Wt, RARα−/− and RARβ2−/− lines than in RA-treated RARγ−/− cells, and represent specific RARγ targets. Other genes, such as Runx1, were expressed at lower levels in both F9 RARβ2−/− and RARγ−/− cell lines then in F9 Wt and RARα−/−. Genes specifically induced by RA at 6h with the protein synthesis inhibitor cycloheximide in F9 Wt, but not in RARγ−/− cells, included Hoxa3, Hoxa5, Gas1, Cyp26a1, Sfrp2, Fbp2, and Emp1. These genes represent specific primary RARγ targets in F9 cells. Several genes in the Wnt signaling pathway were regulated by RARγ. Delineation of the receptor specific actions of RA with respect to cell proliferation and differentiation should result in more effective therapies with this drug.

Keywords: retinoic acid receptor, gene expression profiles, differentiation, retinol, sfrp5, Tie1

1. INTRODUCTION

Retinoids are a group of natural or synthetic derivatives of vitamin A (retinol) which include all-trans-, 9-cis- and 13-cis-retinoic acids (RA, 9cRA and 13cRA, respectively). Synthesis of RA, the main biologically active metabolite of vitamin A, involves the irreversible oxidation of retinol in target cells [1, 2]. Retinoids are involved in multiple physiological processes including reproduction, embryonic development, epithelial differentiation, immune function, and vision [3]. In addition, retinoids modulate both normal and neoplastic cell growth through the regulation of cell differentiation and/or induction of apoptosis [4, 5].

The biological effects of RA are thought to be primarily mediated through interactions with retinoic acid receptors (RARs) or retinoic X receptors (RXRs), which function as transcription factors that activate transcription by binding to RA-response elements (RAREs or RXREs) in the promoter regions of target genes [6]. Both RARs and RXRs exhibit three isotypes, α, β, and γ, which are encoded by different genes. Each isotype has several isoforms produced from differential splicing and alternative use of promoters [7, 8]. All-trans retinoic acid (RA) binds and activates RARs, while 9-cis-retinoic acid binds and activates both RARs and RXRs. The multiple RARs and RXRs are conserved in vertebrate evolution and display distinct spatial-temporal expression patterns in developing embryos and adult tissues [9, 10]. Although gene disruption studies in mice have revealed some functional redundancy among the different RAR isotypes [11, 12], each RAR isotype performs unique functions in development and differentiation which can not be replaced by the actions of the other isotypes [11, 12]. RARγ plays a major role in axial rotation and vertebra specification [13], hindbrain patterning (specification of rhombomere 5/6 territory) [14], epidermal hyperplasia [15, 16], formation of otocysts, pharyngeal arches and the forelimb bud, closure of the primitive gut, cardiac looping morphogenesis [17], alveoli regeneration [18], hematopoietic stem cell self-renewal [19], and inflammatory cytokine production [20]. In addition, activation of RARγ is essential for induction of apoptosis in melanoma cells [21], neuroblastoma cells [22], neoplastic epidermal cells [23], and human pancreatic cancer cells [24]. In melanoma cells, RARγ selective compounds were able to induce apoptosis and differentiation [25]. However, the molecular mechanisms by which RARγ induces apoptosis and growth inhibition in these tumor cells are not well defined.

To delineate the specific roles of RARγ in retinoid signaling, the identification of specific RARγ target genes is crucial. The F9 teratocarcinoma stem cell line is a good model for investigating the mechanism by which RA induces cell differentiation and controls cell proliferation [26]. Upon exposure to RA, F9 Wt cells differentiate into primitive endoderm-like cells, with differentiation markers that resemble those in early embryos [2729]. In our laboratory F9 RARα−/−, RARβ2−/− and RARγ−/− cell lines have been generated by homologous recombination [3033]. The F9 RARγ null cells are unable to differentiate fully [30, 34]. Based on our previous findings and those of other researchers that the expression of some RA-inducible genes, such as Hoxa1, Hoxa3, laminin B1, Cdx1, collagen IV (α1), GATA-4, BMP-2, and Cyp26a1, is reduced in the RARγ−/− F9 cells [11, 30, 34, 35], we wanted to explore the effect of the loss of RARγ on gene transcription on a genome-wide scale. Thus, we compared the global gene expression profiles of F9 Wt versus RARγ−/− cells using Affymetrix oligonucleotide arrays (MG-U74Av2 and MG-430.2). These experiments uncovered new molecular actions of RARγ in F9 cell differentiation and provided insights into the molecular mechanisms by which RA causes cell differentiation.

2. MATERIALS AND METHODS

2.1. Chemicals

RA and cycloheximide were purchased from Sigma, St. Louis, MO.

2.2. Cell Culture, RA, cycloheximide treatment and RNA preparation

F9 Wt, RARγ−/−[30], RARα−/−[32] and RARβ2−/−[31] cells were grown in Dulbecco’s modified Eagle’s (DMEM) supplemented with 5% bovine calf serum, 5% fetal calf serum and 2 mM glutamine. Tissue culture flasks and plates were gelatinized with 0.3% gelatin solution for at least 5 min prior to plating. 2.0 ×106 cells were plated in 100 mm tissue culture plates for 16 hours before RA or vehicle (ethanol) treatment. For cyclohexmide treatment, cells were pretreated with cycloheximide at 1 μg/ml for 20 minutes before RA or vehicle (ethanol) was added for a further incubation for 5.5 h. RA was dissolved in 100% ethanol, filter (0.2 μm) sterilized, and diluted in cell culture medium to a final concentration of 1 × 10−6 M. Control samples were cultured in the same volume of ethanol (vehicle) used for the RA-treated samples. The final concentration of ethanol in all experiments did not exceed 0.1% volume. All experiments involving RA were performed in dim light. Total cellular RNA was extracted using Trizol (Invitrogen, P/N 15596026) by following the manufacturer’s protocol.

2.3. DNA Microarray Analyses

Microarray analysis was carried out according to the Affymetrix Genechip expression analysis technical manual. The extracts of total RNA were cleaned up with RNeasy Mini Kit (QIAGEN) according to the manufactors’s protocol. Total RNA (30 μg) from RA-treated (1 μM RA for 24 h), or RA plus cycloheximide –treated (1 μg/mL for 6 h) F9 Wt and F9 RARγ−/− cells was reverse transcribed into cDNA. After second-strand synthesis, cDNAs were then in vitro transcribed into cRNAs with biotinylated ribonucleotides (Enzo Diagnostics, Farmingdale, NY). cRNA (20 μg) was fragmented by heating at 94°C for 35 minutes. A cocktail containing fragmented cRNA, control oligonucleotide B2, control cRNA (Biotin B, Biotin C, Biotin D, and Cre, Hybridization Control Kit, Affymetrix, P/N 900454),), and herring sperm DNA was hybridized to microarray chip MG-U74Av2 (Affymetrix, P/N 900344), which covers 36,000 mouse transcripts and variants representing 12,488 genes, for 16 hours at 45°C. In a second study, 15 μg of total RNA from vehicle (ethanol) treated or RA (1 μM for 24 h) treated F9 Wt and F9 RARγ−/− cells was reverse transcribed into cDNA and then in vitro transcribed into cRNAs. Fragmented cRNAs (15 μg) were hybridized to microarray chip MG-430 2.0 Array (Affymetrix, P/N 900496) which covers the transcribed mouse genome with over 45,000 transcripts representing 34,000 well-substantiated mouse genes. The hybridized microarray chips were washed and stained in a Fluidics station, and were scanned by the GeneArray Scanner in the Microarray Core Facility at Weill Cornell Medical College. The resultant images were processed with MAS (Microarray Suite) 5.0 software (Affymetrix, Santa Clara, CA). The microarray experiments were repeated two or three times with different RNA preparations, and the overall data from the three independent experiments were further analyzed using the GeneSpring v7.0 (Silicon Genetics) software package. These data have been deposited in the GEO database (www.ncbi.nlm.nih.gov/geo, Accession # GSE8431).

A three-step normalization algorithm was used in the GeneSpring software analysis. In the first step, data transformation of all data less than 0.01 was reset to 0.01. In the second normalization step, each chip was normalized to the 50th percentile intensity of total intensity. In the third step, per gene normalization was done by dividing the median intensity of that gene in several control samples. A Welch’s T-test with a cutoff of a P-value ≤ 0.05 in a cross-gene error model was applied to select those statistically significant genes between two groups.

2.4. Semi-Quantitative and Quantitative Real-time RT-PCR

Total RNA (5 μg) isolated from F9 cells was reverse transcribed in a total 20 μl volume reaction with 200 units of Superscript Reverse transcritase II (Invitrogen Life Technologies) and oligo dT (12–18) primer (Invitrogen Life Technologies). The cDNA thus produced was diluted to 100 μl with diethyl pyrocarbonate/water, of which 2 μl was used in a PCR as follows. The PCR was performed using the following conditions: 94°C for 30 s, 58°C for 30 s, and 72°C for 80 s, with a final extension at 72°C for 10 min. Taq polymerase was from Invitrogen (catalog number 18038-042). The PCR products were subjected to 1% agarose gel electrophoresis. The gel images were stained with ethidium bromide and were recorded with a FluorChem 8800 system (Alpha Innotech, San Leandro, CA). Real-time PCR was performed using Bio-Rad iQ SYBR Green Supermix following the manufacturer’s instructions, on a Bio-Rad MyiQ Single Color Real-time PCR Detection System. Primers used in this study are listed in Appendix 3.

Appendix 3.

Primers used for RT-PCR.

Common Name Sequence Product Size (bp) GeneBank Accession No.
Hoxa1 (sense) 5′ TGG AGG AAG TGA GAA AGT TGG C 3′ 485 NM_010449
 (antisense) 5′ATG GGA GTC GAG AGG TTT CC 3′

FBP-2 (sense) 5′ TCC TGT ATG AAT GCA ATC CTG T 3′ 232 D42083
 (antisense) 5′CAA TTG ACA AAG ACA AAG GGA AG 3′

Tie1 (sense) 5′ CCT TTG CTC AGA TCG CAC TA 3′ 381 X80764
 (antisense) 5′ ATG CTG CTT TAG GTG GAG GA 3′

EMP1 (sense) 5′ CCT CTC CAT CAT CTT CTC CAT C 3′ 620 X98471
 (antisense) 5′ GAC GTC AAG GAA GCA TCA GCA T 3′

EMP3 (sense) 5′ CCT CTT CAT GTT CCA ACT CTA 3′ 238 NM_010129
 (antisense) 5′ TTT CCG CAG GTG GAT GTA GAC 3′

Sfrp2 (sense) 5′ CAA CCT GCT GGG CCA CGA GAC C 3′ 695 NM_009144
 (antisense) 5′ GCT TGC GGA TGC TGC GGG AGA T 3′

Sfrp5 (sense) 5′ CTA TCC CTG TTC CCT CTA CTA C 3′ 240 NM_018780
 (antisense) 5′ AGA ACC CTT CAG TCA AAG AGG G 3′

Coch5B2 (sense) 5′ GGG CAG TCC TAT GAT GAT GT 3′ 243 AF006741
 (antisense) 5′ CCT TGCACG TAT TCC TTG AG 3′

mOTT3 (sense) 5′ ATC TCA TCA TGA CCA CTC AGG G 3′ 267 NM_011022
 (antisense) 5′ TTC TTC GAT GTT CCT GTA CCC A 3′

Slc27a2 (sense) 5′ AGT TCT ACG CAT CCA CTG AAG 3′ 655 AF072757
 (antisense) 5′ TGA CTG TGG GAT TGA AGC CCT CTT 3′

Xlr3b (sense) 5′ AGG CTG CCT TGT GGA GAG 3′ 231 NM_011727
 (antisense) 5′ CTG TTG CCT CTC TGT TCC TG 3′

Runx-1 (sense) 5′ CCA GCA AGC TGA GGA GCG GCG 3′ 348 NM_009821
 (antisense) 5′ CGG ATT TGT AAA GAC GGT GA 3′

Crygc (sense) 5′ CTG ACT ACC AGC AGT GGA TGG G 3′ 171 NM_007775
 (antisense) 5′ CCT CAC TGA GGT GGA AGC GAT C 3′

Zfp503 (sense) 5′ GCC TTT TGT GCA CGC TGT 3′ 243 AK032903
 (antisense) 5′ ACCGAG AGT TTG GAA GA3′

Raet1a (sense) 5′GCC ACT CTA CTT CTA AGA AAG GAT T 3′ 307 NM_009016
 (antisense) 5′GTG AAG CTT ACT GTG GGA CTT 3′

Peg1/Mest(sense) 5′ ATT CGC AAC AAT GAC GGC 3′ 441 BC006639
 (antisense) 5′ TGA GGT GGA CTA TTG TGT CAC C 3′

RARα (sense) 5′ ATC GAG ACC CAG AGC AGC AG 3′ 409 NM_009024
 (antisense) 5′ CCT GGT GCG CTT TGC GAA CC 3′

RARβ (sense) 5′ GCA GAG TTT GAT GGA GTT CGT 3′ 507 NM_011243
 (antisense) 5′ CCC ACT TCA AAG CAC TTC TGC A3′

RARγ (sense) 5′ CAA TAA GGA GAG ACT CTT TGC G 3′ 324 NM_011244
 (antisense) 5′ TAC CAC TAT GGG GTC AGC TCC TGT G 3′

Neo-RARγ (sense) 5′-ATT CGC AGC GCA TCG CCT TCT AT3′ 612 NM_011244
 (antisense) 5′-TTG CTG ACC TTG GTG ATG AGT 3′

Actin (sense) 5′ AAG TGT GAC GTT GAC ATC CG 3′ 222 NM_007393
 (antisense) 5′ GAT CCA CAT CTG CTG GAA GG 3′

3. RESULTS

3.1. Comparison of the gene expression profiles in F9 Wt versus F9 RARγ−/− cell lines after treatment with RA plus a protein synthesis inhibitor, cycloheximide, for 6 h and after treatment with RA for 24 h

To identify the immediate, primary RARγ-regulated genes, F9 Wt and F9 RARγ−/− cells were treated with RA and the protein synthesis inhibitor, cycloheximide, for 6 h, and the gene expression profiles in the two cell lines were then compared using the U74Av2 affymetrix arrays. No untreated F9 Wt or F9 RARγ−/− samples were analyzed (Fig. 1). The rationale for this is that since RARγ is constitutively expressed in F9 cells, the primary target genes of RARγ should be transcriptionally activated by RA even in the absence of new protein synthesis. Thus, these transcripts can be detected in RA plus cycloheximide treated F9 Wt cells, but not in the F9 cells which lack RARγ. A total number of 5885 probe sets which were flagged with Presence in at least 3 chips were subjected to statistical analysis using GeneSpring V7 software. The resulting 94 genes exhibiting a 2-fold or greater net intensity ratio difference (p ≤0.05, Welch’s t-test) are shown (Table 1). These genes are putative RARγ primary target genes.

Figure 1. Design of microarray experiment.

Figure 1

Four conditions were used to examine gene expression in F9 Wt and F9 RARγ−/− cells. A“#” indicates the number of times the microarray analysis was performed under each condition, starting with fresh cells. RA, all-trans retinoic acid; CHM, cycloheximide.

Table 1. RARγ primary target genes identified using microarrays U74Av2.

Wt and RARγ−/− F9 cells were treated for 6 h with 1 μg/ml cycloheximide and 1 μM RA. Only those genes with at least a 2.0-fold change in F9 Wt cells relative to F9 RARγ−/− cells in both sets of microarrays with p values ≤0.05 are shown. Fold change=F9 Wt/F9 RARγ−/−.

Affymetrix ID Fold Changeab Gene Symbol Gene Title
103086_at 10.94 Hoxa5 homeo box A5
102087_at 10.6 Hoxa3 homeo box A3
97426_at 9.359 Emp1 epithelial membrane protein 1
94813_at 7.189 Gas1 growth arrest specific 1
97379_at 5.973 Fbp2 fructose bisphosphatase 2
104378_at 5.521 Pon2 paraoxonase 2
104464_s_at 4.512 Kdelr3 KDEL (Lys-Asp-Glu-Leu) endoplasmic reticulum protein retention receptor 3
101861_at 4.397 Sgce sarcoglycan, epsilon
98320_at 4.226 Cyp26a1 cytochrome P450, family 26, subfamily a, polypeptide 1
104406_at 4.208 Ptges Prostaglandin E synthase
100914_at 4.204 Transcribed sequences
104580_at 4.168 Plcd phospholipase C, delta
93503_at 4.129 Sfrp2 Secreted frizzled-related sequence protein 2
95016_at 4.056 Nrp Neuropilin
102649_s_at 4.051 Raet1a retinoic acid early transcript 1, alpha
103653_at 3.88 Mras muscle and microspikes RAS
99665_at 3.666 Satb1 special AT-rich sequence binding protein 1
92502_at 3.588 Plagl1 pleiomorphic adenoma gene-like 1
161013_f_at 3.435 4930422J18Rik RIKEN cDNA 4930422J18 gene
161817_f_at 3.405 4930422J18Rik RIKEN cDNA 4930422J18 gene
103761_at 3.403 Tcfcp2l1 transcription factor CP2-like 1
104480_at 3.328 Dsg2 desmoglein 2
104701_at 3.322 Bhlhb2 basic helix-loop-helix domain containing, class B2
96662_at 3.195 Ppap2b phosphatidic acid phosphatase type 2B
99045_at 3.152 Eno2 enolase 2, gamma neuronal
95474_at 3.087 F2r coagulation factor II (thrombin) receptor
104375_at 3.062 Spock2 sparc/osteonectin, cwcv and kazal-like domains proteoglycan 2
92501_s_at 2.963 Plagl1 pleiomorphic adenoma gene-like 1
93994_at 2.941 Chpt1 choline phosphotransferase 1
92582_at 2.776 Slc1a5 solute carrier family 1 (neutral amino acid transporter), member 5
94027_at 2.737 CDNA clone IMAGE:30033444, partial cds
97353_at 2.706 Dab2ip Disabled homolog 2 (Drosophila) interacting protein
102712_at 2.706 Saa3 serum amyloid A 3
100011_at 2.575 Klf3 Kruppel-like factor 3 (basic)
160961_at 2.57 Sipa1l2 signal-induced proliferation-associated 1 like 2
102886_at 2.553 Gpc4 glypican 4
92241_at 2.507 1110019L22Rik RIKEN cDNA 1110019L22 gene
100410_at 2.47 C330027G06Rik RIKEN cDNA C330027G06 gene
94418_at 2.467 Elovl6 ELOVL family member 6, elongation of long chain fatty acids (yeast)
98016_at 2.46 D3Wsu161e DNA segment, Chr 3, Wayne State University 161, expressed
100528_at 2.45 Ube2h ubiquitin-conjugating enzyme E2H
95161_at 2.409 Ctdsp2 CTD (carboxy-terminal domain, RNA polymerase II, polypeptide A) small phosphatase 2
98535_at 2.402 Comt catechol-O-methyltransferase
93083_at 2.392 Anxa5 annexin A5
160255_at 2.376 1110004P15Rik RIKEN cDNA 1110004P15 gene
104576_at 2.371 Ski Sloan-Kettering viral oncogene homolog
97509_f_at 2.37 Fgfr1 Fibroblast growth factor receptor 1
97138_at 2.369 9430077C05Rik RIKEN cDNA 9430077C05 gene
92833_at 2.365 Hal histidine ammonia lyase
99994_at 2.335 Cidea Cell death-inducing DNA fragmentation factor, alpha subunit-like effector A
97711_at 2.322 B430320C24Rik RIKEN cDNA B430320C24 gene
160188_at 2.29 Nudt4 nudix (nucleoside diphosphate linked moiety X)-type motif 4
104210_at 2.285 Itga3 integrin alpha 3
103288_at 2.276 Nrip1 nuclear receptor interacting protein 1
96207_at 2.264 Rbms1 RNA binding motif, single stranded interacting protein 1
160393_at 2.167 Etnk1 Ethanolamine kinase 1
92796_at 2.162 Akp2 alkaline phosphatase 2, liver
103614_at 2.128 Nfkb2 nuclear factor of kappa light polypeptide gene enhancer in B-cells 2, p49/p100
92368_at 2.047 Ramp3 receptor (calcitonin) activity modifying protein 3
100475_at 2.044 Trim25 B6-derived CD11 +ve dendritic cells cDNA, RIKEN full-length enriched library, clone:F730014H10 product:unclassifiable, full insert sequence
100962_at 2.039 Nab2 Ngfi-A binding protein 2
97452_at 2.038 H2afy H2A histone family, member Y
160887_at 2.036 Hes1 hairy and enhancer of split 1 (Drosophila)
94408_at 2.025 Nab1 Ngfi-A binding protein 1
103551_at 2 2810431N21Rik RIKEN cDNA 2810431N21 gene
160546_at 0.487 Aldo3 aldolase 3, C isoform
94829_at 0.466 1110020A09Rik RIKEN cDNA 1110020A09 gene
92635_at 0.464 Tuba4 tubulin, alpha 4
98109_at 0.462 Mrpl55 Mitochondrial ribosomal protein L55
103549_at 0.459 Nes Nestin
93483_at 0.458 Hck hemopoietic cell kinase
98544_at 0.455 Guk1 Guanylate kinase 1
93543_f_at 0.452 Gstm1 glutathione S-transferase, mu 1
93836_at 0.439 Bnip3 BCL2/adenovirus E1B 19kDa-interacting protein 1, NIP3
97995_at 0.431 Tcf7 transcription factor 7, T-cell specific
160495_at 0.423 Ahr Aryl-hydrocarbon receptor
103581_at 0.407 Cte1 cytosolic acyl-CoA thioesterase 1
94334_f_at 0.392 Ina internexin neuronal intermediate filament protein, alpha
103405_at 0.388 2610019A05Rik RIKEN cDNA 2610019A05 gene
160085_at 0.384 Tst thiosulfate sulfurtransferase, mitochondrial
92786_at 0.359 Efhd1 EF hand domain containing 1
104652_at 0.349 Kcnk2 potassium channel, subfamily K, member 2
94335_r_at 0.346 Ina internexin neuronal intermediate filament protein, alpha
95669_g_at 0.345 Stmn2 stathmin-like 2
93680_at 0.344 Stk10 serine/threonine kinase 10
99812_at 0.338 Capn3 calpain 3
95670_at 0.296 Stmn2 stathmin-like 2
95603_at 0.268 Gldc glycine decarboxylase
95706_at 0.267 Lgals3 lectin, galactose binding, soluble 3
96244_at 0.239 Uchl1 ubiquitin carboxy-terminal hydrolase L1
104286_at 0.183 Slc38a4 solute carrier family 38, member 4
161068_at 0.12 3830422N12Rik RIKEN cDNA 3830422N12 gene
100967_at 0.0994 Slc27a2 solute carrier family 27 (fatty acid transporter), member 2
104544_at 0.0981 4930517K11Rik RIKEN cDNA 4930517K11 gene
a

Average from three experiments.

b

Some genes have multiple distinct probes on the GeneChip® resulting in different fold-changes from different probes.

The global gene expression patterns in F9 Wt and F9 RARγ−/− cells after a 24-hour exposure to 1 μM RA were also compared on U74 Av2 arrays. The overall presence call was 6171/12422=49.68%. After the Welch T-test, 241 genes with P-values ≤0.05 were selected for further fold change analysis. The results from 66 genes exhibiting a 2.0-fold or greater net intensity ratio difference are shown (Table 2). These genes encode cell membrane proteins, cell-cell adhesion molecules, signal transducers, and surface tyrosine kinases involved in cell development. By comparing the 94 genes (6 h treatment with RA plus cycloheximide, Table 2) with those altered at 24 hour after RA exposure (Table 2), we found that only 8 genes overlapped. These genes are the epithelial memebrane protein (emp)-1, secreted frizzled-related sequence protein 2 (Sfrp2), alkaline phosphatase 2 (AKP2), Stmn2, solute carrier family 38 member 4 (Slc38a4), Slc27a2, and lectin-galactose binding soluble 3 (Lgals3). One obvious reason why the changes in most of the early response genes were not seen at the 24 h RA treatment is that the kinetics of the transcriptional activation of the early vs. late genes induced by RARγ are different.

Table 2. Genes differentially expressed between F9 Wt and F9 RARγ−/− cells after RA treatment for 24 h, identified using U74AV2 microarrays.

Only those genes with at least a 2.0-fold change in F9 Wt cells relative to F9 RARγ−/− cells with p values ≤0.05 are shown. Fold change=F9 Wt/F9 RARγ−/−.

Affymetrix ID Fold Changeab Gene Symbol Gene Ttile
99936_at 25.08 Tie1 tyrosine kinase receptor 1
160943_at 15.3 Stag3 stromal antigen 3
97426_at 13.86 Emp1 epithelial membrane protein 1
93503_at 4.634 Sfrp2 secreted frizzled-related sequence protein 2
101027_s_at 4.142 Pttg1 pituitary tumor-transforming 1
103506_f_at 3.773 Dsc2 Desmocollin 2
104146_at 3.567 Rasip1 Ras interacting protein 1
97379_at 3.534 Fbp2 fructose bisphosphatase 2
99669_at 3.532 Lgals1 Lectin, galactose binding, soluble 1
161482_f_at 3.427 Prph1 Peripherin 1
104580_at 3.385 Plcd phospholipase C, delta
100610_at 2.998 Capns1 calpain, small subunit 1
95426_at 2.946 Echs1 enoyl Coenzyme A hydratase, short chain, 1, mitochondrial
101358_at 2.885 Plcb3 phospholipase C, beta 3
104746_at 2.821 Fkbp7 FK506 binding protein 7
160978_at 2.808 D630035O19Rik RIKEN cDNA D630035O19 gene
96865_at 2.677 Marcks myristoylated alanine rich protein kinase C substrate
98435_at 2.631 Adss adenylosuccinate synthetase, muscle
100828_at 2.325 ---
160388_at 2.305 Sc4mol sterol-C4-methyl oxidase-like
160215_at 2.299 Aes amino-terminal enhancer of split
104232_at 2.289 Gjb3 gap junction membrane channel protein beta 3
96016_at 2.27 2700094K13Rik RIKEN cDNA 2700094K13 gene
102993_at 2.265 Ggta1 glycoprotein galactosyltransferase alpha 1, 3
98535_at 2.247 Comt catechol-O-methyltransferase
92586_at 2.242 Glud Glutamate dehydrogenase
99184_at 2.232 Csad cysteine sulfinic acid decarboxylase
101107_at 2.222 Calu Calumenin
102926_at 2.193 Gfra3 Glial cell line derived neurotrophic factor family receptor alpha 3
93750_at 2.16 Gsn Gelsolin
104673_at 2.137 Epha4 Eph receptor A4
103085_at 2.133 Hebp1 Heme binding protein 1
92607_at 2.103 Peg1 Mus musculus Peg1/MEST protein
93522_at 2.094 Rad9 RAD9 homolog (S. pombe)
95733_at 2.048 Slc29a1 solute carrier family 29 (nucleoside transporters), member 1
97717_at 2.041 Tcf15 transcription factor 15
92796_at 2.04 Akp2 alkaline phosphatase 2, liver
96038_at 2.038 Rnase4 ribonuclease, RNase A family 4
93373_at 2.023 Naglu alpha-N-acetylglucosaminidase (Sanfilippo disease IIIB)
103056_at 0.496 6230425C22Rik RIKEN cDNA 6230425C22 gene
103423_at 0.495 Cyb561 cytochrome b-561
93038_f_at 0.486 Anxa1 annexin A1
95756_at 0.486 Ftsj3 FtsJ homolog 3 (E. coli)
100600_at 0.467 Cd24a CD24a antigen
92306_at 0.465 Ott Ovary testis transcribed
101030_at 0.426 Rhob ras homolog gene family, member B
102841_at 0.414 Similar to ribosomal protein L40 (LOC216818), mRNA
96151_at 0.407 Mocos molybdenum cofactor sulfurase
160236_at 0.383 9630044O09Rik RIKEN cDNA 9630044O09 gene
95954_at 0.379 D7Ertd143e DNA segment, Chr 7, ERATO Doi 143, expressed
93864_s_at 0.36 Enah enabled homolog (Drosophila)
95883_at 0.349 Phf17 PHD finger protein 17
104486_at 0.349 A2m alpha-2-macroglobulin
102344_s_at 0.345 Tcea3 transcription elongation factor A (SII), 3
93680_at 0.343 Stk10 serine/threonine kinase 10
102418_at 0.339 Tex19 testis expressed gene 19
96704_at 0.339 Sfn Stratifin
99577_at 0.33 Kitl kit ligand
104063_at 0.327 Srcasm Src activating and signaling molecule
95670_at 0.308 Stmn2 stathmin-like 2
99034_at 0.266 Irx3 Iroquois related homeobox 3 (Drosophila)
104338_r_at 0.25 1200008D14Rik RIKEN cDNA 1200008D14 gene
93028_at 0.246 H19 H19 fetal liver mRNA
101883_at 0.212 Xlr3b XLR related protein, Mouse A12 mRNA
93294_at 0.211 Ctgf connective tissue growth factor
103317_at 0.189 Coch5B2 Coagulation factor C homolog (Limus polyphemus) cds=(68,1726)
100967_at 0.189 Slc27a2 solute carrier family 27 (fatty acid transporter), member 2
104126_at 0.188 Cstf2t cleavage stimulation factor, 3′ pre-RNA subunit 2, tau
161081_at 0.158 Cpeb2 cytoplasmic polyadenylation element binding protein 2
104286_at 0.106 Slc38a4 solute carrier family 38, member 4
93013_at 0.0853 Idb2 inhibitor of DNA binding 2
95706_at 0.0819 Lgals3 lectin, galactose binding, soluble 3
a

Average from two experiments.

b

Some genes have multiple distinct probes on the GeneChip® resulting in different fold-changes from different probes.

To validate the microarray data, new F9 cell samples were treated with RA for 24 h to confirm the results. Several differentially expressed genes were selected for examination by semi-quantitative RT-PCR. In addition to the F9 Wt and RARγ−/− lines, F9 RARα−/− and RARβ2−/− cell lines were also tested during the validation studies in order to determine if the genes were specifically regulated by RARγ. The mRNA expression of the different RAR isotypesα, β, and γ in each cell line was examined. The RARα−/−, RARβ2−/−, and RARγ−/− cell lines were each established by homologous recombination with a targeting vector to their B domains [3032]. Both alleles of the mutant RAR in each cell line contain the neomycin resistance gene, which disrupts the encoded RARα, RARβ2, and RARγ transcripts and protein. Using primers specific for each isotype which span the neomycin resistance gene, the transcripts from the wild type allele and mutant allele were amplified. Since each of the mutant alleles contains the neomycin resistance gene, the mutant allele gives rise to a larger transcript than that from the wild type allele. RARα mRNA was detected in Wt, RARγ−/− and RARβ2−/− cells, but not in F9 RARα−/− cells (Fig. 2A). The expression of RARβ2 transcripts was strongly induced F9 Wt cells, F9 RARα−/−, and RARγ−/− cells, but not in RA treated RARβ2−/− cells (Fig. 2A). RARγ mRNA could not be detected in F9 RARγ−/− cells, though RARγ mRNA was detected in F9 Wt, RARα−/− and RARβ2−/− cells (Fig. 2A). Transcripts from the mutant RARγ (RARγ-NEO) were only detected in F9 RARγ−/−, but not in F9 Wt, RARα−/− and RARβ2−/− cells (Fig. 2A). Thus, the identities of the cell lines were confirmed (Fig. 2A).

Figure 2. Semi-quantitative RT-PCR analyses of selected genes, FBP-2, Tie1, EMP1, EMP3, Sfrp2, Coch5B2, mOTT3, mFATP2, and Xlr3b, in F9 Wt, RARγ−/−, RARα−/− and RARβ2−/− cell lines in response to RA.

Figure 2

A and B, total RNA was extracted from the F9 Wt, RAR γ−/−, RARα−/−, RARβ2−/− cells cultured in the presence or absence of 1 μM RA for 24 h. An equivalent amount of RNA (5 μg) was subjected to RT-PCR with oligonucleotide primers specific for the indicated genes. The PCR-reactions were stopped between the 22ndand 32nd cycles depending on the gene being tested to ensure linear amplification ranges. PCR-amplified products were electrophoresed in 1% agarose gels and the products were stained with ethidium bromide. The 1:2 dilution of cDNA from RA-treated F9 Wt or RA-treated F9 RARγ−/− samples was used to indicate the linear amplification ranges for genes which showed increased mRNA levels in RARγ−/− cells or decreased levels in RARγ−/− cells, respectively. The sizes of the amplified bands are indicated on the left. These assays were repeated using three different RNA preparations, and similar results were obtained.

Hoxa1, a known RARγ-regulated gene, was examined as a positive control. Hoxa1 mRNA levels increased after exposure to RA in F9 Wt, RARα−/− and RARβ2−/− cells, but the RA associated increase in Hoxa1 mRNA was greatly reduced in F9 RARγ−/− cells as compared to F9 Wt cells (Fig. 2B).

The nine genes chosen for validation by semi-quantitative RT-PCR analyses were FBP-2, Tie1, EMP1, EMP3, Sfrp2, Coch5B2, mOTT3, mFATP2 (Slc27a2), and Xlr3b (Fig. 2B). These genes were selected for validation because they exhibited relatively high fold changes in mRNA levels and/or interesting biological functions.

FBP-2 (fructose 1,6-bisphosphatase), via altering levels of F26P2 (Fructose-2,6-bisphosphate), influences glucose and lipid metabolism and provides cooperative regulation of fuel metabolism [36]. F26P2 can in turn regulate transcription factors and certain key proteins (enzymes) of signaling and/or energy sensing [36]. For example, F26P2 can regulate the amount and/or phosphorylation state of transcription factors such as hepatic nuclear factor 1-alpha (HNF1α), peroxisome proliferator-activated receptor alpha (PPARα), and PPARγ co-activator 1beta (PGC1β), and the kinases Akt and AMP-activated protein kinase (AMPK) [36]. Fbp2 mRNA levels were 5.9 ± 2.0-fold and 3.5 ± 1.1-fold higher in F9 Wt cells than in F9 RARγ−/− cells cultured in the presence of RA plus cycloheximide for 6 h, and in the presence of RA for 24 h, respectively. Semi-quantitative RT-PCR confirmed that Fbp2 mRNA was present at higher levels in F9 Wt than in F9 RARγ−/− cells (Fig. 2B).

Tie1 is a member of the receptor tyrosine kinase family [37, 38]. It is essential for the development and maintenance of vascular vascular system and hematopoiesis [39]. Tie1 mRNA levels were 25.1 ± 8.2-fold higher in F9 Wt than in F9 RARγ−/− cells cultured in the presence of RA for 24 h. Semi-quantitative RT-PCR confirmed that Tie1 mRNA levels were higher in F9 Wt cells than in F9 RARγ−/− cells (Fig. 2B).

Emp1 (epithelial membrane protein 1) and Emp3 (epithelial membrane protein 3) are members of the peripheral myelin protein 22 (PMP22) family [40]. Emp1 mRNA levels were 9.4 ± 2.4-fold and 13.7 ± 3.5-fold higher in F9 Wt cells than in F9 RARγ−/− cells cultured in the presence of RA plus cycloheximide for 6 h, and cultured in the presence of RA for 24 h, respectively. Semi-quantitative RT-PCR confirmed that Emp1 and Emp3 transcripts are present at higher levels in F9 Wt than in F9 RARγ−/− cells (Fig. 2B).

Sfrp2 (secreted frizzled related protein 2) is a secreted Wnt antagonist that directly interacts with the Wnt ligand to inhibit Wnt signaling [41]. Sfrp2 is required for anteroposterior (AP) axis elongation and somitogenesis in the thoracic region during mouse embryogenesis. Sfrp2 mRNA levels were 4.1 ± 1.7-fold higher in F9 Wt than in F9 RARγ−/− cells cultured in the presence of RA plus cycloheximide for 6 h, and 4.6 ± 2.0-fold cultured in the presence of RA for 24h, respectively. Semi-quantitative RT-PCR confirmed that Sfrp2 mRNA levels are higher in F9 Wt cells than in F9 RARγ−/− cells (Fig. 2B).

Coch5B2 encodes a protein with regions highly homologous to the collagen-binding type A domains of von Willebrand factor [42]. Coch5B2 is highly expressed in fetal inner ear structures, the cochlea, and the vestibule [43]. Mutations in Coch5B2 cause DFNA9, a human nonsyndromic deafness with vestibular dysfunction [44, 45]. Coch 5B2 mRNA levels were 5.3 ± 1.4-fold higher in the F9 RARγ−/− cells than in F9 Wt cells cultured in the presence of RA for 24 h, and this was confirmed by semi-quantitative RT-PCR (Fig. 2B).

OTT3 is an X-linked gene expressed in testis and spermatogonia, but not in somatic tissues [46]. A role for OTT3 in splicing regulation has been demonstrated [47]. Ott3 mRNA levels were 2.2 ± 0.7-fold higher in F9 RARγ−/− cells than in F9 Wt cells cultured in the presence of RA for 24 h, and this was validated by semi-quantitative RT-PCR (Fig. 2B).

Slc27a2 (solute carrier family 27 (fatty acid transporter), member 2) belongs to the long chain fatty acid transporter (FATP) family. These proteins facilitate fatty acid uptake from the medium [48]. Slc27a2 mRNA levels were 2.2 ± 0.3-fold higher in F9 RARγ−/− cells than in F9 Wt cells cultured in the presence of RA for 24 h, and this was confirmed by semi-quantitative RT-PCR (Fig. 2B).

Xlr3b (X-linked lymphocyte regulated, 3b) is an X-linked, maternally expressed, imprinted gene [49] which may mediate X-linked imprinting effects on cognitive processes [50, 51]. Xlr3b mRNA levels were 4.7±1.3-fold higher in F9 RARγ−/− cells than in F9 Wt cells cultured in the presence of RA for 24 h and semi-quantitative RT-PCR confirmed that Xlr3b mRNA levels are higher in F9 RARγ−/− cells than in F9 Wt cells (Fig. 2B).

The mRNA levels of FBP-2, Tie1, EMP1, EMP3 and Sfrp2 were lower in F9 RARγ−/− cells than in F9 Wt cells, but these transcripts were not expressed at lower levels in F9 RARα−/− and RARβ2−/− cells, either in the presence or absence of RA. Thus, these genes represent specific RARγ target genes. In contrast, Coch5B2, mOTT3, Slc27a2, and Xlr3b transcripts were expressed at higher levels in F9 RARγ−/− cells than in Wt, RARα−/− (with the exception of Slc27a2), and RARβ2−/− cells. Coch5B2, Xlr3b and mOTT3 transcripts, therefore, are specifically increased in F9 RARγ−/− cells (Fig. 2B). A list of these genes is in the GEO database (www.ncbi.nlm.nih.gov/geo, Accession # GSE8431).

3.2. Comparison of the gene expression profiles associated with F9 Wt and F9 RARγ−/− cells cultured in the absence vs. presence of RA

The initial microarray analyses we performed identified the genes that are differentially expressed between RA-treated F9 Wt and RA-treated F9 RARγ−/− cells at 6 h and 24 h, but no control (untreated) F9 Wt or F9 RARγ−/− samples were analyzed (Fig. 1). Thus, the genes identified in these initial studies also included a subset of genes that were not responsive to RA. In order to identify additional RA responsive genes and to incorporate a larger fraction of the mouse genome into our survey of RARγ-regulated genes, we performed another microarray analysis utilizing Affymetrix MG-430.2 gene chips, which contain 45,037 transcripts representing 34,000 well-substantiated mouse genes.

F9 Wt and F9 RARγ−/− cells were treated with either 1 μM RA or vehicle (ethanol) for 24 hours. The gene expression profiles of RA-treated F9 Wt cells, vehicle–treated (control) F9 Wt cells, RA-treated F9 RARγ−/− cells, and vehicle-treated (control) F9 RARγ−/− cells were then compared using MG-430.2 gene chips. The genes that were differentially expressed between F9 Wt cells and F9 RARγ−/− cells in the presence or absence of RA were identified. Since a protein synthesis inhibitor was not used in this set of experiments, both primary and secondary RARγ dependent RA response genes were identified.

In order to identify the subset of RARγ regulated, RA-responsive genes, a strategy involving multiple two-condition comparisons was employed (Fig. 3). Genes were only considered differentially expressed between the two conditions if they exhibited at least a twofold difference in expression with a p value of less than 0.05 (Welch’s t-test). Genes were defined as RA responsive genes if they exhibited at least a two fold difference from the control (no RA) condition (P≤0.05). For the analysis with MG-430.2 chips, all of the genes with a “Present” call in at least three chips were selected and were subjected to statistical analysis for two condition comparisons using GeneSpring v7.0. The overall presence call (# of genes with a Present call/total # of probe sets on the array) for those samples was 23329/45037 = 51.78 %. For filtering the data for significant changes, the parameteric Welch’s t-test was performed individually for each of the following pair comparisons: Vehicle-treated (control) F9 Wt cells vs vehicle-treated (control) F9 RARγ−/− cells (Wt vs RARγ−/−); RA-treated Wt cells vs RA-treated RARγ−/− cells (Wt +RA vs RARγ−/−+RA); RA-treated Wt cells vs vehicle-treated control Wt cells (Wt +RA vs Wt); resulting in 2151 genes, 2832 genes, and 1909 genes that had statistically significant p values ≤ 0.05, respectively. Genes with statistically significant differences in expression (p< 0.05) were then further subjected to a fold change filter application, resulting in 1058 genes (Wt vs RARγ−/−), 1348 genes (Wt +RA vs RARγ−/−+RA), and 345 genes (Wt +RA vs Wt), respectively, with a differential expression of at least two fold for each pair comparison (Fig. 3).

Figure 3. An overview of the strategy to select differentially expressed genes.

Figure 3

Beginning with all Affymetrix IDs on the MG-430 2.0 GeneChip (45037 genes), the first filter selected the genes with a “Present” call in at least 3 microarrays. The second step filtered out statistically insignificant means based on the Welch’s t- test. The third step filtered out genes with less than a 2-fold difference in expression between the two conditions.

3.3. Distinctive expression profiles associated with the F9 Wt and F9 RARγ−/− cell lines cultured in the absence of RA

There were a total of 1058 probe sets differentially expressed by at least 2-fold between the vehicle-treated (control) F9 Wt and vehicle-treated control F9 RARγ−/− cells at 24 hours (Fig. 3). This indicates a fundamental difference in the gene expression profiles between the two cell types in the absence of RA. The differentially expressed genes with the highest fold changes are shown (Table 3A, B). After a 24 h-RA treatment, a total of 1348 probe sets exhibited at least a 2–fold difference in expression between the RA-treated Wt and RA-treated RARγ−/− F9 cells (Fig. 3), and the differentially expressed genes with the highest fold changes are listed (Table 4A, B).

Table 3. A.

Top 53 genes with at least 2-fold higher expression in F9 Wt cells relative to F9 RARγ−/− cells at 24 h in the absence of RA. Fold change =WT/RARγ−/−.

Affymetrix ID Fold Changeab Gene Symbol Gene Title
1431094_at 43.31 Rtl1 Retrotransposon-like 1
1456250_x_at 42.39 Tgfbi BB533460 RIKEN full-length enriched, 0 day neonate lung Mus musculus cDNA clone E030030E06 3′ similar to L19932 Mouse (beta ig-h3) mRNA, mRNA sequence.
1417426_at 38.65 Prg1 Proteoglycan 1, secretory granule
1452183_a_at 38.62 Gtl2 GTL2, imprinted maternally expressed untranslated mRNA
1448123_s_at 34.09 H2-Ab1 Histocompatibility 2, class II antigen A, beta 1
1422865_at 31.5 Spp1 Secreted phosphoprotein 1
1439380_x_at 29.61 Gtl2 BB093563 RIKEN full-length enriched, 12 days embryo, embryonic body between diaphragm region and neck Mus musculus cDNA clone 9430042P15 3′ similar to Y13832 Mus musculus mRNA for GT12 protein, mRNA sequence.
1415871_at 27.87 H2-Ab1 Histocompatibility 2, class II antigen A, beta 1
1448926_at 27.49 Hoxa5 Homeo box A5
1441075_at 25.77 LOC329416 nitric oxide synthase trafficker
1418215_at 25.05 Mep1b Meprin 1 beta
1432159_a_at 24.87 Il2 Interleukin 2
1452905_at 24.52 Gtl2 GTL2, imprinted maternally expressed untranslated mRNA
1455626_at 23.25 Hoxa9 homeo box A9
1425109_at 20.16 Slc44a3 Solute carrier family 44, member 3
1422864_at 19.68 Spp1 Secreted phosphoprotein 1
1423294_at 18.68 Mest Transcribed sequence with moderate similarity to protein sp:Q9UBF2 (H. sapiens) CPG2_HUMAN Coatomer gamma-2 subunit
1435989_x_at 18.51 Krt2-8 keratin complex 2, basic, gene 8
1427580_a_at 18.48 Rian 15 days embryo head cDNA, RIKEN full-length enriched library, clone:D930050K13 product:unclassifiable, full insert sequence
1426990_at 18.39 Cubn cubilin (intrinsic factor-cobalamin receptor)
1429273_at 18.02 Bmper BMP-binding endothelial regulator
1452400_a_at 17.57 Hoxa11s homeo box A11, opposite strand transcript
1419537_at 17.39 Ctsd Cathepsin D
1460550_at 16.3 BC051083 cDNA sequence BC051083
1417787_at 16.24 Prnd Prion protein dublet
1451332_at 16.03 Zfp521 Zinc finger protein 521; synonyms: Evi3, B930086A16Rik; isoform 2 is encoded by transcript variant 2; ecotropic viral integration site 3; Mus musculus zinc finger protein 521 (Zfp521), transcript variant 2, mRNA.
1448804_at 15.79 Cyp11a1 Cytochrome P450, family 11, subfamily a, polypeptide 1
1430637_at 15.68 2210016H18Rik Adult male stomach cDNA, RIKEN full-length enriched library, clone:2210016H18 product:hypothetical protein, full insert sequence
1418713_at 15.56 Pcbd1 Pterin 4 alpha carbinolamine dehydratase/dimerization cofactor of hepatocyte nuclear factor 1 alpha (TCF1) 1
1431475_a_at 15.11 Hoxa10 Homeo box A10
1458232_at 15.1 Dkk1 dickkopf homolog 1 (Xenopus laevis)
1434647_at 14.92 AU040377 expressed sequence AU040377
1427433_s_at 14.79 5730446D14Rik RIKEN cDNA 5730446D14 gene
1428781_at 13.99 1110014F24Rik RIKEN cDNA 1110014F24 gene
1451687_a_at 12.91 Tcf2 transcription factor 2
1451191_at 12.51 Crabp1 Cellular retinoic acid binding protein I
1418672_at 12.46 Lrrc1 Leucine rich repeat containing 1
1418187_at 12.46 Ramp2 Receptor (calcitonin) activity modifying protein 2
1429693_at 12.41 Dab2 Disabled homolog 2 (Drosophila)
1424393_s_at 12.14 Syt11 Synaptotagmin XI
1453782_at 11.99 3021401C12Rik RIKEN cDNA 0610012A05 gene
1443696_s_at 11.95 Habp2 hyaluronic acid binding protein 2
1433670_at 11.65 Emp2 epithelial membrane protein 2
1450555_at 11.56 Il2 Interleukin 2
1460722_at 11.52 Soat2 Sterol O-acyltransferase 2
1449568_at 11.4 Klb Klotho beta
1440878_at 11.38 Runx1 Runt related transcription factor 1
1420360_at 11.05 Dkk1 Dickkopf homolog 1 (Xenopus laevis)
1420565_at 10.88 Hoxa1 Homeo box A1
1417828_at 10.62 Aqp8 Aquaporin 8
1449088_at 10.47 Eef2 Eukaryotic translation elongation factor 2
1456733_x_at 10.36 Serpinh1 BB329489 RIKEN full-length enriched, 4 days neonate thymus Mus musculus cDNA clone B630008L19 3′, mRNA sequence.
1417920_at 10.22 Creb1 CAMP responsive element binding protein 1
1438558_x_at 9.989 Foxq1 forkhead box Q1
a

Average from three experiments.

b

Some genes have multiple distinct probes on the GeneChip® resulting in different fold-changes from different probes.

Table 3. B.

Top 50 genes with at least 2-fold lower expression in F9 Wt cells relative to F9 RARγ−/− cells after 24 h in the absence of RA. Fold change =WT/RARγ−/−.

Affymetrix ID Fold Changeab Gene Symbol Gene Title
1436796_at 0.307 1110061A14Rik matrin 3
1422557_s_at 0.305 Mt1 Metallothionein 1
1450292_a_at 0.305 Ap3b1 Adaptor-related protein complex 3, beta 1 subunit
1448991_a_at 0.302 Ina Internexin neuronal intermediate filament protein, alpha
1450417_a_at 0.301 Rps20 ribosomal protein S20
1457195_at 0.299 Plekhm1 cDNA sequence BC038943
1424490_at 0.292 Atp5o ATP synthase, H+ transporting, mitochondrial F1 complex, O subunit
1454681_at 0.288 2210008M09Rik cDNA sequence BC031468
1429490_at 0.286 Rif1 Rap1 interacting factor 1 homolog (yeast)
1417751_at 0.285 Mapk7 Mitogen activated protein kinase 7
1418994_at 0.283 2410116G06Rik RIKEN cDNA 2410116G06 gene
1437311_at 0.279 A930034L06Rik RIKEN cDNA A930034L06 gene
1425415_a_at 0.255 Slc1a1 Solute carrier family 1 (neuronal/epithelial high affinity glutamate transporter, system Xag), member 1
1416149_at 0.252 Akt2 Thymoma viral proto-oncogene 2
1436619_at 0.251 H3077A05-3 NIA Mouse 15K cDNA Clone Set Mus musculus cDNA clone H3077A05 3′, mRNA sequence.
1460204_at 0.251 Tsc2 Tuberous sclerosis 2
1427046_at 0.246 1810073N04Rik RIKEN cDNA 1810073N04 gene
1423281_at 0.246 Stmn2 stathmin-like 2
1440997_at 0.246 9930033H14Rik RIKEN cDNA 9930033H14 gene
1455688_at 0.245 AI838577 Adult male urinary bladder cDNA, RIKEN full-length enriched library, clone:9530036D08 product:unknown EST, full insert sequence
1439880_at 0.238 D630023F18Rik Transcribed sequences
1453219_a_at 0.231 L1td1 LINE-1 type transposase domain containing 1
1425035_s_at 0.23 Dnmt3l DNA (cytosine-5-)-methyltransferase 3-like
1421299_a_at 0.229 Lef1 Lymphoid enhancer binding factor 1
1423280_at 0.226 Stmn2 stathmin-like 2
1458599_at 0.223 Transcribed sequences
1443858_at 0.218 Trim34 RIKEN cDNA 9230105E10 gene
1448299_at 0.217 Slc1a1 Solute carrier family 1 (neuronal/epithelial high affinity glutamate transporter, system Xag), member 1
1436388_a_at 0.213 3830406C13Rik RIKEN cDNA 3830406C13 gene
1425926_a_at 0.199 Otx2 Orthodenticle homolog 2 (Drosophila)
1460386_a_at 0.197 Slc1a1 solute carrier family 1 (neuronal/epithelial high affinity glutamate transporter, system Xag), member 1
1418825_at 0.195 Psmd11 Proteasome (prosome, macropain) 26S subunit, non-ATPase, 11
1433789_at 0.158 Rnu17d 18 days pregnant adult female placenta and extra embryonic tissue cDNA, RIKEN full-length enriched library, clone:3830421G02 product:unknown EST, full insert sequence
1445271_at 0.151 9230105E10Rik RIKEN cDNA 9230105E10 gene
1445617_at 0.15 Transcribed sequences
1439207_at 0.149 Similar to paraneoplastic antigen like 5; paraneoplastic antigen family 5 (LOC386569), mRNA
1423327_at 0.133 D730048I06Rik RIKEN cDNA D730048I06 gene
1429203_at 0.131 2410076I21Rik RIKEN cDNA 2410076I21 gene
1433792_at 0.131 AW491344 Transcribed sequence with weak similarity to protein ref:NP_113662.1 (H. sapiens) hypothetical protein DKFZp761G1913 [Homo sapiens]
1448558_a_at 0.118 Pla2g4a Phospholipase A2, group IVA (cytosolic, calcium-dependent)
1422617_at 0.102 Top1 Topoisomerase (DNA) I
1429701_at 0.101 2410003J06Rik RIKEN cDNA 2410003J06 gene
1434739_at 0.0949 3830422N12Rik RIKEN cDNA 3830422N12 gene
1453544_at 0.0876 4933424F23Rik RIKEN cDNA 4933424F23 gene
1449625_at 0.0707 Transcribed sequence with weak similarity to protein ref:NP_570953.1 (M. musculus) CAMP [Mus musculus]
1419241_a_at 0.0671 Aire Autoimmune regulator (autoimmune polyendocrinopathy candidiasis ectodermal dystrophy)
1420357_s_at 0.0663 Fndc6 Fibronectin type III domain containing 6
1447021_at 0.0447 CDNA clone MGC:61032 IMAGE:30024827, complete cds
1448889_at 0.0344 Slc38a4 Solute carrier family 38, member 4
1428111_at 0.028 Slc38a4 Solute carrier family 38, member 4
1423523_at 0.0225 Aass aminoadipate-semialdehyde synthase
a

Average from three experiments.

b

Some genes have multiple distinct probes on the GeneChip® resulting in different fold-changes from different probes.

Table 4, A.

Top 50 genes with at least 2-fold higher expression in RA-treated Wt F9 cells relative to RA-treated RARγ−/− cells at 24 h. Fold change=F9 Wt/F9 RARγ−/−.

Affymetrix ID Fold Changeab Gene Symbol Gene Title
1455498_at 122.6 Transcribed sequence with weak similarity to protein pir:A43932 (H. sapiens) A43932 mucin 2 precursor, intestinal – human
1451332_at 71.76 Zfp521 Zinc finger protein 521; synonyms: Evi3, B930086A16Rik; isoform 2 is encoded by transcript variant 2; ecotropic viral integration site 3; Mus musculus zinc finger protein 521 (Zfp521), transcript variant 2, mRNA.
1441075_at 51 LOC329416 nitric oxide synthase trafficker
1452183_a_at 48.69 Gtl2 GTL2, imprinted maternally expressed untranslated mRNA
1428055_at 47.37 15 days embryo head cDNA, RIKEN full-length enriched library, clone:D930050K13 product:unclassifiable, full insert sequence
1417426_at 35.01 Prg1 Proteoglycan 1, secretory granule
1452905_at 34.73 Gtl2 GTL2, imprinted maternally expressed untranslated mRNA
1432159_a_at 31.56 Il2 Interleukin 2
1450555_at 25.66 Il2 Interleukin 2
1423294_at 25.26 Mest Transcribed sequence with moderate similarity to protein sp:Q9UBF2 (H. sapiens) CPG2_HUMAN Coatomer gamma-2 subunit
1423023_at 24.61 Sfrp5 Secreted frizzled-related sequence protein 5
1439380_x_at 24.04 Gtl2 BB093563 RIKEN full-length enriched, 12 days embryo, embryonic body between diaphragm region and neck Mus musculus cDNA clone 9430042P15 3′ similar to Y13832 Mus musculus mRNA for GT12 protein, mRNA sequence.
1415871_at 24 H2-Ab1 Histocompatibility 2, class II antigen A, beta 1
1460550_at 23.48 BC051083 cDNA sequence BC051083
1429273_at 23.48 Bmper BMP-binding endothelial regulator
1448926_at 22.68 Hoxa5 Homeo box A5
1437347_at 21.85 Ednrb endothelin receptor type B
1430637_at 20.7 2210016H18Rik Adult male stomach cDNA, RIKEN full-length enriched library, clone:2210016H18 product:hypothetical protein, full insert sequence
1421952_at 19.86 Capn6 calpain 6
1421224_a_at 19.8 Tcf2 Transcription factor 2
1456250_x_at 19.4 Tgfbi BB533460 RIKEN full-length enriched, 0 day neonate lung Mus musculus cDNA clone E030030E06 3′ similar to L19932 Mouse (beta ig-h3) mRNA, mRNA sequence.
1436172_at 19.3 Adult male testis cDNA, RIKEN full-length enriched library, clone:4921508F21 product:similar to HISTOCOMPATIBILITY 2, CLASS II ANTIGEN E BETA [Mus musculus], full insert sequence
1455626_at 17.4 Hoxa9 homeo box A9
1426758_s_at 17.38 Gtl2 GTL2, imprinted maternally expressed untranslated mRNA
1422866_at 17.33 Col13a1 Procollagen, type XIII, alpha 1
1436713_s_at 16.96 L0922F05-3 NIA Mouse Newborn Kidney cDNA Library (Long) Mus musculus cDNA clone NIA:L0922F05 IMAGE:30002176 3′, mRNA sequence.
1425273_s_at 15.6 Emp2 Epithelial membrane protein 2
1440878_at 15.18 Runx1 Runt related transcription factor 1
1459742_at 14.46 BB800744 RIKEN full-length enriched, 15 days embryo brain Mus musculus cDNA clone G630032A08 3′, mRNA sequence.
1420603_s_at 14.36 Sgk3 Serum/glucocorticoid regulated kinase 3
1422865_at 12.97 Spp1 Secreted phosphoprotein 1
1450759_at 12.89 BC019943 CDNA sequence BC019943
1433428_x_at 12.67 Tgm2 transglutaminase 2, C polypeptide
1416855_at 12.61 Gas1 BB550400 RIKEN full-length enriched, 2 days pregnant adult female oviduct Mus musculus cDNA clone E230022A16 3′ similar to X65128 M. musculus gas1 mRNA, mRNA sequence.
1448123_s_at 12.58 H2-Ab1 Histocompatibility 2, class II antigen A, beta 1
1421917_at 12.28 Pdgfra platelet derived growth factor receptor, alpha polypeptide
1452400_a_at 12.15 Hoxa11s homeo box A11, opposite strand transcript
1438558_x_at 11.59 Foxq1 forkhead box Q1
1432673_at 11.11 2300010F08Rik 12 days embryo embryonic body between diaphragm region and neck cDNA, RIKEN full-length enriched library, clone:9430087B15 product:unclassifiable, full insert sequence
1450624_at 10.81 Surf4 Surfeit gene 4
1429177_x_at 10.6 Mafg V-maf musculoaponeurotic fibrosarcoma oncogene family, protein G (avian)
1423805_at 10.3 Dab2 Disabled homolog 2 (Drosophila)
1424649_a_at 10.22 Tspan8 Tetraspanin 8
1449088_at 10.12 Eef2 Eukaryotic translation elongation factor 2
1429693_at 9.983 Dab2 Disabled homolog 2 (Drosophila)
1451191_at 9.949 Crabp1 Cellular retinoic acid binding protein I
1433615_at 9.937 B930062P21Rik RIKEN cDNA B930062P21 gene
1417787_at 9.648 Prnd Prion protein dublet
1429310_at 9.583 Flrt3 fibronectin leucine rich transmembrane protein 3
1426341_at 9.02 Slc1a3 solute carrier family 1 (glial high affinity glutamate transporter), member 3
a

Average from three experiments.

b

Some genes have multiple distinct probes on the GeneChip® resulting in different fold-changes from different probes.

Table 4, B.

Top 50 genes with at least 2-fold lower expression in RA-treated Wt F9 cells relative to RA-treated RARγ−/− cells at 24 h. Fold change=F9 Wt/F9 RARγ−/−.

Affymetrix ID Fold Changeab Gene Symbol Gene Title
1419227_at 0.195 Cct6b Chaperonin subunit 6b (zeta)
1424531_a_at 0.191 Tcea3 Transcription elongation factor A (SII), 3
1449005_at 0.188 Itgav Integrin alpha V
1436657_at 0.187 LOC380738 (LOC380738), mRNA
1455015_at 0.187 4933431N12Rik RIKEN cDNA 4933431N12 gene
1452384_at 0.187 Enpp3 ectonucleotide pyrophosphatase/phosphodiesterase 3
1455881_at 0.186 2610524G09Rik RIKEN cDNA 2610524G09 gene
1416626_at 0.185 Pla2g1b Phospholipase A2, group IB, pancreas
1441971_at 0.184 Transcribed sequences
1436677_at 0.184 1810032O08Rik sialyltransferase 7 ((alpha-N-acetylneuraminyl 2,3-beta-galactosyl-1,3)-N-acetyl galactosaminde alpha-2,6-sialyltransferase) B
1416953_at 0.183 Ctgf Connective tissue growth factor
1426808_at 0.181 Lgals3 Lectin, galactose binding, soluble 3
1450989_at 0.18 Tdgf1 teratocarcinoma-derived growth factor
1428942_at 0.178 Mt2 metallothionein 2
1434719_at 0.175 A2m Alpha-2-macroglobulin
1434094_at 0.162 6330530A05Rik RIKEN cDNA 6330530A05 gene
1423281_at 0.161 Stmn2 stathmin-like 2
1453219_a_at 0.149 L1td1 LINE-1 type transposase domain containing 1
1425035_s_at 0.144 Dnmt3l DNA (cytosine-5-)-methyltransferase 3-like
1431633_x_at 0.142 4930526L06Rik Adult male testis cDNA, RIKEN full-length enriched library, clone:4930526L06 product:unknown EST, full insert sequence
1423280_at 0.137 Stmn2 stathmin-like 2
1439207_at 0.135 Similar to paraneoplastic antigen like 5; paraneoplastic antigen family 5 (LOC386569), mRNA
1433789_at 0.13 Rnu17d 18 days pregnant adult female placenta and extra embryonic tissue cDNA, RIKEN full-length enriched library, clone:3830421G02 product:unknown EST, full insert sequence
1450943_at 0.128 Ndufa11 NADH dehydrogenase (ubiquinone) 1 alpha subcomplex 11
1439746_at 0.126 C130085G02Rik RIKEN cDNA C130085G02 gene
1423327_at 0.122 D730048I06Rik RIKEN cDNA D730048I06 gene
1423378_at 0.115 Adam23 a disintegrin and metalloprotease domain 23
1436388_a_at 0.114 3830406C13Rik RIKEN cDNA 3830406C13 gene
1417156_at 0.104 Krt19 Keratin 19
1437786_at 0.095 C80008 Transcribed sequences
1453874_at 0.0878 4933401B06Rik RIKEN cDNA 4933401B06 gene
1456035_at 0.0827 Transcribed sequences
1425926_a_at 0.0799 Otx2 Orthodenticle homolog 2 (Drosophila)
1450177_at 0.0773 Ngfr Nerve growth factor receptor (TNFR superfamily, member 16)
1417122_at 0.0767 Vav3 Vav 3 oncogene; synonyms: AA986410, MGC27838, A530094I06Rik; isoform 2 is encoded by transcript variant 2; Mus musculus vav 3 oncogene (Vav3), transcript variant 2, mRNA.
1454254_s_at 0.072 Itgb1 Integrin beta 1 (fibronectin receptor beta)
1431711_a_at 0.07 Apbb2 Amyloid beta (A4) precursor protein-binding, family B, member 2
1429701_at 0.0689 2410003J06Rik RIKEN cDNA 2410003J06 gene
1434739_at 0.0629 3830422N12Rik RIKEN cDNA 3830422N12 gene
1429203_at 0.06 2410076I21Rik RIKEN cDNA 2410076I21 gene
1423523_at 0.0587 Aass aminoadipate-semialdehyde synthase
1436799_at 0.0559 D230005D02Rik RIKEN cDNA D230005D02 gene
1447021_at 0.0509 CDNA clone MGC:61032 IMAGE:30024827, complete cds
1419540_at 0.0496 Clec5a C-type lectin domain family 5, member a
1420357_s_at 0.0474 Fndc6 Fibronectin type III domain containing 6
1448889_at 0.0334 Slc38a4 Solute carrier family 38, member 4
1419241_a_at 0.0192 Aire Autoimmune regulator (autoimmune polyendocrinopathy candidiasis ectodermal dystrophy)
1453544_at 0.0184 4933424F23Rik RIKEN cDNA 4933424F23 gene
1428111_at 0.0165 Slc38a4 Solute carrier family 38, member 4
a

Average from three experiments.

b

Some genes have multiple distinct probes on the GeneChip® resulting in different fold-changes from different probes.

The mRNA levels of these genes were greater than 10-fold higher in F9 Wt cells than in F9 RARγ−/− cells cultured in the absence of RA: retrotransposon-like 1, meprin 1 beta, nitric oxide synthase trafficker, runt related transcription factor 1(Runx-1), transcription factor 2, Zfp521 (Evi3), GTL2 (the imprinted maternally expressed untranslated mRNA), prg (proteoglycan, secretory granule), tex13 (testis expressed gene 13), Mest, Tgfbi, Bmper (BMP-binding endothelial regulator), Dab2, Col13a1, BMP6, Foxq1, aquaporin 8, EMP2 and Amot (angiomotin). Since some of these genes, such as Dab2, BMP6, Foxq1, Hoxa1, and Col13a1, play critical roles in F9 endoderm differentiation, the lower expression of these genes in the F9 RARγ−/− cells relative to F9 Wt cells could reflect a reduced differentiation potential of the F9 RARγ−/− cells. The F9 RARγ−/− cells have been shown to exhibit reduced RA associated endoderm differentiation [30].

Genes exhibiting greater than 5-fold higher mRNA levels in F9 RARγ−/− cells than in F9 Wt cells in the absence of RA included Aass (aminoadipate-semialdehyde synthase), Slc38a4, Xlr3a, Aire, RIKEN cDNA 3830422N12 gene, RIKEN cDNA 4933424F23 gene, topoisomerase (DNA) I, Pla2g4a (Phospholipase A2, group IVA (cytosolic, calcium-dependent)), Ifi1 (Proteasome (prosome, macropain) 26S subunit, non-ATPase, 11), Slc1a1, and Otx2.

The number of genes expressed at higher levels in the F9 Wt cells relative to the F9 RARγ−/− cells was 5-fold greater than the number of genes expressed at reduced levels (894 versus 164) in the control condition (vehicle treated, 24 h) (Fig. 3). This could indicate that RARγ can mediate a substantial level of transcriptional activation in the absence of RA. A complete list of these genes is in the GEO database (www.ncbi.nlm.nih.gov/geo, Accession # GSE8431).

3.4. Distinctive Expression Profiles in the F9 Wt cell line following RA addition: RA-regulated genes

RA-responsive genes in this F9 stem cell differentiation system were identified by comparing F9 Wt cells treated with 1 μM RA for 24 h versus F9 Wt cells treated with vehicle only (ethanol). Of the 45,037 gene transcripts, 345 gene transcripts (0.76 %) exhibited at least a two-fold difference in expression after RA treatment (p ≤ 0.05, Welch’s t-test). Among these, 190 (0.42%) of the gene transcripts are up-regulated and 155 (0.34%) of the gene transcripts are down-regulated by at least two-fold after RA addition. The complete list of these 345 genes, including ESTs, can be found in the GEO database (www.ncbi.nlm.nih.gov/geo, Accession # GSE8431).

Many known RA responsive genes with RAREs were identified in this analysis. They include Cdx1, a number of Hox genes (Hox-a1, b1, a4), Cyp26A1, CRABPII, and RARβ (appendix 1). The list also includes genes known to be RA-responsive, but which lack a well characterized RARE; they include AKP2, Stra6, CRABP1, Ptge1, CRBPII, (RBP2), KcNK6, Kitl, Lgals3, Lgals1, PDGFα, and Pik3r1(P85α) [52]. We also found that some genes whose expression was known to be regulated by RA in other cell lines are also regulated by RA in F9 Wt cells. These genes include Gas1 [53], IL12b, Zfp503 [54], PMP22 [53, 55], Nrip1 (Rip140) [56], C3 complement [57], keratin8, Gfra3, and Myosin light regulatory polypeptide 7.

Appendix 1. RA responsive genes in the F9 Wt cells identified using MG-430.2.

F9 Wt cells were treated for 24 h with 1 μM RA or vehicle control. Only those genes with at least a 2.0-fold change in RA treated F9 Wt cells relative to control treated cells with p values ≤0.05 are shown. Fold change=F9 Wt+RA/F9 Wt control.

Affymetrix ID Fold Changeab Gene Symbol Gene Title
1449582_at 43.69 Cdx1 Caudal type homeo box 1
1427354_at 40.16 Hoxa4 homeo box A4
1449397_at 35.68 Hoxb2 Homeo box B2
1418415_at 26.59 Il12b Interleukin 12b
1455498_at 23.18 Transcribed sequence with weak similarity to protein pir:A43932 (H. sapiens) A43932 mucin 2 precursor, intestinal-human
1423836_at 21.98 Zfp503 zinc finger protein 503
1422723_at 16.94 Stra6 Stimulated by retinoic acid gene 6
1422674_s_at 15.55 Crygb Crystallin, gamma B
1453501_at 13.08 Il12b Interleukin 12b
1422617_at 11.5 Top1 Topoisomerase (DNA) I
1435693_at 10.2 BC012256 cDNA sequence BC012256
1451191_at 9.91 Crabp1 Cellular retinoic acid binding protein I
1454906_at 8.227 Rarb retinoic acid receptor, beta
1419430_at 7.714 Cyp26a1 Cytochrome P450, family 26, subfamily a, polypeptide 1
1456229_at 7.222 H3122A02-3 NIA Mouse 15K cDNA Clone Set Mus musculus cDNA clone H3122A02 3′, mRNA sequence.
1416855_at 7.03 Gas1 BB550400 RIKEN full-length enriched, 2 days pregnant adult female oviduct Mus musculus cDNA clone E230022A16 3′ similar to X65128 M. musculus gas1 mRNA, mRNA sequence.
1420603_s_at 6.49 Sgk3 Serum/glucocorticoid regulated kinase 3
1418880_at 6.354 Gfra3 Glial cell line derived neurotrophic factor family receptor alpha 3
1448926_at 6.102 Hoxa5 Homeo box A5
1460379_at 6.055 Hoxb4 homeo box B4
1420568_at 5.652 Msc Musculin
1455037_at 5.247 Plxna2 plexin A2
1422007_at 5.069 Krt8 Keratin 8
1420565_at 5.057 Hoxa1 Homeo box A1
1451481_s_at 5.049 Cul7 Cullin 7
1433428_x_at 4.912 Tgm2 transglutaminase 2, C polypeptide
1436075_at 4.842 Sfrp5 secreted frizzled-related sequence protein 5
1416149_at 4.695 Akt2 Thymoma viral proto-oncogene 2
1427233_at 4.662 Sdccag33 serologically defined colon cancer antigen 33
1446358_at 4.603 0 day neonate cerebellum cDNA, RIKEN full-length enriched library, clone:C230033K16 product:unknown EST, full insert sequence
1418084_at 4.478 Nrp1 Neuropilin 1
1438512_at 4.414 LOC210321 epididymal protein Av381126
1418445_at 4.405 Slc16a2 solute carrier family 16 (monocarboxylic acid transporters), member 2
1423023_at 4.404 Sfrp5 Secreted frizzled-related sequence protein 5
1421474_a_at 4.372 Capzb Capping protein (actin filament) muscle Z-line, beta
1448494_at 4.321 Gas1 BB550400 RIKEN full-length enriched, 2 days pregnant adult female oviduct Mus musculus cDNA clone E230022A16 3′ similar to X65128 M. musculus gas1 mRNA, mRNA sequence.
1449089_at 4.31 Nrip1 synonyms: RIP140, 9630050P12, 6030458L20Rik; go_component: nucleus [goid 0005634] [evidence IDA] [pmid 9774688]; go_function: transcription co-repressor activity [goid 0003714] [evidence IDA] [pmid 9774688]; go_function: protein binding [goid 0005515] [evidence IPI] [pmid 9774688]; go_process: negative regulation of transcription from Pol II promoter [goid 0000122] [evidence IDA] [pmid 9774688]; Mus musculus nuclear receptor interacting protein 1 (Nrip1), mRNA.
1437277_x_at 4.262 Tgm2 transglutaminase 2, C polypeptide
1434384_at 4.237 8430438I05Rik nuclear receptor interacting protein 1
1423835_at 4.207 Zfp503 zinc finger protein 503
1418379_s_at 4.133 1700034M03Rik RIKEN cDNA 1700034M03 gene
1418496_at 4.116 Foxa2 Forkhead box A2
1435261_at 4.087 4732416N19Rik Adult male epididymis cDNA, RIKEN full-length enriched library, clone:9230114I18 product:unknown EST, full insert sequence
1427433_s_at 4.024 5730446D14Rik RIKEN cDNA 5730446D14 gene
1460605_at 4.02 AA606869 Similar to Homeobox protein goosecoid-like (GSC-2) (LOC381848), mRNA
1435184_at 4.013 B430320C24Rik 0 day neonate lung cDNA, RIKEN full-length enriched library, clone:E030020K21 product:weakly similar to HYPOTHETICAL 13.8 KDA PROTEIN [Homo sapiens], full insert sequence
1422099_a_at 3.99 Oprl1 Opioid receptor-like 1
1448327_at 3.961 AI747699 Expressed sequence AI747699
1417937_at 3.932 Ctsb Cathepsin B
1439341_at 3.903 Transcribed sequences
1422008_a_at 3.886 Krt8 Keratin 8
1417133_at 3.777 Pmp22 Peripheral myelin protein
1439561_at 3.759 2010012O05Rik BB322051 RIKEN full-length enriched, adult male adrenal gland Mus musculus cDNA clone B330007D01 3′, mRNA sequence.
1420753_at 3.719 Kitl Kit ligand
1419735_at 3.71 C3 Complement component 3
1449370_at 3.619 Sox4 SRY-box containing gene 4
1447211_at 3.611 Nrip1 nuclear receptor interacting protein 1
1449071_at 3.572 Myl7 Myosin, light polypeptide 7, regulatory
1422597_at 3.572 Mmp15 Matrix metallopeptidase 15
1452303_at 3.485 Arhgef10 ceroid-lipofuscinosis, neuronal 8
1426927_at 3.473 Ap3b2 adaptor-related protein complex 3, beta 2 subunit
1418446_at 3.453 Slc16a2 solute carrier family 16 (monocarboxylic acid transporters), member 2
1453286_at 3.445 Plxna2 BB085537 RIKEN full-length enriched, adult male diencephalon Mus musculus cDNA clone 9330200E06 3′, mRNA sequence.
1440143_at 3.434 F630022B06Rik RIKEN cDNA F630022B06 gene
1447845_s_at 3.404 Vnn1 vanin 1
1454677_at 3.373 Timp2 tissue inhibitor of metalloproteinase 2
1417500_a_at 3.367 Tgm2 Transglutaminase 2, C polypeptide
1418469_at 3.359 Nrip1 synonyms: RIP140, 9630050P12, 6030458L20Rik; go_component: nucleus [goid 0005634] [evidence IDA] [pmid 9774688]; go_function: transcription co-repressor activity [goid 0003714] [evidence IDA] [pmid 9774688]; go_function: protein binding [goid 0005515] [evidence IPI] [pmid 9774688]; go_process: negative regulation of transcription from Pol II promoter [goid 0000122] [evidence IDA] [pmid 9774688]; Mus musculus nuclear receptor interacting protein 1 (Nrip1), mRNA.
1433662_s_at 3.353 Timp2 tissue inhibitor of metalloproteinase 2
1429134_at 3.347 2900056N03Rik RIKEN cDNA 2900056N03 gene
1430425_at 3.328 5330435L01Rik RIKEN cDNA 5330435L01 gene
1417956_at 3.325 Cidea Cell death-inducing DNA fragmentation factor, alpha subunit-like effector A
1420150_at 3.316 4930422J18Rik RIKEN cDNA 4930422J18 gene
1450460_at 3.233 Krt8 Keratin 8
1437347_at 3.217 Ednrb endothelin receptor type B
1448754_at 3.201 Rbp1 Retinol binding protein 1, cellular
1420831_at 3.19 Klhl22 Kelch-like 22 (Drosophila)
1456329_at 3.171 A230098A12Rik RIKEN cDNA A230098A12 gene
1456481_at 3.169 D9Ertd280e hypothetical protein D930024E11
1418486_at 3.073 Edg2 Endothelial differentiation, lysophosphatidic acid G-protein-coupled receptor, 2
1449364_at 3.066 Aurkc Aurora kinase C
1438682_at 3.054 Pik3r1 Hypothetical protein C530050K14 (C530050K14), mRNA
1432269_a_at 2.995 Sh3kbp1 SH3-domain kinase binding protein 1
1439747_at 2.987 Ptges prostaglandin E synthase
1460204_at 2.974 Tsc2 Tuberous sclerosis 2
1450624_at 2.955 Surf4 Surfeit gene 4
1419959_s_at 2.882 C330003B14Rik RIKEN cDNA C330003B14 gene
1435743_at 2.882 C130068N17Rik RIKEN cDNA C130068N17 gene
1449925_at 2.88 Cxcr3 Chemokine (C-X-C motif) receptor 3
1416674_at 2.862 Lyzs Lysozyme
1438333_at 2.833 A230098A12Rik RIKEN cDNA A230098A12 gene
1424194_at 2.82 Rcsd1 RCSD domain containing 1
1448000_at 2.804 Cdca3 AV352659 RIKEN full-length enriched, 11 days embryo gonad Mus musculus cDNA clone 7030413D12 3′, mRNA sequence.
1429074_at 2.771 Eif5a2 Eukaryotic translation initiation factor 5A2
1428097_at 2.771 2510009E07Rik RIKEN cDNA 2510009E07 gene, mRNA (cDNA clone IMAGE:6491720), partial cds
1426589_at 2.722 Gab3 growth factor receptor bound protein 2-associated protein 3
1460287_at 2.712 Timp2 Tissue inhibitor of metalloproteinase 2
1453622_s_at 2.699 Mllt3 Myeloid/lymphoid or mixed lineage-leukemia translocation to 3 homolog (Drosophila); synonyms: Af9, D4Ertd321e, 2210011H10Rik, 2610012I03Rik, 3830408D16Rik; isoform 2 is encoded by transcript variant 2; Mus musculus myeloid/lymphoid or mixed lineage-leukemia translocation to 3 homolog (Drosophila) (Mllt3), transcript variant 2, mRNA.
1424437_s_at 2.696 Mtap4 Microtubule-associated protein 4
1457829_at 2.681 Clgn Calmegin
1430240_a_at 2.654 Clgn Calmegin
1447326_s_at 2.638 Zfp261 zinc finger protein 261
1415921_a_at 2.617 Tnf Tumor necrosis factor
1419106_at 2.611 Tmem97 Transmembrane protein 97
1427195_at 2.61 Transcribed sequence with weak similarity to protein ref:NP_081764.1 (M. musculus) RIKEN cDNA 5730493B19 [Mus musculus]
1419155_a_at 2.597 Sox4 SRY-box containing gene 4
1429959_at 2.585 6620401D04Rik 12 days embryo female ovary cDNA, RIKEN full-length enriched library, clone:6620401D04 product:interleukin 10 receptor, beta, full insert sequence
1439734_at 2.58 Transcribed sequence with weak similarity to protein pir:S12207 (M. musculus) S12207 hypothetical protein
1448470_at 2.578 Sod2 Superoxide dismutase 2, mitochondrial
1443882_at 2.575 Transcribed sequence with weak similarity to protein ref:NP_081764.1 (M. musculus) RIKEN cDNA 5730493B19 [Mus musculus]
1448825_at 2.568 Pdk2 Pyruvate dehydrogenase kinase, isoenzyme 2
1451277_at 2.551 Zfp131 Zinc finger protein 131
1431602_a_at 2.547 1810064L21Rik RIKEN cDNA A230065J02 gene
1455958_s_at 2.538 9130017A15Rik RIKEN cDNA 9130017A15 gene
1420832_at 2.535 Klhl22 Kelch-like 22 (Drosophila)
1452141_a_at 2.534 Sepp1 Selenoprotein P, plasma, 1
1452302_at 2.534 Arhgef10 ceroid-lipofuscinosis, neuronal 8
1449450_at 2.532 Ptges Prostaglandin E synthase
1419602_at 2.524 Hoxa2 Homeo box A2
1458798_at 2.501 Transcribed sequences
1424334_at 2.497 Selpl Selectin, platelet (p-selectin) ligand
1460337_at 2.454 Sh3kbp1 SH3-domain kinase binding protein 1
1455796_x_at 2.441 Olfm1 olfactomedin 1
1448024_at 2.44 B430320C24Rik 0 day neonate lung cDNA, RIKEN full-length enriched library, clone:E030020K21 product:weakly similar to HYPOTHETICAL 13.8 KDA PROTEIN [Homo sapiens], full insert sequence
1417605_s_at 2.411 St3gal2 ST3 beta-galactoside alpha-2,3-sialyltransferase 2
1460206_at 2.406 Mybbp1a MYB binding protein (P160) 1a
1450673_at 2.398 Col9a2 Procollagen, type IX, alpha 2
1429483_at 2.395 Ndp52l1 H3048G12-3 NIA Mouse 15K cDNA Clone Set Mus musculus cDNA clone H3048G12 3′, mRNA sequence.
1435740_at 2.377 E330020C23 hypothetical protein E330020C23
1454752_at 2.371 AI606861 Similar to dJ259A10.1 (ssDNA binding protein (SEB4D)) (LOC380843), mRNA
1448147_at 2.365 Tnf Tumor necrosis factor
1433844_a_at 2.362 Dusp9 dual specificity phosphatase 9
1439810_s_at 2.358 Pramel7 preferentially expressed antigen in melanoma like 7
1420924_at 2.353 Timp2 Tissue inhibitor of metalloproteinase 2
1433795_at 2.352 Tgfbr3 18-day embryo whole body cDNA, RIKEN full-length enriched library, clone:1110036H20 product:unknown EST, full insert sequence
1420774_a_at 2.343 4930570C03Rik RIKEN cDNA 4930570C03 gene
1434856_at 2.342 A130096K20 hypothetical protein A130096K20
1432331_a_at 2.338 Prrx2 Paired related homeobox 2
1418517_at 2.323 Gja4 Gap junction membrane channel protein alpha 4
1454877_at 2.315 Sertad4 16 days embryo head cDNA, RIKEN full-length enriched library, clone:C130018M11 product:unclassifiable, full insert sequence
1454727_at 2.296 AI173486 expressed sequence AI173486
1425212_a_at 2.289 Tnf Tumor necrosis factor
1417625_s_at 2.281 Cmkor1 Chemokine orphan receptor 1
1433575_at 2.281 Sox4 SRY-box containing gene 4
1448727_at 2.277 Bcl10 B-cell leukemia/lymphoma 10
1419276_at 2.255 Enpp1 Ectonucleotide pyrophosphatase/phosphodiesterase 1
1452861_at 2.245 2010300C02Rik RIKEN cDNA 2010300C02 gene
1456210_at 2.241 Transcribed sequences
1433845_x_at 2.226 Dusp9 dual specificity phosphatase 9
1430650_at 2.223 Zfp191 zinc finger protein 191
1460570_at 2.217 2900019M05Rik piggyBac transposable element derived 5
1447537_at 2.217 1500032P08Rik properdin factor, complement
1419301_at 2.216 Fzd4 602109129F1 NCI_CGAP_Kid14 Mus musculus cDNA clone IMAGE:4237519 5′, mRNA sequence.
1419309_at 2.213 Pdpn Podoplanin
1417794_at 2.197 Casp2 Caspase 2
1427401_at 2.185 Chrna5 cholinergic receptor, nicotinic, alpha polypeptide 5
1458878_at 2.175 Yes Yamaguchi sarcoma viral (v-yes) oncogene homolog
1434277_a_at 2.171 6430570G24 Adult male olfactory brain cDNA, RIKEN full-length enriched library, clone:6430570G24 product:unknown EST, full insert sequence
1435342_at 2.17 Kcnk6 potassium inwardly-rectifying channel, subfamily K, member 6
1450839_at 2.167 D0H4S114 DNA segment, human D4S114
1419157_at 2.159 Sox4 SRY-box containing gene 4
1434528_at 2.15 Aard alanine and arginine rich domain containing protein
1449110_at 2.146 App Amyloid beta (A4) precursor protein
1454737_at 2.143 Dusp9 dual specificity phosphatase 9
1419693_at 2.143 Colec12 Collectin sub-family member 12
1450992_a_at 2.136 Meis1 myeloid ecotropic viral integration site 1
1426514_at 2.133 Agrin Agrin
1417604_at 2.13 St3gal2 ST3 beta-galactoside alpha-2,3-sialyltransferase 2
1418709_at 2.126 Adarb1 Adenosine deaminase, RNA-specific, B1
1426785_s_at 2.124 Mgll monoglyceride lipase
1449449_at 2.118 Ptges Prostaglandin E synthase
1453836_a_at 2.112 Mgll Monoglyceride lipase
1422088_at 2.102 Lmyc1 lung carcinoma myc related oncogene 1
1424951_at 2.089 Cacna1d Calcium channel, voltage-dependent, L type, alpha 1D subunit
1454082_a_at 2.076 Giyd2 GIY-YIG domain containing 2
1419156_at 2.073 Sox4 SRY-box containing gene 4
1452888_at 2.061 1110034G24Rik RIKEN cDNA 1110034G24 gene
1417129_a_at 2.059 Mrg1 Myeloid ecotropic viral integration site-related gene 1
1418617_x_at 2.05 Clgn Calmegin
1426869_at 2.05 Boc biregional cell adhesion molecule-related/down-regulated by oncogenes (Cdon) binding protein
1460378_a_at 2.04 Myb; Tes Myeloblastosis oncogene; synonyms: TESS, Tes1, Tes2, testin2, D6Ertd352e; isoform 2 is encoded by transcript variant 2; testin 2; Mus musculus testis derived transcript (Tes), transcript variant 2, mRNA.
1418505_at 2.036 Ogdh Oxoglutarate dehydrogenase (lipoamide)
1434069_at 2.035 BC067047 Adult male testis cDNA, RIKEN full-length enriched library, clone:4930413J11 product:mitochondria located 1 homolog (human), full insert sequence
1442180_at 2.026 BC038059 cDNA sequence BC038059
1455642_a_at 2.023 Fbxo23 F-box only protein 23
1426991_at 2.016 1810048J11Rik RIKEN cDNA 1810048J11 gene
1459818_x_at 2.014 Zfp261 zinc finger protein 261
1434756_at 2.013 5430421B17 frizzled homolog 4 (Drosophila)
1456475_s_at 2.01 Prkar2b protein kinase, cAMP dependent regulatory, type II beta
1442308_at 2.006 Smyd4 SET and MYND domain containing 4
1418872_at 0.499 Arsk Arylsulfatase K
1416967_at 0.499 Sox2 SRY-box containing gene 2
1458277_at 0.498 Ccl25 chemokine (C-C motif) ligand 25
1424652_at 0.498 Kctd10 Potassium channel tetramerisation domain containing 10
1435436_at 0.497 Transcribed sequences
1443822_s_at 0.497 D10Ertd214e DNA segment, Chr 10, ERATO Doi 214, expressed
1450728_at 0.496 Fjx1 AV230815 RIKEN full-length enriched, 0 day neonate skin Mus musculus cDNA clone 4632401J20 3′, mRNA sequence.
1450282_at 0.496 Fgf4 Fibroblast growth factor 4
1421052_a_at 0.492 Sms Spermine synthase
1430780_a_at 0.491 Pmm1 phosphomannomutase 1
1428775_at 0.491 1110008L16Rik RIKEN cDNA 1110008L16 gene
1416605_at 0.49 Prkcbp1 Protein kinase C binding protein 1
1416505_at 0.489 Nr4a1 Nuclear receptor subfamily 4, group A, member 1
1447703_x_at 0.489 3110024A21Rik AV214133 RIKEN full-length enriched, ES cells Mus musculus cDNA clone 2410133J07 3′, mRNA sequence.
1447934_at 0.488 9630033F20Rik RIKEN cDNA 9630033F20 gene
1420964_at 0.488 Enc1 ectodermal-neural cortex 1
1451026_at 0.487 Ftsj3 FtsJ homolog 3 (E. coli)
1459658_at 0.487 Mcm5 minichromosome maintenance deficient 5, cell division cycle 46 (S. cerevisiae)
1428869_at 0.486 Nolc1 nucleolar and coiled-body phosphoprotein 1
1421260_a_at 0.484 Pgk1 Phosphoglycerate kinase 1
1428069_at 0.483 Ubqln1 Ubiquilin 1
1444883_at 0.482 Tmem19 L0079C06-3 NIA Mouse E12.5 Female Mesonephros and Gonads cDNA Library Mus musculus cDNA clone L0079C06 3′, mRNA sequence.
1430433_at 0.481 4933406J08Rik RIKEN cDNA 4933406J08 gene
1450928_at 0.481 Idb4 inhibitor of DNA binding 4
1445924_at 0.48 Transcribed sequence with weak similarity to protein ref:NP_060730.1 (H. sapiens) hypothetical protein FLJ10891 [Homo sapiens]
1441243_at 0.478 Transcribed sequence with weak similarity to protein ref:NP_081764.1 (M. musculus) RIKEN cDNA 5730493B19 [Mus musculus]
1452609_at 0.477 1190005I06Rik RIKEN cDNA 1190005I06 gene
1418649_at 0.477 Egln3 EGL nine homolog 3 (C. elegans)
1448272_at 0.477 Btg2 B-cell translocation gene 2, anti-proliferative
1444390_at 0.476 Transcribed sequences
1453251_at 0.476 Tyrp1 Tyrosinase-related protein 1
1435367_at 0.476 Mapk4 mitogen-activated protein kinase 4
1417725_a_at 0.475 Mtvr2 Mammary tumor virus receptor 2
1430208_at 0.474 Pir Pirin
1421113_at 0.474 Whrn Whirlin
1448566_at 0.469 Cdc42 Cell division cycle 42 homolog (S. cerevisiae)
1415938_at 0.469 Spink3 Serine peptidase inhibitor, Kazal type 3
1422786_at 0.469 Slc30a1 Solute carrier family 30 (zinc transporter), member 1
1431193_at 0.464 2610524B04Rik RIKEN cDNA 4932409F03 gene
1454197_a_at 0.464 Calb1 Calbindin-28K
1427640_a_at 0.463 Runx1t1 Runt-related transcription factor 1; translocated to, 1 (cyclin D-related)
1424880_at 0.463 Trib1 tribbles homolog 1 (Drosophila)
1451123_at 0.462 D19Wsu12e DNA segment, Chr 19, Wayne State University 12, expressed
1429937_at 0.461 AI662478; BB238373; 1700010I21Rik RIKEN cDNA D530033C11 gene
1417780_at 0.459 Lass4 longevity assurance homolog 4 (S. cerevisiae)
1428377_at 0.459 Btbd11 BTB (POZ) domain containing 11
1429123_at 0.457 Rab27a RAB27A, member RAS oncogene family
1419700_a_at 0.455 Prom1 Prominin 1
1436865_at 0.454 F630021I08Rik RIKEN cDNA F630021I08 gene
1433959_at 0.454 9630048M01Rik RIKEN cDNA 9630048M01 gene
1456296_at 0.453 5832426L23Rik RIKEN cDNA 5832426L23 gene
1455890_x_at 0.453 Snrpn small nuclear ribonucleoprotein N
1444292_at 0.453 D7Ertd143e NACHT, LRR and PYD containing protein 12
1437554_at 0.452 Plec1 plectin 1
1436319_at 0.452 Sulf1 sulfatase 1
1423748_at 0.452 B830012B01; D530020C15Rik pyruvate dehydrogenase kinase, isoenzyme 1
1423259_at 0.451 Idb4 inhibitor of DNA binding 4
1434815_a_at 0.45 AI874665 mq46b10.x1 Soares_thymus_2NbMT Mus musculus cDNA clone IMAGE:581755 3′, mRNA sequence.
1419708_at 0.45 Wnt6 Wingless-related MMTV integration site 6
1427023_at 0.449 Phyhipl RIKEN cDNA 4921522K17 gene
1449484_at 0.449 Hfe2 Hemochromatosis type 2 (juvenile) (human homolog)
1449187_at 0.448 Pdgfa BB371842 RIKEN full-length enriched, 16 days embryo head Mus musculus cDNA clone C130061I20 3′, mRNA sequence.
1447181_s_at 0.446 Slc7a7 solute carrier family 7 (cationic amino acid transporter, y+ system), member 7
1424310_at 0.446 Mocs2 molybdenum cofactor synthesis 2
1443435_at 0.445 Transcribed sequences
1423786_at 0.444 Eif2c3 Eukaryotic translation initiation factor 2C, 3
1424643_at 0.444 Tcof1 Treacher Collins Franceschetti syndrome 1, homolog
1428549_at 0.443 Ccdc3 coiled-coil domain containing 3
1426041_a_at 0.442 Bre; Fgd4 Brain and reproductive organ-expressed protein; synonyms: Frabp, ZFYVE6, 9030023J02Rik, 9330209B17Rik; isoform beta is encoded by transcript variant beta; frabin; Fgd1-related F-actin-binding protein; Mus musculus FYVE, RhoGEF and PH domain containing 4 (Fgd4), transcript variant beta, mRNA.; isoform gamma is encoded by transcript variant gamma; Mus musculus FYVE, RhoGEF and PH domain containing 4 (Fgd4), transcript variant gamma, mRNA.
1449996_a_at 0.441 Hspa8 Heat shock protein 8
1449031_at 0.441 Cited1 Cbp/p300-interacting transactivator with Glu/Asp-rich carboxy-terminal domain 1
1420086_x_at 0.436 Fgf4 fibroblast growth factor 4
1429442_at 0.433 4921503C21Rik RIKEN cDNA 4921503C21 gene
1422887_a_at 0.432 Ctbp2
1449146_at 0.431 Notch4 Notch gene homolog 4 (Drosophila)
1427537_at 0.428 EPIPL; EPIPL1; 6230424I18Rik epiplakin 1
1443241_at 0.427 13 days embryo stomach cDNA, RIKEN full-length enriched library, clone:D530023N15 product:unclassifiable, full insert sequence
1417392_a_at 0.426 My+lat1; AI790233 solute carrier family 7 (cationic amino acid transporter, y+ system), member 7
1417061_at 0.426 Cdc42 Cell division cycle 42 homolog (S. cerevisiae)
1427364_a_at 0.423 Odc1 Ornithine decarboxylase, structural 1
1448931_at 0.421 F2rl1 Coagulation factor II (thrombin) receptor-like 1
1448568_a_at 0.418 Keap1 Kelch-like ECH-associated protein 1
1434705_at 0.417 D7Wsu87e TRAF-binding protein
1451527_at 0.417 Pcolce2 Procollagen C-endopeptidase enhancer 2
1428283_at 0.412 Cyp2s1 Cytochrome P450, family 2, subfamily s, polypeptide 1
1434239_at 0.411 AA408556 expressed sequence AA408556
1453072_at 0.411 Gpr160 G protein-coupled receptor 160
1452384_at 0.411 Enpp3 ectonucleotide pyrophosphatase/phosphodiesterase 3
1415944_at 0.411 Sdc1 syndecan 1
1427302_at 0.409 Enpp3 ectonucleotide pyrophosphatase/phosphodiesterase 3
1423747_a_at 0.409 B830012B01; D530020C15Rik pyruvate dehydrogenase kinase, isoenzyme 1
1418744_s_at 0.407 Musk Muscle, skeletal, receptor tyrosine kinase
1448688_at 0.406 Podxl Podocalyxin-like
1436227_at 0.4 Ebaf endometrial bleeding associated factor
1431425_a_at 0.399 4930535B03Rik RIKEN cDNA 4930535B03 gene
1451502_at 0.394 Pla2g10 Phospholipase A2, group X
1444458_at 0.393 Transcribed sequence with moderate similarity to protein pir:S12207 (M. musculus) S12207 hypothetical protein
1422912_at 0.392 Bmp4 Bone morphogenetic protein 4
1433733_a_at 0.39 Cry1 cryptochrome 1 (photolyase-like)
1434458_at 0.388 Fst follistatin
1438160_x_at 0.379 Slco4a1 AV348121 RIKEN full-length enriched, adult male olfactory bulb Mus musculus cDNA clone 6430631M08 3′, mRNA sequence.
1449455_at 0.375 Ehd4 EH-domain containing 4
1442379_at 0.37 Transcribed sequences
1455692_x_at 0.37 1700097N02Rik Adult male testis cDNA, RIKEN full-length enriched library, clone:1700122G02 product:unknown EST, full insert sequence
1449109_at 0.365 Socs2 Suppressor of cytokine signaling 2
1420085_at 0.36 Fgf4 fibroblast growth factor 4
1436562_at 0.359 Ddx58 RIKEN cDNA 6430573D20 gene
1416316_at 0.355 Slc27a2 Solute carrier family 27 (fatty acid transporter), member 2
1438861_at 0.349 Bnc2 RIKEN cDNA 5031434M05 gene
1449231_at 0.349 1110008L16Rik RIKEN cDNA 1110008L16 gene
1432418_a_at 0.342 Ckmt1 Creatine kinase, mitochondrial 1, ubiquitous
1421385_a_at 0.339 Zfp191 Zinc finger protein 191
1424531_a_at 0.331 Tcea3 Transcription elongation factor A (SII), 3
1452004_at 0.325 Calca Calcitonin/calcitonin-related polypeptide, alpha
1443786_at 0.321 Utf1 BB000218 RIKEN full-length enriched, ES cells XhoI/SstI Mus musculus cDNA clone 2410014O14 3′ similar to NM_009482 Mus musculus undifferentiated embryonic cell transcription factor 1 (Utf1), mRNA sequence.
1438084_at 0.32 Transcribed sequences
1450771_at 0.319 Dgcr2 DiGeorge syndrome critical region gene 2
1417745_at 0.318 Hnrpu Heterogeneous nuclear ribonucleoprotein U
1437004_at 0.318 C78977 Similar to tripartite motif-containing 43 (LOC236469), mRNA
1416899_at 0.317 Utf1 Undifferentiated embryonic cell transcription factor 1
1426673_at 0.315 Cdh3 Cadherin 3
1425284_a_at 0.314 Ash; 2210402C08Rik; 2410003M20Rik; 4933437C11Rik RAB27A, member RAS oncogene family
1422952_at 0.308 Ng23
1440563_at 0.299 15 days embryo head cDNA, RIKEN full-length enriched library, clone:D930010N06 product:unknown EST, full insert sequence
1426431_at 0.294 Jag2 AV264681 RIKEN full-length enriched, adult male testis (DH10B) Mus musculus cDNA clone 4930502C11 3′ similar to Y14331 Mus musculus partial mRNA for jagged2 protein, clone MB16, mRNA sequence.
1416630_at 0.289 Id3 Inhibitor of DNA binding 3
1432018_at 0.287 Mas1 MAS1 oncogene
1439260_a_at 0.286 Enpp3 BB039510 RIKEN full-length enriched, 13 days embryo male testis Mus musculus cDNA clone 6030443A19 3′, mRNA sequence.
1459469_at 0.28 C78516 Transcribed sequences
1437588_at 0.279 Pou4f2 POU domain, class 4, transcription factor 2
1426808_at 0.278 Lgals3 Lectin, galactose binding, soluble 3
1419930_at 0.271 D15Ertd55e Transcribed sequences
1450194_a_at 0.27 c-myb; M16449; AI550390; MGC18531; Myb myeloblastosis oncogene; synonyms: c-myb, M16449, AI550390, MGC18531; isoform 2 is encoded by transcript variant 2; myb protein; tumor-specific myb protein; Mus musculus myeloblastosis oncogene (Myb), transcript variant 2, mRNA.
1438068_at 0.269 BB251859 RIKEN full-length enriched, 7 days neonate cerebellum Mus musculus cDNA clone A730048G08 3′, mRNA sequence.
1450989_at 0.269 Tdgf1 teratocarcinoma-derived growth factor
1425926_a_at 0.269 Otx2 Orthodenticle homolog 2 (Drosophila)
1423232_at 0.256 Map2k5 Mitogen activated protein kinase kinase 5
1440867_at 0.251 Spry4 sprouty homolog 4 (Drosophila)
1439746_at 0.246 C130085G02Rik RIKEN cDNA C130085G02 gene
1421317_x_at 0.245 c-myb; M16449; AI550390; MGC18531; Myb myeloblastosis oncogene; synonyms: c-myb, M16449, AI550390, MGC18531; isoform 2 is encoded by transcript variant 2; myb protein; tumor-specific myb protein; Mus musculus myeloblastosis oncogene (Myb), transcript variant 2, mRNA.
1422021_at 0.244 Spry4 sprouty homolog 4 (Drosophila)
1438824_at 0.234 Slc20a1 solute carrier family 20, member 1
1424942_a_at 0.219 Myc Myelocytomatosis oncogene
1445669_at 0.217 Spry4 sprouty homolog 4 (Drosophila)
1450177_at 0.217 Ngfr nerve growth factor receptor (TNFR superfamily, member 16)
1449064_at 0.212 Mab21l2 Mab-21-like 2 (C. elegans)
1430086_at 0.202 Chrna9 cholinergic receptor, nicotinic, alpha polypeptide 9
1436657_at 0.202 LOC380738 (LOC380738), mRNA
1453345_at 0.196 3830408G10Rik RIKEN cDNA 3830408G10 gene
1454974_at 0.151 Ntn1 syntaxin 8
1423378_at 0.135 Adam23 a disintegrin and metalloprotease domain 23
1444823_at 0.131 Transcribed sequences
1420463_at 0.0989 Clnk Cytokine-dependent hematopoietic cell linker
1457402_at 0.0939 0 day neonate lung cDNA, RIKEN full-length enriched library, clone:E030031B17 product:unknown EST, full insert sequence
1431711_a_at 0.0812 Apbb2 Amyloid beta (A4) precursor protein-binding, family B, member 2

We identified novel RA-regulated genes which exhibited greater than 2-fold changes compared to the levels of expression in untreated F9 Wt cells, such as Serum/glucocorticoid regulated kinase 3, Topoisomerase (DNA) I, Musculin, plexin A2, Cullin 7, Akt2, serologically defined colon cancer antigen 33, Neuropilin 1, epididymal protein Av381126, Opioid receptor-like 1, Cathepsin B, Matrix metallopeptidase 15, ceroid-lipofuscinosis neuronal 8, vanin 1, endothelin receptor type B, Tuberous sclerosis 2, Chemokine (C-X-C motif) receptor 3, Eukaryotic translation initiation factor 5A2, growth factor receptor bound protein 2-associated protein 3, Microtubule-associated protein 4, and zinc finger protein 261 (Appendix 1).

3.5. Expression Profiles in the F9 Wt and RARγ−/− cell lines following RA addition: RA-regulated genes

To identify RARγ-mediated, RA responsive genes, we compared the differentially expressed genes between the two cell types to the RA-responsive genes in the F9 Wt cells in a Venn diagram (Fig. 4). Among the 345 probe sets which were identified as RA-responsive by at least a two-fold change in expression levels after exposure to RA in F9 Wt cells, 161 probe sets were found to be differentially expressed between F9 Wt and RARγ−/− cells after a 24 h exposure to RA (Appendix 2).

Figure 4. Venn diagram of 1058 genes (Wt vs RARγ−/−), 1348 genes (Wt+RA vs RARγ−/−+RA) and 345 genes (Wt+RA vs Wt).

Figure 4

Genes from Figure 3 were analyzed to determine which of the RA-responsive genes were also differentially expressed between the F9 Wt cells and F9 RARγ−/− cells.

Appendix 2.

RA-responsive genes with at least a 2-fold change in expression in F9 Wt cells relative to F9 RARγ−/− cells after RA treatment for 24 h. Fold change = F9 Wt +RA/F9 RARγ−/− +RA.

Affymetrix ID Fold Changeab Gene Symbol Gene Title
1455498_at 122.6 Transcribed sequence with weak similarity to protein pir:A43932 (H. sapiens) A43932 mucin 2 precursor, intestinal – human
1423023_at 24.61 Sfrp5 secreted frizzled-related sequence protein 5
1448926_at 22.68 Hoxa5 Homeo box A5
1437347_at 21.85 Ednrb endothelin receptor type B
1420603_s_at 14.36 Raet1a retinoic acid early transcript alpha
1433428_x_at 12.67 Tgm2 transglutaminase 2, C polypeptide
1416855_at 12.61 Gas1 BB550400 RIKEN full-length enriched, 2 days pregnant adult female oviduct Mus musculus cDNA clone E230022A16 3′ similar to X65128 M.musculus gas1 mRNA, mRNA sequence.
1450624_at 10.81 Bhmt betaine-homocysteine methyltransferase
1451191_at 9.949 Crabp2 cellular retinoic acid binding protein II
1436075_at 8.714 Sfrp5 secreted frizzled-related sequence protein 5
1437277_x_at 8.36 Tgm2 transglutaminase 2, C polypeptide
1453501_at 8.34 Hoxb1 Homeo box B1
1448470_at 7.705 Fbp1 fructose bisphosphatase 1
1421385_a_at 7.631 Myo7a Myosin VIIa
1430425_at 6.958 5330435L01Rik RIKEN cDNA 5330435L01 gene
1450839_at 6.684 D0H4S114 DNA segment, human D4S114
1420924_at 6.492 Timp2 tissue inhibitor of metalloproteinase 2
1428097_at 6.483 2510009E07Rik RIKEN cDNA 2510009E07 gene, mRNA (cDNA clone IMAGE:6491720), partial cds
1438512_at 6.477 LOC210321 epididymal protein Av381126
1426785_s_at 6.38 Mgll monoglyceride lipase
1439810_s_at 6.263 Pramel7 preferentially expressed antigen in melanoma like 7
1433662_s_at 5.664 Timp2 tissue inhibitor of metalloproteinase 2
1427433_s_at 5.661 5730446D14Rik RIKEN cDNA 5730446D14 gene
1454677_at 5.545 Timp2 tissue inhibitor of metalloproteinase 2
1448494_at 5.482 Gas1 BB550400 RIKEN full-length enriched, 2 days pregnant adult female oviduct Mus musculus cDNA clone E230022A16 3′ similar to X65128 M.musculus gas1 mRNA, mRNA sequence.
1417500_a_at 5.389 Tgm2 transglutaminase 2, C polypeptide
1453836_a_at 5.373 Mgll monoglyceride lipase
1423835_at 5.279 Zfp503 Zinc finger protein 503
1451481_s_at 5.22 D630035O19Rik RIKEN cDNA D630035O19 gene
1448825_at 5.09 Pdk2 pyruvate dehydrogenase kinase, isoenzyme 2
1449397_at 5.047 Hoxb2 Homeo box B2
1450992_a_at 5.013 Meis1 myeloid ecotropic viral integration site 1
1433795_at 4.994 Tgfbr3 18-day embryo whole body cDNA, RIKEN full-length enriched library, clone:1110036H20 product:unknown EST, full insert sequence
1419430_at 4.84 Cyp26a1 cytochrome P450, family 26, subfamily a, polypeptide 1
1447845_s_at 4.806 Vnn1 Vanin 1
1453286_at 4.747 Plxna2 BB085537 RIKEN full-length enriched, adult male diencephalon Mus musculus cDNA clone 9330200E06 3′, mRNA sequence.
1438682_at 4.54 Pik3r1 Hypothetical protein C530050K14 (C530050K14), mRNA
1420831_at 4.52 Qscn6 quiescin Q6
1419602_at 4.455 Hoxa2 Homeo box A2
1417937_at 4.35 Dact1 dapper homolog 1, antagonist of beta-catenin (xenopus)
1449449_at 4.129 Ptges prostaglandin E synthase
1448754_at 4.118 Rbp1 Retinol binding protein 1, cellular
1440143_at 4.067 F630022B06Rik RIKEN cDNA F630022B06 gene
1418486_at 3.89 Vnn1 Vanin 1
1422674_s_at 3.854 Crygc crystallin, gamma C
1435342_at 3.836 Kcnk6 potassium inwardly-rectifying channel, subfamily K, member 6
1451277_at 3.782 C530046K17Rik RIKEN cDNA C530046K17 gene
1460287_at 3.777 Timp2 tissue inhibitor of metalloproteinase 2
1422597_at 3.726 Mmp15 Matrix metalloproteinase 15
1420565_at 3.686 Hoxa1 Homeo box A1
1454906_at 3.557 Rarb retinoic acid receptor, beta
1415938_at 3.518 Spink3 serine protease inhibitor, Kazal type 3
1419309_at 3.429 Gp38 glycoprotein 38
1435184_at 3.393 B430320C24Rik 0 day neonate lung cDNA, RIKEN full-length enriched library, clone:E030020K21 product:weakly similar to HYPOTHETICAL 13.8 KDA PROTEIN [Homo sapiens], full insert sequence
1452302_at 3.393 Arhgef10 Ceroid-lipofuscinosis, neuronal 8
1422723_at 3.387 Stra6 Stimulated by retinoic acid gene 6
1420753_at 3.375 Tll Tolloid-like
1420832_at 3.279 Qscn6 quiescin Q6
1456481_at 3.272 D9Ertd280e hypothetical protein D930024E11
1419301_at 3.228 Fzd4 602109129F1 NCI_CGAP_Kid14 Mus musculus cDNA clone IMAGE:4237519 5′, mRNA sequence.
1418709_at 3.199 Cox7a1 cytochrome c oxidase, subunit VIIa 1
1424194_at 3.175 BC025872 cDNA sequence BC025872
1434069_at 3.041 BC067047 Adult male testis cDNA, RIKEN full-length enriched library, clone:4930413J11 product:mitochondria located 1 homolog (human), full insert sequence
1452303_at 2.966 Arhgef10 Ceroid-lipofuscinosis, neuronal 8
1424334_at 2.938 Fbxo23 F-box only protein 23
1449450_at 2.922 Ptges prostaglandin E synthase
1420774_a_at 2.863 4930583H14Rik RIKEN cDNA 4930583H14 gene
1455642_a_at 2.809 Fbxo23 F-box only protein 23
1419693_at 2.805 Colec12 collectin sub-family member 12
1434277_a_at 2.711 6430570G24 Adult male olfactory brain cDNA, RIKEN full-length enriched library, clone:6430570G24 product:unknown EST, full insert sequence
1452141_a_at 2.71 Sepp1 selenoprotein P, plasma, 1
1435693_at 2.62 BC012256 cDNA sequence BC012256
1448024_at 2.559 B430320C24Rik 0 day neonate lung cDNA, RIKEN full-length enriched library, clone:E030020K21 product:weakly similar to HYPOTHETICAL 13.8 KDA PROTEIN [Homo sapiens], full insert sequence
1419155_a_at 2.549 Sox4 SRY-box containing gene 4
1433575_at 2.506 Sox4 SRY-box containing gene 4
1460605_at 2.486 AA606869 Similar to Homeobox protein goosecoid-like (GSC-2) (LOC381848), mRNA
1418084_at 2.454 Nrp Neuropilin
1419157_at 2.418 Sox4 SRY-box containing gene 4
1455796_x_at 2.418 Olfm1 olfactomedin 1
1443882_at 2.393 Transcribed sequence with weak similarity to protein ref:NP_081764.1 (M.musculus) RIKEN cDNA 5730493B19 [Mus musculus]
1427195_at 2.36 Transcribed sequence with weak similarity to protein ref:NP_081764.1 (M.musculus) RIKEN cDNA 5730493B19 [Mus musculus]
1424951_at 2.347 1300006M19Rik RIKEN cDNA 1300006M19 gene
1417133_at 2.329 Pmp22 peripheral myelin protein
1448727_at 2.293 Tle6 Transducin-like enhancer of split 6, homolog of Drosophila E(spl)
1439341_at 2.261 Transcribed sequences
1435743_at 2.244 C130068N17Rik RIKEN cDNA C130068N17 gene
1416674_at 2.235 Ptpru Protein tyrosine phosphatase, receptor type, U
1418415_at 2.225 Hoxb5 Homeo box B5
1417605_s_at 2.209 Camk1 calcium/calmodulin-dependent protein kinase I
1456329_at 2.081 A230098A12Rik RIKEN cDNA A230098A12 gene
1417956_at 2.055 Cidea cell death-inducing DNA fragmentation factor, alpha subunit-like effector A
1418445_at 2.006 Slc16a2 solute carrier family 16 (monocarboxylic acid transporters), member 2
1457829_at 2.004 Clgn Calmegin
1422912_at 0.497 Bmp4 Bone morphogenetic protein 4
1424643_at 0.483 Tcof1 Treacher Collins Franceschetti syndrome 1, homolog
1438824_at 0.466 Slc20a1 solute carrier family 20, member 1
1460204_at 0.465 Tec cytoplasmic tyrosine kinase, Dscr28C related (Drosophila)
1444390_at 0.444 Transcribed sequences
1426041_a_at 0.426 Fgd4 FYVE, RhoGEF and PH domain containing 4
1435436_at 0.405 Transcribed sequences
1430433_at 0.405 4933406J08Rik RIKEN cDNA 4933406J08 gene
1428283_at 0.401 Cyp2s1 cytochrome P450, family 2, subfamily s, polypeptide 1
1424310_at 0.399 Mocs2 molybdenum cofactor synthesis 2
1433733_a_at 0.396 Cry1 cryptochrome 1 (photolyase-like)
1430780_a_at 0.394 Pmm1 Phosphomannomutase 1
1424942_a_at 0.391 Myc Myelocytomatosis oncogene
1437588_at 0.391 Pou4f2 POU domain, class 4, transcription factor 2
1419735_at 0.39 Csnk Casein kappa
1429937_at 0.379 D530033C11Rik RIKEN cDNA D530033C11 gene
1416967_at 0.374 Sox2 SRY-box containing gene 2
1419930_at 0.372 D15Ertd55e Transcribed sequences
1422887_a_at 0.364 Ctbp2
1438861_at 0.357 Bnc2 RIKEN cDNA 5031434M05 gene
1431425_a_at 0.354 4930535B03Rik RIKEN cDNA 4930535B03 gene
1451527_at 0.345 Pcolce2 procollagen C-endopeptidase enhancer 2
1454197_a_at 0.341 D19Ertd678e unnamed protein product; hypothetical protein (evidence: rsCDS,ProCrest,decoder,NCBI CDS Predictor, Longest-ORF) putative; Mus musculus adult male testis cDNA, RIKEN full-length enriched library, clone:4933411H20 product:hypothetical protein, full insert sequence.
1436865_at 0.335 F630021I08Rik RIKEN cDNA F630021I08 gene
1449455_at 0.333 Hck hemopoietic cell kinase
1423259_at 0.332 Idb4 inhibitor of DNA binding 4
1416899_at 0.329 Utf1 undifferentiated embryonic cell transcription factor 1
1434705_at 0.329 D7Wsu87e TRAF-binding protein
1416505_at 0.321 Nr4a1 nuclear receptor subfamily 4, group A, member 1
1421317_x_at 0.314 Myb myeloblastosis oncogene
1443786_at 0.313 Utf1 BB000218 RIKEN full-length enriched, ES cells XhoI/SstI Mus musculus cDNA clone 2410014O14 3′ similar to NM_009482 Mus musculus undifferentiated embryonic cell transcription factor 1 (Utf1), mRNA sequence.
1423748_at 0.312 D530020C15Rik RIKEN cDNA D530020C15 gene
1428775_at 0.311 1110008L16Rik RIKEN cDNA 1110008L16 gene
1449231_at 0.31 Zfp296 Zinc finger protein 296
1455692_x_at 0.308 1700097N02Rik Adult male testis cDNA, RIKEN full-length enriched library, clone:1700122G02 product:unknown EST, full insert sequence
1418649_at 0.302 Egln3 EGL nine homolog 3 (C. elegans)
1438160_x_at 0.301 Slco4a1 AV348121 RIKEN full-length enriched, adult male olfactory bulb Mus musculus cDNA clone 6430631M08 3′, mRNA sequence.
1452609_at 0.298 1190005I06Rik RIKEN cDNA 1190005I06 gene
1416605_at 0.292 Nola2 nucleolar protein family A, member 2
1434239_at 0.288 AA408556 expressed sequence AA408556
1442379_at 0.287 Transcribed sequences
1432018_at 0.281 Ascl2 achaete-scute complex homolog-like 2 (Drosophila)
1420086_x_at 0.279 Fgf4 fibroblast growth factor 4
1450282_at 0.277 Fgf4 fibroblast growth factor 4
1449484_at 0.276 Stc2 stanniocalcin 2
1416316_at 0.272 Slc27a2 solute carrier family 27 (fatty acid transporter), member 2
1444292_at 0.27 D7Ertd143e NACHT, LRR and PYD containing protein 12
1440563_at 0.269 15 days embryo head cDNA, RIKEN full-length enriched library, clone:D930010N06 product:unknown EST, full insert sequence
1451026_at 0.267 Ftsj3 FtsJ homolog 3 (E. coli)
1453345_at 0.26 3830408G10Rik RIKEN cDNA 3830408G10 gene
1452004_at 0.259 Calca calcitonin/calcitonin-related polypeptide, alpha
1426431_at 0.252 Jag2 AV264681 RIKEN full-length enriched, adult male testis (DH10B) Mus musculus cDNA clone 4930502C11 3′ similar to Y14331 Mus musculus partial mRNA for jagged2 protein, clone MB16, mRNA sequence.
1438084_at 0.221 Transcribed sequences
1418744_s_at 0.215 Tesc Tescalcin
1417745_at 0.21 Cpn1 carboxypeptidase N, polypeptide 1
1420085_at 0.208 Fgf4 fibroblast growth factor 4
1453072_at 0.208 Gpr160 G protein-coupled receptor 160
1449146_at 0.199 Notch4 Notch gene homolog 4 (Drosophila)
1424531_a_at 0.191 Tcea3 transcription elongation factor A (SII), 3
1436657_at 0.187 LOC380738 (LOC380738), mRNA
1452384_at 0.187 Enpp3 Ectonucleotide pyrophosphatase/phosphodiesterase 3
1426808_at 0.181 Lgals3 Lectin, galactose binding, soluble 3
1450989_at 0.18 Tdgf1 teratocarcinoma-derived growth factor
1439746_at 0.126 C130085G02Rik RIKEN cDNA C130085G02 gene
1423378_at 0.115 Adam23 a disintegrin and metalloprotease domain 23
1425926_a_at 0.0799 Otx2 orthodenticle homolog 2 (Drosophila)
1450177_at 0.0773 Ngfr Nerve growth factor receptor (TNFR superfamily, member 16)
1431711_a_at 0.07 9030409G11Rik RIKEN cDNA 9030409G11 gene
a

Average from three experiments.

b

Some genes have multiple distinct probes on the GeneChip® resulting in different fold-changes from different probes.

Among the 1348 probe sets which were differentially expressed between the F9 Wt and RARγ−/− cell lines by at least two fold after 24 h-RA exposure, only a total of 161 probe sets were responsive to RA. These results indicate that 88% of these 1348 probe sets are not responsive to RA, or exhibit a less than two-fold change after RA addition to F9 Wt cells. The changes in mRNA levels of these genes between the two cell types indicate that the majority of genes regulated by RARγ are not RA-regulated. However, some of the genes designated as RA non-responsive genes may be partially RA responsive. Their responsiveness to RA may be less than two fold, or the responsiveness to RA may occur at a later time point. Genes in this category include GATA4, GATA6, and laminin B1. Thus, the genes listed (Appendix 2) represent only the putative RARγ-mediated, RA responsive genes at 24 h after RA addition.

3.6. Real-time RT-PCR analyses of selected genes in F9 Wt, RARγ−/−, RARα−/− and RARβ2−/− cell lines in response to RA: Peg1/mest2, tie1, sfrp5, crygc, Zfp503, Runx-1 and Raet1a

To validate the MG-430.2 microarray results and to determine if these RA-regulated genes are specifically regulated through RARγ, the mRNA levels of several of these genes were examined in the F9 Wt, RARγ−/−, RARα−/−, and RARβ2−/− cell lines. The four cell lines were cultured in the presence or absence of RA for 6 h, 24 h, or 48 h. β-actin mRNA was used for comparison. The mRNA levels of Peg1/mest2, sfrp5, crygc, Zfp503, Runx-1 and Raet1a were examined by real-time RT-PCR (Fig. 5). Tie1, a RARγ-regulated gene identified in our earlier gene chip (U74Av2) analyses, was also assessed by real-time quantitative RT-PCR (Fig. 5). These genes were selected for validation because they had relatively high fold changes and/or were found to have interesting biological functions.

Figure 5. Real-time RT-PCR analyses of Tie1, Peg1/mest2, Runx-1, crygc, sfrp5, Zfp503, and Raet1a, in F9 Wt, RARγ−/−, RARα−/−, and RARβ2−/− cell lines in response to RA.

Figure 5

Total RNA was extracted from the F9 Wt, RAR γ−/−, RARα−/−, RARβ2−/− cells cultured in the presence or absence of 1 μM RA for 6, 24, or 48 h. An equivalent amount of RNA (5 μg) was subjected to RT-PCR with primers specific for the indicated genes. Real-time quantitative (SYBR green) PCR of the indicated genes was performed and the mRNA levels were normalized to β-actin mRNA levels. The data are shown as fold increases in mRNA levels relative to the β-actin mRNA levels at each condition. Y axis, relative mRNA expression level. The results shown here are derived from three independent experiments with triplicates. The normalized mRNA levels of F9 RARα−/−, RARβ2−/−, RARγ−/− cell samples were compared to F9 Wt cell samples at the respective time points and drug treatments for statistical significance. The unpaired student’s t-test was used to determine the p values. bars, S.E. *, significant (P<0.05), **, very significant (P<0.001) in comparisons of the RNA values in the mutants with those in F9 Wt treated under the same condition.

Peg1/mest2 encodes “Paternally expressed 1 (Peg1)/mesoderm specific transcript (Mest)”, and is an imprinted gene which is only transcribed from the paternal (father’s) allele [58]. It is expressed predominantly in cells of the mesodermal lineage [59]. Our microarray data indicate that Peg1/mest mRNA levels are not regulated by RA in Wt F9 cells at 24 h (Appendix 2). Peg1/mest2 transcripts exhibit 18.6 ± 3.7-fold and 25.2 ± 6.9-fold higher levels in F9 Wt than in RARγ−/− cells in control and RA treated samples, respectively. Peg1/mest mRNA levels are lower in F9 RARα−/−, RARβ2−/−, and RARγ−/− than in F9 Wt by 45.6-fold (P<0.001), 0.37- fold (P>0.05), and 50.2-fold (P<0.001) at 6 h; 17.9-fold (P<0.001), 0.8-fold (P>0.05), and 77.8-fold (P<0.001) at 24 h; 16.2-fold (P<0.001), 0.92-fold (P>0.05), and 91.9-fold (P<0.001) at 48 h, respectively, by quantitative real time RT-PCR (Fig. 5). Thus, Peg1/mest2 is a putative target gene of both RARγ and RARα.

Runx-1 belongs to the Runt-related (Runx) transcription factor family [60]. Runx1 regulates the differentiation of hematopoietic cells. Mutations in Runx1 are closely associated with human acute myeloid leukemia [61]. Runx1 is also involved in neuronal cell fate specification and the formation of axonal projections [62]. Our microarray data indicate that Runx1 mRNA levels were not responsive to RA in F9 Wt cells at 24 h, and were 11.3 ± 3.2-fold and 15.1 ± 4.5-fold higher in F9 Wt cells than in RARγ−/− cells in control and RA treated samples, respectively. Runx-1 mRNA levels were increased by RA by 2.6 ± 0.6-fold in F9 Wt cells at 48 h (Fig. 4). A RA-associated increase in Runx1 mRNA levels of 4.2 ± 0.5-fold was also seen in RARα−/− cells at 48 h (Fig. 4). In addition, Runx-1 mRNA levels were 5- to 13-fold higher in F9 Wt than in RARγ−/− and RARβ2−/− cells at all of the time points examined (Fig. 5). These results indicate that Runx-1 is a putative target gene of both RARγ and RARβ2.

Crygc encodes gamma C-crystallin, a major component of the eye lens [63]. Mutations in the mouse Crygs gene were demonstrated to cause cataracts [64]. Our microarray data indicate that RA increased Crygc mRNA levels by 15.5 ± 2.2-fold in F9 Wt cells and that Crygc mRNA levels were 3.8 ± 0.9-fold higher in RA-treated F9 Wt cells than in RA-treated F9 RARγ−/− cells at 24 h. Real-time PCR assays confirmed that RA increased Crygc mRNA levels in F9 Wt cells by 16.9 ± 2.4-fold (P<0.001) and 36.3 ± 2.1-fold (P<0.001) at 24 h and 48 h, respectively (Fig. 5). Crygc mRNA levels were higher in F9 Wt than in F9 RARα−/−, RARβ−/−, and RARγ−/− cells by 3.8-fold (P<0.05), 12.7-fold (P<0.001), and 7.4-fold (P<0.001) respectively, after a 24 h- RA treatment (Fig. 5).

Sfrp5 is a member of the Secreted frizzled-related protein (Sfrp) gene family [41]. The secreted Sfrp proteins can bind Wnt molecules and inhibit Wnt signaling [41]. Sfrp5 is expressed in the anterior visceral endoderm (AVE) and the ventral foregut endoderm of the early mouse embryo [65, 66], though genetic evidence indicates that sfrp5 is not essential for axis development [67]. Abnormal down-regulation of Sfrp5 due to abnormal promoter methylation has been suggested in certain tumors [6870]. The microarray data indicate that RA treatment increased Sfrp5 mRNA levels by 4.0 ± 1.2-fold in Wt F9 cells, and Sfrp5 mRNA levels were 24.6 ± 5.2-fold higher in RA-treated F9 Wt than in RA-treated F9 RARγ−/− cells at 24 h. Real-time RT-PCR data confirmed that RA increased Sfrp5 mRNA levels by 2.2 ± 0.1-fold (P<0.05) and 16.2 ± 2.9-fold (P<0.001), at 24 h and 48 h, respectively, in F9 Wt cells (Fig. 5). Sfrp5 mRNA levels in F9 RARα−/−, RARβ2−/−, and RARγ−/− cells were 2.6-fold (P>0.05), 9.1-fold (P<0.001), and 13.0-fold (P<0.001) lower, respectively, than in F9 Wt cells after a 24 h-RA treatment (Fig. 5) and 1.3-fold (P>0.05), 7.9-fold (P<0.05), and 1.8-fold (P<0.05) lower respectively, than F9 Wt cells after a 48 h- RA treatment (Fig. 5).

Zfp503 (also called Nolz-1) is a member of the nocA/elB/tlp-1 family [71]. Nolz-1 mRNA was dramatically induced upon neural induction of P19 embryonal carcinoma cells by RA [54]. Our microarray data indicate that RA increased Zfp503 mRNA levels by 21.9 ± 3.7-fold in F9 Wt cells, and Zfp503 mRNA levels were 5.2 ± 1.8-fold higher in RA-treated F9 Wt cells than in RA-treated F9 RARγ−/− cells at 24 h. RA increased Zfp503 mRNA levels by 2.2 ± 0.1-fold (P<0.05) and 16.2 ± 2.9-fold (P<0.001) at 24 h and 48 h, respectively, in F9 Wt cells as measured by real-time RT-PCR (Fig. 5). Zfp503 mRNA levels were 2.5-fold (P<0.05) and 4.2-fold (P<0.001) higher in F9 Wt than in F9 RARβ2−/− and RARγ−/− cells, respectively, after a 24 h-RA treatment as measured by real time RT-PCR (Fig. 5). These results indicate that Zfp503 is a putative target gene of both RARγ and RARβ2.

Raet1a encodes a glycoprotein “retinoic acid early inducible protein 1 alpha precursor” (RAE-1alpha). The Rae proteins are anchored on the cell surface by a glycosyl phosphatidylinositol (GPI)-tail [72], and act as ligands for a natural killer cell lectin-like receptor [73]. Rae proteins are not expressed in normal, healthy tissues in adult mice, but are induced by viral infection or cellular transformation, implying a role for Rae proteins in the regulation of immunity [74]. Our microarray data indicate that RA increased Raet1a mRNA levels by 6.4 ± 1.5-fold in F9 Wt cells, and Raet1a mRNA levels were 14.3 ± 2.6-fold higher in RA-treated F9 Wt cells than in RA-treated F9 RARγ−/− cells at 24 h. RA increased Raet1a mRNA levels in F9 Wt cells by 4.9 ± 1.1-fold (P<0.001) and 11.1 ± 2.8-fold (P<0.001) at 24 h and 48 h, respectively, by real-time RT-PCR (Fig. 5). After a 24 h-RA treatment, Raet1a mRNA levels were 6.2-fold (P<0.001), 2.4-fold (P<0.05), and 7.1-fold (P<0.001) lower, respectively, in F9 RARα−/−, RARβ2−/−, and RARγ−/− cells than in F9 Wt cells. These data indicate that all of these RARs are involved in the regulation of Raet1a.

Tie1 mRNA levels were 25.0 ± 8.2-fold higher in F9 Wt cells than in F9 RARγ−/− cells after a 24 h RA treatment (Table 3). Tie1 mRNA levels were also higher in F9 Wt cells than in F9 RARγ−/− cells as assessed by semi-quantitative RT-PCR (Fig. 2). Tie1 mRNA levels increased by 4.1 ± 0.1-fold (p<0.05) and by 18.2 ±3.1-fold (p<0.05) at 24 h and 48 h after RA addition, respectively, in F9 Wt cells. Tie1 mRNA levels were 3.9-fold and 3.3- fold higher in F9 Wt cells than in F9 RARγ−/− cells at 24 h and 48 h after RA addition, respectively (Fig. 5). From these data (Fig. 5), the Tie1 gene has the characteristics of an RARγ specific target gene.

3.7. Further Analysis Using DAVID

For the purpose of understanding how the loss of RARγ affects the biology of F9 cells, we chose to identify the biological processes associated with genes that show reduced or increased expression independently. DAVID identifies categories that are over-represented in the gene list relative to the representation within the genome of a given species. First, the functional annotation clustering tool displays similar annotations together in clusters. The grouping algorithm is based on the hypothesis that similar annotations should have similar gene members. The degree of common genes between two annotations is measured by Kappa statistics, and the Kappa values are then used to classify groups of similar annotations. Next, DAVID calculates the chances of over-representation of the clusters using the Fisher Exact test. The main benefit of over-representation analysis is to order categories associated with a gene list in order to focus on those processes most likely associated with the biological phenomenon under study; in this case, the biological phenomenon is the lack of RARγ function in F9 cells.

The major biological processes that were enriched in either F9 RARγ−/− cells or in F9 Wt cells, based on the differentially expressed genes, are shown (Table 5). Based on the genes that were expressed at higher levels in the control F9 RARγ−/− cells, the biological processes pattern specification, odentogenesis, and organic acid transport were the top terms to be enriched. Four organic acid transporter genes were expressed at higher mRNA levels in F9 RARγ−/− cells than in F9 Wt cells, including Slc43, Slc1a1, Slc38a4, and Riken cDNA 9130023d20. The control F9 RARγ−/− cells were highly enriched in genes involved in pattern specification, such as Otx2, Lef1, Fst, Mib1, and FGF4. The RA treated F9 RARγ−/− cells were highly enriched in genes involved in ribosome biogenesis and cell migration, suggesting that RARγ activity plays a role in regulating ribosome biogenesis and cell migration in F9 cells. Five ribosome genes were expressed at higher mRNA levels in F9 RARγ−/− cells than in F9 Wt cells after a 24 h exposure to RA, including RpL36a, RpL27, mRpL22, RpS24, and RpS20. Genes involved in regulation of ribosome biogenesis including Ftsj3, Gtpb4, and Rrs1, were also expressed at higher mRNA levels in F9 RARγ−/− cells than in F9 Wt cells after a 24 h exposure to RA. The cell migration related genes that had higher mRNA levels in F9 RARγ−/− cells than in F9 Wt cells include Pou4f2, Ngfr, Tdgf, Rtn4, jagged 2, enabled homolog, Ctgf, Tek, and PECAM-1.

Table 5.

Biological processes that are changed in F9 RARγ−/− cells as compared to F9 Wt cells at 24 h in the presence of RA or absence of RA, as indicated by DAVID analyses. “−”, reduced processes; “+”, increased processes.

Change in F9 RARγ−/− cells
Change # of genes P*
24 h-Control Morphogenesis 73 4.4E-10
Lipid Biosynthesis 22 2.7E-6
Zinc ion homeostasis + 3 8.5E-4
Translational initiation + 3 4.6E-2
A-P Pattern specification + 4 4.5E-3
Odontogenesis + 3 7.2E-3
24 h-RA Vasculature development 14 5.6E-3
Neurogenesis 17 1.4E-2
Ribosome biogenesis and assembly + 13 7.0E-5
Cell migration + 19 1.6E-3
*

one-tail Fisher Exact Probability Value

The control F9 Wt cells were highly enriched in genes involved in cellular morphogenesis, such as Smo, Gjb3, and Dkk1, suggesting that RARγ activity is important for normal F9 cell morphogenesis. Genes up-regulated in 24 h-RA treated F9 Wt cells were found to be enriched in the biological processes, cell differentiation, nervous system development, vasculature development, and angiogenesis categories, which indicates that F9 Wt cells are induced by RA to differentiate into certain cell lineages, and that these developmental programs require RARγ activity to proceed normally.

4. DISCUSSION

4.1. Microarray analyses of F9 Wt vs. F9 RARγ−/− cells

F9 teratocarcinoma stem cells normally undergo RA-induced differentiation by activating the transcription of genes involved in cellular differentiation and development, and repressing the transcription of stem cell genes [75]. However, how gene transcription on a genome-wide scale is affected when RARγ expression is abolished is poorly understand. The goal of this research was to identify genes which showed altered expression in the absence of RARγ in F9 cells. We chose to study mRNA levels at the genome-wide level by microarray analysis.

Despite the similarity of F9 Wt and F9 RARγ−/− cells in morphology and growth characteristics [76], the gene expression profiles in the two cell lines are distinctive. A total of 1058 probe sets were differentially expressed by greater than 2-fold between the F9 Wt and RARγ−/− cell lines in the absence of exogenously added RA, suggesting that RARγ controls gene transcription in the absence of its ligand. According to the current model of gene regulation by retinoids established by Dilworth et al. [77], unliganded and DNA-bound retinoid receptors repress transcription through the recruitment of the corepressors NCoR and SMRT [78]. However, transient transfection reporter assays have demonstrated that RARγ mediated substantial levels of transcriptional activation in the absence of RA, as did RARβ [79]. Our microarray data demonstrate that a far greater number of genes are expressed at a higher level rather than a lower level in the vehicle-treated, control F9 Wt cells relative to the vehicle-treated F9 RARγ−/− cells (Fig. 3). Our data are consistent with the idea that unliganded RARγ, a nuclear transcription factor, can activate gene transcription. Unliganded RARγ only weakly interacts with SMRT [79]. The intramolecular interactions between helix 3 and helix 12 of the ligand binding domain of RARγ prevent corepressor binding [80]. Unliganded RARα, in contrast, has been shown to interact strongly with the nuclear corepressor SMRT, and RARα does repress transcription [79]. Recent studies indicate that the transcriptional activity of RARγ can also be modified by RARγ phosphorylation by ubiquitin-mediated proteasomal degradation of RARγ [81].

4.2. Comparison of this microarray research in F9 Wt cells with previous studies

There is a high similarity between the RA-responsive gene we identified in F9 Wt cells as compared to those identified by Eifert et al [82], who used 0.5 μM of RA to treat F9 Wt cells for 24 h and used Affymetrix U74AV2 arrays for analysis. However, Eifert et al. detected a greater number of genes changed by 2-fold or more in expression by RA than we did, even though the murine U74Av2 arrays they used contained a lower number of probe sets. One reason is that the data selection criteria for the two experiments were different. We employed a filter first to select genes that gave a “present” call in at least 3 chips out of a total of 12 chips for further analysis. Thus, genes that were not expressed or were weakly expressed across more than nine microarray chips were not selected, even though their mRNA fold changes between RA-treated and vehicle-treated samples were large. An examination of the data analysis protocol described by Eifert et al. suggests that they did not apply such a filter in their analyses; all of the genes that were differentially expressed were subjected to fold change analyses. Second, different concentrations of RA were used in the two studies, which may affect subsets of genes that are sensitive to RA concentration. There is another report describing microarray analysis of F9 RARγ-mediated RA responsive genes [76]. By comparison of our data with this study, there are some overlapping genes. However, because of the differences in the manner in which the microarray analyses were performed, the Parrella et al. report and our experimental design emphasized different aspects of the differentiation process. In our experiments we compared the differentially expressed genes between the F9 Wt and F9 RARγ−/− cells in the presence vs. the absence of RA, while Parrella et al. (2006) only compared differences in gene inducibility by RA between the two cell lines. These researchers also did not emphasize the quantitative differences in gene expression between the two cell lines. Quantitative differences in gene expression have important biological consequences, as demonstrated by the different developmental outcome resulting from a <2-fold variation in the expression of Oct4 in mouse ES cells [83]. Thus, the comparison of quantitative differences in gene expression between F9 Wt and F9 RARγ−/− cells in the presence versus the absence of RA, coupled with bioinformatics analyses concerning gene functions, is much more helpful in understanding the phenotypic differences between the F9 Wt and RARγ−/− cell lines.

4.3. Complex molecular mechanisms of actions of RARs

Over the last twenty years, more than 532 genes have been put forward as either direct or indirect targets of RA in permissive cellular contexts (reviewed by Balmer and Blomhoff, 2002). Our laboratory was the first to clone the Hoxa1 cDNA and show that it was a direct, primary RA target gene [84]. Transcriptional control of Hoxa1 by RA is driven by an RAR/RXR heterodimer bound to a DR5 RARE [85, 86]. Using chromatin immunoprecipitation, Gillespie and Gudas (2007) showed that RA increased the RXRα’s association with the Hoxa1 RARE [87, 88]. In the past few years, additional genes were reported to be regulated by RARs through non-classical RAREs, via coordination of other transcription factors, or through indirect pathways which involved the actions of other transcription factors or through multiple phosphorylation sites of RAR [8991]. As examined using TESS (Transcription Element Esearching Program, www.cbil.upenn.edu/tess), few of the RARγ-regulated genes identified in our microarray analyses contain classical RAREs in their 5′ or 3′ flanking regions. It is possible that RAREs are located further 5′ or 3′ of the genes, or that some of the genes are “secondary” target genes rather than “primary” targets of the RARs. Thus, more research is required to understand the detailed transcriptional mechanisms by which these genes are regulated by RARγ.

Acknowledgments

We thank members of the Gudas laboratory for scientific discussion. We thank Dr. Xiaohan Tang and Vasundhra Kashyap for reading and reviewing this manuscript and Karl Ecklund for editorial assistance. This research was supported by the National Cancer Institute (LJG) and in part by DOD predoctoral fellowship # W81XWH-04-1-0440 to DS.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Hayden LJ, Hawk SN, Sih TR, Satre MA. Metabolic conversion of retinol to retinoic acid mediates the biological responsiveness of human mammary epithelial cells to retinol. J Cell Physiol. 2001;186:437–447. doi: 10.1002/1097-4652(2000)9999:999<000::AID-JCP1043>3.0.CO;2-5. [DOI] [PubMed] [Google Scholar]
  • 2.Napoli JL. A gene knockout corroborates the integral function of cellular retinol-binding protein in retinoid metabolism. Nutr Rev. 2000;58:230–236. doi: 10.1111/j.1753-4887.2000.tb01870.x. [DOI] [PubMed] [Google Scholar]
  • 3.Blomhoff R, Blomhoff HK. Overview of retinoid metabolism and function. J Neurobiol. 2006;66(7):606–30. doi: 10.1002/neu.20242. [DOI] [PubMed] [Google Scholar]
  • 4.Love JM, Gudas LJ. Vitamin A, differentiation and cancer. Curr Op Cell Biol. 1994;6:825–831. doi: 10.1016/0955-0674(94)90051-5. [DOI] [PubMed] [Google Scholar]
  • 5.Altucci L, Rossin A, Raffelsberger W, Reitmair A, Chomienne C, Gronemeyer H. Retinoic acid-induced apoptosis in leukemia cells is mediated by paracrine action of tumor-selective death ligand TRAIL. Nat Med. 2001;7(6):680–6. doi: 10.1038/89050. [DOI] [PubMed] [Google Scholar]
  • 6.Chambon P. A decade of molecular biology of retinoic acid receptors. FASEB J. 1996;10:940–954. [PubMed] [Google Scholar]
  • 7.Kastner P, Mark M, Chambon P. Nonsteroid nuclear receptors: what are genetic studies telling us about their role in real life? Cell. 1995;83:859–869. doi: 10.1016/0092-8674(95)90202-3. [DOI] [PubMed] [Google Scholar]
  • 8.Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schütz G, Umesono K, et al. The nuclear receptor superfamily: the second decade. Cell. 1995;83:835–840. doi: 10.1016/0092-8674(95)90199-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Gudas LJ. Retinoids and vertebrate development. J Biol Chem. 1994;269:15399–15402. [PubMed] [Google Scholar]
  • 10.Means AL, Gudas LJ. The roles of retinoids in vertebrate development. Annu Rev Biochem. 1995;64:201–33. doi: 10.1146/annurev.bi.64.070195.001221. [DOI] [PubMed] [Google Scholar]
  • 11.Taneja R, Bouillet P, Boylan JF, Gaub MP, Roy B, Gudas LJ, et al. Reexpression of retinoic acid receptor (RAR)γ or RARβ in RARγ-null F9 cells reveals a partial functional redundancy between the three RAR types. Proc Natl Acad Sci. 1995;92:7854–7858. doi: 10.1073/pnas.92.17.7854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Mark M, Ghyselinck NB, Chambon P. Function of retinoid nuclear receptors: lessons from genetic and pharmacological dissections of the retinoic acid signaling pathway during mouse embryogenesis. Annu Rev Pharmacol Toxicol. 2006;46:451–80. doi: 10.1146/annurev.pharmtox.46.120604.141156. [DOI] [PubMed] [Google Scholar]
  • 13.Allan D, Houle M. Bouchard N, Meyer BI, Gruss P and Lohnes D, RARgamma and Cdx1 interactions in vertebral patterning. Dev Biol. 2001;240(1):46–60. doi: 10.1006/dbio.2001.0455. [DOI] [PubMed] [Google Scholar]
  • 14.Wendling O, Ghyselinck NB, Chambon P, Mark M. Roles of retinoic acid receptors in early embryonic morphogenesis and hindbrain patterning. Development. 2001;128:2031–2038. doi: 10.1242/dev.128.11.2031. [DOI] [PubMed] [Google Scholar]
  • 15.Ghyselinck NB, Chapellier B, Calleja C, Kumar Indra A, Li M, Messaddeq N, et al. [Genetic dissection of retinoic acid function in epidermis physiology] Ann Dermatol Venereol. 2002;129(5 Pt 2):793–9. [PubMed] [Google Scholar]
  • 16.Chapellier B, Mark M, Messaddeq N, Calleja C, Warot X, Brocard J, et al. Physiological and retinoid-induced proliferations of epidermis basal keratinocytes are differently controlled. Embo J. 2002;21(13):3402–13. doi: 10.1093/emboj/cdf331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Iulianella A, Lohnes D. Chimeric analysis of retinoic acid receptor function during cardiac looping. Dev Biol. 2002;247(1):62–75. doi: 10.1006/dbio.2002.0685. [DOI] [PubMed] [Google Scholar]
  • 18.McGowan S, Jackson SK, Jenkins-Moore M, Dai HH, Chambon P, Snyder JM. Mice bearing deletions of retinoic acid receptors demonstrate reduced lung elastin and alveolar numbers. Am J Respir Cell Mol Biol. 2000;23(2):162–7. doi: 10.1165/ajrcmb.23.2.3904. [DOI] [PubMed] [Google Scholar]
  • 19.Purton LE, Dworkin S, Olsen GH, Walkley CR, Fabb SA, Collins SJ, et al. RARgamma is critical for maintaining a balance between hematopoietic stem cell self-renewal and differentiation. J Exp Med. 2006;203(5):1283–93. doi: 10.1084/jem.20052105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Dzhagalov I, Chambon P, He YW. Regulation of CD8+ T lymphocyte effector function and macrophage inflammatory cytokine production by retinoic acid receptor gamma. J Immunol. 2007;178(4):2113–21. doi: 10.4049/jimmunol.178.4.2113. [DOI] [PubMed] [Google Scholar]
  • 21.Spanjaard RA, Ikeda M, Lee PJ, Charpentier B, Chin WW, Eberlein TJ. Specific activation of retinoic acid receptors (RARs) and retinoid X receptors reveals a unique role for RARgamma in induction of differentiation and apoptosis of S91 melanoma cells. J Biol Chem. 1997;272(30):18990–9. doi: 10.1074/jbc.272.30.18990. [DOI] [PubMed] [Google Scholar]
  • 22.Meister B, Fink FM, Hittmair A, Marth C, Widschwendter M. Antiproliferative activity and apoptosis induced by retinoic acid receptor-gamma selectively binding retinoids in neuroblastoma. Anticancer Res. 1998;18(3A):1777–86. [PubMed] [Google Scholar]
  • 23.Hatoum A, El-Sabban ME, Khoury J, Yuspa SH, Darwiche N. Overexpression of retinoic acid receptors alpha and gamma into neoplastic epidermal cells causes retinoic acid-induced growth arrest and apoptosis. Carcinogenesis. 2001;22(12):1955–63. doi: 10.1093/carcin/22.12.1955. [DOI] [PubMed] [Google Scholar]
  • 24.Pettersson F, Dalgleish AG, Bissonnette RP, Colston KW. Retinoids cause apoptosis in pancreatic cancer cells via activation of RAR-gamma and altered expression of Bcl-2/Bax. Br J Cancer. 2002;87(5):555–61. doi: 10.1038/sj.bjc.6600496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Siegrist W, Hintermann E, Roggo CN, Apfel CM, Klaus M, Eberle AN. Melanoma cell growth inhibition and melanocortin receptor downregulation induced by selective and non-selective retinoids. Melanoma Res. 1998;8(2):113–22. doi: 10.1097/00008390-199804000-00003. [DOI] [PubMed] [Google Scholar]
  • 26.Strickland S, Smith KK, Marotti KR. Hormonal induction of differentiation in teratocarcinoma stem cells: generation of parietal endoderm by retinoic acid and dibutyryl cAMP. Cell. 1980;21(2):347–55. doi: 10.1016/0092-8674(80)90471-7. [DOI] [PubMed] [Google Scholar]
  • 27.Wang S-Y, Gudas LJ. Isolation of cDNA clones specific for collagen IV and laminin from mouse teratocarcinoma cells. Proc Natl Acad Sci. 1983;80:5880–5884. doi: 10.1073/pnas.80.19.5880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Wang SY, LaRosa GJ, Gudas LJ. Molecular cloning of gene sequences transcriptionally regulated by retinoic acid and dibutyryl cyclic AMP in cultured mouse teratocarcinoma cells. Dev Biol. 1985;107(1):75–86. doi: 10.1016/0012-1606(85)90377-x. [DOI] [PubMed] [Google Scholar]
  • 29.Hogan BLM, Barlow D, Tilly R. F9 teratocarcinoma cells as a model for the differentiation of parietal and visceral endoderm in the mouse. Cancer Surv. 1983;2:115–140. [Google Scholar]
  • 30.Boylan JF, Lohnes D, Taneja R, Chambon P, Gudas LJ. Loss of retinoic acid receptor gamma function in F9 cells by gene disruption results in aberrant Hoxa-1 expression and differentiation upon retinoic acid treatment. Proc Natl Acad Sci U S A. 1993;90(20):9601–5. doi: 10.1073/pnas.90.20.9601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Faria TN, Mendelsohn C, Chambon P, Gudas LJ. The targeted disruption of both alleles of RARbeta(2) in F9 cells results in the loss of retinoic acid-associated growth arrest. J Biol Chem. 1999;274(38):26783–8. doi: 10.1074/jbc.274.38.26783. [DOI] [PubMed] [Google Scholar]
  • 32.Boylan JF, Lufkin T, Achkar CC, Taneja R, Chambon P, Gudas LJ. Targeted disruption of retinoic acid receptor alpha (RAR alpha) and RAR gamma results in receptor-specific alterations in retinoic acid-mediated differentiation and retinoic acid metabolism. Mol Cell Biol. 1995;15(2):843–51. doi: 10.1128/mcb.15.2.843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Zhuang Y, Faria TN, Chambon P, Gudas LJ. Identification and characterization of retinoic acid receptor beta2 target genes in F9 teratocarcinoma cells. Mol Cancer Res. 2003;1(8):619–30. [PubMed] [Google Scholar]
  • 34.Boylan JF, Lufkin T, Achkar CC, Taneja R, Chambon P, Gudas LJ. Targeted disruption of retinoic acid receptor α(RAR α) and RAR γ results in receptor-specific alterations in retinoic acid-mediated differentiation and retinoic acid metabolism. Mol Cell Biol. 1995;15:843–851. doi: 10.1128/mcb.15.2.843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Abu-Abed SS, Beckett BR, Chiba H, Chithalen JV, Jones G, Metzger D, et al. Mouse P450RAI (CYP26) expression and retinoic acid-inducible retinoic acid metabolism in F9 cells are regulated by retinoic acid receptor gamma and retinoid X receptor alpha. J Biol Chem. 1998;273(4):2409–15. doi: 10.1074/jbc.273.4.2409. [DOI] [PubMed] [Google Scholar]
  • 36.Wu C, Khan SA, Peng LJ, Lange AJ. Roles for fructose-2,6-bisphosphate in the control of fuel metabolism: beyond its allosteric effects on glycolytic and gluconeogenic enzymes. Adv Enzyme Regul. 2006;46:72–88. doi: 10.1016/j.advenzreg.2006.01.010. [DOI] [PubMed] [Google Scholar]
  • 37.Korhonen J, Lahtinen I, Halmekyto M, Alhonen L, Janne J, Dumont D, et al. Endothelial-specific gene expression directed by the tie gene promoter in vivo. Blood. 1995;86(5):1828–35. [PubMed] [Google Scholar]
  • 38.Puri MC, Rossant J, Alitalo K, Bernstein A, Partanen J. The receptor tyrosine kinase TIE is required for integrity and survival of vascular endothelial cells. Embo J. 1995;14(23):5884–91. doi: 10.1002/j.1460-2075.1995.tb00276.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Eklund L, Olsen BR. Tie receptors and their angiopoietin ligands are context-dependent regulators of vascular remodeling. Exp Cell Res. 2006;312(5):630–41. doi: 10.1016/j.yexcr.2005.09.002. [DOI] [PubMed] [Google Scholar]
  • 40.Liehr T, Kuhlenbaumer G, Wulf P, Taylor V, Suter U, Van Broeckhoven C, et al. Regional localization of the human epithelial membrane protein genes 1, 2, and 3 (EMP1, EMP2, EMP3) to 12p12.3, 16p13.2, and 19q13.3. Genomics. 1999;58(1):106–8. doi: 10.1006/geno.1999.5803. [DOI] [PubMed] [Google Scholar]
  • 41.Jones SE, Jomary C. Secreted Frizzled-related proteins: searching for relationships and patterns. Bioessays. 2002;24(9):811–20. doi: 10.1002/bies.10136. [DOI] [PubMed] [Google Scholar]
  • 42.Boulassel MR, Tomasi JP, Deggouj N, Gersdorff M. COCH5B2 is a target antigen of anti-inner ear antibodies in autoimmune inner ear diseases. Otol Neurotol. 2001;22(5):614–8. doi: 10.1097/00129492-200109000-00009. [DOI] [PubMed] [Google Scholar]
  • 43.Robertson NG, Skvorak AB, Yin Y, Weremowicz S, Johnson KR, Kovatch KA, et al. Mapping and characterization of a novel cochlear gene in human and in mouse: a positional candidate gene for a deafness disorder, DFNA9. Genomics. 1997;46(3):345–54. doi: 10.1006/geno.1997.5067. [DOI] [PubMed] [Google Scholar]
  • 44.Robertson NG, Lu L, Heller S, Merchant SN, Eavey RD, McKenna M, et al. Mutations in a novel cochlear gene cause DFNA9, a human nonsyndromic deafness with vestibular dysfunction. Nat Genet. 1998;20(3):299–303. doi: 10.1038/3118. [DOI] [PubMed] [Google Scholar]
  • 45.Eavey RD, Manolis EN, Lubianca J, Merchant S, Seidman JG, Seidman C. Mutations in COCH (formerly Coch5b2) cause DFNA9. Adv Otorhinolaryngol. 2000;56:101–2. doi: 10.1159/000059070. [DOI] [PubMed] [Google Scholar]
  • 46.Wang PJ, McCarrey JR, Yang F, Page DC. An abundance of X-linked genes expressed in spermatogonia. Nat Genet. 2001;27(4):422–6. doi: 10.1038/86927. [DOI] [PubMed] [Google Scholar]
  • 47.Hiriart E, Gruffat H, Buisson M, Mikaelian I, Keppler S, Meresse P, et al. Interaction of the Epstein-Barr virus mRNA export factor EB2 with human Spen proteins SHARP, OTT1, and a novel member of the family, OTT3, links Spen proteins with splicing regulation and mRNA export. J Biol Chem. 2005;280(44):36935–45. doi: 10.1074/jbc.M501725200. [DOI] [PubMed] [Google Scholar]
  • 48.Hirsch D, Stahl A, Lodish HF. A family of fatty acid transporters conserved from mycobacterium to man. Proc Natl Acad Sci U S A. 1998;95(15):8625–9. doi: 10.1073/pnas.95.15.8625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Raefski AS, O’Neill MJ. Identification of a cluster of X-linked imprinted genes in mice. Nat Genet. 2005;37(6):620–4. doi: 10.1038/ng1567. [DOI] [PubMed] [Google Scholar]
  • 50.Davies W, Isles A, Smith R, Karunadasa D, Burrmann D, Humby T, et al. Xlr3b is a new imprinted candidate for X-linked parent-of-origin effects on cognitive function in mice. Nat Genet. 2005;37(6):625–9. doi: 10.1038/ng1577. [DOI] [PubMed] [Google Scholar]
  • 51.Banerjee A, Xu HJ, Hu SX, Araujo D, Takahashi R, Stanbridge EJ, et al. Changes in growth and tumorigenicity following reconstitution of retinoblastoma gene function in various human cancer cell types by microcell transfer of chromosome 13. Cancer Res. 1992;52(22):6297–304. [PubMed] [Google Scholar]
  • 52.Bastien J, Plassat JL, Payrastre B, Rochette-Egly C. The phosphoinositide 3-kinase/Akt pathway is essential for the retinoic acid-induced differentiation of F9 cells. Oncogene. 2006;25(14):2040–7. doi: 10.1038/sj.onc.1209241. [DOI] [PubMed] [Google Scholar]
  • 53.Wang L, Mear JP, Kuan CY, Colbert MC. Retinoic acid induces CDK inhibitors and growth arrest specific (Gas) genes in neural crest cells. Dev Growth Differ. 2005;47(3):119–30. doi: 10.1111/j.1440-169X.2005.00788.x. [DOI] [PubMed] [Google Scholar]
  • 54.Chang CW, Tsai CW, Wang HF, Tsai HC, Chen HY, Tsai TF, et al. Identification of a developmentally regulated striatum-enriched zinc-finger gene, Nolz-1, in the mammalian brain. Proc Natl Acad Sci U S A. 2004;101(8):2613–8. doi: 10.1073/pnas.0308645100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Wulf P, Suter U. Embryonic expression of epithelial membrane protein 1 in early neurons. Brain Res Dev Brain Res. 1999;116(2):169–80. doi: 10.1016/s0165-3806(99)00092-9. [DOI] [PubMed] [Google Scholar]
  • 56.Kerley JS, Olsen SL, Freemantle SJ, Spinella MJ. Transcriptional activation of the nuclear receptor corepressor RIP140 by retinoic acid: a potential negative-feedback regulatory mechanism. Biochem Biophys Res Commun. 2001;285(4):969–75. doi: 10.1006/bbrc.2001.5274. [DOI] [PubMed] [Google Scholar]
  • 57.Scantlebury T, Sniderman AD, Cianflone K. Regulation by retinoic acid of acylation-stimulating protein and complement C3 in human adipocytes. Biochem J. 2001;356(Pt 2):445–52. doi: 10.1042/0264-6021:3560445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Kaneko-Ishino T, Kuroiwa Y, Miyoshi N, Kohda T, Suzuki R, Yokoyama M, et al. Peg1/Mest imprinted gene on chromosome 6 identified by cDNA subtraction hybridization. Nat Genet. 1995;11(1):52–9. doi: 10.1038/ng0995-52. [DOI] [PubMed] [Google Scholar]
  • 59.Reule M, Krause R, Hemberger M, Fundele R. Analysis of Peg1/Mest imprinting in the mouse. Dev Genes Evol. 1998;208(3):161–3. doi: 10.1007/s004270050168. [DOI] [PubMed] [Google Scholar]
  • 60.Ito Y. Oncogenic potential of the RUNX gene family: ‘overview’. Oncogene. 2004;23(24):4198–208. doi: 10.1038/sj.onc.1207755. [DOI] [PubMed] [Google Scholar]
  • 61.de Bruijn MF, Speck NA. Core-binding factors in hematopoiesis and immune function. Oncogene. 2004;23(24):4238–48. doi: 10.1038/sj.onc.1207763. [DOI] [PubMed] [Google Scholar]
  • 62.Yoshikawa M, Senzaki K, Yokomizo T, Takahashi S, Ozaki S, Shiga T. Runx1 selectively regulates cell fate specification and axonal projections of dorsal root ganglion neurons. Dev Biol. 2007;303(2):663–74. doi: 10.1016/j.ydbio.2006.12.007. [DOI] [PubMed] [Google Scholar]
  • 63.Graw J. The crystallins: genes, proteins and diseases. Biol Chem. 1997;378(11):1331–48. [PubMed] [Google Scholar]
  • 64.Graw J, Neuhauser-Klaus A, Klopp N, Selby PB, Loster J, Favor J. Genetic and allelic heterogeneity of Cryg mutations in eight distinct forms of dominant cataract in the mouse. Invest Ophthalmol Vis Sci. 2004;45(4):1202–13. doi: 10.1167/iovs.03-0811. [DOI] [PubMed] [Google Scholar]
  • 65.Finley KR. Tennessen J and Shawlot W, The mouse secreted frizzled-related protein 5 gene is expressed in the anterior visceral endoderm and foregut endoderm during early post-implantation development. Gene Expr Patterns. 2003;3(5):681–4. doi: 10.1016/s1567-133x(03)00091-7. [DOI] [PubMed] [Google Scholar]
  • 66.Kemp C, Willems E, Abdo S, Lambiv L, Leyns L. Expression of all Wnt genes and their secreted antagonists during mouse blastocyst and postimplantation development. Dev Dyn. 2005;233(3):1064–75. doi: 10.1002/dvdy.20408. [DOI] [PubMed] [Google Scholar]
  • 67.Leaf I, Tennessen J, Mukhopadhyay M, Westphal H, Shawlot W. Sfrp5 is not essential for axis formation in the mouse. Genesis. 2006;44(12):573–8. doi: 10.1002/dvg.20248. [DOI] [PubMed] [Google Scholar]
  • 68.Roman-Gomez J, Cordeu L, Agirre X, Jimenez-Velasco A, San Jose-Eneriz E, Garate L, et al. Epigenetic regulation of Wnt-signaling pathway in acute lymphoblastic leukemia. Blood. 2007;109(8):3462–9. doi: 10.1182/blood-2006-09-047043. [DOI] [PubMed] [Google Scholar]
  • 69.Nojima M, Suzuki H, Toyota M, Watanabe Y, Maruyama R, Sasaki S, et al. Frequent epigenetic inactivation of SFRP genes and constitutive activation of Wnt signaling in gastric cancer. Oncogene. 2007 doi: 10.1038/sj.onc.1210259. [DOI] [PubMed] [Google Scholar]
  • 70.Marsit CJ, McClean MD, Furniss CS, Kelsey KT. Epigenetic inactivation of the SFRP genes is associated with drinking, smoking and HPV in head and neck squamous cell carcinoma. Int J Cancer. 2006;119(8):1761–6. doi: 10.1002/ijc.22051. [DOI] [PubMed] [Google Scholar]
  • 71.Cheah PY, Meng YB, Yang X, Kimbrell D, Ashburner M, Chia W. The Drosophila l(2)35Ba/nocA gene encodes a putative Zn finger protein involved in the development of the embryonic brain and the adult ocellar structures. Mol Cell Biol. 1994;14(2):1487–99. doi: 10.1128/mcb.14.2.1487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Nomura M, Zou Z, Joh T, Takihara Y, Matsuda Y, Shimada K. Genomic structures and characterization of Rae1 family members encoding GPI-anchored cell surface proteins and expressed predominantly in embryonic mouse brain. J Biochem (Tokyo) 1996;120(5):987–95. doi: 10.1093/oxfordjournals.jbchem.a021517. [DOI] [PubMed] [Google Scholar]
  • 73.Cerwenka A, Bakker AB, McClanahan T, Wagner J, Wu J, Phillips JH, et al. Retinoic acid early inducible genes define a ligand family for the activating NKG2D receptor in mice. Immunity. 2000;12(6):721–7. doi: 10.1016/s1074-7613(00)80222-8. [DOI] [PubMed] [Google Scholar]
  • 74.Oppenheim DE, Roberts SJ, Clarke SL, Filler R, Lewis JM, Tigelaar RE, et al. Sustained localized expression of ligand for the activating NKG2D receptor impairs natural cytotoxicity in vivo and reduces tumor immunosurveillance. Nat Immunol. 2005;6(9):928–37. doi: 10.1038/ni1239. [DOI] [PubMed] [Google Scholar]
  • 75.Gudas LJ, Sporn MB, Roberts AB. Cellular biology and biochemistry of the retinoids. In: Sporn MB, Roberts AB, Goodman DS, editors. The Retinoids: Biology, Chemistry, and Medicine. Raven Press; New York: 1994. pp. 443–520. [Google Scholar]
  • 76.Parrella E, Gianni M, Fratelli M, Barzago MM, Raska I, Jr, Diomede L, et al. Antitumor activity of the retinoid-related molecules (E)-3-(4′-hydroxy-3′-adamantylbiphenyl-4-yl)acrylic acid (ST1926) and 6-[3-(1-adamantyl)-4-hydroxyphenyl]-2-naphthalene carboxylic acid (CD437) in F9 teratocarcinoma: Role of retinoic acid receptor gamma and retinoid-independent pathways. Mol Pharmacol. 2006;70(3):909–24. doi: 10.1124/mol.106.023614. [DOI] [PubMed] [Google Scholar]
  • 77.Dilworth FJ, Chambon P. Nuclear receptors coordinate the activities of chromatin remodeling complexes and coactivators to facilitate initiation of transcription. Oncogene. 2001;20(24):3047–54. doi: 10.1038/sj.onc.1204329. [DOI] [PubMed] [Google Scholar]
  • 78.Bastien J, Rochette-Egly C. Nuclear retinoid receptors and the transcription of retinoid-target genes. Gene. 2004;328:1–16. doi: 10.1016/j.gene.2003.12.005. [DOI] [PubMed] [Google Scholar]
  • 79.Hauksdóttir H, Privalsky ML. DNA recognition by the aberrant retinoic acid receptors implicated in human acute promyelocytic leukemia. Cell Growth Diff. 2001;12:85–98. [PMC free article] [PubMed] [Google Scholar]
  • 80.Farboud B, Hauksdottir H, Wu Y, Privalsky ML. Isotype-restricted corepressor recruitment: a constitutively closed helix 12 conformation in retinoic acid receptors beta and gamma interferes with corepressor recruitment and prevents transcriptional repression. Mol Cell Biol. 2003;23(8):2844–58. doi: 10.1128/MCB.23.8.2844-2858.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Bruck N, Bastien J, Bour G, Tarrade A, Plassat JL, Bauer A, et al. Phosphorylation of the retinoid X receptor at the omega loop, modulates the expression of retinoic-acid-target genes with a promoter context specificity. Cell Signal. 2005;17(10):1229–39. doi: 10.1016/j.cellsig.2004.12.006. [DOI] [PubMed] [Google Scholar]
  • 82.Eifert C, Sangster-Guity N, Yu LM, Chittur SV, Perez AV, Tine JA, et al. Global gene expression profiles associated with retinoic acid-induced differentiation of embryonal carcinoma cells. Mol Reprod Dev. 2006;73(7):796–824. doi: 10.1002/mrd.20444. [DOI] [PubMed] [Google Scholar]
  • 83.Niwa H, Miyazaki J, Smith AG. Quantitative expression of Oct-3/4 defines differentiation, dedifferentiation or self-renewal of ES cells. Nat Genet. 2000;24(4):372–6. doi: 10.1038/74199. [DOI] [PubMed] [Google Scholar]
  • 84.LaRosa GJ, Gudas LJ. An early effect of retinoic acid: cloning of an mRNA (Era-1) exhibiting rapid and protein synthesis-independent induction during teratocarcinoma stem cell differentiation. Proc Natl Acad Sci U S A. 1988;85(2):329–33. doi: 10.1073/pnas.85.2.329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Langston AW, Thompson JR, Gudas LJ. Retinoic acid-responsive elements located 3′ of the Hox A and Hox B homeobox gene clusters: functional analysis. J Biol Chem. 1997;272:2167–2175. doi: 10.1074/jbc.272.4.2167. [DOI] [PubMed] [Google Scholar]
  • 86.Langston AW, Gudas LJ. Identification of a retinoic acid responsive enhancer 3′ of the murine homeobox gene Hox 1.6. Mech Dev. 1992;38:217–228. doi: 10.1016/0925-4773(92)90055-o. [DOI] [PubMed] [Google Scholar]
  • 87.Gillespie RF, Gudas LJ. RAR isotype specificity in F9 teratocarcinoma stem cells results from the differential recruitment of coregulators to retinoic acid response elements. J Biol Chem. 2007 doi: 10.1074/jbc.M704845200. [DOI] [PubMed] [Google Scholar]
  • 88.Gillespie RF, Gudas LJ. Retinoid Regulated Association of Transcriptional Co-regulators and the Polycomb Group Protein SUZ12 with the Retinoic Acid Response Elements of Hoxa1, RARbeta(2), and Cyp26A1 in F9 Embryonal Carcinoma Cells. J Mol Biol. 2007 doi: 10.1016/j.jmb.2007.06.079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Bastien J, Adam-Stitah S, Riedl T, Egly JM, Chambon P, Rochette-Egly C. TFIIH interacts with the retinoic acid receptor gamma and phosphorylates its AF-1-activating domain through cdk7. J Biol Chem. 2000;275(29):21896–904. doi: 10.1074/jbc.M001985200. [DOI] [PubMed] [Google Scholar]
  • 90.Cerignoli F, Guo X, Cardinali B, Rinaldi C, Casaletto J, Frati L, et al. retSDR1, a short-chain retinol dehydrogenase/reductase, is retinoic acid-inducible and frequently deleted in human neuroblastoma cell lines. Cancer Res. 2002;62:1196–1204. [PubMed] [Google Scholar]
  • 91.Kopf E, Plassat JL, Vivat V, de The H, Chambon P, Rochette-Egly C. Dimerization with retinoid X receptors and phosphorylation modulate the retinoic acid-induced degradation of retinoic acid receptors alpha and gamma through the ubiquitin-proteasome pathway. J Biol Chem. 2000;275(43):33280–8. doi: 10.1074/jbc.M002840200. [DOI] [PubMed] [Google Scholar]

RESOURCES