Abstract
Thyroid hormone receptors (TRs) are critical in regulating gene expression in normal physiological processes. Decreased expression and/or somatic mutations of TRs have been shown to be associated several types of human cancers including liver, breast, lung, and thyroid. To understand the molecular mechanisms by which mutated TRs promote carcinogenesis, an animal model of follicular thyroid carcinoma (FTC) (Thrbpv/pv mice) was used in the present study. The Thrbpv/pv mouse harbors a knockin dominant negative PV mutation, identified in a patient with resistance to thyroid hormone. To understand whether oncogenic actions of PV involve not only the loss of normal TR functions but also gain-of-function activities, we compared the gene expression profiles of thyroid lesions in Thrbpv/pv mice and Thra1-/- Thrb-/- mice that also spontaneously develop FTC, but with less severe malignancy. Analysis of the cDNA microarray data derived from microdissected thyroid tumor cells of these two mice showed contrasting global gene expression profiles. With stringent selection using 2.5-fold change (p<0.01) in cDNA microarray analysis, 241 genes with altered gene expression were identified. Nearly half of the genes (n=103: 42.7% of total) with altered gene expression in thyroid tumor cells of Thrbpv/pv mice were associated with tumorigenesis and metastasis; some of these genes function as oncogenes in human thyroid cancers. The remaining genes were found to function in transcriptional regulation, RNA processing, cell proliferation, apoptosis, angiogenesis, and cytoskeleton modification. These results indicate that the more aggressive thyroid tumor progression in Thrbpv/pv mice was not due simply to the loss of tumor suppressor functions of TR via mutation but also, importantly, to gain-of-function in the oncogenic activities of PV to drive thyroid carcinogenesis. Thus, the present study identifies a novel mechanism by which a mutated TRβ evolves with an oncogenic advantage to promote thyroid carcinogenesis.
Keywords: Mutant TR, thyroid cancer, mouse model, microarray, gene expression
Introduction
Thyroid cancer is the most common malignancy of endocrine organs, and its incidence rate is steadily increasing [1]. Prognostic factors of thyroid cancer patients include tumor histologi-cal type, tumor size, the presence of lymph node metastasis, extrathyroidal extension, distance metastasis, and the existence of oncogenes [1]. Amongthese factors, the main cause of mortality in thyroid cancer is due to distant disease with about 50% survival at 3.5 years [2].
Thyroid hormone receptors (TRs), encoded by thyroid hormone receptor a (THRA) and b (THRB) genes, mediate the action of the thyroid hormone (T3) in embryonic development, cell growth, development, differentiation, and metabolic homeostasis. They are ligand-dependent transcription factors that bind to thyroid hormone response elements (TREs) in the promoter regions of target genes [3]. In view of the vital biological roles of TRs, it is reasonable to expect that their mutations could lead to deleterious effects. Indeed, mutations of the THRB gene are known to cause a genetic disease, resistance to thyroid hormone (RTH). However, whether mutations of the THRB gene also play a role in cancer development has not been clear. Loss or reduced expression of the THRB gene is reported to be closely associated with human malignancies such as breast, liver, thyroid, pituitary, colon, and renal cancers [4, 5]. High frequencies of somatic deletions, gene rearrangements, and/or loss of heterozygosity of chromosome 3p where the THRB gene is located were detected in many neoplasms [4]. In addition, somatic mutations leading to aberrant TRβ functions were identified in hepatocellular carcinomas [6], thyroid carcinomas [7], renal clear cell carcinomas [8], and pituitary tumors [9]. These observations led to a converging proposition that TRβ could function as a tumor suppressor. Indeed, cell-based studies and xenograft models have demonstrated that TRβ is a suppressor of ras-mediated cell proliferation, transformation, and tumorigenesis [10]. Moreover, TRβ disrupts the mitogenic action of growth factors by suppressing activation of extracellular signal-regulated kinase and phosphatidylinositol 3-kinase (PI3K) signaling pathways to suppress tumor cell invasiveness and metastasis [11].
That TRβ could function as a tumor suppressor is further strengthened by the compelling in vivo evidence that mice harboring a TRβ mutation spontaneously develop follicular thyroid carcinoma (FTC) similar to human thyroid cancer (Thrbpv/pv mice; [12, 13]). The PV mutation was identified in a patient with resistance to thyroid hormone (RTH) [14]. It has a frame-shift mutation in the C-terminal 14 amino acids, resulting in the complete loss of T3 binding activity and transcription capacity [15]. The phenotypic manifestation of the Thrbpv/pv mouse is reminiscent of cancer patients with somatic mutations in TRβ which have lost T3 binding and transcrip-tional capacity [6-8]. Using the Thrbpv/pv mouse, we have shown that PV functions as an oncogene by suppressing the expression and activity of a tumor suppressor, such as the peroxisome proliferator activating receptor γ (PPARγ) [16], and by activating tumor promoters such as cyclin D1, β-catenin, PI3K, AKT, and pituitary tumor transforming gene [17-21]. However, a critical question has been whether the oncogenic activity of PV is due simply to the loss of the wild -type (WT) TR tumor suppressor functions or also results from gain-of-function activities.
To address this question in vivo, we took advantage of another mutant mouse that is deficient in all functional WT TRs (Thra1-/-Thrb-/- mouse). In further support of the idea that TRs can function as a tumor suppressor, this mouse, which lacks both TRβ and TRal, also spontaneously develops FTC [22]. The Thrbpv/pv and Thra1-/- Thrb-/- mice exhibit similarly elevated serum levels of thyroid stimulating hormone (TSH) and thyroid hormones [23], but intriguingly the Thra1-/-Thrb-/- mouse develops FTC with a slower progression and a less aggressive malignant phenotype [22-25]. These observations led us to hypothesize that in addition to the loss of normal tumor suppressor functions of WT TRβ, PV could acquire additional oncogenic activity via gain-of-function through mutation. To test this hypothesis, using cDNA microarrays, we compared gene expression profiles in microdis-sected thyroid tumor lesions of age- and gender-matched Thrbpv/pv and Thra1-/-Thrb-/- mice. We found that the gene expression profiles in the thyroid tumor cells differed between Thrbpv/pv and Thra1-/-Thrb-/- mice, indicating that PV has acquired additional functions beyond simply the loss of normal TR functions. Thus, the more aggressive thyroid tumor progression in Thrbpv/pv than in Thra1-/-Thrb-/- mice resulted from dual oncogenic activity of PV. The present findings uncover a novel mechanism by which a mutated TRβ could exert deleterious effect, leading to cancer and other diseases.
Materials and methods
Animals
Mice harboring the two mutated alleles of the Thrb gene (Thrbpv/pv mice) and mice with double knockout of Thr genes (Thra1-/-Thrb-/- mice) were generated as previously described [12, 23]. As with the WT mice, these mutant mice were given normal chow and tap water and housed under 12-h light/12-h dark cycles at 22°C. Thyroid tissues or tumors were dissected out from mice around 10- to 12-months-old and kept frozen at -80°C until further use. The animal protocols used in the study were approved by the Animal Care and Use Committee at NCI.
Laser capture microdissection
Laser capture microdissection was performed on 5- to 8-μm-thick cryosections of histologically proven thyroid tissues of WT, Thra1-/-Thrb-/-, and Thrbpv/pv mice by using an ArcturusXT (Arcturus Engineering, Inc.) or Veritas machine (Arcturus Engineering, Inc). Captured cells were further processed for RNA extraction by using a Pi-coPure kit (Cat. No. 0202, Arcturus Biosciences, Inc) according to the manufacturer's instructions. The extracted total RNA was then amplified with a MessageAmp™II aRNA Amplification Kit (AM 1751, Ambion). Briefly, 0.1-1.0 ng of RNA was subjected to two rounds of amplification, and enriched aRNA was labeled with biotin-11-UTP for microarray hybridization. The quantity and quality of biotinylated aRNA were analyzed by a Nanodrop (Thermo Scientific) and 2100 Bioanalyzer (Agilent Technologies).
Microarray hybridization and data analysis
For microarray analysis, biotinylated-aRNA from three replicates of each group were used in hybridization of the GeneChip Mouse Genome 430 2.0 array (Affymetrix, Santa Clara, CA) and scanned on Affymetrix GeneChip scanner 3000. Data were collected using Affymetrix GCOS software. Statistical and clustering analysis was performed with Partek Genomics Suite software using a random multiple access normalization algorithm. Differentially expressed genes were identified with ANOVA analysis. Genes that were up- or down-regulated more than 2.5-fold with a p<0.01 were considered significant. Significant genes were analyzed for pathway enrichment and for functional annotation by using Ingenuity Pathway Analysis software (Ingenuity Systems, Inc., Redwood City, CA) and the DAVID bioinformatics database (http://david.abcc.ncifcrf.gov), respectively.
Validation of array analysis by real time RT-PCR
Real time RT-PCR was applied to confirm the differential expression of selected genes from microarray analysis. Briefly, a total 50 ng of RNA extracted from thyroid tissue of Thra1-/-Thrb-/- or Thrbpv/pv mice was used in each reaction of the real-time RT-PCR. The reactions were performed with a QuantiTech SYBR RT-PCR kit (Qiagen, Germantown, MD) on LightCycler (Roche, Indianapolis, IN). For the comparison of gene expression between Thra1-/-Thrb-/- or Thrbpv/pv mice, three or four samples were tested on each target gene. The Student's t-test was performed using GraphPad Prism version 5.00 for Mac (GraphPad Software, San Diego CA).
Results
Analysis of gene expression profiles in thyroid tumor cells of Thrbpv/pv and Thra1-/-Thrtb-/- mice
Array data were obtained from laser capture microdissected normal thyrocytes of age-matched male WT mice and thyroid lesions of ThrbPV/PV and Thra1-/-Thrb-/- mice (n=3 for each genotype of mice). Figure 1A shows the results of principal component analysis (PCA) of gene expression profiles from the three groups of mice. The three-dimensional projection of the top three principal components of PCA, capturing 70.09% of total variance, shows clear separation of the three groups (Figure 1A). The well-segregated three clusters of data derived from WT, Thrbpv/pv, and Thra1-/-Thrb-/- mice allowed us to compare changes in gene expression due to the effects of PV mutation (compare WT with Thrbpv/pv mice) or due to the lack of functional TRs (compare WT with Thra1-/-Thrb-/- mice). Subsequent comparison of gene expression profiles between the PV-mediated effects (loss of TR suppressor functions and/or oncogenic activity via gain-of-function) and the effects due to the lack of TR tumor suppressor functions allowed us to sort out the gene expression profiles due to the gain-of-function of PV critical for tumor-promoting activity.
Figure 1.
A. Principal Component Analysis (PC) of the gene expression profiles in the microdissected thyroid cells of wild-type (WT) mice and tumor cells of Thrbpv/pv and Thra1-/-Thrb-/- mice. 3D Projection of the top three principal components of PCA in (A) which captures 70.09% of total variance, shows clear separation of the three groups. B. Hierarchical clustering (average of Euclidean distance) analysis of the top 50 genes, which were filtered by the adjusted P values <0.01 and a minimum 2.5-fold change in the expression level between between Thrbpv/pv mice and Thra1-/-Thrb-/- mice.
Analysis of array data derived from the Affymetrix GeneChip, representing approximately 14,000 well-substantiated mouse genes, identified 241 genes with a significant difference in fold change >2.5 (adjusted p<0.01). Among these genes with altered expression, 97 were up-regulated and 144 were down-regulated in thyroid tumor cells of Thrbpv/pv mice as compared with those of Thra1-/-Thrb-/- mice. Hierarchical clustering analysis of the top 50 genes with altered expression profiles is shown in the Figure 1B. Contrasting expression patterns were clearly evident in the tumor cells between Thrbpv/pv and Thra1-/-Thrb-/- mice, indicating that the expression of genes mediated by mutant PV in Thrbpv/pv mice did not simply reflect from the loss of normal TR functions.
Functional classification of genes with distinct expression in thyroid tumor cells of Thrbpv/pv mice
To understand how the distinctly expressed genes were involved in the gain-of-function activity mediated by PV, we analyzed the gene ontology and functional annotation using the Database for Annotation, Visualization and Integrated Discovery (DAVID) v6.7 (http:// david.abcc.ncifcrf.gov/) and Ingenuity Pathway Analysis software (Ingenuity Systems, Inc., Redwood City, CA). In addition, we searched the GO Ontology database, as well as the PubMed data-base, for each gene, using the terms tumor, metastasis, invasion, thyroid hormone receptor, thyroid tumor, and metabolism, to identify the functions of genes. We then grouped the genes in three categories: tumor and/or metastasis-related (Table 1), thyroid or thyroid tumor-related (Table 2), and other related biological functions (Table 3). Since it is not possible within the allowed journal space to detail the cellular functions of each of 241 genes that had altered expression, only the genes that have been well studied in other cancers are highlighted in each category, as shown in the following sections. Readers should refer to the Tables for the genes that they are interested in.
Table 1-a.
Genes with altered expression having roles in tumorigenesis/metastasis (n=100)
| Symbol | Gene name | Accession | Fold change | Tumor-related | Metastasis-related | Type(s) |
|---|---|---|---|---|---|---|
| Fgg | fibrinogen gamma chain | NM_133862 | 13.1 | * | other | |
| Gfra1 | glial cell line derived neurotrophic factor family receptor alpha 1 | AV221299 | 11.78 | * | * | transmembrane receptor |
| Plac8 | placenta-specific 8 | AF263458 | 11.31 | * | other | |
| Slc16a1 | solute carrier family 16 (monocarboxylic acid transporters), member 1 | NM_009196 | 9.48 | * | transporter | |
| Eid1 | EP300 interacting inhibitor of differentiation 1 | BC010712 | 7.78 | * | transcription regulator | |
| Ap1s3 | adaptor-related protein complex AP-1, sigma 3 | AW259574 | 6.56 | * | transporter | |
| Camk2n2 | calcium/calmodulin-dependent protein kinase II inhibitor 2 | AK013788 | 6.1 | * | other | |
| Shh | sonic hedgehog | AV304616 | 4.66 | * | peptidase | |
| Masp1 | mannan-binding lectin serine peptidase 1 | AB049755 | 4.42 | * | peptidase | |
| Arsb | arylsulfatase B | BI440651 | 4.35 | * | enzyme | |
| Rassf4 | Ras association (RalGDS/AF-6) domain family member 4 | AV291679 | 4.32 | * | * | other |
| Lad1 | ladinin | NM_133664 | 4.28 | * | other | |
| Bach2 | BTB and CNC homology 2 | BB529913 | 3.99 | * | transcription regulator | |
| Usp2 | ubiquitin specific peptidase 2 | AI553394 | 3.88 | * | peptidase | |
| Adi1 | acireductone dioxygenase 1 | NM_134052 | 3.7 | * | enzyme | |
| Timp2 | tissue inhibitor of metalloproteinase 2 | BF168458 | 3.69 | * | * | other |
| C4bp | complement component 4 binding protein | NM_007576 | 3.56 | * | other | |
| Gng10 | guanine nucleotide binding protein (G protein), gamma 10 | NM_025277 | 3.56 | * | enzyme | |
| Sirpa | signal-regulatory protein alpha | AB018194 | 3.53 | * | phosphatase | |
| Idh1 | isocitrate dehydrogenase 1 (NADP+), soluble | NM_010497 | 3.44 | * | * | enzyme |
| B4galnt2 | beta-l,4-N-acetyl-galactosaminyl transferase 2 | AI593864 | 3.34 | * | enzyme | |
| Prkcsh | protein kinase C substrate 80K-H | NM_008925 | 3.34 | * | enzyme | |
| Mt3 | metallothionein 3 | NM_013603 | 3.28 | * | other | |
| Ccl6 | chemokine (C-C motif) ligand 6 | AV084904 | 3.21 | * | cytokine | |
| Muc4 | mucin 4 | AF218265 | 3.14 | * | * | growth factor |
| Itpr1 | inositol 1,4,5-triphosphate receptor 1 | NM_010585 | 2.99 | * | ion channel | |
| Prdx3 | peroxiredoxin 3 | NM_007452 | 2.83 | * | enzyme | |
| Ube2a | ubiquitin-conjugating enzyme E2A, RAD6 homolog(S. cerevisiae) | BG868960 | 2.83 | * | enzyme | |
| Thy1 | thymus cell antigen 1, theta | AV028402 | 2.82 | * | other | |
| Fdft1 | farnesyl diphosphate farnesyl transferase 1 | BB028312 | 2.81 | * | enzyme | |
| Pnkd | paroxysmal nonkinesiogenic dyskinesia | NM_019999 | 2.79 | * | * | other |
| Sema4d | sema domain, immunoglobulin domain (Ig), transmembrane domain (TM) and short cytoplasmic domain, (semaphorin) 4D | AV256403 | 2.78 | * | * | other |
| Slc9a3r1 | solute carrier family 9 (sodium/hydrogen exchanger), member 3 regulator 1 | BG066200 | 2.78 | * | other | |
| Uqcrh | ubiquinol-cytochrome c reductase hinge protein | AK019085 | 2.74 | * | ||
| Aebp1 | AE binding protein 1 | NM_009636 | 2.73 | * | peptidase | |
| Rpa1 | replication protein Al | BB491281 | 2.72 | * | other | |
| Dnajc15 | DnaJ (Hsp40) homolog, subfamily C, member 15 | NM_025384 | 2.7 | * | other | |
| Hnf1b | HNF1 homeobox B | AI987804 | 2.7 | * | transcription regulator | |
| Kctd12 | potassium channel tetramerisation domain containing 12 | BM220945 | 2.7 | * | ion channel | |
| Mobk13 | MOB1, Mps One Binder kinase activator-like 3 (yeast) | AK011829 | 2.69 | * | other | |
| Pigy | phosphatidylinositol glycan anchor biosynthesis, class Y | AK003713 | 2.65 | * | other | |
| Nudt2 | nudix (nucleoside diphosphate linked moiety X)-type motif 2 | NM_025539 | 2.64 | * | phosphatase | |
| Esr1 | estrogen receptor 1 (alpha) | AI646838 | 2.61 | * | * | ligand-dependent nuclear receptor |
| Mrpl38 | mitochondrial ribosomal protein L38 | B1135190 | 2.6 | * | other | |
| Prdm2 | PR domain containing 2, with ZNF domain | BM226301 | 2.6 | * | transcription regulator | |
| Rnf4 | ring finger protein 4 | AV045658 | 2.6 | * | transcription regulator | |
| Ccdc56 | coiled-coil domain containing 56 | C77389 | 2.58 | * | other | |
| Uba1 | ubiquitin-like modifier activating enzyme 1 | NM_009457 | 2.53 | * | enzyme | |
| Plekha8 | pleckstrin homology domain containing, family A (phosphoinositide binding specific) member 8 | AI324154 | 2.52 | * | other | |
| Bmp5 | bone morphogenetic protein 5 | NM_007555 | -2.61 | * | growth factor | |
| Ggnbp2 | gametogenetin binding protein 2 | BG078867 | -2.67 | * | other | |
| Tbl1xr1 | transducin (beta)-like lX-linked receptor 1 | BG071620 | -2.69 | * | transcription regulator | |
| Creb1 | cAMP responsive element binding protein 1 | AK014391 | -2.71 | * | transcription regulator | |
| Rbm5 | RNA binding motif protein 5 | BE446879 | -2.74 | * | * | other |
| Mcf2l | mcf.2 transforming sequence-like | AV293368 | -2.75 | * | other | |
| Lair1 | leukocyte-associated Ig-like receptor 1 | BB667693 | -2.76 | * | transmembrane receptor | |
| Abcc5 | ATP-binding cassette, sub-family C (CFTR/MRP), member 5 | BB436535 | -2.79 | * | transporter | |
| Khdrbs1 | KH domain containing, RNA binding, signal transduction associated 1 | AV117555 | -2.79 | * | transcription regulator | |
| Rgnef | Rho-guanine nucleotide exchange factor | BB530298 | -2.79 | * | * | other |
| Ccdc68 | coiled-coil domain containing 68 | AV378320 | -2.81 | * | other | |
| Birc6 | baculoviral IAP repeat-containing 6 | BB527646 | -2.82 | * | enzyme | |
| Fus | fusion, derived from t(12;16) malignant liposarcoma (human) | BE985138 | -2.83 | * | transcription regulator | |
| Bach1 | BTB and CNC homology 1 | NM_007520 | -2.86 | * | transcription regulator | |
| Efnb2 | ephrin B2 | U30244 | -2.87 | * | other | |
| Elf2 | E74-like factor 2 | BC027739 | -2.9 | * | transcription regulator | |
| Notch2 | Notch gene homolog 2 (Drosophila) | AI787996 | -2.9 | * | transcription regulator | |
| Bnip2 | BCL2/adenovirus E1B interacting protein 2 | AV144704 | -2.93 | * | other | |
| Alcam | activated leukocyte cell adhesion molecule | BB534113 | -3.05 | * | * | other |
| Kcnq1ot1 | KCNQ1 overlapping transcript 1 | BG063584 | -3.44 | * | other | |
| Bean | brevican | BB335613 | -3.59 | * | other | |
| Mirhg1 | microRNA host gene 1 (non-protein coding) | AK017164 | -3.6 | * | other | |
| Nfia | nuclear factor I/A | AF326553 | -3.6 | * | transcription regulator | |
| Sp4 | trans-acting transcription factor 4 | NM_009239 | -3.76 | * | transcription regulator | |
| Cpeb3 | cytoplasmic polyadenylation element binding protein 3 | BB281000 | -3.78 | * | other | |
| Bmf | BCL2 modifying factor | BB212341 | -3.82 | * | other | |
| Sfpq | splicing factor proline/glutamine rich (polypyrimidine tract binding protein associated); similar to PTB-associated splicing factor | BI738328 | -3.92 | * | other | |
| Crtc3 | CREB regulated transcription coactivator3 | AI429792 | -3.96 | * | other | |
| Krit1 | KRIT1, ankyrin repeat containing | BI793814 | -4.02 | * | other | |
| Srd5a1 | steroid 5 alpha-reductase 1 | AA530749 | -4.08 | * | enzyme | |
| Vegfc | vascular endothelial growth factor C | NM_009506 | -4.17 | * | * | growth factor |
| Cckrs | CDC2-related kinase, arginine/serine-rich | BG070845 | -4.25 | * | enzyme | |
| Ednrb | endothelin receptor type B | BB451714 | -4.35 | * | G-protein coupled receptor | |
| Itsn2 | intersectin 2 | AI326108 | -4.36 | * | other | |
| Igf1r | insulin-like growth factor 1 receptor | AV374369 | -4.42 | * | transmembrane receptor | |
| Nr2f2 | similar to COUP-TFI; nuclear receptor subfamily 2, group F, member 2 | AI463873 | -4.44 | * | * | ligand-dependent nuclear receptor |
| Nmt2 | N-myristoyltransferase 2 | BB409982 | -4.5 | * | enzyme | |
| Smc4 | structural maintenance of chromosomes 4 | BI665568 | -4.79 | * | transporter | |
| Rtn4 | reticulon 4 | BG072267 | -4.91 | * | other | |
| Mtss1 | metastasis suppressor 1 | BB157298 | -5.09 | * | * | other |
| Vps37a | vacuolar protein sorting 37A (yeast) | AK008752 | -5.78 | * | * | other |
| Dnmt3a | DNA methyltransf erase 3A | BB795491 | -5.97 | * | enzyme | |
| Git2 | G protein-coupled receptor kinase-interactor 2 | AK017943 | -6.4 | * | other | |
| Sdha | succinate dehydrogenase complex, subunit A, flavoprotein (Fp) | AK005350 | -6.86 | * | enzyme | |
| Cspg4 | chondroitin sulfate proteoglycan 4 | BB377873 | -7.13 | * | * | other |
| Cpd | carboxypeptidase D | NM_007754 | -8.93 | * | peptidase | |
| Bclaf1 | BCL2-associated transcription factor 1 | BE853331 | -10.45 | * | transcription regulator | |
| Wnt4 | wingless-related MMTV integration site 4 | AW047257 | -10.61 | * | other | |
| Abca3 | ATP-binding cassette, sub-family A (ABC1), member 3 | BC006932 | -11.71 | * | * | transporter |
| Ddx6 | DEAD (Asp-Glu-Ala-Asp) box polypeptide 6 | BC021452 | -15.13 | * | enzyme | |
| Sik1 | salt inducible kinase 1 | AI648260 | -21.3 | * | * | kinase |
Table 2.
Thyroid cancer-related genes with altered expression in Thrbpv/pv mice (n=19)
| Symbol | Gene name | Accession No. | Fold change | Tumor-related | Metastasis-related | Thyroid/thyroid tumor-related | Note |
|---|---|---|---|---|---|---|---|
| Gfra1 | glial cell line derived neurotrophic factor family receptor alpha 1 | AV22129 9 | 11.78 | * | * | * | medullary thyroid carcinomas (MTC)-and RET oncogene-related |
| Chrdl1 | chordin-like 1 | AV14414 5 | 5.54 | * | down-regulation in FTC | ||
| Shh | sonic hedgehog | AV30461 6 | 4.66 | * | * | thyroid development | |
| Minpp1 | multiple inositol polyphosphate histidine phosphatase 1 | AV33936 6 | 4.07 | * | mutation found in FTC | ||
| Thra | thyroid hormone receptor alpha; similar to thyroid hormone receptor | BI07668 9 | 3.55 | * | |||
| Idh1 | isocitrate dehydrogenase 1 (NADP+), soluble | NM 0104 97 | 3.44 | * | * | * | mutation in FTC and ATC |
| Slc5a5 | solute carrier family 5 (sodium iodide symporter), member 5 | AF38035 3 | 3.15 | * | iodide transporter | ||
| Muc4 | mucin 4 | AF21826 5 | 3.14 | * | * | * | thyroid tumor |
| Hnf1b | HNF1 homeobox B | AI987804 | 2.70 | * | * | regulatory factor for TBG genes and nicotinamide N-methyltransferase (NNMT) gene in PTC | |
| Esr1 | estrogen receptor 1 (alpha) | AI646838 | 2.61 | * | * | * | polymorphism associated with thyroid cancers |
| Casq2 | calsequestrin 2 | NM 0098 14 | -2.66 | * | up-regulation in Grave's ophthalmopathy patients | ||
| Tbl1xr1 | transducin (beta)-like 1X-linked receptor 1 | BG07162 0 | -2.69 | * | * | co-repressor of TRs | |
| Efnb2 | ephrin B2 | U30244 | -2.87 | * | * | diagnostic marker for malignant thyroid tumors | |
| Mirhg1 | microRNA host gene 1 (non-protein coding) | AK01716 4 | -3.60 | * | * | miR-17-92 cluster in ATCs | |
| Vegfc | vascular endothelial growth factor C | NM_0095 06 | -4.17 | * | * | * | thyroid tumor lymphatic vessel formation |
| Igf1r | insulin-like growth factor 1 receptor | AV37436 9 | -4.42 | * | * | overexpression in human thyroid cancers | |
| Nr2c2 | nuclear receptor subfamily 2, group C, member 2 | AU06692 0 | -4.49 | * | modulator of TR-targeted genes | ||
| Sdha | succinate dehydrogenase complex, subunit A, flavoprotein (Fp) | AK00535 0 | -6.86 | * | * | thyroid tumors | |
| Abca3 | ATP-binding cassette, sub-family A (ABC1), member 3 | BC00693 2 | -11.71 | * | * | * | TTF-1- and SREBP-target gene |
Table 3-a.
Genes with altered expression in Thrbpv/pv mice related to biological functions other than tumorigenesis or metastasis (n=135)
| Symbol | Gene name | Accession No. | Fold change | Note(s) |
|---|---|---|---|---|
| Prpf19 | PRP19/PSO4 pre-mRNA processing factor 19 homolog (S. cerevisiae) | BC004070 | 5.25 | ubiquitin ligase activity; mRNA processing; DNA repair |
| Fam163a | family with sequence similarity 163, member A | BB183509 | 4.16 | aka, neuroblastoma-derived secretory protein (NDSP) |
| Tmed6 | transmembrane emp24 protein transport domain containing 6 | NM_025458 | 4.07 | ER transport |
| II10rb | interleukin 10 receptor, beta | NM_008349 | 3.96 | interleukin-10 receptor activity |
| Sc4mol | sterol-C4-methyl oxidase-like | AK005441 | 3.93 | putative function in cholesterol biosynthesis |
| Slmo1 | slowmo homolog 1 (Drosophila) | BB835597 | 3.82 | aka erythroid differentiation and denucleation factor 1; function unknown |
| Kpna1 | karyopherin (importin) alpha 1 | U20619 | 3.80 | nuclear import; regulation of DNA recombination |
| Dlat | dihydrolipoamideS-acetyltransferase | AV336908 | 3.72 | accept acetyl groups formed by the oxidative decarboxylation of pyruvate and transfers them to coenzyme A |
| Tmem213 | transmembrane protein 213 | AI315206 | 3.57 | unknown |
| Mgl1 | macrophage galactose N-acetyl-galactosamine specific lectin 1 | NM_010796 | 3.50 | sugar binding |
| Pigx | phosphatidylinositol glycan anchor biosynthesis, class X | BC002202 | 3.47 | GPI anchor biosynthetic process |
| Ogdh | oxoglutarate dehydrogenase (lipoamide) | BC013670 | 3.38 | a subunit of the 2-oxoglutarate dehydrogenase complex; convert 2-oxoglutarate to succinyl-CoA during the Krebs cycle |
| Lcmt1 | leucine carboxyl methyltransferase 1 | NM_025304 | 3.36 | protein modification |
| Ktil2 | KTI12 homolog, chromatin associated (S. cerevisiae) | NM_029571 | 3.29 | chromatin modification |
| Mrps21 | mitochondrial ribosomal protein S21 | NM_078479 | 3.24 | mitochondrial small ribosomal subunit |
| Med8 | mediator of RNA polymerase II transcription, subunit8 homolog (yeast) | AK011080 | 3.13 | a subunit of the mediator complex |
| Grinl1a | glutamate receptor, ionotropic, N-methyl D-aspartate-like 1A | AK002571 | 3.11 | transcriptional regulation (?); maintenance of ER location |
| Aldh1a2 | aldehyde dehydrogenase family 1, subfamily A2 | NM_009022 | 3.06 | an enzyme that catalyzes the synthesis of retinoic acid (RA) from retinaldehyde |
| Atp5j2 | ATP synthase, H+ transporting, mitochondrial FO complex, subunitf, isoform 2 | BG794445 | 3.03 | the f subunit of the FO complex of the proton channel of mitochondrial ATP synthase |
| Rshl2a | radial spokehead-like 2A; radial spokehead-like 2B | AA544511 | 3.03 | abundantly expressed in tissues rich in highly ciliated cells, such as olfactory sensory neurons with unknown function |
| Nubp2 | nucleotide binding protein 2 | AV035368 | 2.99 | microtubule organizing center |
| Ube2f | ubiquitin-conjugating enzyme E2F (putative) | AK007937 | 2.99 | Nedd8 ligase activity; protein neddylation (ubiquitination-like) |
| Tmem45a | transmembrane protein 45a | NM_019631 | 2.98 | unknown |
| Dusp14 | dual specificity phosphatase 14 | AK009744 | 2.97 | phosphatase |
| Gstm5 | glutathione S-transferase, mu 5 | NM_010360 | 2.94 | glutathione transferase activity |
| Mrps2 | mitochondrial ribosomal protein S2 | AV031454 | 2.91 | mitochondrial small ribosomal subunit |
| Hbs1l | Hbsl-like (S. cerevisiae) | BG067949 | 2.84 | translation elongation factor activity |
| Morn2 | MORN repeat containing 2 | BF319573 | 2.84 | membrane occupation and recognition nexus (MORN) motif protein in testis; function unknown |
| Fut9 | fucosyltransf erase 9 | AU067636 | 2.75 | synthesizes the LeX oligosaccharide, which is expressed in organ buds progressing in mesenchyma during human embryogenesis |
| Nsdhl | NAD(P) dependent steroid dehydrogenase-like | BCO19945 | 2.70 | a sterol dehydrogenase involved in the removal of C-4 methyl groups in one of the later steps of cholesterol biosynthesis. |
| Suclg2 | succinate-Coenzyme A ligase, GDP-forming, beta subunit | BF608645 | 2.70 | a GTP-specific beta subunit of succinyl-CoA synthetase |
| Cisd1 | CDGSH iron sulfur domain 1 | NM_134007 | 2.69 | a member of the CDGSH domain-containing family and may play a role in the regulation of mitochondrial oxidative capacity |
| Twf2 | twinfilin, actin-binding protein, homolog 2 (Drosophila) | AK002699 | 2.68 | intracellular signaling |
| Slc35a3 | solute carrier family 35, member 3 | AW822833 | 2.67 | UDP-GlcNAc transporter |
| Zscan22 | zinc finger and SCAN domain containing 22 | BB811893 | 2.67 | transcription regulation |
| Zcchc17 | zinc finger, CCHC domain containing 17 | BG962152 | 2.65 | ribonucleoprotein complex |
| Bat1a | HLA-B-associated transcript 1A | BB145254 | 2.64 | pre-mRNA processing |
| Coq9 | coenzyme Q9 homolog (yeast) | AK004527 | 2.64 | one of several enzymes involved in biosynthesis of CoQIO and likely functions in modification of the benzoquinone ring |
| Cacnb3 | calcium channel, voltage-dependent, beta 3 subunit | NM_007581 | 2.64 | calcium ion transport |
| Mrps15 | mitochondrial ribosomal protein S15 | NM_025544 | 2.64 | mitochondrial small ribosomal subunit |
| Pcbd1 | pterin 4 alpha carbinolamine dehydratase | NM_025273 | 2.62 | phenylalanine hydroxylation; regulate HNF1 dimerization |
| Socs1 | suppressor of cytokine signaling 1 | AB000710 | 2.61 | cytokine-mediated signaling pathway; fat cell differentiation. |
| Ndufa12 | NADH dehydrogenase (ubiquinone) 1 alpha subcomplex, 12 | BF152811 | 2.60 | an accessary subunit of mitochondrial complex 1 |
| Naglu | alpha-N-acetylglucosaminidase (Sanfilippo disease 1MB) | NM_013792 | 2.53 | hydrolase activity, acting on glycosyl bonds |
| Map6d1 | MAP6 domain containing 1 | BB762333 | 2.52 | negative regulation of microtubule depolymerization |
| Srrm1 | serine/arginine repetitive matrix 1 | BE199719 | -2.55 | pre-mRNA processing |
| Etohd2 | ethanol decreased 2 | BB214299 | -2.56 | unknown |
| Mtf2 | metal response element binding transcription factor 2 | BG066919 | -2.57 | transcription regulation |
| Gfod1 | glucose-fructose oxidoreductase domain containing 1 | BB538651 | -2.57 | oxidoreductase activity |
| Zfp871 | zinc finger protein 871 | BB008634 | -2.61 | unknown |
| Atl3 | atlastin GTPase 3 | BC017138 | -2.64 | ER/Golgi-bound GTPase |
| Zfp40 | zinc finger protein 40 | AI450803 | -2.65 | transcription regulation |
| Taf1a | TATA box binding protein (Tbp)-associated factor, RNA polymerase 1, A | NM_021466 | -2.66 | a component of SL1/TIFIB complex involved in the assembly of the preinitiation complex on RNA polymerase 1 promoter |
| Ccdc76 | coiled-coil domain containing 76 | BF147713 | -2.75 | methyltransferase activity; tRNA processing |
| Ankrd50 | ankyrin repeat domain 50 | BM119343 | -2.81 | unknown |
| Prrc1 | proline-rich coiled-coil 1 | BB481914 | -2.81 | unknown |
| Trim41 | tripartite motif-containing 41 | BE685711 | -2.82 | ligase activity |
| Clasp1 | CLIP associating protein 1 | BB190028 | -2.83 | cell cycle-related; microtubule-binding |
| Zcchc14 | zinc finger, CCHC domain containing 14 | BB223737 | -2.83 | cell communication; phosphoinositide binding |
| Impa1 | inositol (myo)-l(or 4)-monophosphatase 1 | AV154049 | -2.87 | inositol phosphate dephosphorylation |
| Rapgef4 | Rap guanine nucleotide exchange factor (GEF) 4 | AK004874 | -2.87 | calcium ion-dependent exocytosis; cAMP-mediated signaling |
| Atrx | alpha thalassemia/mental retardation syndrome X-linked homolog (human) | BF165715 | -2.93 | DNA helicase; chromatin remodeling |
| Dgkq | diacylglycerol kinase, theta | BB818538 | -2.94 | GPCR signaling; co-regulator of NR-targeted gene transcription |
| Rbm26 | RNA binding motif protein 26 | AV134514 | -2.96 | pre-mRNA processing |
| Fst | follistatin | NM_008046 | -3.00 | BMP signaling pathway |
| Tcf20 | transcription factor 20 | AW552808 | -3.05 | transcription regulation |
| Tcfe2a | transcription factor E2a | AF352579 | -3.06 | transcription regulation; histone acetylation |
| Ash1l | ashl (absent, small, or homeotic)-like (Drosophila) | BG694892 | -3.11 | chromatin remodeling; cell junction |
| Gate | glutamyl-tRNA(Gln) amidotransferase, subunit C homolog (bacterial) | AI452045 | -3.11 | mitochondrial tRNA modification |
| Stc2 | stanniocalcin 2 | AF031035 | -3.12 | hormone activity |
| Epb4.1 | erythrocyte protein band 4.1 | AI606195 | -3.14 | actin cytoskeleton organization |
| Gm5124 | predicted gene 5124 | BB049966 | -3.21 | unknown |
| Sbno1 | sno, strawberry notch homolog 1 (Drosophila) | BB147192 | -3.22 | hydrolase activity |
| Sfrs12 | splicing factor, arginine/serine-rich 12 | AV012790 | -3.24 | pre-mRNA processing |
| Neu1 | neuraminidase 1 | AI649303 | -3.26 | lysosomal hydrolase activity; acting on glycosyl bonds |
| Npc2 | Niemann Pick type C2 | BB556874 | -3.27 | lysosome-located protein; cholesterol and lipid transport from lysosome to other cell compartment (?) |
| Scaper | S phase cyclin A-associated protein in the ER | AV319713 | -3.27 | unknown |
| Cugbp1 | CUG triplet repeat, RNA binding protein 1 | BI412951 | -3.29 | pre-mRNA processing; embryonic development |
| Heatr7a | HEAT repeat containing 7A | BB142087 | -3.30 | cellular component (?) |
| Cflar | CASP8 and FADD-like apoptosis regulator | BE284491 | -3.30 | anti-apoptosis |
| Slc4a4 | solute carrier family 4 (anion exchanger), member 4 | BE655147 | -3.33 | sodium ion transport |
| Gmeb1 | glucocorticoid modulatory element binding protein 1 | BB039426 | -3.35 | transcription regulation |
| Phf21a | PHD finger protein 21A | BB094173 | -3.35 | chromatin modification |
| Fgfr1op2 | FGFR1 oncogene partner | AB041650 | -3.36 | unknown |
| Spnb2 | spectrin beta 2 | BM213516 | -3.37 | actin filament capping; SMAD protein nuclear translocation |
| Myh9 | myosin, heavy polypeptide 9, non-muscle | BM121854 | -3.39 | cell morphology; cell-cell adhesion; establishment of meiotic spindle localization |
| Daam1 | dishevelled associated activator of morphogenesis 1 | BB794633 | -3.39 | actin-binding protein; non-canonical Wnt mediated planar cell polarity (PCP) signaling pathway (?) |
| Sorbs1 | sorbin and SH3 domain containing 1 | BB372866 | -3.40 | signaling molecule in insulin-stimulated glucose uptake (?) |
| Olfml2a | olfactomedin-like 2A | AW489058 | -3.40 | extracellular matrix organization |
| Trove2 | TROVE domain family, member 2 | BQ176653 | -3.41 | transcription from RNA polymerase III promoter |
| Serinc3 | serine incorporator 3 | BM239368 | -3.41 | induction of apoptosis |
| Lpcat1 | lysophosphatidylcholine acyltransferase 1 | BG068664 | -3.42 | surfactant phospholipid synthesis |
| Cgn | cingulin | AK018143 | -3.45 | apical junction complex |
| Pcmtd2 | protein-L-isoaspartate (D-aspartate) 0-methyltransferase domain containing 2 | BM117243 | -3.54 | protein methyltransferase |
| Phip | pleckstrin homology domain interacting protein | BB473157 | -3.55 | insulin receptor signaling pathway; negative regulation of apoptosis |
| Dnaja2 | DnaJ (Hsp40) homolog, subfamily A, member 2 | BB324466 | -3.81 | protein folding |
| Pafah1b1 | platelet-activating factor acetylhydrolase, isoform lb, subunit 1 | L25109 | -3.81 | actin cytoskeleton organization; establishment of mitotic spindle orientation |
| Scfd1 | Seel family domain containing 1 | BB200692 | -3.82 | regulation of ER to Golgi vesicle-mediated transport |
| Plaa | phospholipase A2, activating protein | BB532258 | -3.93 | inflammatory response; phospholipase A2 activator activity |
| Gdpd5 | glycerophosphodiester phosphodiesterase domain containing 5 | BC024955 | -3.95 | glycerol metabolism; it catalyzes degradation of glycerophosphocholine (GPC) |
| Tmcc1 | transmembrane and coiled coil domains 1 | BB470329 | -3.95 | unknown |
| Xylb | xylulokinase homolog (H. influenzae) | BB431728 | -3.95 | energy metabolism (?) |
| Flnb | filamin, beta | BM206272 | -4.13 | cytoskeleton; stress fiber; acting binding |
| Eif4g3 | eukaryotic translation initiation factor 4 gamma, 3 | BM212250 | -4.13 | translation regulation |
| Apobec2 | apolipoprotein B mRNA editing enzyme, catalytic polypeptide 2 | AI666693 | -4.17 | pre-mRNA processing |
| Sfrs18 | splicingfactor, arginine/serine-rich 18 | BG277020 | -4.22 | nuclear speck |
| Phf7 | PHD finger protein 7 | AK005673 | -4.31 | unknown |
| Atp6v1h | ATPase, H+ transporting, lysosomal VI subunit H | AI849595 | -4.33 | protein transport |
| Lnpep | leucyl/cystinyl aminopeptidase | BE850004 | -4.34 | proteolysis; cell-cell signaling |
| Saps3 | SAPS domain family, member 3 | AK018652 | -4.57 | regulation of phosphoprotein phosphatase activity |
| Rfx7 | regulatory factor X, 7 | BB148972 | -4.60 | DNA-dependent transcriptional regulation |
| Fnbp4 | formin binding protein 4 | BG091626 | -4.67 | cellular component (?) |
| Stx17 | syntaxin 17 | AK018158 | -4.69 | vesicle-mediated transport |
| lno80d | IN080 complex subunit D | BE197105 | -4.87 | chromatin remodeling |
| Slc12a9 | solute carrier family 12 (potassium/chloride transporters), member 9 | BB668140 | -5.00 | ion transporter |
| Klhdc2 | kelch domain containing 2 | BB105408 | -5.16 | transcrioptional co-repressor |
| Rhbdl2 | rhomboid, veinlet-like 2 (Drosophila) | AV378451 | -5.17 | membrane protease |
| Rnf125 | ring finger protein 125 | BB667823 | -5.18 | ligase activity |
| C1qtnf3 | Clq and tumor necrosis factor related protein 3 | NM_030888 | -5.24 | an adipokine of the Clq/TNF molecular superfamily; secreted by the adipocytes of mice and humans |
| Nipbl | Nipped-B homolog (Drosophila) | BG070859 | -5.24 | cell cycle; embryonic development |
| Abca4 | ATP-binding cassette, sub-family A (ABC1), member 4 | NM_007378 | -5.47 | ATPase, photon-transduction |
| Fhdc1 | FH2 domain containing 1 | BB736539 | -5.62 | actin cytoskeleton organization |
| Rpgrip1 | retinitis pigmentosa GTPase regulator interacting protein 1 | AK015037 | -5.69 | eye photoreceptor cell development |
| Mef2c | myocyte enhancer factor 2C | BB280300 | -5.72 | transcription regulation |
| Cnot10 | CCR4-NOT transcription complex, subunit 10 | BB297961 | -5.73 | a subunit of the CCR4-NOT complex; transcriptional repression; mRNA metabolism |
| Plekhg3 | pleckstrin homology domain containing, family G, member 3 | BB280013 | -5.78 | Rho guanyl-nucleotide exchange factor activity |
| Kremen1 | kringle containing transmembrane protein 1 | BB373408 | -5.89 | Wnt receptor signaling pathway |
| Ccdc58 | coiled-coil domain containing 58 | BG064532 | -6.29 | mitochondrion component (?) |
| Fam13a | family with sequence similarity 13, member A | BB745929 | -6.34 | unknown |
| Klf7 | Kruppel-like factor 7 (ubiquitous) | BB524597 | -6.70 | transcriptional co-activator |
| Eif2s2 | eukaryotic translation initiation factor 2, subunit 2 (beta) | BG066754 | -6.84 | translation initiation complex |
| Kctd14 | potassium channel tetramerisation domain containing 14 | AW553424 | -7.20 | potassium ion transport |
| Senp7 | SUMOl/sentrin specific peptidase 7 | BM238538 | -8.52 | acts as a SUMO-2/3-specific protease that is likely to regulate the metabolism of poly-SUMO-2/3 |
| Camk2d | calcium/calmodulin-dependent protein kinase II, delta | BG074866 | -9.58 | calcium ion transport; cell cycle-related |
| Snhg3 | small nucleolar RNA host gene (non-protein coding) 3 | BI082172 | -17.34 | unknown |
Identification of genes involved in tumorigenesis mediated by PV to drive thyroid tumor progression of Thrbpv/pv mice
Among the total 241 genes with altered expression in thyroid tumor cells of Thrbpv/pv mice, 100 (41.5%) genes have been reported to be associated with carcinogenesis or tumor metastasis. Of these 100 genes, 49 (49%) were up-regulated (2.5- to 13.1-fold) and 51 were down-regulated (2.6- to 21.3-fold). These genes were originally involved in many different cellular functions including transcriptional regulation, RNA processing, chromatin modification, cell development, cell proliferation, apoptosis, embryonic development, angiogenesis, signaling transduction, and cell adhesion (Table 1). At the top of the list with activated expression (Table 1-a) are fibrinogen gamma chain (Fgg; 13.1-fold), glial cell line derived neurotrophic factor family receptor alpha 1 (Gfra1; 11.78-fold), and placenta-specific 8 (Plac8; 11.31-fold). Use of real time RT-PCR confirmed mRNA levels were increased in thyroid tumor cells of Thrbpv/pv mice as compared with Thra1-/-Thrb-/- mice. As shown in Figures 2A-C, 5-, 155-, and 4.5-fold increased expressions were found for Fgg, Gfra, and Plac8, respectively. The expressions of several relatively down-regulated genes including DEAD (Asp-Glu-Ala-Asp) box polypeptide 6 (Ddx6), BCL2-associated transcription factor 1 (Bclaf1), and nuclear receptor subfamily 2, group F, member 2 (COUP-TFII, Nr2f2) were also examined by real time RT-PCR. As shown in Figures 2D-F, 45%, 50%, and 50% repressions of the mRNA expression were found for Ddx6, Bclaf1, and Nr2f2, respectively. These results show a concordance in gene expression as determined by RT-PCR and microarray analyses.
Figure 2.
Validation of altered gene expression in the thyroids of Thrbpv/pv and Thra1-/-Thrbpv/pv mice by real time RT-PCR. Total RNA was extracted from the mouse thyroid at the age of 9-12 months, and real time RT-PCR was performed as described in Materials and Methods. Fold of changes to the mRNA level of Thra1pv/pvThrbpv/pv mice is shown. An “*” indicates p<0.05; “**”, p<0.01; and “***”, p<0.001, by the Student's t test.
It is important to note that the 100 genes, 45% of the total 241 genes with altered expression mediated by PV exhibited diverse and broad cellular functions relevant to tumorigenesis and metastasis as revealed by the functional annotation (Table 1-b). These functions include transcription, RNA and DNA metabolic processes, cell development, differentiation, proliferation, and apoptosis. Moreover, genes involved in embryogenesis, angiogenesis, and cell adhesion were also detected. Thus, PV exerts extensive effects via gain-of-function on critical cellular functions to drive carcinogenesis.
Table 1-b.
Functional annotation analysis in genes relevant to tumorigenesis/metastasis in Thrbpv/pv mice
| Function | Count | Genes |
|---|---|---|
| Regulation of transcription | 22 | Khdrbs1,Fus,Crtc3,Bach1,Eid1,Tbl1xr1,Dnmt3a,Bclaf1,Hnf1b,Aebp1,Elf2,Bach2,Creb1,Esr1,Shh,Notch2,Rnf4,Sfpq,Sp4,Prdm2,Nr2f2,Nfia |
| Regulation of RNA metabolic process | 15 | Bach1,Fus,Eid1,Dnmt3a,Tbl1xr1,Aebp1,Hnf1b,Elf2,Bach2,Creb1,Esr1,Rbm5,Shh,Nr2f2,Nfia |
| Regulation of DNA metabolic process | 6 | Rpa1,Dnmt3a,Ube2a,Sfpq,Kcnqlot1,Nfia |
| Regulation of cell development/differentiati on | 5 | Rtn4,Wnt4,Shh,Thy1,Mt3,Vegfc,Sema4d |
| Positive regulation of cell proliferation | 5 | Rpa1,Vegfc,Esr1,Birc6,Shh |
| Regulation of apoptosis | 7 | Notch2,Bclaf1,Nudt2,Esr1,Rbm5,Birc6,Bmf |
| Embryonic development | 5 | Rpa1,Ube2a,Hnf1b,Birc6,Shh,Nr2f2 |
| Blood vessel morphogenesis/angiogenesis | 6 | Rtn4,Vegfc,Cspg4,Nr2f2,Shh,Thy1 |
| Signal | 26 | Arsb,Aebp1,Masp1,Cspg4,Bcan,Timp2,Prkcsh,Shh,Ccl6,Alcam,Ednrb,lgf1r,Wnt4,Fgg,Lair1,Efnb2,Sirpa,Muc4,Thy1,Notch2,Vegfc,C4bp,Gfra1,Sema4d,Cpd,Bmp5 |
| Small GTPase regulator activity | 6 | Krit1,Git2,ltsn2,Rgnef,Mcf21,Thy1 |
| Oxidation reduction | 7 | Adi1,Sdha,Uqcrh,ldh1,Srd5a1,Prdx3,Fdft1 |
| Vesicle-mediated transport | 4 | Apis3,ltsn2,Sirpa,Shh |
| Cell adhesion | 6 | Alcam,Aebp1,Bcan,Sirpa,Muc4,Thy1 |
Uncovering genes specifically affected by PV via gain-of-function in the thyroid
Microarray analyses uncovered 11 genes involved in thyroid carcinoma that exhibited altered expression in thyroid tumor cells of Thrbpv/pv mice (Table 2). Among these genes, increased expression of the GFRA1 gene was reported in medullary thyroid carcinoma [26]. Consistent with this observation, microarrays also showed an increase in its expression (11.78-fold; Table 2) in the thyroid lesions of Thrbpv/pv mice as compared with Thra1-/-Thrb-/- mice. Array analysis identified another thyroid cancer-related gene, Minpp1 (multiple inositol polyphosphate histidine phosphatase 1), which is a phosphatase with an overlapping function with that of PTEN. Its gene localization (10q23.3) is near to the PTEN gene. Loss of heterozygosity in this locus is frequent in follicular thyroid tumors [27]. Intriguingly, the expression of this gene was up-regulated (4.07-fold) according to the array analysis. Another key thyroid cancer-related gene, Vegfc (vascular endothelial factor C), was also identified by the array analysis. Vegfc is a key regulator in stimulating proliferation of lymphatic vessels and thus is related to lymphatic metastasis of tumors [28]. Moreover, it is known that serum VEGF (s-VEGF) and VEGF-c (s-VEGF-C) are elevated in patients with recurrence of papillary thyroid cancer (PTC) and correlated significantly with the presence of nodal metastases and advanced tumor stages [29, 30]. Paradoxically, decreased expression of the Vegfc gene was detected in the tumor cells of Thrbpv/pv mice as compared with Thra1-/-Thrb-/- mice (Table 2). Array analysis indicated a 4.17-fold decrease, a finding confirmed by RT-PCR (4.5-fold decrease; Figure 2G). In addition, Table 2 also shows that several transcriptional co-regulators of TR or TR-targeted genes, such as the HNF1 homeobox B gene (Hnf1b; 2.7-fold up-regulation), transducin (beta)-like 1X-linked receptor 1 (Tbl1xr1; 2.69-fold down-regulation), and nuclear receptor subfamily 2, group C, member 2 (TR4, Nr2c2; 4.49-fold down-regulation), also showed altered expression in the tumor cells of Thrbpv/pv mice. The expressions of the Hnf1b, Tbl1xr1, and Nr2c2 genes were confirmed by RT-PCR, as shown in Figure 2H, I and J with 10-fold up-regulation; 2.2-fold and 2-fold down-regulation, respectively.
Alteration in the expression of genes with diverse functions collaborates with the PV oncogenic actions
In addition to the genes involved in tumorigenesis or metastasis shown in Tables 1 and 2, array analysis identified 135 genes (56% of total genes) with altered expression related to other biological functions. This large group includes 45 genes that were up-regulated (ranging from 2.52- to 5.25-fold) and 90 genes that were down-regulated (ranging from 2.55- to 17.34-fold). These 135 genes have broad and diverse cellular functions (see Table 3-a) including transcription regulation, cytoskeleton remodeling, molecule transport, cell signaling, RNA processing, protein translation, protein-protein interaction, cell signaling, and metabolism (Table 3-b). That PV could invoke the changes in the expression of an extensive array of genes with diverse cellular functions would suggest that its oncogenic actions would require that genes not be exclusively involved in tumorigenesis, but also collaborate with genes involved in other cellular functions. How some of these genes could network with each other to carry out the oncogenic functions of PV via gain-of-function is further elucidated in the next section.
Table 3-b.
Summary of functions in genes with altered expression in Thrbpv/pv mice
| Biological function | Count | Genes |
|---|---|---|
| Transcriptional regulation | 19 | Apobec2,Atp6v1h,Cnot10,Dgkq,Gmeb1,lno80d,Klf7,Klhdc2,Kti12,Med8,Mtf2,Phf21a,Rfx7,Taf1a,Tcf20,Tcfe2a,Trove2,Zfp40,Zscan22 |
| Intracellular non-membrane-bounded organelle | 17 | Dgkq,Taf1a,Twf2,Mrps15,Mrps21,Myh9,Zcchc17,Flnb,Mrps2,Atrx,Cgn,Nubp2,Spnb2,Epb4.1,Clasp1,Pafah1b1,Map6d1 |
| Metabolism | 17 | Aldh1a2,Atp5j2,Cisd1,Coq9,Dlat,Fut9,Gdpd5,Lpcat1,Ndufa12,Npc2,Nsdhl,Ogdh,Pcbd1,Suclg2,Sc4mol,Sorbs1,Xylb |
| Cytoskeleton | 13 | Dgkq,Twf2,Nubp2,Cgn,Spnb2,Epb4.1,Pafah1b1,Clasp1,Myh9,Flnb,Map6d1,Fhdc1,Daam1 |
| Ribonucleoprotein complex | 9 | Prpf19,Trove2,Mrps15,Batla,Srrm1,Mrps21,Zcchc17,Mrps2,Sfrs12 |
| Protein catabolic process | 7 | Rnf125,Lnpep,Med8,Socs1,Ube2f,Myh9,Senp7 |
| Translation | 6 | Eif4g3,Mrps15,Hbs1l,Eif2s2,Mrps21,Mrps2 |
| Intracellular transport | 5 | Stx17,Spnb2,Pafah1b1,Myh9,Kpna1 |
| Tightjunction | 5 | Cgn,Spnb2,Epb4.1,Ash1l,Myh9 |
| mRNA processing | 5 | Prpf19,Apobec2,Batla,Srrma,Sfrs12 |
| Calmodulin binding | 4 | Spnb2,Camk2d,Myh9,Map6d1 |
| Methyltransferase | 4 | Ccdc76,Lcmt1,Pcmtd2,Ash1l |
| Cell-cell junction | 4 | Sorb1,Cgn,Ash1l,Myh9 |
| Negative regulation of protein complex disassembly | 3 | Spnb2,Clasp1,Map6d1 |
| Nuclear speck | 3 | Sfrs18,Dgkq,Srrm1 |
| Cell signaling | 11 | Dgkq,Dusp14,Fst,ll10rb,Kremen1,Phip,Plaa,Plekhg3,Saps3,Socs1,Twf2 |
| Transporter | 11 | Abca4,Atp6v1h,Cacnb3,Camk2d,Kctd14,Kpnal,Rapgef4,Scfd1,Slc12a9,Slc35a3,Slc4a4, |
Gene interaction network analysis implicates involvement of multiple signaling pathways
Via Ingenuity Pathway Analysis software, 25 networks that could function coordinately to bring out the tumorigenic phenotypic manifestation were predicted from the genes identified with altered expression. With the highest score of gene correlation, the top four networks are shown in Figure 3. In Network 1, a group of genes related to tissue development such as lymphoid tissue formation was linked (Figure 3A). Among them, we noted Vegfc, Nr2f2, and ephrin B2 (Efnb2). Vegfc and Nr2f2 are known to be essential in lymphangiogenesis whereas Efnb2 has been related to lymph node metastasis [31, 32]. All of these genes showed a reduced expression level in the thyroid of Thrbpv/pv mice, which might account for the observations that in Thrbpv/pv mice the metastatic target sites (lung and heart) are via the vascular system, rather than via lymphatic pathways. However, Network 1 shows an increased mRNA level of metallothionein 3 (Mt3) which may contribute to the angiogenesis by encoding a small peptide that can significantly induce Vegf mRNA and protein expression in mouse brain capillary endothelial cell line bEND.3 cells during hypoxia [33]. Gene-like bone morphogenetic protein 5 (Bmp5) in Network 1 is associated with cardiovascular system development, and its transcription is under the regulation of Rbm5 (RNA-binding motif protein 5), a tumor suppressor whose mechanism involves potentiating apoptosis and inhibition of the cell cycle [34]. The coordinated down-regulation of Bmp5 and Rbm5 outlined in Network 1 would suggest that the repression of Rbm5 could lead to decreased apoptosis and increased tumor cell proliferation of thyroid tumor cells.
Figure 3.
Network prediction of differentially expressed genes in thyroid tumor cells between Thrbpv/pv and Thra1-/-Thrb-/- mice. The four top-scoring networks (A-D) are displayed graphically. As explained in the legend (A-b), different shapes represent different types of proteins while the relationships between proteins are indicated by the solid bar (direct interaction), dotted bar (indirect relationship), or arrow (acting on). The increased (symbol with vertical lines) and decreased expression (hatched symbol) is marked as indicated in the legends (A-b). (Refer to next page for Figure 3B, C and D).
In Network 2, genes involved in DNA replication, DNA repair, and cell signaling were crosslinked (Figure 3B). In our previous study, we discovered chromosomal aberrations in cell lines derived from thyroid tumors of Thrbpv/pv mice. These cell lines exhibit abnormal karyotypes and a variety of structural chromosomal aberrations, including common recurrent translocations and deletions, raising the possibility that induction of chromosomal instability may contribute to the thyroid carcinogenesis in Thrbpv/pv mice [35]. There is also an aberrant accumulation of a critical mitotic checkpoint protein, Pttg1 (pituitary tumor-transforming gene 1), that helps hold sister chromatids together before entering anaphase, and thus impedes mitotic progression in cells expressing PV [36]. From Network 2, we uncovered additional genes (Smc4, Sfpq, Fus, Pafah1b1, Rpa1) that play critical roles in DNA replication, cell cycle regulation, maintenance of chromosome stability, and/ or DNA repair, which were also deregulated in Thrbpv/pv mice. Smc4 (structural maintenance of chromosomes 4) is an essential component of the condensin complex. The Smc complex is vital for a wide range of processes including chromosome structure and dynamics, gene regulation, and DNA repair [37]. Sfpq (splicing factor proline/ glutamine rich) has functions in pre-mRNA processing [38], transcriptional regulation, and DNA repair by complexes with its paralog named NONO/ p54nrb or RAD51D/ RAD51C/XRCC2 during different DNA repair events [39, 40]. Depletion of SFPQ can lead to increased chromosomal aberrations and substantial sister chromatid cohesion defects [40]. Fus (fusion, derived from t (12;16) malignant liposarcoma gene), an oncogenic RNA binding protein, is also related to DNA repair as evidenced by the fact that mice lacking the protein FUS are hypersensitive to ionizing radiation and FUS is a phosphorylation target of the ATM (ataxia-telangiectasia mutated) signaling pathway in DNA repair [41-43]. Pafah1b1 (platelet-activating factor acetyl-hydrolase, isoform 1b, subunit 1) plays a pivotal role in microtubule regulation and mitotic spindle orientation of neuroepitheiial stem cell proliferation [44]. Remarkably, expression of these four genes was decreased in a coordinated manner in Thrbpv/pv mice. This reduction could further weaken the chromosomal stability in Thrbpv/pv mice. Replication protein A (Rpa1), a major eukaryotic ssDNA binding protein, is required for cell viability and plays essential roles in DNA replication, repair, and recombination [45]. A higher RPA expression has been reported in breast and colon cancers as a factor in promoting cell proliferation during tumor growth and progression [46, 47]. In Thrbpv/pv mice, the mRNA level of Rpa1 was also higher than in Thra-/-Thrb-/- mice. In this network, we also noted several key regulators in the Wnt/b-catenin signaling pathway, including Wnt4 (wingless-related MMTV integration site 4) and Daam1 (dishevelled associated activator of morphogenesis 1), that could crosstalk with the p38 MAPK pathway. Previously, we showed dys-regulation of both signaling cascades (Wnt/b-catenin and p38 MAPK) in thyroid carcinogenesis of Thrbpv/pv mice [17, 48]. However, it was unknown whether these two pathways could be linked via PV to affect thyroid carcinogenesis. Thus, the possibility that the two pathways can crosstalk could potentially open a new area of study to gain further insights into thyroid carcinogenesis.
In Network 3, genes participating in the cancer-related signaling pathways are shown. Several key nodes are Akt, Mapk, Ras, and IGF1R. Aberrant activation of the signaling involving these key regulators has been shown to be associated with human follicular thyroid carcinoma. Consistent with human cancer, PI3K-Akt is also activated during thyroid carcinogenesis of Thrbpv/pv mice [18, 19]. One of the genes with altered expression identified in Network 3 that merits further investigation is estrogen receptor (ER). A long-standing question in the understanding of human thyroid cancers is the preponderance of female patients in the ratio of 3- or 4-to-1 over male patients. Network 3 shows the extensive down-stream regulators and effectors of the PV-activated ER signaling. These ER downstream regulators need to be further studied to understand the gender disparity in thyroid cancer.
Network 4 shows sets of genes grouped by their common functions in cytoskeleton organization, molecular transport, and small molecule biochemistry (Figure 3D). In this group, several genes related to cytoskeleton structure or cell adhesion (e.g., erythrocyte protein band 4.1, Epb4.1, MCF.2 cell line derived transforming sequence-like, Mcf2l, myosin, heavy polypeptide 9, non-muscle, Myh9, filamin, beta, Flnb, cingulin, Cgn) were identified. The differential expression of these actin-related genes might give insights into how cellular behaviors in vascular invasion differed between Thrbpv/pv and Thra1-/-Thrb-/-mice.
Discussion
We have previously shown that both Thrbpv/pv and Thra1-/-Thrb-/- mice spontaneously develop FTC similar to human thyroid cancer [13, 22]. However, it is important to point out that FTC is more aggressive in Thrbpv/pv than Thra1-/-Thrb-/- mice. The size of thyroid tumors is larger and the pathological progression is faster with a higher frequency of metastasis. This observation is intriguing in view of the fact that Thrbpv/pv mice harbor a mutated TRβ that was expected to function as a dominant negative mutant, leading only to the loss of normal TR suppressor functions. The phenotypic manifestation of thyroid tumors in Thrbpv/pv mice was expected to be similar to that of Thra1-/-Thrb-/- mice that have lost TR suppressor functions due to deficiency in TRs. That FTC is more aggressive in Thrbpv/pv than in Thra1-/-Thrb-/- mice prompted us to test the hypothesis that PV, in addition to the loss of normal TR functions, could also act via gain-of-function. Indeed, global gene expression profiling of the microdissected thyroid tumor cells from the two groups of age- and gender-matched mutant mice shows contrasting expression patterns of 241 genes. Although the majority of the genes with contrasting expression are involved in tumorigenesis and metastasis, the remaining genes play key roles in broad and diverse cellular functions. These findings indicate that PV, in addition to acting as a mutant that has lost normal TR functions, could also act via gain-of-function to promote thyroid carcinogenesis.
Extensive analysis of the gene expression data using gene ontology, functional annotation, and gene network prediction has provided new insights into the molecular signaling pathways that lead to a more aggressive thyroid malignancy in Thrbpv/pv mice than in Thra1-/-Thrb-/- mice. Previously, we identified several pathways including PI3K/Akt, Wnt/β-catenin, and p38 MAPK signaling that are dysregulated in Thrbpv/pv mice [17, 19, 48]. In Thra1-/-Thrb-/- mice, activation of PI3K/Akt signaling was also observed [22]. The present studies showed that different subsets of effectors/regulators were implicated in the activation of the same pathways that could further modulate the magnitudes of the responses. The different subsets of genes could also function in different pathways to regulate the extent or alter the outcome of signaling. Gfra1 was one such gene uncovered in the present studies. Its mRNA expression was highly elevated in the thyroid tumors of Thrbpv/pv mice (Table 1-a and Figure 2B). This gene encodes a glycosylphosphatidylinositol (GPI)-linked co-receptor of proto-oncogenic receptor tyrosine kinase RET (rearranged during transfection) in mediating extracellular signal from glial cell line-derived neurotrophic factors (GDNFs) via Ras/ MAPK, PI3K/Akt, and PLCγ pathways [49, 50]. Gfra1 also directly complexes with neural cell adhesion molecule (NCAM) and activates Fyn kinase to regulate cell migration neuronal morphology, and synapse formation [51]. Gfra1 can also mediate cell adhesion uniquely in a ligand-dependent manner [52]. In addition, Gfra1 can be shed from the cell surface by the action of membrane-associated phospholipases. The soluble Gfra1 stably binds to GDNF to act at a distance to promote neuronal survival and neurite outgrowth on RET-expressing neurons/ axons [53, 54]. In thyroid cancers, increased expression level of Gfra1 or constitutive activation of RET signaling has been reported due to the rearrangement of RET (RET/PTC) in papillary thyroid cancer (PTC) or medullary thyroid carcinoma patients [26, 55]. This raises the possibility that a highly increased expression level of Gfra1 in Thrbpv/pv mice could be involved in the activation of the RET signaling in these mice. If this possibility proves to be true in future studies, Thrbpv/pv mice would become a promising preclinical animal model in testing novel RET inhibitors for thyroid cancer treatment.
In addition to the identification of Gfra1 as a novel modulator of Ras/MAPK, PI3K/Akt signaling in thyroid carcinogenesis of Thrbpv/pv mice, we also detected the highly activated expression of Shh (sonic hedgehog) that could also function to modulate the PI3K/Akt and Wnt/β-catenin pathways. Shh encodes a signaling peptide functioning via the Patched (Ptc)-Smoothened (Smo) receptor complex and glioblastoma (Gli) family of transcription factors in controlling cell proliferation and differentiation. The Shh pathway crosstalks with PI3K-Akt and Wnt/β-catenin pathways at multiple levels and coordinates developmental transitions in mammals [56]. In thyroid, knockout of the Shh gene leads to hemiagenesis of thyroid and ectopic expression [57]. In addition to Shh, several Shh-signaling-related genes such as Aldh1a2, Mtss1, and Nsdhl were also identified in the analysis. Aldh1a2 (aldehyde dehydrogenase 1 family, member A2) catalyzes the synthesis of retinoic acid, which regulates early mouse embryonic forelimb development by controlling sonic hedgehog signaling [58]. The increased mRNA level of Aldhla2 (3.7 fold, Table 3-a) found in Thrbpv/pv mice may enhance the activated Shh signaling. Another gene that regulates hedgehog signaling is Nsdh1 (NADH sterol dehydrogenase-like), which was found to be up-regulated (2.7-fold, Table 4-a) in Thrbpv/pv mice as well. This gene encodes a sterol dehydrogenase involved in the removal of C-4 methyl groups in one of the later steps of cholesterol biosynthesis. In Nsdhl-deficient mice, the abnormality of placental development is related to the hedgehog signaling pathway [59]. This may be due to the decrease of sterol synthesis that directly affects one or more proteins (including Shh) associated with sterols. This could also be due to the alteration of the properties of the plasma membrane which may secondarily affect Shh signaling pathway [60, 61]. Another gene involved in Shh signaling is Mtss1 (metastasis suppressor 1). Mtss1 is a direct target gene of Shh-Gli signaling and its product can potentiate Gli-dependent transcription [62]. Mtss1 is also able to bind monomeric actin and behaves as a tumor suppressor in cell proliferation [63, 64]. In our study, the expression of Mtss1 (5-fold; Table 1-a) is not consistent with the increased mRNA level of Shh in Thrbpv/pv mice, which suggests that Mtss1 may not be regulated by Shh in thyrocytes but affects thyroid tumorigenesis via its tumor suppressor function.
That thyroid tumors of Thrbpv/pv mice are larger than those of Thrb-/-Thrb-/- mice could be understood further from the identification of several down-regulated genes encoding pro-apoptotic factors (Bclaf1, Bmf, and Bnip2) related to Bcl2 in the present studies. The suppression of apoptosis by lowering the expression of these pro-apoptotic molecules sustains malignant cell proliferation in thyroids of Thrbpv/pv mice. Thus, while the proliferation of thyroid tumor cells of Thrbpv/pv mice and Thra1-/-Thrb-/- mice is stimulated, the decreased apoptosis due to the suppression of apoptotic activity in thyroid tumor cells of Thrbpv/pv mice gains additional proliferation advantage. The contribution of the apoptotic pathways to the more aggressive thyroid malignancy due to PV via the gain-of -function suggests new therapeutic strategies that would use these apoptotic genes as molecular targets.
Ever since the identification of dominantly negative TRβ mutations in RTH patients, the molecular mechanisms by which these mutants function in the pathogenesis of RTH have been extensively investigated. While it has long been accepted that these TRβ mutations act in a dominant negative fashion to interfere with the functions of WT TRs, whether they could also function via gain-of-function was less extensively explored. The contrasting gene expression profiles in the thyroids of Thrbpv/pv and Thra1-/-Thrb-/- mice clearly show that PV can act beyond the dominant negative mode. It is possible that this gain-of-function of PV can also operate in other target tissues besides the thyroid, thereby further contributing to the pathogenesis of RTH. The verification of this possibility awaits further studies.
Acknowledgments
This research was supported by the Intramural Research Program of the Center for Cancer Research. National Cancer Institute, NIH. We thank Drs. Jaime Rodriguez-Canales and Jeffrey Hanson, for assistance with the laser capture microdissection experiments.
References
- 1.Sipos JA, Mazzaferri EL. Thyroid cancer epidemiology and prognostic variables. Clin Oncol (R Coll Radiol) 2010;22:395–404. doi: 10.1016/j.clon.2010.05.004. [DOI] [PubMed] [Google Scholar]
- 2.Sampson E, Brierley JD, Le LW, Rotstein L, Tsang RW. Clinical management and outcome of papillary and follicular (differentiated) thyroid cancer presenting with distant metastasis at diagnosis. Cancer. 2007;110:1451–1456. doi: 10.1002/cncr.22956. [DOI] [PubMed] [Google Scholar]
- 3.Cheng SY. Multiple mechanisms for regulation of the transcriptional activity of thyroid hormone receptors. Rev Endocr Metab Disord. 2000;1:9–18. doi: 10.1023/a:1010052101214. [DOI] [PubMed] [Google Scholar]
- 4.Gonzalez-Sancho JM, Garcia V, Bonilla F, Munoz A. Thyroid hormone receptors/THR genes in human cancer. Cancer Lett. 2003;192:121–132. doi: 10.1016/s0304-3835(02)00614-6. [DOI] [PubMed] [Google Scholar]
- 5.Cheng SY. Thyroid hormone receptor mutations in cancer. Mol Cell Endocrinol. 2003;213:23–30. doi: 10.1016/j.mce.2003.10.051. [DOI] [PubMed] [Google Scholar]
- 6.Lin KH, Shieh HY, Chen SL, Hsu HC. Expression of mutant thyroid hormone nuclear receptors in human hepatocellular carcinoma cells. Mol Carcinog. 1999;26:53–61. doi: 10.1002/(sici)1098-2744(199909)26:1<53::aid-mc7>3.0.co;2-z. [DOI] [PubMed] [Google Scholar]
- 7.Puzianowska-Kuznicka M, Krystyniak A, Madej A, Cheng SY, Nauman J. Functionally impaired TR mutants are present in thyroid papillary cancer. J Clin Endocrinol Metab. 2002;87:1120–1128. doi: 10.1210/jcem.87.3.8296. [DOI] [PubMed] [Google Scholar]
- 8.Kamiya Y, Puzianowska-Kuznicka M, McPhie P, Nauman J, Cheng SY, Nauman A. Expression of mutant thyroid hormone nuclear receptors is associated with human renal clear cell carcinoma. Carcinogenesis. 2002;23:25–33. doi: 10.1093/carcin/23.1.25. [DOI] [PubMed] [Google Scholar]
- 9.Ando S, Sarlis NJ, Oldfield EH, Yen PM. Somatic mutation of TRbeta can cause a defect in negative regulation of TSH in a TSH-secreting pituitary tumor. J Clin Endocrinol Metab. 2001;86:5572–5576. doi: 10.1210/jcem.86.11.7984. [DOI] [PubMed] [Google Scholar]
- 10.Garcia-Silva S, Aranda A. The thyroid hormone receptor is a suppressor of ras-mediated transcription, proliferation, and transformation. Mol Cell Biol. 2004;24:7514–7523. doi: 10.1128/MCB.24.17.7514-7523.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Martinezlglesias O, Garcia-Silva S, Tenbaum SP, Regadera J, Larcher F, Paramio JM, Vennstrom B, Aranda A. Thyroid hormone receptor betal acts as a potent suppressor of tumor invasiveness and metastasis. Cancer Res. 2009;69:501–509. doi: 10.1158/0008-5472.CAN-08-2198. [DOI] [PubMed] [Google Scholar]
- 12.Kaneshige M, Kaneshige K, Zhu X, Dace A, Garrett L, Carter TA, Kazlauskaite R, Pankratz DG, Wynshaw-Boris A, Refetoff S, Weintraub B, Willingham MC, Barlow C, Cheng S. Mice with a targeted mutation in the thyroid hormone beta receptor gene exhibit impaired growth and resistance to thyroid hormone. Proc Natl Acad Sci U S A. 2000;97:13209–13214. doi: 10.1073/pnas.230285997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Suzuki H, Willingham MC, Cheng SY. Mice with a mutation in the thyroid hormone receptor beta gene spontaneously develop thyroid carcinoma: a mouse model of thyroid carcinogenesis. Thyroid. 2002;12:963–969. doi: 10.1089/105072502320908295. [DOI] [PubMed] [Google Scholar]
- 14.Weiss RE, Refetoff S. Resistance to thyroid hormone. Rev Endocr Metab Disord. 2000;1:97–108. doi: 10.1023/a:1010072605757. [DOI] [PubMed] [Google Scholar]
- 15.Parrilla R, Mixson AJ, McPherson JA, McClaskey JH, Weintraub BD. Characterization of seven novel mutations of the c-erbA beta gene in unrelated kindreds with generalized thyroid hormone resistance. Evidence for two “hot spot” regions of the ligand binding domain. J Clin Invest. 1991;88:2123–2130. doi: 10.1172/JCI115542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Kato Y, Ying H, Zhao L, Furuya F, Araki O, Willingham MC, Cheng SY. PPARgamma insufficiency promotes follicular thyroid carcinogenesis via activation of the nuclear factor-kappaB signaling pathway. Oncogene. 2006;25:2736–2747. doi: 10.1038/sj.onc.1209299. [DOI] [PubMed] [Google Scholar]
- 17.Guigon CJ, Zhao L, Lu C, Willingham MC, Cheng SY. Regulation of beta-catenin by a novel nongenomic action of thyroid hormone beta receptor. Mol Cell Biol. 2008;28:4598–4608. doi: 10.1128/MCB.02192-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Furuya F, Lu C, Willingham MC, Cheng SY. Inhibition of phosphatidylinositol 3-kinase delays tumor progression and blocks metastatic spread in a mouse model of thyroid cancer. Carcinogenesis. 2007;28:2451–2458. doi: 10.1093/carcin/bgm174. [DOI] [PubMed] [Google Scholar]
- 19.Furuya F, Hanover JA, Cheng SY. Activation of phosphatidylinositol 3-kinase signaling by a mutant thyroid hormone beta receptor. Proc Natl Acad Sci U S A. 2006;103:1780–1785. doi: 10.1073/pnas.0510849103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Kim CS, Ying H, Willingham MC, Cheng SY. The pituitary tumor-transforming gene promotes angiogenesis in a mouse model of follicular thyroid cancer. Carcinogenesis. 2007;28:932–939. doi: 10.1093/carcin/bgl231. [DOI] [PubMed] [Google Scholar]
- 21.Kim CS, Vasko VV, Kato Y, Kruhlak M, Saji M, Cheng SY, Ringel MD. AKT activation promotes metastasis in a mouse model of follicular thyroid carcinoma. Endocrinology. 2005;146:4456–4463. doi: 10.1210/en.2005-0172. [DOI] [PubMed] [Google Scholar]
- 22.Zhu XG, Zhao L, Willingham MC, Cheng SY. Thyroid hormone receptors are tumor suppressors in a mouse model of metastatic follicular thyroid carcinoma. Oncogene. 2010;29:1909–1919. doi: 10.1038/onc.2009.476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Furumoto H, Ying H, Chandramouli GV, Zhao L, Walker RL, Meltzer PS, Willingham MC, Cheng SY. An unliganded thyroid hormone beta receptor activates the cyclin D1/cyclin-dependent kinase/retinoblastoma/E2F pathway and induces pituitary tumorigenesis. Mol Cell Biol. 2005;25:124–135. doi: 10.1128/MCB.25.1.124-135.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kaneshige M, Suzuki H, Kaneshige K, Cheng J, Wimbrow H, Barlow C, Willingham MC, Cheng S. A targeted dominant negative mutation of the thyroid hormone alpha 1 receptor causes increased mortality, infertility, and dwarfism in mice. Proc Natl Acad Sci U S A. 2001;98:15095–15100. doi: 10.1073/pnas.261565798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Gothe S, Wang Z, Ng L, Kindblom JM, Barros AC, Ohlsson C, Vennstrom B, Forrest D. Mice devoid of all known thyroid hormone receptors are viable but exhibit disorders of the pituitary-thyroid axis, growth, and bone maturation. Genes Dev. 1999;13:1329–1341. doi: 10.1101/gad.13.10.1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Frisk T, Farnebo F, Zedenius J, Grimelius L, Hoog A, Wallin G, Larsson C. Expression of RET and its ligand complexes, GDNF/GFRalpha-1 and NTN/GFRalpha-2, in medullary thyroid carcinomas. Eur J Endocrinol. 2000;142:643–649. doi: 10.1530/eje.0.1420643. [DOI] [PubMed] [Google Scholar]
- 27.Gimm O, Chi H, Dahia PL, Perren A, Hinze R, Komminoth P, Dralle H, Reynolds PR, Eng C. Somatic mutation and germline variants of MINPP1, a phosphatase gene located in proximity to PTEN on 10q23.3, in follicular thyroid carcinomas. J Clin Endocrinol Metab. 2001;86:1801–1805. doi: 10.1210/jcem.86.4.7419. [DOI] [PubMed] [Google Scholar]
- 28.Achen MG, Stacker SA. Molecular control of lymphatic metastasis. Ann N Y Acad Sci. 2008;1131:225–234. doi: 10.1196/annals.1413.020. [DOI] [PubMed] [Google Scholar]
- 29.Yu XM, Lo CY, Lam AK, Lang BH, Leung P, Luk JM. The potential clinical relevance of serum vascular endothelial growth factor (VEGF) and VEGF-C in recurrent papillary thyroid carcinoma. Surgery. 2008;144:934–940. doi: 10.1016/j.surg.2008.07.027. discussion 940-931. [DOI] [PubMed] [Google Scholar]
- 30.Yu XM, Lo CY, Lam AK, Leung P, Luk JM. Serum vascular endothelial growth factor C correlates with lymph node metastases and high-risk tumor profiles in papillary thyroid carcinoma. Ann Surg. 2008;247:483–489. doi: 10.1097/SLA.0b013e31815fa447. [DOI] [PubMed] [Google Scholar]
- 31.Lin FJ, Chen X, Qin J, Hong YK, Tsai MJ, Tsai SY. Direct transcriptional regulation of neu-ropilin-2 by COUP-TFII modulates multiple steps in murine lymphatic vessel development. J Clin Invest. 2010;120:1694–1707. doi: 10.1172/JCI40101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Tachibana M, Tonomoto Y, Hyakudomi R, Hyakudomi M, Hattori S, Ueda S, Kinugasa S, Yoshimura H. Expression and prognostic significance of EFNB2 and EphB4 genes in patients with oesophageal squamous cell carcinoma. Dig Liver Dis. 2007;39:725–732. doi: 10.1016/j.dld.2007.05.013. [DOI] [PubMed] [Google Scholar]
- 33.Zhu Y, Sun Y, Xie L, Jin K, Sheibani N, Greenberg DA. Hypoxic induction of endoglin via mitogen-activated protein kinases in mouse brain microvascular endothelial cells. Stroke. 2003;34:2483–2488. doi: 10.1161/01.STR.0000088644.60368.ED. [DOI] [PubMed] [Google Scholar]
- 34.Mourtada-Maarabouni M, Keen J, Clark J, Cooper CS, Williams GT. Candidate tumor suppressor LUCA-15/RBM5/H37 modulates expression of apoptosis and cell cycle genes. Exp Cell Res. 2006;312:1745–1752. doi: 10.1016/j.yexcr.2006.02.009. [DOI] [PubMed] [Google Scholar]
- 35.Zimonjic DB, Kato Y, Ying H, Popescu NC, Cheng SY. Chromosomal aberrations in cell lines derived from thyroid tumors spontaneously developed in TRbetaPV/PV mice. Cancer Genet Cytogenet. 2005;161:104–109. doi: 10.1016/j.cancergencyto.2005.02.007. [DOI] [PubMed] [Google Scholar]
- 36.Ying H, Furuya F, Zhao L, Araki O, West BL, Hanover JA, Willingham MC, Cheng SY. Aberrant accumulation of PTTG1 induced by a mutated thyroid hormone beta receptor inhibits mitotic progression. J Clin Invest. 2006;116:2972–2984. doi: 10.1172/JCI28598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Losada A, Hirano T. Dynamic molecular linkers of the genome: the first decade of SMC proteins. Genes Dev. 2005;19:1269–1287. doi: 10.1101/gad.1320505. [DOI] [PubMed] [Google Scholar]
- 38.Patton JG, Porro EB, Galceran J, Tempst P, Nadal-Ginard B. Cloning and characterization of PSF, a novel pre-mRNA splicing factor. Genes Dev. 1993;7:393–406. doi: 10.1101/gad.7.3.393. [DOI] [PubMed] [Google Scholar]
- 39.Yang YS, Hanke JH, Carayannopoulos L, Craft CM, Capra JD, Tucker PW. NonO, a non-POU -domain-containing, octamer-binding protein, is the mammalian homolog of Drosophila non-Adiss. Mol Cell Biol. 1993;13:5593–5603. doi: 10.1128/mcb.13.9.5593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Rajesh C, Baker DK, Pierce AJ, Pittman DL. The splicing-factor related protein SFPQ/PSF interacts with RAD51D and is necessary for homology-directed repair and sister chromatid cohesion. Nucleic Acids Research. 2010 doi: 10.1093/nar/gkq738. 10.1093/nar/gkql738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Crozat A, Aman P, Mandahl N, Ron D. Fusion of CHOP to a novel RNA-binding protein in human myxoid liposarcoma. Nature. 1993;363:640–644. doi: 10.1038/363640a0. [DOI] [PubMed] [Google Scholar]
- 42.Kuroda M, Sok J, Webb L, Baechtold H, Urano F, Yin Y, Chung P, de Rooij DG, Akhmedov A, Ashley T, Ron D. Male sterility and enhanced radiation sensitivity in TLS(-/-) mice. EMBO J. 2000;19:453–462. doi: 10.1093/emboj/19.3.453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Gardiner M, Toth R, Vandermoere F, Morrice NA, Rouse J. Identification and characterization of FUS/TLS as a new target of ATM. Biochem J. 2008;415:297–307. doi: 10.1042/BJ20081135. [DOI] [PubMed] [Google Scholar]
- 44.Yingling J, Youn YH, Darling D, Toyo-Oka K, Pramparo T, Hirotsune S, Wynshaw-Boris A. Neuroepithelial stem cell proliferation requires LIS1 for precise spindle orientation and symmetric division. Cell. 2008;132:474–486. doi: 10.1016/j.cell.2008.01.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Binz SK, Sheehan AM, Wold MS. Replication protein A phosphorylation and the cellular response to DNA damage. DNA Repair (Amst) 2004;3:1015–1024. doi: 10.1016/j.dnarep.2004.03.028. [DOI] [PubMed] [Google Scholar]
- 46.Givalos N, Gakiopoulou H, Skliri M, Bousboukea K, Konstantinidou AE, Korkolopoulou P, Lelouda M, Kouraklis G, Patsouris E, Karatzas G. Replication protein A is an independent prognostic indicator with potential therapeutic implications in colon cancer. Mod Pathol. 2007;20:159–166. doi: 10.1038/modpathol.3800719. [DOI] [PubMed] [Google Scholar]
- 47.Tomkiel JE, Alansari H, Tang N, Virgin JB, Yang X, VandeVord P, Karvonen RL, Granda JL, Kraut MJ, Ensley JF, Fernandez-Madrid F. Autoimmunity to the M(r) 32,000 subunit of replication protein A in breast cancer. Clin Cancer Res. 2002;8:752–758. [PMC free article] [PubMed] [Google Scholar]
- 48.Lu C, Zhao L, Ying H, Willingham MC, Cheng SY. Growth activation alone is not sufficient to cause metastatic thyroid cancer in a mouse model of follicular thyroid carcinoma. Endocrinology. 2010;151:1929–1939. doi: 10.1210/en.2009-1017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Ibanez CF. Beyond the cell surface: new mechanisms of receptor function. Biochem Biophys Res Commun. 2010;396:24–27. doi: 10.1016/j.bbrc.2010.01.136. [DOI] [PubMed] [Google Scholar]
- 50.Wells SA., Jr and Santoro M. Targeting the RET pathway in thyroid cancer. Clin Cancer Res. 2009;15:7119–7123. doi: 10.1158/1078-0432.CCR-08-2742. [DOI] [PubMed] [Google Scholar]
- 51.Paratcha G, Ledda F, Ibanez CF. The neural cell adhesion molecule NCAM is an alternative signaling receptor for GDNF family ligands. Cell. 2003;113:867–879. doi: 10.1016/s0092-8674(03)00435-5. [DOI] [PubMed] [Google Scholar]
- 52.Ledda F, Paratcha G, Sandoval-Guzman T, Ibanez CF. GDNF and GFRalphal promote formation of neuronal synapses by ligand-induced cell adhesion. Nat Neurosci. 2007;10:293–300. doi: 10.1038/nn1855. [DOI] [PubMed] [Google Scholar]
- 53.Paratcha G, Ledda F, Baars L, Coulpier M, Besset V, Anders J, Scott R, Ibanez CF. Released GFRalphal potentiates downstream signaling, neuronal survival, and differentiation via a novel mechanism of recruitment of c-Ret to lipid rafts. Neuron. 2001;29:171–184. doi: 10.1016/s0896-6273(01)00188-x. [DOI] [PubMed] [Google Scholar]
- 54.Crone SA, Lee KF. The bound leading the bound: target-derived receptors act as guidance cues. Neuron. 2002;36:333–335. doi: 10.1016/s0896-6273(02)01009-7. [DOI] [PubMed] [Google Scholar]
- 55.Santoro M, Melillo RM, Fusco A. RET/PTC activation in papillary thyroid carcinoma: European Journal of Endocrinology Prize Lecture. Eur J Endocrinol. 2006;155:645–653. doi: 10.1530/eje.1.02289. [DOI] [PubMed] [Google Scholar]
- 56.Elia D, Madhala D, Ardon E, Reshef R, Halevy O. Sonic hedgehog promotes proliferation and differentiation of adult muscle cells: Involvement of MAPK/ERK and PI3K/Akt pathways. Biochim Biophys Acta. 2007;1773:1438–1446. doi: 10.1016/j.bbamcr.2007.06.006. [DOI] [PubMed] [Google Scholar]
- 57.Fagman H, Grande M, Gritli-Linde A, Nilsson M. Genetic deletion of sonic hedgehog causes hemiagenesis and ectopic development of the thyroid in mouse. Am J Pathol. 2004;164:1865–1872. doi: 10.1016/S0002-9440(10)63745-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Niederreither K, Vermot J, Schuhbaur B, Chambon P, Dolle P. Embryonic retinoic acid synthesis is required forforelimb growth and anter-oposterior patterning in the mouse. Development. 2002;129:3563–3574. doi: 10.1242/dev.129.15.3563. [DOI] [PubMed] [Google Scholar]
- 59.Jiang F, Herman GE. Analysis of Nsdhl-deficient embryos reveals a role for Hedgehog signaling in early placental development. Hum Mol Genet. 2006;15:3293–3305. doi: 10.1093/hmg/ddl405. [DOI] [PubMed] [Google Scholar]
- 60.Mann RK, Beachy PA. Novel lipid modifications of secreted protein signals. Annu Rev Biochem. 2004;73:891–923. doi: 10.1146/annurev.biochem.73.011303.073933. [DOI] [PubMed] [Google Scholar]
- 61.Chen MH, Li YJ, Kawakami T, Xu SM, Chuang PT. Palmitoylation is required for the production of a soluble multimeric Hedgehog protein complex and long-range signaling in vertebrates. Genes Dev. 2004;18:641–659. doi: 10.1101/gad.1185804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Callahan CA, Ofstad T, Horng L, Wang JK, Zhen HH, Coulombe PA, Oro AE. MIM/BEG4, a Sonic hedgehog-responsive gene that potentiates Gli-dependent transcription. Genes Dev. 2004;18:2724–2729. doi: 10.1101/gad.1221804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Mattila PK, Salminen M, Yamashiro T, Lappalainen P. Mouse MIM, a tissue-specific regulator of cytoskeletal dynamics, interacts with ATP-actin monomers through its C-terminal WH2 domain. J Biol Chem. 2003;278:8452–8459. doi: 10.1074/jbc.M212113200. [DOI] [PubMed] [Google Scholar]
- 64.Lee YG, Macoska JA, Korenchuk S, Pienta KJ. MIM, a potential metastasis suppressor gene in bladder cancer. Neoplasia. 2002;4:291–294. doi: 10.1038/sj.neo.7900231. [DOI] [PMC free article] [PubMed] [Google Scholar]



