Abstract
Human pituitary adenomas are the most common intracranial neoplasms. Approximately 5% of them are familial adenomas. Patients with familial tumors carry germline mutations in predisposition genes, including AIP, MEN1 and PRKAR1A. These mutations are extremely rare in sporadic pituitary adenomas, which therefore are caused by different mechanisms. Multiple tumor suppressive genes linked to sporadic tumors have been identified. Their inactivation is caused by epigenetic mechanisms, mainly promoter hypermethylation, and can be placed into two groups based on their functional interaction with tumor suppressors RB or p53. The RB group includes CDKN2A, CDKN2B, CDKN2C, RB1, BMP4, CDH1, CDH13, GADD45B and GADD45G; AIP and MEN1 genes also belong to this group. The p53 group includes MEG3, MGMT, PLAGL1, RASSF1, RASSF3 and SOCS1. We propose that the tumor suppression function of these genes is mainly mediated by the RB and p53 pathways. We also discuss possible tumor suppression mechanisms for individual genes.
Keywords: Sporadic pituitary adenoma, Tumor suppression, Genetic mutation, Epigenetic, RB, p53
1. Introduction
Human pituitary adenomas are the most common intracranial tumors, typically arising from hormone secreting cells in the anterior pituitary (Ezzat et al., 2004). Tumors secreting excess pituitary hormones and causing characteristic phenotypic syndromes in patients are classified as clinically functioning adenomas, and include growth hormone (GH) secreting, prolactin (PRL) secreting, adrenocorticotropin (ACTH) secreting and thyrotropin (TSH) secreting tumors. Rarely, tumors producing gonadotropins, luteining hormone (LH) and follicle stimulating hormone (FSH), can cause clinical symptoms of gonadotropin excess. Tumors which do not lead to clinical symptoms are known as clinically non-functioning adenomas (NFAs). NFAs are derived from all types of anterior pituitary hormone secreting cells, but most commonly from gonadotroph cells (Chaidarun and Klibanski, 2002). NFAs can grow very large in size and can cause mass effect resulting in hypopituitarism, headache, vision impairment, and neurologic dysfunction in patients. Despite recent advances in understanding the pathogenesis of these tumors, the underlying cause of the majority of pituitary tumors remains elusive.
Studies have demonstrated that genetic and epigenetic mutations play a determining role in the development of human neoplasms (Peltomaki, 2012). Normal cells contain an intrinsic tumor suppression mechanism which consists of multiple pathways maintaining normal cell homeostasis (Lowe et al., 2004). These tumor suppression pathways regulate many checkpoints which dictate the fate of cells in response to genotoxic, metabolic and other stress stimuli. Cells with excessive damage are prevented from further proliferation by induction of permanent arrest, such as cellular senescence, or programmed death, such as apoptosis. Each tumor suppression pathway is composed of multiple components forming a regulatory cascade. For normal cells to become tumorous, these tumor suppression barriers have to be circumvented by mutation of one or more pathway components (Hanahan and Weinberg, 2013).
The first evidence indicating that pituitary tumors may be caused by somatic mutations, genetic or epigenetic, came from early tumor clonality studies. We investigated X-chromosome inactivation patterns in NFAs from female patients by analyzing restriction fragment length polymorphisms and methylation status of the phosphoglycerate kinase (PGK) and hypoxanthine phosphoribosyltransferase (HPRT) genes (Alexander et al., 1990). We found that individual NFAs contain only one type of X-inactivation, paternal, or, maternal, never both. This observation was confirmed by other independent studies (Herman et al., 1990; Jacoby et al., 1990). In addition, similar results were also found in ACTH- secreting adenomas (Biller et al., 1992; Gicquel et al., 1992, 1994; Herman et al., 1990; Zahedi et al., 2001), GH-secreting adenomas (Herman et al., 1990; Jacoby et al., 1990), and prolactinomas (Herman et al., 1990; Jacoby et al., 1990; Ma et al., 2002). These data indicate that human pituitary adenomas are monoclonal in origin suggesting that individual tumors are derived from single cells driven by a somatic gene mutation or mutations.
The second line of evidence came from studies of patients with presumed germ-line mutations in familial pituitary adenomas (FPAs), including multiple endocrine neoplasia type I (MEN1), Carney complex (CNC) and familial isolated pituitary adenoma (FIPA). Patients with these conditions carry germline mutations, and pituitary tumors from these patients display loss of heterozygosity (LOH) in the affected locus. For example, genetic linkage analyses indicate that the genetic defect causing MEN1 is located on chromosome 11q13 (Larsson et al., 1988). Subsequently, the germline mutations in the multiple endocrine neoplasia type I (MEN1) gene were identified from this region. Similarly the germline mutation in cAMP-dependent protein kinase type I-alpha regulatory subunit (PRKAR1A) was identified in CNC patients (Kirschner et al., 2000) and aryl hydrocarbon receptor interacting protein (AIP) in FIPA patients (Vierimaa et al., 2006). In addition, germline mutations in cyclin-dependent kinase inhibitor 1B (CDKN1B) were identified in patients with an MEN1-like syndrome, but without a MEN1 gene mutation (Pellegata et al., 2006).
Genetic mutations clearly contribute to the development of FPAs. However, FPA accounts for only ∼5% of all pituitary tumors. The genetic mutations identified in FPAs do not play a significant role in the pathogenesis of sporadic tumors. Thus far, GNAS, encoding the stimulatory guanine nucleotide-binding protein (Gαs) (Landis et al., 1989; Lyons et al., 1990), is the only gene confirmed which is linked to a subset (∼40%) of GH-secreting adenomas (Freda et al., 2007), suggesting that novel somatic DNA mutations, if any, have yet to be identified in vast majority of sporadic tumors. Interestingly, no difference has been found in clinical characteristics, hormone levels or response to therapy in patients with tumors with or without Gαs mutations.
In recent years, emerging evidence indicates that epigenetic modifications are the major alternative force altering the expression of genes involved in neoplastic development (Dawson and Kouzarides, 2012; You and Jones, 2012), including pituitary tumorigenesis (Tateno et al., 2010). Epigenetic changes include aberrant DNA methylation and histone modification (Jaenisch and Bird, 2003; Peltomaki, 2012). Importantly, DNA methylation has been observed to be the main cause for gene inactivation in pituitary tumors (Yacqub-Usman et al., 2012b). DNA methylation involves DNA methlytransferases (DNMTs), which transfer a methyl group from S-adenosylmethionine to the 5-position of cytosine residues in DNA. Four major DNMTs have been identified so far: DNMT1, DNMT3A, DNMT3B and DNMT3L. DNMT3A and DNMT3B specifically recognize unmethylated DNA and establish de novo methylation patterns (Okano et al., 1999, 1998). DNMT3L does not possess enzyme activity, but functions as a co-factor to stimulate activities of DNMT3A and DNMT3B (Suetake et al., 2004). DNMT1 is universally expressed and responsible for methylation maintenance during DNA replication and repair (Leonhardt et al., 1992; Mortusewicz et al., 2005). Among the three enzymatically active DNMTs, only DNMT3B has been shown to be up regulated in human pituitary tumors by a histone modification mechanism (Zhu et al., 2008). Although DNMT3B was suggested as a putative mediator of epigenetic control in pituitary adenomas (Zhu et al., 2008), it remains to be determined whether it is responsible for gene silencing in these tumors. Over the years, a number of genes have been found to be inactivated in pituitary tumors by genetic or epigenetic mechanisms (Table 1). They are functionally linked to the two most important tumor suppressors, RB and p53. In this review we will discuss recent progress on these tumor suppressive genes involved in the development of human pituitary adenomas.
Table 1.
List of tumor suppressive genes inactivated in sporadic pituitary adenomas.a
| Gene | Description |
|---|---|
| AIP | Aryl hydrocarbon receptor interacting protein, predisposition gene for FIPA, inactivated in 3.8% sporadic pituitary tumors by genetic mutations (number of tumors examined, n = 1875) |
| BMP-4 | Bone morphogenetic protein 4, cytokine of the TGFβ superfamily, down regulated in 45% of pituitary tumors (n = 107) |
| CDKN2A | Cyclin-dependent kinase inhibitor 2A, p16Ink4a, inhibits CDK4/6 and activates RB, silenced by promoter methylation in 56% of pituitary tumors (n = 426) |
| CDKN2A | Alternative open reading frame of CDKN2A transcripts, p14Arf, inhibits MDM2 and activates p53, silenced by promoter methylation in 12% of pituitary tumors (n = 69) |
| CDKN2B | Cyclin-dependent kinase inhibitor 2B, p15Ink4b, inhibits CDK4/6 and activates RB, silenced by promoter methylation in 38% of pituitary tumors (n = 76) |
| CDKN2C | Cyclin-dependent kinase inhibitor 2C, p18Ink4c, inhibits CDK4/6 and activates RB, silenced by promoter methylation in 20% of pituitary tumors (n = 83) |
| CDKN1A | Cyclin-dependent kinase inhibitor 1A, p21Cip1, a p53 target gene, inhibits CDK2, CDK4 and activates RB, component of the RB pathway, down regulated in 47% of pituitary tumors (n = 111) |
| CDKN1B | Cyclin-dependent kinase inhibitor 1B, p27Kip1, a predisposition gene for MEN4, inhibits CDK2, CDK4 and activates RB, down regulated in 45% of sporadic pituitary adenomas (n = 404) |
| CDH1 | E-cadherin, classical epithelial cadherin, cell adhesion protein, inhibitor of epithelial–mesenchymal transition, silenced by promoter methylation in 36% of pituitary tumors (n = 85) |
| CDH13 | H-cadherin, a nonclassical cadherin, silenced by promoter methylation in 30% of tumors (n = 69) |
| GADD45B | Growth arrest and DNA-damage-inducible protein GADD45β, inhibits CDK1/cyclin B1 and interacts with PCNA, down regulated in pituitary tumors (n = 14) |
| GADD45G | Growth arrest and DNA-damage-inducible protein GADD45γ, inhibits CDK1/cyclin B1, interacts with p21Cip1 and PCNA, silenced by promoter methylation in 58% of tumors (n = 33) |
| MEG3 | Maternally expressed gene 3, a long non-coding RNA, a p53 activator, silenced in 96% of NFAs (n = 79), down regulated in 30% of functioning adenomas (n = 68) |
| MEN1 | Predisposition gene for multiple endocrine neoplasia type I syndrome, encodes menin, inactivated in 3% of sporadic pituitary tumors by genetic mutations (n = 535) |
| MGMT | O-6-methylguanine–DNA methyltransferase, involved in DNA repair, silenced by promoter methylation in 18% of tumors (n = 93) |
| PLAGL1 | Pleiomorphic adenoma gene-like 1, also known as ZAC (zinc finger protein which regulates apoptosis and cell cycle arrest), down regulated in 64% ofpituitary tumors by RT-PCR (n = 47) |
| RASSF1 | Ras association (RalGDS/AF-6) domain family member 1, a p53 activator, silenced by promoter methylation in 38% of pituitary tumors (n = 52) |
| RASSF3 | Ras association (RalGDS/AF-6) domain family member 3, also known as RASSF5, a p53 activator, silenced by promoter methylation in all pituitary tumors (n = 27) |
| RB1 | Retinoblastoma 1, the core member of the RB pathway, a negative cell cycle regulator, silenced by promoter methylation in 27% of pituitary tumors (n = 158) |
| SOCS1 | Suppressor of cytokine signaling 1, inhibitor of JAK/STAT pathways, a p53 activator, silenced by promoter methylation in 51% of pituitary tumors (n = 57) |
Percentages of tumors with genetic or epigenetic mutations for each individual gene are calculated based on data obtained from references cited in the main text.
2. The RB connection
The tumor suppressor RB plays a central role in regulating cell cycle progression (Henley and Dick, 2012). The RB pathway is also involved in the regulation of differentiation, apoptosis and maintenance of genome stability (Dick and Rubin, 2013; Indovina et al., 2013). This pathway is composed of the RB family of proteins, E2Fs, cyclins, cyclin-dependent kinases (CDKs) and CDK inhibitors. When active, RB arrests the cell cycle at G1 by interacting with chromosome remodeling factors (CRFs) and transcription factors E2Fs, thereby inhibiting expression of genes required for cell cycle progression (Henley and Dick, 2012). Phosphorylation of RB releases CRFs and frees E2Fs, which activates expression of genes permitting cell cycle progression. RB is mainly phosphorylated by CDK-cyclin complexes, including cyclin D-CDK4/6 and cyclin E/CDK2, which are negatively regulated by the INK4 and CIP/KIP families of inhibitors. The INK4 family of CDK inhibitors, namely p15Ink4b, p16Ink4a, p18Ink4c and p19Ink4d, specifically inhibit cyclin D/CDK4 or 6, whereas the CIP/KIP family of CDK inhibitors, namely p21Cip1, p27Kip1 and p57Kip2, inhibit a broader range of CDKs (Sherr and Roberts, 1999). The RB pathway is compromised if a mutation inactivates one or more components of the pathway. Therefore, the vast majority of human malignancies harbor mutations in the RB pathway (Di Fiore et al., 2013; Nevins, 2001). In human pituitary adenomas, genetic mutations of the RB pathway components are very rare. However, abundant evidence indicates that inactivation of the RB pathway is mediated by epigenetic mutations, which play a significant role in pituitary tumorigenesis.
2.1. The RB pathway and sporadic pituitary adenomas
Genetic mutations of genes in the RB pathway are extremely rare in human pituitary adenomas (Cryns et al., 1993; Ikeda et al., 1997; Jaffrain-Rea et al., 1999; Takeuchi et al., 1998). To date, only a small number of genetic mutations of RB1, p15Ink4b and p16Ink4a have been reported in sporadic tumors (Ogino et al., 2005; Simpson et al., 2001). Most studies have focused on epigenetic modifications, mainly promoter methylation. Very few studies include the investigation of cyclins and CDKs. These data are also inconsistent. For example, Saeger et al. (2001) reported that expression of cyclin D1 was detected in 1 out of 60 tumors by immunohistochemical staining. In contrast, a study by Simpson et al. (2001) indicated that 22 out of 45 tumors were positive for cyclin D1. Jordan et al. (2000) reported that over expression of cyclin D1 was detected in NFAs and cyclin E in ACTH-secreting tumors, respectively. CDK4 carrying mutations at codon R24 are resistant to the INK4 family of CDK inhibitors and have been identified as germline mutations in familial melanoma patients (Soufir et al., 1998; Zuo et al., 1996). However, such CDK4 mutations have not been found in pituitary tumors (Honda et al., 2003; Ogino et al., 2005; Simpson et al., 2001; Vax et al., 2003). These data suggest that cyclins and CDKs may not play a significant role in pituitary tumorigenesis.
The most frequently investigated CDK inhibitor of the CIP/KIP family is p27Kip1 encoded by the CDKN1B gene, which has been identified as a predisposition gene in MEN-1 like patients (Pellegata et al., 2006). No genetic mutations of the CDKN1B gene have been found in sporadic pituitary tumors (Dahia et al., 1998; Igreja et al., 2009; Ikeda et al., 1997; Takeuchi et al., 1998). However, the expression of p27Kip1 is generally lower in pituitary tumors compared to that in normal pituitaries (Bamberger et al., 1999; Komatsubara et al., 2001; Korbonits et al., 2002; Lidhar et al., 1999). The CDKN1A gene, encoding p21Cip1, has also been investigated and no genetic mutations in this gene were found in pituitary tumors (Ikeda et al., 1997; Yoshino et al., 2007). Expression of p21Cip1 was down regulated in approximately 47% of tumors compared to normal pituitary tissue (Hiyama et al., 2002; Suliman et al., 2001). It appears that promoter hypermethylation does not play a role in down regulation of p21Cip1 and p27Kip1 in pituitary tumors (Yoshino et al., 2007). Therefore, the mechanism for their down regulation remains to be determined.
In contrast, promoter hypermethylation is frequently associated with the INK4 family of CDK inhibitors and the RB1 gene in pituitary tumors. More than 400 pituitary tumors have been examined for p16Ink4a methylation (Abd El-Moneim and Abd El-Rehim, 2009; Bello et al., 2006; Jaffrain-Rea et al., 1999; Ogino et al., 2005; Ruebel et al., 2001; Seemann et al., 2001; Simpson et al., 1999, 2001, 2004; Woloschak et al., 1997; Yoshino et al., 2007). Promoter hypermethylation of thep16Ink4a gene are found in 56% of pituitary tumors. It occurs most frequently in NFAs (69%) and least frequently in GH-secreting tumors (29%). Compared to p16Ink4a, p15Ink4b and p18Ink4c promoter methylation occur less frequently, 38% and 20% of pituitary tumors, respectively (Hossain et al., 2009; Kirsch et al., 2009; Ogino et al., 2005; Yoshino et al., 2007). Loss of p16Ink4a expression is found in 65% of pituitary tumors (Abd El-Moneim and Abd El-Rehim, 2009; Seemann et al., 2001; Simpson et al., 1999, 2001, 2004; Woloschak et al., 1996). RB1 promoter methylation is found in 27% of pituitary tumors (Bello et al., 2006; Ogino et al., 2005; Simpson et al., 2001, 2000; Yoshino et al., 2007), and loss of RB1 gene expression occurs in 29% of tumors (Ogino et al., 2005; Simpson et al., 2001, 2000). Two observations need to be pointed out. First, promoter methylation correlates closely with the loss of gene expression (Abd El-Moneim and Abd El-Rehim, 2009; Ogino et al., 2005; Simpson et al., 2004), indicating that gene inactivation in pituitary tumors is likely due to promoter methylation. Second, the methylation of p16Ink4a and RB1 promoters is mutually exclusive in pituitary tumors (Bello et al., 2006; Ogino et al., 2005; Yoshino et al., 2007). Data from two studies indicate that the percentage of pituitary tumors with at least one RB pathway gene silenced due to promoter methylation is approximately 90% (Ogino et al., 2005; Yoshino et al., 2007). Because any one gene inactivation is likely to compromise the function of the RB pathway, these studies strongly suggest that inactivation of the RB pathway contributes to the development of pituitary adenomas.
Animal models provide further evidence supporting the hypothesis that the RB pathway plays a causal role in pituitary tumor development. Mice carrying a heterozygous deletion of the Rb1 gene rapidly grow pituitary tumors with 100% penetrance (Hu et al., 1994; Jacks et al., 1992; Leung et al., 2004). Mice with mutations of p27Kip1 or p18Ink4c develop pituitary hyperplasia as well as pituitary tumors (Fero et al., 1996; Franklin et al., 1998; Kiyokawa et al., 1996; Nakayama et al., 1996). Furthermore, mice with a double knockout of both p27Kip1 and p18Ink4c grew pituitary tumors at a much younger age compared to mice with individual gene deletions (Franklin et al., 1998), indicating that these two CDK inhibitors function synergistically in tumor suppression. These knockout mice grew melanotroph tumors in the intermediate lobe (Nikitin and Lee, 1996); no tumors grew in the anterior lobe where human hormone secreting cells reside. This is likely due to the genetic background of the animals which were mixed between C57BL/6 and 129/Sv or its derivatives. It has been demonstrated that Rb1+/−mice on a 129/Sv background favors the development of tumors in the intermediate lobe of the pituitary. It was later found that the intermediate lobe of the pituitary is inherently abnormal in 129/Sv mice (Leung et al., 2004). Switching to a C57BL/6 background, by backcrossing, increases anterior lobe tumor development in the Rb1+/− mice (Leung et al., 2004). In the aggregate, these studies further demonstrate the critical role that the RB pathway plays in suppression of pituitary tumorigenesis.
Not all human pituitary tumors have p16Ink4a or RB inactivation (Bello et al., 2006; Ogino et al., 2005; Yoshino et al., 2007), suggesting that inactivation of other genes also contributes to pituitary tumor growth in humans. The RB pathway functions as an effector in cell cycle control in response to growth stimulatory or inhibitory signals, such as growth factors, oncogenic stresses, oxidative stresses and DNA damage (Courtois-Cox et al., 2008; Macleod, 2008; Rayess et al., 2012; Sperka et al., 2013). It also regulates its downstream target genes involved in a broad range of cellular activities (Indovina et al., 2013). Therefore, it is likely that inactivation of inhibitory signaling regulating the RB pathway may also contribute to pituitary tumorgenesis.
2.2. Aryl hydrocarbon receptor interacting protein (AIP)
The AIP gene was identified in FIPA patients from two families in Northern Finland (Vierimaa et al., 2006). It consists of 6 exons and encodes a protein of 330 amino acids, containing a peptidylprolyl cis-trans isomerase-like (PPIase-like) domain (amino acids 31-121), a tetratricopeptide repeat (TPR) domain with three TPR motifs (aa 179-298) and a c-terminal α-7 helix (Cα-7h) (Trivellin and Korbonits, 2011). Sequencing DNA from the patient's normal tissues uncovered a nonsense mutation at nucleotide position 40 in the coding region of the AIP gene where a cytosine was substituted by a thymidine (c.40 C > T) converting a Gln codon to a stop codon (Q14X) (Vierimaa et al., 2006). Importantly, the wild type allele of the AIP gene was found to be lost in all patient tumors. Patients with AIP mutations are much younger than the AIP mutation negative patients (Vierimaa et al., 2006). All these fit well with the classic “two-hit” tumor suppressor model proposed originally by Knudson (Knudson, 1971), suggesting that the AIP gene is a tumor suppressor.
Since the first AIP mutations in FIPA were reported in 2006, more than 75 different types of mutations in the AIP gene have been identified (Beckers et al., 2013; Cazabat et al., 2012; Cuny et al., 2013). Beckers et al. (2013) reported a comprehensive list of AIP mutations in 215 patients reported in literature in a 2013 review. The AIP mutation occurs in 26% of familial adenomas based on four studies including 341 tumors (Daly et al., 2007; Georgitsi et al., 2008b; Igreja et al., 2010; Leontiou et al., 2008). Most familial adenomas with AIP mutations are GH- secreting and GH/PRL-secreting tumors, which account for 41% and 19%, respectively. Prolactinoma and NFA account for 9% and 16%, respectively. The AIP mutation occurs much less frequently in sporadic pituitary adenomas; 3.8% based on 13 independent studies which have examined 1875 tumors (Barlier et al., 2007; Buchbinder et al., 2008; Cazabat et al., 2012; Cuny et al., 2013; DiGiovanni et al., 2007; Georgitsi et al., 2008a, 2007; Iwata et al., 2007; Leontiou et al., 2008; Occhi et al., 2010; Raitila et al., 2007; Tichomirowa et al., 2011; Yu et al., 2006). No sporadic GH/PRL-secreting tumors have been reported in these studies. AIP mutations occur in 5.4%, 4.4%, 3.3% and 2.4% of PRL-, GH-, ACTH-secreting tumors and NFAs, respectively. Approximately two thirds of those mutations are germline. The types of mutations in the AIP gene are very heterogeneous. They include nonsense mutations, missense mutations, small in-frame deletions, large deletions and the full gene deletion as well as mutations in introns that affect RNA splicing. These data suggest that mutations in the AIP gene are likely a random event in sporadic pituitary tumors and AIP inactivation plays an equally important role in the development of these tumors.
To investigate the role of the AIP gene in tumorigenesis, an Aip knockout mouse line was generated by inserting a gene trap cassette in intron 2. The insertion results in a truncated AIP protein without the TPR domain and the c-terminal alpha-7 helix (Raitila et al., 2010). A homozygous Aip inactivation was embryonic lethal. This is consistent with the finding in another Aip knockout mouse model by Lin et al. (2007) who observed that Aip−/− embryos died between E10.5 and E14.5 due to cardiovascular defects. Heterozygous Aip mice grew normally but developed multiple pituitary tumors as early as 6 months of age. All mice developed pituitary tumors at 15 months. The vast majority of the tumors (88%) were GH-secreting. Tumors secreting PRL or GH/PRL are also observed. In addition, the wild type Aip allele was found to be lost in all GH secreting tumors judged by LOH analysis and immunochemical staining (Raitila et al., 2010). The tumor spectrum in Aip mice resembles that found in FIPA patients with AIP mutations. Data from Aip mice have thus provided further support that mutation of the AIP gene predisposes to pituitary tumor development in humans.
The consensus regarding the molecular mechanisms whereby AIP suppresses pituitary tumor development is through its interactions with a large number of partner proteins. AIP interacts with partners at its TPR and Cα-7h domains (Morgan et al., 2012; Trivellin and Korbonits, 2011). Approximately 90% of AIP mutations result in loss of AIP protein expression, AIP proteins without TPR and/or Cα-7h domains, or AIP proteins with mutations in both domains (Beckers et al., 2013). At least 16 proteins have been identified and confirmed to interact with AIP as has been reviewed in detail by Trivellin and Korbonits (2011) and Beckers et al. (2013). The centerpiece of the AIP interacting proteins is aryl hydrocarbon receptor (AHR) (Carver and Bradfield, 1997; Ma and Whitlock, 1997). AHR was originally identified as a ligand dependent transcription factor activated by environmental toxins such as 2,3,7,8-tetrachloro-p-dioxin (TCDD) and is involved in xenobiotic metabolism (Denison and Nagy, 2003). AHR is a multiple function protein regulating a variety of cellular activities. When in the inactive state, AHR resides in the cytoplasm where it is bound to AIP, heat shock protein 90 (HSP90) and p23. Ligand binding activates AHR by translocation of the AHR complex to the nucleus. There, AHR is freed from its binding proteins and forms a heterodimer with aryl hydrocarbon receptor nuclear translocator (ARNT) to regulate expression of its target genes which are involved in xenobiotic metabolism, cellular homeostasis, cell proliferation and development (Denison et al., 2011).
A likely mechanism whereby AIP suppresses tumor growth is by regulating AHR function. Indeed, AIP interaction has been shown to stabilize AHR by preventing it from ubiquitin mediated degradation (Kazlauskas et al., 2000; LaPres et al., 2000). In addition, AIP interaction retains AHR in the cytoplasm and enhances its ligand binding (Kazlauskas et al., 2000; LaPres et al., 2000; Petrulis et al., 2000). AIP interaction is essential for AHR function. For example, deletion of Aip led to a reduction in AHR levels. In addition, AHR mediated hepatotoxicity is abolished in the absence of AIP (Nukaya et al., 2010). Finally, Cai et al. (2011) reported that AIP formed complexes with ERα, which are recruited to the promoter of the target gene to inhibit gene transcription. Because AHR binds ERα and inhibits its function (Madak-Erdogan and Katzenellenbogen, 2012), it is likely that AIP inhibition of ERα is mediated by AHR. Taken together, these data support the concept that AIP suppresses tumor growth via AHR signaling.
Overwhelming evidence indicates that AHR plays a role in tumor suppression, primarily by activation of the RB pathway. AHR activated by ligands, such as TCDD, 3-methylcholanthrene, or baicalein, inhibits proliferation or induces apoptosis in several human cell lines via a number of mechanisms. For example, it has been shown that ligand activated AHR (1) induces CDK inhibitors p27Kip1 and p21Cip1 (Kolluri et al., 1999; Pang et al., 2008), (2) inhibits E2F mediated transcription through displacement of or direct interaction with coactivator p300 (Marlowe et al., 2004, 2008), and (3) activates RB by physical interaction (Elferink et al., 2001; Puga et al., 2000) or by induction of RB hypophosorylation (Barhoover et al., 2010; Cheng et al., 2012). However, AHR promotion of cell proliferation has also been reported. This discrepancy appears to be due to the experimental approaches used. AHR activation by a ligand usually inhibits cell proliferation (Barhoover et al., 2010; Cheng et al., 2012; Elferink et al., 2001; Pang et al., 2008). The conclusion that AHR promotes cell proliferation is usually based on data obtained by ectopic overexpression of AHR (Wong et al., 2009) or knockdown of AHR by RNAi (Dever and Opanashuk, 2012; Kalmes et al., 2011; Yin et al., 2013). One interpretation of these data obtained from experiments not using ligand treatment is that AHR may have a ligand independent function which is important in the maintenance of cellular homeostasis. Therefore, these studies do not necessarily contradict the hypothesis that AHR mediates ligand induced inhibition of cell proliferation.
2.3. Multiple endocrine neoplasia Type I (MEN1)
MEN1 syndrome is manifested by at least two concurrent endocrine related tumors including parathyroid adenomas, entero-pancreatic endocrine tumors, and pituitary adenomas (Brandi et al., 2001). Sporadic MEN1 accounts for approximately 10% of all patients diagnosed with MEN1 (Trump et al., 1996). Pituitary adenomas are common in MEN1 patients; up to 40% of MEN1 patients are diagnosed with pituitary adenomas (O'Brien et al., 1996; Oberg et al., 1989; Trump et al., 1996; Verges et al., 2002). The MEN1 gene responsible for the MEN1 phenotype is 9 kb long, containing 10 exons which encode a protein of 610-amino acids, named menin (Chandrasekharappa et al., 1997). MEN1 gene germline mutations are very common in MEN1 patients. Lemos and Thakker (2008) reviewed in detail MEN1 mutations reported for the first 10 years after the MEN1 gene was identified (Lemos and Thakker, 2008). LOH has been detected in all types of MEN1 tumors ranging from 10% to 100% (Bystrom et al., 1990; Debelenko et al., 1997; Friedman et al., 1989; Lubensky et al., 1996; Thakker et al., 1989). The loss of MEN1- both germline and in tumor tissue is consistent with the two hit model articulated by Knudsen (Knudson, 1971), indicating that MEN1 is a tumor suppressor. MEN1 mutations in sporadic pituitary tumors are very rare; a total of 15 tumors with MEN1 mutations have been identified from 535 sporadic adenomas and only one is a germline mutation (Asa et al., 1998; Bergman et al., 2000; Cuny et al., 2013; Farrell et al., 1999; Fukino et al., 1999; Poncin et al., 1999; Prezant et al., 1998; Schmidt et al., 1999; Tanaka et al., 1998; Wenbin et al., 1999; Zhuang et al., 1997). The extremely low mutation rate suggests that MEN1 mutations do not play a significant role in sporadic pituitary tumors.
Data from Men1 knockout mouse models further support the hypothesis that MEN1 is a tumor suppressor. Several Men1 mouse models have been created by various schemes (Bertolino et al., 2003a, 2003b; Biondi et al., 2002; Crabtree et al., 2001; Harding et al., 2009; Loffler et al., 2007). These Men1 mice share a similar phenotype. Homozygous mice died in utero at E11.5 to E13.5 (Bertolino et al., 2003b; Crabtree et al., 2001). Heterozygous mice developed normally. They grew tumors in a spectrum resembling that found in human MEN1. The percent of Men1+/− mice developing pituitary tumors varies depending on the study, ranging from 31% to 91% (Bertolino et al., 2003b; Harding et al., 2009; Loffler et al., 2007). Genetic analyses indicate that all pituitary tumors in the mice have lost the wild-type allele of the Men1 gene (Harding et al., 2009). These data provide further evidence that MEN1 is a bona fide tumor suppressor gene.
Menin plays a role in the regulation of cell proliferation both in vitro and in vivo. Ectopic expression of menin inhibits proliferation of several human and mouse cell lines in vitro (Bazzi et al., 2008; Gao et al., 2009; Kim et al., 1999; La et al., 2004; Schnepp et al., 2006; Shi et al., 2013; Stalberg et al., 2004; Wu et al., 2010) and in nude mice (Kim et al., 1999), and pituitary tumor cells in Men1 knockout mice (Walls et al., 2012). Conversely, inactivation of menin by RNAi knockdown or deletion of the Men1 gene leads to an increase in cell proliferation in cultured cells and in mice pancreatic islets (Milne et al., 2005b; Ratineau et al., 2004; Schnepp et al., 2006, 2004). Menin has also been shown to mediate growth inhibition induced by activin and TGFβ (Canaff et al., 2012; Kaji et al., 2001; Lacerte et al., 2004). Taken together, cell proliferation inhibition by menin contributes, at least in part, to tumor suppression by the MEN1 gene.
One plausible mechanism whereby menin inhibits cell proliferation and suppresses tumor growth is by activating the RB pathway. Menin up regulates expression of p27Kip1 and p18Ink44c by recruiting MLL to their respective promoter regions (Karnik et al., 2005; Milne et al., 2005b). MLL, a menin interacting protein, is a histone H3 lysine 4 specific histone methlytransferase complex, which activates transcription by histone modification (Milne et al., 2002, 2005a; Yokoyama et al., 2004). Transcription activation by MLL also requires the recruitment of LEDGF by menin (Yokoyama and Cleary, 2008). In contrast, menin represses cyclin B2 expression by interfering with promoter binding of several transcription factors including NF-Y, E2F and αCBP (Wu et al., 2010). Menin does so by recruiting histone H3 deacetylase 3 (HDAC3) to the gene, which inhibits transcription by reduction of histone H3 acetylation. Interestingly, methylation of histone H3 lysine 4 is also reduced (Wu et al., 2010). How does menin recruit MLL and HDAC to specific genes? La et al. (2004) found that menin can physically bind to double stranded DNAs and regulate cell proliferation. This could explain how menin may recognize certain sequences in p27Kip1, p18Ink4c and cyclin B2 genes thereby recruiting MLL and HDAC to their respective locations. Furthermore, Ivo et al. (2011) reported that menin increases the cellular concentration of RB protein via unknown mechanisms. Taken together, the RB pathway is likely a direct downstream target of menin signaling.
2.4. Bone morphogenic protein 4 (BMP-4)
BMP-4, a member of the TGFβ superfamily of cytokines, regulates diverse biological activities including development, differentiation and homeostasis. BMP-4 signals through its type I and type II serine/threonine kinase receptors. Ligand bound type I receptors are activated by the type II receptor via phosphorylation. Activated type I receptors phosphorylate R-Smads including Samds 1, 5 and 8, which subsequently associate with Smad4 and translocate to the nucleus where they transcriptionally activate their target genes (Miyazono et al., 2005). Evidence indicates that BMP-4 plays a role in tumor suppression via activation of the RB pathway. For example, BMP-4 inhibits growth of colorectal cancer stem cells by induction of terminal differentiation through up regulation of E-cadherin and down regulation of cyclin D1 (Lombardo et al., 2011). Su et al. (2011) reported that BMP-4 inhibits proliferation of lung cancer cells by induction of premature senescence involving stimulating expression of p16Ink4a and p21Cip1. In addition, BMP-4 inhibits growth of diffuse-type gastric carcinoma cells and prostate cancer cells by RB activation through up regulation of p21Cip1 (Brubaker et al., 2004; Shirai et al., 2011). In addition to activation of RB, BMP4 was shown to induce p53 dependent apoptosis by up regulation of PUMA expression and enhancement of Bax endopasmic reticulum translocation (Fukuda et al., 2006), suggesting that p53 also plays a role in mediating BMP4 tumor suppression.
The pituitary gland is a BMP-4 target, which regulates pituitary cell proliferation and hormone secretion in a cell type specific manner (Labeur et al., 2010). A link between BMP-4 and human pituitary tumorigenesis was first reported by Takeda et al. (2003), who investigated activin/BMP system in pituitary adenomas including 5 NFAs and 3 gonadotropinomas. No BMP-4 was detected in any tumor by RT-PCR. Recently, Yacqub-Usman et al. (2012a) found that down regulation of BMP-4 occurred in the majority of pituitary tumors when BMP-4 mRNA was determined by RT-PCR (overall 75%, n = 36). BMP-4 down regulation occurred most frequently in GH-secreting tumors (100%, n = 4), followed by NFAs (88%, n = 16), ACTH- secreting tumors (72%, n = 7), and prolactinomas (44%, n = 9). There are two CpG clusters in the BMP-4 gene. Bisulfite sequencing did not detect any differences in methylation of these CpGs between normal pituitary and pituitary tumors, indicating that DNA methylation does not play a significant role in BMP-4 down regulation. Instead, the level of histone H3 lysine 27 trimethylation (H3K27Me3), which is associated with gene repression (Bernstein et al., 2006; Cao et al., 2002), is significantly elevated in NFAs and GH- secreting tumors. In contrast, H3 lysine 9 acetylation (H3K9Ac), which is associated with gene activation (Karmodiya et al., 2012), is significantly reduced (Yacqub-Usman et al., 2012a), suggesting that the BMP-4 down regulation in pituitary tumors is due to histone modification.
Conflicting data have been obtained when BMP-4 expression is measured at the protein level. In Yacqub-Usman's study, the BMP-4 protein levels, measured by ELISA, were only down regulated in NFA and GH-secreting tumors compared to normal controls (Yacqub-Usman et al., 2012a). Paez-Pereda et al. (2003) reported that BMP-4 protein levels, measured by western blotting, were not down regulated in any pituitary tumor type compared to normal pituitary tissue. Interestingly, both studies showed that among tumor types, BMP-4 protein levels were the highest in prolactinomas. This high BMP-4 expression was believed to promote prolactinoma growth in mice (Paez-Pereda et al., 2003). Giacomini et al. (2006) reported that BMP-4 expression, measured by immunostaining, was significantly down regulated in 13 out of 15 human ACTH-secreting tumors. They demonstrated that BMP-4 inhibited proliferation of AtT20, a murine corticotroph tumor cell line, in culture and tumor growth in nude mice. The cause of these discrepancies is not known. However, it raises the possibility that BMP-4 function is cell type dependent and may promote prolactinoma growth, but suppress ACTH-secreting adenoma development.
2.5. Cadherin 1 (CDH1)
The CDH1 gene encodes E-cadherin, a member of the cadherin family of proteins, which play a major role in maintaining cell–cell adhesion and communication in normal tissues (Angst et al., 2001; Meng and Takeichi, 2009). There are several studies investigating E-cadherin expression in human pituitary adenomas. Immunohistochemical staining is the most common method used to detect E-cadherin. Because the antibodies and staining scoring system used in individual studies vary significantly, it is difficult to compare data between studies. However, the consensus is that E-cadherin expression is lost or significantly reduced in pituitary adenomas compared to normal pituitary tissue regardless of tumor type. For example, Elston et al. (2009) reported that E-cadherin is lost or significantly reduced in 96% of pituitary tumors. A similar result was also reported by Kawamoto et al. (1997). Studies by Qian et al. (2007) found a somewhat lower percentage of tumors with reduced E-cadhein expression, 62% of 69 tumors. Evang et al. (2011) investigated 48 ACTH-secreting tumors and found that 77% of them had lost or had significantly reduced E-cadherin. Fougner et al. (2010) found that the percentage was 47% in GH-tumors (n = 83). However, data from Xu et al. (2002) who studied 127 pituitary adenomas including all tumor types showed that the reduced expression is found only in GH tumors with prominent fibrous bodies. In general, data from most studies do not show any significant differences in E-cadherin expression between tumor types (Elston et al., 2009; Kawamoto et al., 1997; Qian et al., 2007). However, E-cadherin expression is usually significantly lower in invasive tumors compared to non-invasive tumors (Elston et al., 2009; Qian et al., 2007). E-cadherin expression also negatively correlates with tumor size (Fougner et al., 2010). These results indicate that loss of E-cadherin may play an important role in promoting pituitary tumor growth.
The methylation status of the CDH1 promoter was investigated by methylation specific PCR (MSP). Qian et al. (2007) found that approximately 36% of pituitary tumors had CDH1 promoter methylation. Interestingly, the number of tumors with CDH1 methylation increases with tumor grade: 11% of grade I tumors contained CDH1 methylation, while 63% of grade IV tumors contained CDH1 methylation. Therefore, the percentage of tumors with promoter methylation is much lower than those with E-cadherin down regulation. This may be due to the limitation of MSP which may not be able to detect all methylation sites in the promoter. Another possibility is that additional mechanisms may be responsible for down regulation of E-cadherin in tumors without CDH1 methylation. In 16 GH-secreting tumors with prominent fibrous bodies which were negative for E-cadherin expression, Xu et al. (2002) found that 6 had promoter methylation. In contrast, none of the 10 GH-secreting tumors without prominent fibrous bodies, which expressed E-cadherin, showed promoter methylation. These studies indicate that promoter methylation plays a significant role in silencing the CDH1 gene in pituitary adenomas.
Classical cadherins, which are transmembrane proteins, are key structural components of the adherens junction (Meng and Takeichi, 2009). E-cadherin is a classical type I cadherin found in epithelial tissues. It consists of a calcium binding extracellular domain, which is responsible for calcium-dependent homophilic and heterophilic cell interactions, and a cytoplasmic domain, which binds to catenins regulating intracellular signaling (van Roy and Berx, 2008). E-cadherin plays an important role in tumor suppression (Berx and van Roy, 2009; Jeanes et al., 2008). It is one of the major factors inhibiting epithelial–mesenchymal transition (EMT), which converts epithelial cells to migratory and invasive cells (De Craene and Berx, 2013). Down-regulation of E-cadherin has been shown to increase EMT, promoting invasiveness and metastasis (Osada et al., 1996; Perl et al., 1998; Tiwari et al., 2012; Tryndyak et al., 2010; Vleminckx et al., 1991). Lekva et al. (2012) reported that down regulation of E-cadherin is associated with increases in tumor size and invasiveness of GH-secreting tumors. There are several possible mechanisms whereby E-cadherin suppresses tumor growth. E-cadherin inhibits metastatic spreading of tumor cells by promoting cell–cell adhesion and development of normal epithelial architecture (Jeanes et al., 2008); E-cadherin inhibits mitogenic signaling by sequestering catenins with its cytoplasmic tail (Jeanes et al., 2008; Soto et al., 2008); and finally, E-cadherin inhibits cell proliferation by activation of RB via up regulation of the CDK inhibitor p27Kip1 (St Croix et al., 1998).
E-cadherin plays a crucial role in mediating the function of several tumor suppressors, including p53, RB and ESRP1. Wang et al. (2009) found that p53 up regulates E-cadherin expression by promoting degradation of transcription factor Slug, which is known to repress E-cadherin expression (Bolos et al., 2003; Hajra et al., 2002). Conversely, inhibition of p53 results in repression of E-cadherin expression by increasing DNMT1 expression, which epigenetically silences the CDH1 gene (Cheng et al., 2011). RB inhibits the EMT phenotype by regulation of E-cadherin expression (Arima et al., 2008). RB interacts with transcription factor AP-2α and binds to the CDH1 promoter to activate its transcription (Arima et al., 2008; Batsche et al., 1998). Conversely, RB inactivation down-regulates E-cadherin expression (Arima et al., 2008). ESRP1, the epithelial splicing regulatory protein 1, was identified as one of the two RNA binding proteins regulating splicing of the FGFR2 gene (Warzecha et al., 2009). Thought to function as a tumor suppressor in colorectal cancer (Leontieva and Ionov, 2009), ESRP1 is a key regulator of EMT, coordinating a complex epithelial alternative splicing network (Dittmar et al., 2012; Warzecha et al., 2010). Down-regulation of ESRP1 induces EMT phenotypes (Reinke et al., 2012; Warzecha et al., 2009). Very recently, ESRP1 was identified as a master regulator of EMT in human GH-secreting adenomas (Lekva et al., 2012, 2013). ESRP1 is positively correlated with E-cadherin expression in GH-secreting adenomas and its depletion by RNAi results in down regulation of E-cadherin in a GH-adenoma cell line (Lekva et al., 2012). Taken together, these studies suggest that down-regulation of E-cadherin plays a significant role in promoting pituitary tumorigenesis.
2.6. Cadherin 13 (CDH13)
The CDH13 gene encodes H-cadherin, also known as T-cadherin. Qian et al. (2007) found that more than half of pituitary tumors (54%) have lost or significantly reduced expression of H-cadherin. Similar to patterns seen with E-cadherin, H-cadherin down regulation was found more frequently in invasive tumors (74%) than that in non-invasive tumors (38%). Approximately 30% of all pituitary tumors contain hypermethylation in the CDH13 gene. There is no significant difference in promoter methylation between tumor types. However, CDH13 methylation is found more frequently in invasive tumors (42%) than in non-invasive tumors (19%) (Qian et al., 2007), consistent with H-cadherin expression. As is the case seen with the E-cadherin study, the number of tumors with CDH13 promoter methylation is less frequent compared to those with H-cadherin down regulation. Again, this may be due to the technical limitation of MSP or suggests that other, yet to be determined mechanisms, are responsible for the down regulation of H-cadherin in human pituitary tumors.
H-cadherin is a non-classical adherin, as it does not contain transmembrane and cytoplasmic domains but has a glycosylphosphatidylinositol anchor. H-cadherin functions as an adiponectin receptor and may not be involved in cell–cell adhesion (Philippova et al., 2009). Nevertheless, down regulation of H-cadherin by epigenetic silencing has been found in many types of human cancers and H-cadherin inhibits tumor cell proliferation (Andreeva and Kutuzov, 2010). Re-expression of H-cadherin induces G2/M arrest in human cancer cell lines including those derived from gliomas and hepatocellular carcinomas (Chan et al., 2008; Huang et al., 2003). This may be attributed to the induction of p21Cip1 expression by H-cadherin (Huang et al., 2003). In addition, H-cadherin has been shown to inhibit angiogenesis by inhibiting endothelial cell migration (Rubina et al., 2007) or insulin-dependent activation of PI3K/Akt/mTOR signaling (Philippova et al., 2012). These studies indicate that tumor suppression is a function of H-cadherin and that epigenetic down-regulation of the CDH13 gene may contribute to pituitary tumorigenesis.
2.7. Growth arrest and DNA-damage-inducible, beta (GADD45B) and gamma (GADD45G)
The GADD45 gene family consists of three members, GADD45A, GADD45B, and GADD45G, encoding GADD45α, GADD45β, and GADD45γ, respectively. They are stress response genes. GADD45 expression is normally low, and rapidly increases in response to a wide range of stress signals such as UV and ionizing radiation, inflammatory and pro-apoptotic cytokines, mitogenic stimuli and xenobiotics (Moskalev et al., 2012). Loss of GADD45 expression has been observed in human cancers from a variety of tissue origins (Tamura et al., 2012). Interestingly, the large majority of human cancers do not contain GADD45 gene mutations. Instead, promoter methylation is the major cause of GADD45 down regulation (Guo et al., 2013a,b; Tamura et al., 2012). Among the three GADD45 genes, GADD45G is the most frequently inactivated, followed by GADD45B and GADD45A, in human cancers (Tamura et al., 2012).
To search for genes involved in pituitary tumorigenesis, we compared gene expression profiles between normal pituitary tissue and NFAs using cDNA-representational difference analysis (cDNA-RDA) (Zhang et al., 2002). GADD45G was identified as the most frequently detected gene by RDA. Using RT-PCR, we examined GADD45γ expression in four normal pituitaries and 36 pituitary tumors. GADD45γ was detected in all the normal pituitaries. In contrast, it was only detected in four out of 36 tumors (Zhang et al., 2002), indicating that the vast majority of pituitary tumors have lost expression of GADD45γ. This finding was subsequently confirmed by three independent studies (Bahar et al., 2004; Mezzomo et al., 2011; Michaelis et al., 2011). So far a total of 121 sporadic pituitary adenomas including 62 NFA and 59 functioning adenomas have been reported. Loss of or significantly reduced GADD45γ expression is found in 74% of all pituitary tumors. Down regulation of GADD45γ is more frequently found in NFAs than in functioning tumors (90% and 58%, respectively) (Bahar et al., 2004; Mezzomo et al., 2011; Michaelis et al., 2011; Zhang et al., 2002). To investigate the mechanism of GADD45γ down regulation, Bahar et al. (2004) analyzed the methylation status in the GADD45G promoter region using MSP. They showed that methylation of the GADD45G promoter was detected in 58% of tumors. Among GADD45γ negative tumors, 82% had promoter methylation. Conversely, among GADD45γ positive tumors, 91% did not have promoter methylation (Bahar et al., 2004). These data strongly indicate that promoter methylation is the major cause for the loss of GADD45γ expression in human pituitary tumors. Michaelis et al. (2011) also evaluated GADD45α and GADD45β expression in pituitary tumors. Expression of GADD45α was similar between normal pituitary tissue and pituitary tumors. In contrast, GADD45β was down regulated in pituitary tumors by 68-fold judged by microarray data analyzed with the Ingenuity Pathway Analysis program. The GADD45β down regulation in tumors was subsequently confirmed by qPCR and immunoblotting. Surprisingly, hypermethylation in the GADD45B promoter was not detected in those tumors using bisulfite sequencing (Michaelis et al., 2011). The cause for GADD45B down-regulation in pituitary tumors remains to be determined.
The three GADD45 members share a high degree of protein homology. However, they are not functionally identical, differ in tissue distribution and may respond to different stress signals (Liebermann et al., 2011; Tamura et al., 2012). Nevertheless, evidence supports the concept that GADD45β and GADD45γ have tumor suppression functions (Liebermann et al., 2011; Tamura et al., 2012). Re-expression of GADD45β and GADD45γ inhibits proliferation of both human and rodent pituitary cell lines. We introduced GADD45γ into PDFS cells, derived from a human NFA, and observed a significant inhibition in cell proliferation (Zhang et al., 2002). Similar results were also observed in rat pituitary GH4 and mouse pituitary AtT20 cells (Zhang et al., 2002). Michaelis et al. (2011) re-expressed GADD45β in LβT2, which is a mouse gonadotroph tumor cell line, and repressed LβT2 xenograft tumor growth in nude mice. One mechanism whereby GADD45β and GADD45γ inhibit cell proliferation is by arresting the cell cycle at the G1/S or G2/M phase (Fan et al., 1999). G2/M arrest is due to inhibition of CDK1/cyclin B1 (Chung et al., 2003; Fan et al., 1999; Vairapandi et al., 2002). G1 arrest is likely mediated by up regulation of p21Cip1, inhibition of CDK2/cyclin E and induction of RB hypophosphorylation (Fan et al., 1999). Interaction with PCNA may also play a role in GADD45β and GADD45γ induced cell cycle arrest (Azam et al., 2001; Vairapandi et al., 1996). Therefore, these data indicate that GADD45B and GADD45G genes may suppress pituitary tumor growth by activation of the RB pathway.
3. The p53 connection
Tumor suppressor TP53 is a stress response gene which plays a central role in maintaining cellular homeostasis and tumor suppression (Vousden and Prives, 2009). Normally, p53 is maintained at a low level by MDM2 and MDM4. MDM2 functions as an E3 ubiquitin ligase and initiates ubiquitination of p53, thereby mediating its degradation. MDM4 does not possess enzyme activity, but assists MDM2 binding to p53. A variety of stress signals activate p53 by posttranslational modification and stabilization via blocking MDM2/4 mediated degradation (Kruse and Gu, 2009; Perry, 2010; Wade et al., 2010). P53 is a potent transcription factor, regulating expression of genes which mediate its tumor suppression function, such as cell cycle arrest, DNA repair, senescence, and apoptosis (Vousden and Prives, 2009). TP53 is one of the most commonly inactivated genes in human cancers (Olivier et al., 2010; Soussi, 2011). However, it is very rarely mutated in human pituitary adenomas (Herman et al., 1993; Levy et al., 1994; Tanizaki et al., 2007). To date, only one case of a confirmed TP53 somatic mutation has been identified in an atypical corticotroph adenoma (Kawashima et al., 2009). This raises the question of whether p53 is involved in human pituitary tumorigenesis. In mice, deletion of both Trp53 (the mouse p53 gene) alleles leads to development of a wide spectrum of tumors, which do not include pituitary tumors. However, Trp53 deletion dramatically accelerates pituitary tumor growth in Rb+/− mice. For example, Harvey et al. (1995) observed that the earliest age when pituitary tumors were observed in Rb+/− mice was 38 weeks and some tumors were not detected until week 71. In contrast, all p53−/−Rb+/− mice died of tumors by 25 weeks of age and 33% of them had pituitary tumors. Williams et al. (1994) reported that the mean age of survival for Rb+/− mice was 47 weeks and 100% of mice grew pituitary tumors. The mean age of survival for p53−/−Rb+/− mice was 17 weeks and 94% of them grew pituitary tumors. These results indicate that p53 indeed plays a role in tumor suppression in the pituitary. However, loss of p53 does not initiate pituitary tumor development. Instead, p53 plays a role in promoting pituitary tumorigenesis. It must be emphasized that p53 is regulated by and responds to a variety of stress signaling pathways, and that p53 function is mediated by its target genes. Therefore, p53 is as “functionally effective” as its regulators and target genes. Thus, for example, promoter methylation of p14ARF, which is well known to activate p53 by inhibition of MDM2, has been found in a small percentage (12%) of pituitary tumors in several studies (Bello et al., 2006; Kirsch et al., 2009; Yoshino et al., 2007). Significant down regulation of p14ARF expression was also observed in pituitary tumors by Western blotting (Michaelis et al., 2011). This indicates that the signaling to p53 mediated by p14ARF is interrupted in some pituitary tumors and suggests that inactivation of p53 signaling indeed contributes to human pituitary tumorigenesis. In addition to p14ARF, several genes whose expression are lost or significantly reduced have been identified in human pituitary adenomas (Table 1). We will discuss how they may contribute to tumor suppression via p53.
3.1. Maternally expressed gene 3 (MEG3)
The association between the MEG3 gene and human pituitary tumors was first established by comparing gene expression profiles between normal pituitary and NFAs using cDNA-RDA (Zhang et al., 2002). We identified MEG3 as one of the most under expressed genes in NFAs. In contrast, the level of MEG3 expression is very high in the normal human pituitary. No MEG3 RNA was detected in 11 human pituitary adenomas, including 9 NFAs and 3 GH-secreting adenomas by RT-PCR (Zhang et al., 2002). To confirm this finding, we and others have examined additional tumor samples for MEG3 expression. A total of 147 sporadic pituitary adenomas including 79 NFAs have been investigated (Cheunsuchon et al., 2011; Gejman et al., 2008; Mezzomo et al., 2011; Zhang et al., 2003; Zhao et al., 2005). Using conventional RT-PCR, only three out of 54 NFAs contained detectable MEG3 (Mezzomo et al., 2011). In contrast, MEG3 was detected in 35 out of 49 functioning tumors. These data were confirmed by qRT-PCR. The overall MEG3 expression in NFA (0.02 ± 0.02, n = 25, p = 0.006, t-test) is significantly lower compared to that in normal pituitary (1.0 ± 0.88, n = 10). In contrast, there is no significant difference in MEG3 expression between functioning adenomas (0.55 ± 0.47, n = 19, p = 0.16) and normal human pituitary tissue (Cheunsuchon et al., 2011). These data indicate that MEG3 is specifically lost in NFAs and its inactivation may play a significant role in the development of NFAs but not functioning adenomas.
MEG3, an imprinted gene, belongs to the DLK1-MEG3 imprinting locus located on 14q32 and consists of 10 exons. We first determined whether gene deletions are responsible for loss of MEG3 expression in NFAs (Zhao et al., 2005). Two micro satellite markers flanking exons 4 to 9 of the MEG3 gene were quantified using qPCR. No differences in both markers were found between NFAs and normal pituitary tissues, indicating that these tumors do not contain genomic deletions of the MEG3 gene. We next sequenced each individual exon of the MEG3 gene and did not find any small deletions or point mutations in these tumors (Zhao et al., 2005). These data exclude genetic mutations as a mechanism for MEG3 loss in NFAs. Expression of the MEG3 gene is regulated by two differentially methylated regions, IG-DMR and MEG3-DMR, which are methylated in the paternal but not the maternal allele (da Rocha et al., 2008). The IG-DMR is approximately 13 kb upstream of the MEG3 gene. The MEG3-DMR is ∼2 kb long overlapping with the MEG3 promoter and extends into intron 1. We determined the methylation status in both the IG-DMR and MEG3-DMR by bisulfite sequencing. We found that methylation was significantly increased in both the IG-DMR and MEG3-DMR in NFAs compared to normal pituitary tissue (Gejman et al., 2008; Zhao et al., 2005), suggesting that epigenetic changes in these two regulatory region are responsible for silencing of the MEG3 gene in NFAs. This mechanism is also supported by another finding. The MEG3 promoter contains a cAMP response element (CRE), which is required for expression of the MEG3 gene (Zhao et al., 2006). CRE sequences contain a CpG dinucleotide. Its methylation status directly determines CRE mediated transcription. For example, the CD86 gene promotor contains two CREs. No CD86 expression was detected in cells with methylated CREs. In contrast, CD86 was readily detected in cells with unmethylated CREs (Romero-Tlalolini et al., 2013). The mouse insulin promoter also contains a CRE. Kuroda et al. (2009) found that CRE methylation alone reduced promoter activity by 50%. In addition, chromatin immunoprecipitation analysis indicates that CpG methylation blocks binding of transcription factors to endogenous CREs (Kuroda et al., 2009; Romero-Tlalolini et al., 2013). These data suggest that CRE methylation likely plays a role in MEG3 gene down regulation.
Strong evidence suggests that MEG3 is novel tumor suppressor gene (Zhou et al., 2012). It encodes a long non-coding RNA (lncRNA) (Zhang et al., 2010; Zhou et al., 2007). In addition to NFAs, MEG3 expression is lost or significantly down regulated in a number of human cancers. Re-expression of MEG3 inhibits proliferation of several human cancer cell lines (Zhou et al., 2012). It is particularly interesting to note that p53 is the only known MEG3 target. MEG3 induces p53 accumulation, which is in part mediated by down regulation of MDM2, and activates expression of p53 target genes (Zhou et al., 2007). In mice, maternal deletion of the Meg3 gene results in perinatal death (Zhou et al., 2010). Furthermore, we reported that Meg3 inactivation stimulates expression of angiogenesis promoting genes and increases angiogenesis activity (Gordon et al., 2010), suggesting that MEG3 inhibits angiogenesis. Because p53 is a well known angiogenesis inhibitor (Teodoro et al., 2007), it is possible that MEG3 inhibits angiogenesis via activation of p53. Whitson et al. (2013) recently demonstrated that GDF15, a MEG3 activated p53 target gene (Zhou et al., 2007), suppresses angiogenesis (Whitson et al., 2013). MEG3 plays an important role in suppression of human NFAs, likely through activation of p53.
3.2. O-6-methylguanine-DNA methyltransferase (MGMT)
Genomic DNA can be alkylated by chemotherapeutic and environmental compounds at the O6-position of the DNA base guanine, which results in O6-alkylguanines (O6G), including O6-methylguanine (O6MeG), O(6)-[4-oxo-4-(3-pyridyl)butyl]guanine (O6pobG), and O6-chloroethylguanine (O6ClG) (Christmann et al., 2011). MGMT is the main repair enzyme and transfers the alkyl group to a cysteine residue in its active site (Pegg et al., 1995). In the absence of MGMT, the base pair O6G:C would become O6G:T after one round of replication, which causes a transition from G:C to A:T and results in a point mutation after further replication. Therefore, MGMT is the main defense mechanism against mutation driven carcinogenesis caused by O6-alkylating agents. Conversely, MGMT activity may be needed for tumor cell survival. In the absence of MGMT, O6-alkylguanine caused O6G:T mismatch activates DNA mismatch repair (MMR), leading to DNA double strain breaks, which in turn triggers apoptosis leading to cell death (Kaina et al., 2007). Therefore, it is not surprising to note the inconsistency of reported MGMT expression in human cancers. MGMT expression is high in some reports but inactivated in others (Christmann et al., 2011; Sharma et al., 2009). MGMT status may be tissue type and tumor stage specific.
There are very few studies reporting MGMT status in human pituitary tumors. McCormack et al. first reported MGMT was down regulated in a small percentage of pituitary tumors; only 13% of tumors (n = 88) had low MGMT expression. The tumor samples include all types of pituitary adenomas. No difference in MGMT expression levels was found among tumor types and degree of invasiveness (McCormack et al., 2009). In contrast, Fealey et al. (2010) reported that 78% of silent subtype 3 (SS3) pituitary adenomas (n = 23) have lost MGMT expression judged by immunohistochemical staining. Lau et al. (2010) examined MGMT immunoexpression of 30 pituitary carcinomas and 30 pituitary adenomas. Low MGMT expression was found in 57% of carcinomas and 50% of adenomas. Salehi et al. (2011) examined MGMT expression in 12 SS3 tumors and 10 pituitary carcinomas. They found that 11 of SS3 tumors stained negative or low (less than 10% cells positive) for MGMT protein. Five carcinomas exhibited low MGMT staining. Although the number of reports is small, inactivation of MGMT in pituitary adenomas, especially in aggressive tumors, appears to be a frequent event.
Promoter methylation was investigated in three studies. McCormack et al. (2009) reported that 9% of pituitary tumors had MGMT promoter methylation (n = 46). Bello et al. (2006) found the promoter methylation in 23% of tumors (n = 35). Salehi et al. (2011) reported that MGMT methylation was detected in 5 out of 12 aggressive SS3 pituitary adenomas. These results indicate that promoter methylation is not a major mechanism responsible for MGMT down regulation in human pituitary adenomas. There is strong evidence indicating that p53 positively regulates MGMT expression. Ionizing radiation up-regulates MGMT expression in mouse fibroblasts and rat hepatoma cell lines expressing wt p53, but fails to do so in those cells lacking wt p53 (Grombacher et al., 1998). In addition, inactivation of p53 by knockout techniques reduces MGMT expression by more than 90% in neonatal murine astrocytes (Blough et al., 2007). Knockdown of p53 expression by RNAi also reduces MGMT expression in SF767 human astrocytic glioma cells (Blough et al., 2007). These data indicate that p53 plays a crucial role in regulation of MGMT expression. Lost or low MGMT expression levels could be explained by the possibility that p53 signaling may have been compromised in human pituitary tumors.
In mouse models, high MGMT expression levels protect mice from alkylating agent induced cancers (Becker et al., 1997; Dumenco et al., 1993). Conversely, MGMT deficient mice are prone to alkylating agent induced tumor growth (Iwakuma et al., 1997). These studies suggest that MGMT is critical in suppressing tumor development caused by alkylating agent induced DNA mutations. Considering the fact that the loss of MGMT expression encourages DNA point mutations, it is likely that MGMT plays an important role in suppression of pituitary tumors.
3.3. Pleiomorphic adenoma gene-like 1 (PLAGL1)
PLAGL1, also known as ZAC, is an imprinted gene expressed only from the paternal allele (Kamiya et al., 2000). PLAGL1 is highly expressed in the normal anterior pituitary. However, its expression was either lost or significantly reduced in 59% of 41 tumors investigated by Pagotto et al. (1999). PLAGL1 down regulation was found in all NFAs (including null adenomas and gonadotropinomas, n = 22). In contrast, its down regulation was observed in only 11% of functioning tumors (n = 19) (Pagotto et al., 2000). Using RT-PCR, Michaelis et al. (2011) found PLAGL1 was readily detectable in normal pituitary tissue. In contrast, PLAGL1 was not detected in four of the six gonadotropinomas tested and the remaining two tumors had barely detectable PLAGL1. No LOH was detected in pituitary tumor tissues (Pagotto et al., 2000), indicating that a genetic mutation does not play a role in the loss of expression. It has been demonstrated that DNA methylation and histone deacetylation cause the loss of PLAGL1 expression in breast and ovarian cancers (Abdollahi et al., 1997, 2003; Kamikihara et al., 2005). Thus, epigenetic changes are most likely the mechanism responsible for the reduced expression of PLAGL1 in human NFAs.
There is evidence indicating that the PLAGL1 gene is a tumor suppressor. In addition to pituitary adenomas, PLAGL1 expression has been found to be lost or significantly reduced in tumors from many other tissues, such as ovary (Kamikihara et al., 2005), breast (Bilanges et al., 1999), adrenal gland (Jarmalaite et al., 2011), blood (Valleley et al., 2010), central nervous system (Lemeta et al., 2007), and skin (Basyuk et al., 2005). Re-expression of PLAGL1 induces cell cycle arrest and apoptosis and inhibits tumor growth in nude mice (Spengler et al., 1997). Inhibition of PLAGL1 by anti-sense treatment increase proliferation of pituitary tumor cells (Pagotto et al., 1999). In addition, induction of PLAGL1 plays a role in mediating growth inhibition by the somatostatin analog octreotide (Theodoropoulou et al., 2006). These studies suggest that PLAGL1 suppresses tumor growth by inhibition of cell proliferation.
PLAGL1 is a zinc-finger transcription factor with an affinity to GC rich DNA (Hoffmann et al., 2003). The GC-rich region was later identified to contain an Sp1 responsive element (Liu et al., 2011). PLAGL1 functions as a cofactor physically binding to several transcription factors and co-factors, including p53 (Huang et al., 2001), AP1 (Wang et al., 2011), Sp1 (Liu et al., 2011), histone acetyltransferase PCAF (Hoffmann and Spengler, 2008) and histone deacetylase 1 (HDAC1) (Liu et al., 2008). A likely mechanism by which PLAGL1 activates transcription of its target genes is through binding to GC rich or Sp1 responsive elements in the promoter where PLAGL1 interacts with p53, Sp1 or AP1 and recruits PCAF and p300 to the sites (Hoffmann and Spengler, 2008; Liu et al., 2011; Wang et al., 2011). PLAGL1 also interacts with HDAC1 to inhibit its histone deacetylation activity (Liu et al., 2008). Two target genes have been identified to mediate PLAGL1 induced growth inhibition: p21Cip1 (Hoffmann and Spengler, 2008; Huang et al., 2007; Liu et al., 2008, 2011; Wang et al., 2011) and PPARγ (Barz et al., 2006). As a p53 target gene, CDK inhibitor p21Cip1 is known to induce cell cycle arrest (Warfel and El-Deiry, 2013). Interestingly, p21Cip1 is also a target gene of PPARγ and has been shown to mediate PPARγ growth inhibition (Bonofiglio et al., 2008). These data indicate that p21Cip1 is a direct or indirect target of PLAGL1. In sum, PLAGL1 is lost or significantly reduced in NFAs. PLAGL1 inhibits cell proliferation which is likely mediated by up regulation of p21Cip1. Taken together, therefore, inactivation of PLAGL1 may be one of the mechanisms leading to pituitary tumor growth.
3.4. Ras association (RalGDS/AF-6) domain family member 1 (RASSF1)
RASSF1, also known as RASSF1A, belongs to a family of Ras effectors, which consist of a conserved motif, the RalGDS/AF6 Ras association (RA) domain (van der Weyden and Adams, 2007). There is strong evidence indicating that RASSF1 is a tumor suppressor (Donninger et al., 2007) and it is inactivated in many human cancers, mainly through promoter hypermethylation. It is involved in maintenance of genomic stability, and regulates cell cycle progression and apoptosis (Donninger et al., 2007). To determine RASSF1 status in human pituitary tumors, Qian et al. (2005) examined 52 pituitary adenomas for promoter methylation using MSP and RASSF1 expression using RT-PCR. They found RASSF1 promoter hypermethylation in 20 tumors (38%). RASSF1 expression was lost or significantly reduced in 22 tumors (42%), 18 of which have promoter hypermethylation (82%). There are no differences in promoter hypermethylation and RASSF1 expression between NFA and functioning adenomas (Qian et al., 2005). These data indicate that down regulation of RASSF1 is common in human pituitary tumors and that promoter hypermethylation plays a major role in silencing the RASSF1 gene.
There are several possible mechanisms whereby RASSF1 contributes to tumor suppression in the pituitary. RASSF1 controls mitosis progression by inhibition of APC-CDC20. Loss of RASSF1 causes cell division defects manifested by centrosome abnormalities and multipolar spindles (Song et al., 2004). RASSF1 also arrests cell cycle at G1 by inhibition of cyclin D1 accumulation (Shivakumar et al., 2002). Another possibility is that RASSF1 activates p53 by promoting MDM2 degradation, which results in cell cycle arrest (Song et al., 2008). In addition, RASSF1 induces cell cycle arrest by induction of p21Cip1 independent of p53 (Thaler et al., 2009). Finally, RASSF1 represses expression of ERα, inhibiting estrogen independent signaling (Thaler et al., 2012). These data indicate that RASSF1 may suppress tumor growth via multiple pathways.
3.5. Ras association (RalGDS/AF-6) domain family member 3 (RASSF3)
RASSF3, also known as RASSF5, NORE1, NORE1A and NORE1B, is another member of the RASSF family of Ras effectors. As with RASSF1, RASSF3 is inactivated in many human tumors (Djos et al., 2012; Geli et al., 2007, 2008; Lee et al., 2010; Macheiner et al., 2006). Recently, Peng et al. (2013) investigated DNA methylation in GH-secreting adenomas using HG18 CpG plus promoter microarray and found RASSF3 gene promoter methylation in all tumors examined (n = 27). In addition, RASSF3 expression was significantly reduced in all tumors compared to normal controls, suggesting that an epigenetic change is responsible for the gene silencing (Peng et al., 2013). There are several ways by which RASSF3 may suppress tumor growth. Similar to RASSF1, RASSF3 activates p53 by direct interaction with MDM2 and promotes its degradation via ubiquitination. Activation of p53 by RASSF3 induces cell cycle arrest and apoptosis (Kudo et al., 2012). RASSF3 also stimulates p21Cip1 expression by promoting p53 nuclear localization (Calvisi et al., 2009) and suppresses cell growth by ERK pathway inhibition (Moshnikova et al., 2006). Finally, RASSF3 mediates TNF-α induced apoptosis (Park et al., 2010). Therefore, RASSF3 may function as a tumor suppressor in the pituitary.
3.6. Suppressor of cytokine signaling 1 (SOCS1)
Buslei et al. (2006) reported that expression of SOCS1 is down regulated in the majority of pituitary adenomas due to promoter methylation, which was found in over 86% of NFAs. It occurred less frequently in ACTH- and GH-secreting tumors, 33% and 18%, respectively. Methylation was not found in prolactinomas. SOCS1 is a negative regulator of cytokine signaling. Cytokines regulate cell proliferation, survival and differentiation by phosphorylation of Janus kinases (JAK), which in turn phosphorylate signal transduction and activation of transcription (STAT) proteins. Phosphorylated STAT proteins dimerize and translocate to the nucleus to activate the target genes. SOCS1 negatively regulates the JAK/STAT signaling by binding to JAK and blocking STAT phosphorylation and is also a part of a ubiquitin ligase E3 complex, which targets proteins involved in cellular signaling, such as JAK and FAK, for ubiquitin mediated degradation (Mallette et al., 2010; Zhang et al., 2012). Therefore, SOCS1 is thought to function as a tumor suppressor due to its ability to blocking STAT activation (Zhang et al., 2012). Its down regulation in pituitary adenomas suggests that SOCS1 plays a role in pituitary tumor suppression by inhibition of JAK/STAT signaling.
In addition to JAK/STAT inhibition, SOCS1 may suppress tumor growth by activation of p53. Calabrese et al. (2009) showed that STAT5-induced senescence in normal cells is mediated by SOCS1 activation of p53. SOCS1 directly interacts and forms a complex with both p53 and DNA damage regulated kinases ATM/ATR, where p53 is stabilized by phosphorylation at serine 15 (Calabrese et al., 2009). These data indicate that activation of p53 by SOCS1 plays an important role in senescence induction, which is a well known tumor suppression mechanism.
4. Conclusion
Multiple genes are involved in the development of sporadic pituitary adenomas. Although this may reflect the heterogeneity of these tumors, it may also indicate that inactivation of multiple genes is necessary to transform pituitary cells. We have included the 20 genes in this review we felt were most pertinent to understanding pituitary tumorigenesis, particularly in sporadic tumors. However, we have not included several other genes whose expression is also down regulated in human pituitary adenomas, including cAMP-dependent protein kinase type I-alpha regulatory subunit (PRKAR1A) (Kirschner et al., 2000), fibroblast growth factor receptor 2 (FGFR2) (Zhu et al., 2007b), IKAROS family zinc finger 1 (Ikaros) (Zhu et al., 2007a), and lectin, galactoside-binding, soluble, 3 (LGALS3) (Ruebel et al., 2005). The PRKAR1A gene has been linked to CNC only. No mutation in this gene has been found in sporadic tumors. We did not discuss FGFR2, Ikaros and LGALS3 because they were reported in one study, respectively, with a limited number of tumors examined. More tumors need to be investigated before a link between these genes and sporadic pituitary adenomas can be established.
Overall, pathways leading to sporadic pituitary development can be generally grouped into two categories: one category is functionally connected to the RB pathway and the other to p53, although this categorization is not absolute (see Fig. 1). Data from studies with animal models, human pituitary tumors and in vitro cell line models suggest that inactivation of the RB pathway may play an initiation role in the development of sporadic pituitary adenomas; while inactivation of genes which function via the p53 pathway may play a promoting role. Pituitary adenomas are benign tumors. They very rarely develop into carcinomas. When dispersed into primary culture, pituitary tumor cells typically only can survive a few passages before they stop growing and die. All these observations indicate that certain components of the stress response system are still functional in benign pituitary tumors. Because p53 is the major stress response gene, these data suggest that the p53 pathway has been compromised but not completely abolished, consistent with the fact that p53 is rarely mutated in pituitary tumors. What signals cause down regulation of these tumor suppressive genes? What are the exact downstream target genes affected by inactivation of these genes? How do these genes cooperate to suppress pituitary tumors? Clearly there are many questions that need to be addressed before our understanding of the underlying pathogenesis of pituitary tumors may be used as the foundation to develop new therapies for patients with these tumors.
Fig. 1.

Schematic illustration of RB and p53 connections with tumor suppressive genes which are genetically or epigenetically inactivated in human sporadic pituitary adenomas. The upper panel includes genes functionally connected to the RB pathway. The lower panel includes genes functionally connected to p53. The cross talk between p53 and the RB pathway is mediated by p21Cip1. Several genes, including BMP-4, E-Cadherin, PLAGL1, RASSF1, and RASSF3 are functionally connected to both p53 and RB. Circles on pink background are components of the RB pathway. Circles on yellow background are components of the classic p53 pathway. Blue with names in red are genes down regulated in pituitary tumors. Green arrows, positively regulation; Red “T” lines, negatively regulation.
Acknowledgments
This work was supported by grants from the National Institutes of Health (A.K., R01DK40947), the Guthart Family Foundation, and the Jarislowsky Foundation.
Abbreviations
- AHR
aryl hydrocarbon receptor
- AIP
aryl hydrocarbon receptor interacting protein
- BMP-4
bone morphogenetic protein 4
- CDK
cyclin-dependent kinases
- CDKN1A
cyclin-dependent kinase inhibitor 1A
- CDKN1B
cyclin-dependent kinase inhibitor 1B
- CDKN2A
cyclin-dependent kinase inhibitor 2A
- CDKN2B
cyclin-dependent kinase inhibitor 2B
- CDKN2C
cyclin-dependent kinase inhibitor 2C
- CDH1
cadherin 1, type 1, E-cadherin (epithelial)
- CDH13
cadherin 13, H-cadherin (heart)
- CNC
Carney complex
- DNMT1
DNA methlytransferase 1
- DNMT3A
DNA (cytosine-5-)-methyltransferase 3 alpha
- DNMT3B
DNA (cytosine-5-)-methyltransferase 3 beta
- DNMT3L
DNA (cytosine-5-)-methyltransferase 3-like
- EMT
epithelial–mesenchymal transition
- ESRP1
epithelial splicing regulatory protein 1
- FPA
familial pituitary adenoma
- FIPA
familial isolated pituitary adenoma
- GADD45B
growth arrest and DNA-damage-inducible, beta
- GADD45G
growth arrest and DNA-damage-inducible, gamma
- GNAS
guanine nucleotide-binding protein G(s) subunit alpha
- HDAC3
histone deacetylase 3
- MEG3
maternal expression gene 3
- MEN1
multiple endocrine neoplasia type I
- MGMT
O-6-methylguanine-DNA methyltransferase
- MLL
myeloid/lymphoid or mixed-lineage leukemia protein 1
- NFA
clinically non-functioning pituitary adenoma
- PLAGL1
pleiomorphic adenoma gene-like 1
- PRKAR1A
protein kinase, cAMP-dependent, regulatory, type I, alpha
- RASSF1
Ras association (RalGDS/AF-6) domain family member 1
- RASSF3
Ras association (RalGDS/AF-6) domain family member 3
- SOCS1
suppressor of cytokine signaling 1
- TCDD
2,3,7,8-tetrachloro-p-dioxin
References
- Abd El-Moneim HM, Abd El-Rehim D. Immunohistochemical and molecular study of p16INK4A expression in pituitary adenoma. J Egypt Natl Canc Inst. 2009;21:351–360. [PubMed] [Google Scholar]
- Abdollahi A, Godwin AK, Miller PD, Getts LA, Schultz DC, Taguchi T, Testa JR, Hamilton TC. Identification of a gene containing zinc-finger motifs based on lost expression in malignantly transformed rat ovarian surface epithelial cells. Cancer Res. 1997;57:2029–2034. [PubMed] [Google Scholar]
- Abdollahi A, Pisarcik D, Roberts D, Weinstein J, Cairns P, Hamilton TC. LOT1 (PLAGL1/ZAC1), the candidate tumor suppressor gene at chromosome 6q24-25, is epigenetically regulated in cancer. J Biol Chem. 2003;278:6041–6049. doi: 10.1074/jbc.M210361200. [DOI] [PubMed] [Google Scholar]
- Alexander JM, Biller BM, Bikkal H, Zervas NT, Arnold A, Klibanski A. Clinically nonfunctioning pituitary tumors are monoclonal in origin. J Clin Invest. 1990;86:336–340. doi: 10.1172/JCI114705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Andreeva AV, Kutuzov MA. Cadherin 13 in cancer. Genes Chromosom Cancer. 2010;49:775–790. doi: 10.1002/gcc.20787. [DOI] [PubMed] [Google Scholar]
- Angst BD, Marcozzi C, Magee AI. The cadherin superfamily: diversity in form and function. J Cell Sci. 2001;114:629–641. doi: 10.1242/jcs.114.4.629. [DOI] [PubMed] [Google Scholar]
- Arima Y, Inoue Y, Shibata T, Hayashi H, Nagano O, Saya H, Taya Y. Rb depletion results in deregulation of E-cadherin and induction of cellular phenotypic changes that are characteristic of the epithelial-to-mesenchymal transition. Cancer Res. 2008;68:5104–5112. doi: 10.1158/0008-5472.CAN-07-5680. [DOI] [PubMed] [Google Scholar]
- Asa SL, Somers K, Ezzat S. The MEN-1 gene is rarely down-regulated in pituitary adenomas. J Clin Endocrinol Metab. 1998;83:3210–3212. doi: 10.1210/jcem.83.9.5138. [DOI] [PubMed] [Google Scholar]
- Azam N, Vairapandi M, Zhang W, Hoffman B, Liebermann DA. Interaction of CR6 (GADD45gamma) with proliferating cell nuclear antigen impedes negative growth control. J Biol Chem. 2001;276:2766–2774. doi: 10.1074/jbc.M005626200. [DOI] [PubMed] [Google Scholar]
- Bahar A, Bicknell JE, Simpson DJ, Clayton RN, Farrell WE. Loss of expression of the growth inhibitory gene GADD45gamma, in human pituitary adenomas, is associated with CpG island methylation. Oncogene. 2004;23:936–944. doi: 10.1038/sj.onc.1207193. [DOI] [PubMed] [Google Scholar]
- Bamberger CM, Fehn M, Bamberger AM, Ludecke DK, Beil FU, Saeger W, Schulte HM. Reduced expression levels of the cell-cycle inhibitor p27Kip1 in human pituitary adenomas. Eur J Endocrinol. 1999;140:250–255. doi: 10.1530/eje.0.1400250. [DOI] [PubMed] [Google Scholar]
- Barhoover MA, Hall JM, Greenlee WF, Thomas RS. Aryl hydrocarbon receptor regulates cell cycle progression in human breast cancer cells via a functional interaction with cyclin-dependent kinase 4. Mol Pharmacol. 2010;77:195–201. doi: 10.1124/mol.109.059675. [DOI] [PubMed] [Google Scholar]
- Barlier A, Vanbellinghen JF, Daly AF, Silvy M, Jaffrain-Rea ML, Trouillas J, Tamagno G, Cazabat L, Bours V, Brue T, Enjalbert A, Beckers A. Mutations in the aryl hydrocarbon receptor interacting protein gene are not highly prevalent among subjects with sporadic pituitary adenomas. J Clin Endocrinol Metab. 2007;92:1952–1955. doi: 10.1210/jc.2006-2702. [DOI] [PubMed] [Google Scholar]
- Barz T, Hoffmann A, Panhuysen M, Spengler D. Peroxisome proliferators-activated receptor gamma is a Zac target gene mediating Zac antiproliferation. Cancer Res. 2006;66:11975–11982. doi: 10.1158/0008-5472.CAN-06-1529. [DOI] [PubMed] [Google Scholar]
- Basyuk E, Coulon V, Le Digarcher A, Coisy-Quivy M, Moles JP, Gandarillas A, Journot L. The candidate tumor suppressor gene ZAC is involved in keratinocyte differentiation and its expression is lost in basal cell carcinomas. Mol Cancer Res. 2005;3:483–492. doi: 10.1158/1541-7786.MCR-05-0019. [DOI] [PubMed] [Google Scholar]
- Batsche E, Muchardt C, Behrens J, Hurst HC, Cremisi C. RB and c-Myc activate expression of the E-cadherin gene in epithelial cells through interaction with transcription factor AP-2. Mol Cell Biol. 1998;18:3647–3658. doi: 10.1128/mcb.18.7.3647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bazzi W, Renon M, Vercherat C, Hamze Z, Lacheretz-Bernigaud A, Wang H, Blanc M, Roche C, Calender A, Chayvialle JA, Scoazec JY, Cordier-Bussat M. MEN1 missense mutations impair sensitization to apoptosis induced by wild-type menin in endocrine pancreatic tumor cells. Gastroenterology. 2008;135:1698–1709. doi: 10.1053/j.gastro.2008.07.031. e1692. [DOI] [PubMed] [Google Scholar]
- Becker K, Gregel CM, Kaina B. The DNA repair protein O6-methylguanine-DNA methyltransferase protects against skin tumor formation induced by antineoplastic chloroethylnitrosourea. Cancer Res. 1997;57:3335–3338. [PubMed] [Google Scholar]
- Beckers A, Aaltonen LA, Daly AF, Karhu A. Familial isolated pituitary adenomas (FIPA) and the pituitary adenoma predisposition due to mutations in the aryl hydrocarbon receptor interacting protein (AIP) gene. Endocr Rev. 2013;34:239–277. doi: 10.1210/er.2012-1013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bello MJ, De Campos JM, Isla A, Casartelli C, Rey JA. Promoter CpG methylation of multiple genes in pituitary adenomas: frequent involvement of caspase-8. Oncol Rep. 2006;15:443–448. [PubMed] [Google Scholar]
- Bergman L, Boothroyd C, Palmer J, Grimmond S, Walters M, Teh B, Shepherd J, Hartley L, Hayward N. Identification of somatic mutations of the MEN1 gene in sporadic endocrine tumours. Br J Cancer. 2000;83:1003–1008. doi: 10.1054/bjoc.2000.1385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bernstein BE, Mikkelsen TS, Xie X, Kamal M, Huebert DJ, Cuff J, Fry B, Meissner A, Wernig M, Plath K, Jaenisch R, Wagschal A, Feil R, Schreiber SL, Lander ES. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell. 2006;125:315–326. doi: 10.1016/j.cell.2006.02.041. [DOI] [PubMed] [Google Scholar]
- Bertolino P, Radovanovic I, Casse H, Aguzzi A, Wang ZQ, Zhang CX. Genetic ablation of the tumor suppressor menin causes lethality at midgestation with defects in multiple organs. Mech Dev. 2003a;120:549–560. doi: 10.1016/s0925-4773(03)00039-x. [DOI] [PubMed] [Google Scholar]
- Bertolino P, Tong WM, Galendo D, Wang ZQ, Zhang CX. Heterozygous Men1 mutant mice develop a range of endocrine tumors mimicking multiple endocrine neoplasia type 1. Mol Endocrinol. 2003b;17:1880–1892. doi: 10.1210/me.2003-0154. [DOI] [PubMed] [Google Scholar]
- Berx G, van Roy F. Involvement of members of the cadherin superfamily in cancer. Cold Spring Harb Perspect Biol. 2009;1:a003129. doi: 10.1101/cshperspect.a003129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bilanges B, Varrault A, Basyuk E, Rodriguez C, Mazumdar A, Pantaloni C, Bockaert J, Theillet C, Spengler D, Journot L. Loss of expression of the candidate tumor suppressor gene ZAC in breast cancer cell lines and primary tumors. Oncogene. 1999;18:3979–3988. doi: 10.1038/sj.onc.1202933. [DOI] [PubMed] [Google Scholar]
- Biller BM, Alexander JM, Zervas NT, Hedley-Whyte ET, Arnold A, Klibanski A. Clonal origins of adrenocorticotropin-secreting pituitary tissue in Cushing's disease. J Clin Endocrinol Metab. 1992;75:1303–1309. doi: 10.1210/jcem.75.5.1358909. [DOI] [PubMed] [Google Scholar]
- Biondi C, Gartside M, Tonks I, Paterson C, Hayward NK, Kay GF. Targeting and conditional inactivation of the murine Men1 locus using the Cre recombinase: loxP system. Genesis. 2002;32:150–151. doi: 10.1002/gene.10061. [DOI] [PubMed] [Google Scholar]
- Blough MD, Zlatescu MC, Cairncross JG. O6-methylguanine-DNA methyltransferase regulation by p53 in astrocytic cells. Cancer Res. 2007;67:580–584. doi: 10.1158/0008-5472.CAN-06-2782. [DOI] [PubMed] [Google Scholar]
- Bolos V, Peinado H, Perez-Moreno MA, Fraga MF, Esteller M, Cano A. The transcription factor Slug represses E-cadherin expression and induces epithelial to mesenchymal transitions: a comparison with Snail and E47 repressors. J Cell Sci. 2003;116:499–511. doi: 10.1242/jcs.00224. [DOI] [PubMed] [Google Scholar]
- Bonofiglio D, Qi H, Gabriele S, Catalano S, Aquila S, Belmonte M, Ando S. Peroxisome proliferator-activated receptor gamma inhibits follicular and anaplastic thyroid carcinoma cells growth by upregulating p21Cip1/WAF1 gene in a Sp1-dependent manner. Endocr Relat Cancer. 2008;15:545–557. doi: 10.1677/ERC-07-0272. [DOI] [PubMed] [Google Scholar]
- Brandi ML, Gagel RF, Angeli A, Bilezikian JP, Beck-Peccoz P, Bordi C, Conte-Devolx B, Falchetti A, Gheri RG, Libroia A, Lips CJ, Lombardi G, Mannelli M, Pacini F, Ponder BA, Raue F, Skogseid B, Tamburrano G, Thakker RV, Thompson NW, Tomassetti P, Tonelli F, Wells SA, Jr, Marx SJ. Guidelines for diagnosis and therapy of MEN type 1 and type 2. J Clin Endocrinol Metab. 2001;86:5658–5671. doi: 10.1210/jcem.86.12.8070. [DOI] [PubMed] [Google Scholar]
- Brubaker KD, Corey E, Brown LG, Vessella RL. Bone morphogenetic protein signaling in prostate cancer cell lines. J Cell Biochem. 2004;91:151–160. doi: 10.1002/jcb.10679. [DOI] [PubMed] [Google Scholar]
- Buchbinder S, Bierhaus A, Zorn M, Nawroth PP, Humpert P, Schilling T. Aryl hydrocarbon receptor interacting protein gene (AIP) mutations are rare in patients with hormone secreting or non-secreting pituitary adenomas. Exp Clin Endocrinol Diabetes. 2008;116:625–628. doi: 10.1055/s-2008-1065366. [DOI] [PubMed] [Google Scholar]
- Buslei R, Kreutzer J, Hofmann B, Schmidt V, Siebzehnrubl F, Hahnen E, Eyupoglu IY, Fahlbusch R, Blumcke I. Abundant hypermethylation of SOCS-1 in clinically silent pituitary adenomas. Acta Neuropathol. 2006;111:264–271. doi: 10.1007/s00401-005-0009-9. [DOI] [PubMed] [Google Scholar]
- Bystrom C, Larsson C, Blomberg C, Sandelin K, Falkmer U, Skogseid B, Oberg K, Werner S, Nordenskjold M. Localization of the MEN1 gene to a small region within chromosome 11q13 by deletion mapping in tumors. Proc Natl Acad Sci USA. 1990;87:1968–1972. doi: 10.1073/pnas.87.5.1968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cai W, Kramarova TV, Berg P, Korbonits M, Pongratz I. The immunophilin-like protein XAP2 is a negative regulator of estrogen signaling through interaction with estrogen receptor alpha. PLoS One. 2011;6:e25201. doi: 10.1371/journal.pone.0025201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Calabrese V, Mallette FA, Deschenes-Simard X, Ramanathan S, Gagnon J, Moores A, Ilangumaran S, Ferbeyre G. SOCS1 links cytokine signaling to p53 and senescence. Mol Cell. 2009;36:754–767. doi: 10.1016/j.molcel.2009.09.044. [DOI] [PubMed] [Google Scholar]
- Calvisi DF, Donninger H, Vos MD, Birrer MJ, Gordon L, Leaner V, Clark GJ. NORE1A tumor suppressor candidate modulates p21CIP1 via p53. Cancer Res. 2009;69:4629–4637. doi: 10.1158/0008-5472.CAN-08-3672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Canaff L, Vanbellinghen JF, Kaji H, Goltzman D, Hendy GN. Impaired transforming growth factor-beta (TGF-beta) transcriptional activity and cell proliferation control of a menin in-frame deletion mutant associated with multiple endocrine neoplasia type 1 (MEN1) J Biol Chem. 2012;287:8584–8597. doi: 10.1074/jbc.M112.341958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cao R, Wang L, Wang H, Xia L, Erdjument-Bromage H, Tempst P, Jones RS, Zhang Y. Role of histone H3 lysine 27 methylation in Polycomb-group silencing. Science. 2002;298:1039–1043. doi: 10.1126/science.1076997. [DOI] [PubMed] [Google Scholar]
- Carver LA, Bradfield CA. Ligand-dependent interaction of the aryl hydrocarbon receptor with a novel immunophilin homolog in vivo. J Biol Chem. 1997;272:11452–11456. doi: 10.1074/jbc.272.17.11452. [DOI] [PubMed] [Google Scholar]
- Cazabat L, Bouligand J, Salenave S, Bernier M, Gaillard S, Parker F, Young J, Guiochon-Mantel A, Chanson P. Germline AIP mutations in apparently sporadic pituitary adenomas: prevalence in a prospective single-center cohort of 443 patients. J Clin Endocrinol Metab. 2012;97:E663–670. doi: 10.1210/jc.2011-2291. [DOI] [PubMed] [Google Scholar]
- Chaidarun SS, Klibanski A. Gonadotropinomas. Semin Reprod Med. 2002;20:339–348. doi: 10.1055/s-2002-36708. [DOI] [PubMed] [Google Scholar]
- Chan DW, Lee JM, Chan PC, Ng IO. Genetic and epigenetic inactivation of T-cadherin in human hepatocellular carcinoma cells. Int J Cancer. 2008;123:1043–1052. doi: 10.1002/ijc.23634. [DOI] [PubMed] [Google Scholar]
- Chandrasekharappa SC, Guru SC, Manickam P, Olufemi SE, Collins FS, Emmert-Buck MR, Debelenko LV, Zhuang Z, Lubensky IA, Liotta LA, Crabtree JS, Wang Y, Roe BA, Weisemann J, Boguski MS, Agarwal SK, Kester MB, Kim YS, Heppner C, Dong Q, Spiegel AM, Burns AL, Marx SJ. Positional cloning of the gene for multiple endocrine neoplasia-type 1. Science. 1997;276:404–407. doi: 10.1126/science.276.5311.404. [DOI] [PubMed] [Google Scholar]
- Cheng JC, Auersperg N, Leung PC. Inhibition of p53 represses E-cadherin expression by increasing DNA methyltransferase-1 and promoter methylation in serous borderline ovarian tumor cells. Oncogene. 2011;30:3930–3942. doi: 10.1038/onc.2011.117. [DOI] [PubMed] [Google Scholar]
- Cheng YH, Li LA, Lin P, Cheng LC, Hung CH, Chang NW, Lin C. Baicalein induces G1 arrest in oral cancer cells by enhancing the degradation of cyclin D1 and activating AhR to decrease Rb phosphorylation. Toxicol Appl Pharmacol. 2012;263:360–367. doi: 10.1016/j.taap.2012.07.010. [DOI] [PubMed] [Google Scholar]
- Cheunsuchon P, Zhou Y, Zhang X, Lee H, Chen W, Nakayama Y, Rice KA, Tessa Hedley-Whyte E, Swearingen B, Klibanski A. Silencing of the imprinted DLK1-MEG3 locus in human clinically nonfunctioning pituitary adenomas. Am J Pathol. 2011;179:2120–2130. doi: 10.1016/j.ajpath.2011.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Christmann M, Verbeek B, Roos WP, Kaina B. O(6)-Methylguanine-DNA methyltransferase (MGMT) in normal tissues and tumors: enzyme activity, promoter methylation and immunohistochemistry. Biochim Biophys Acta. 2011;1816:179–190. doi: 10.1016/j.bbcan.2011.06.002. [DOI] [PubMed] [Google Scholar]
- Chung HK, Yi YW, Jung NC, Kim D, Suh JM, Kim H, Park KC, Song JH, Kim DW, Hwang ES, Yoon SH, Bae YS, Kim JM, Bae I, Shong M. CR6-interacting factor 1 interacts with Gadd45 family proteins and modulates the cell cycle. J Biol Chem. 2003;278:28079–28088. doi: 10.1074/jbc.M212835200. [DOI] [PubMed] [Google Scholar]
- Courtois-Cox S, Jones SL, Cichowski K. Many roads lead to oncogeneinduced senescence. Oncogene. 2008;27:2801–2809. doi: 10.1038/sj.onc.1210950. [DOI] [PubMed] [Google Scholar]
- Crabtree JS, Scacheri PC, Ward JM, Garrett-Beal L, Emmert-Buck MR, Edgemon KA, Lorang D, Libutti SK, Chandrasekharappa SC, Marx SJ, Spiegel AM, Collins FS. A mouse model of multiple endocrine neoplasia, type 1, develops multiple endocrine tumors. Proc Natl Acad Sci USA. 2001;98:1118–1123. doi: 10.1073/pnas.98.3.1118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cryns VL, Alexander JM, Klibanski A, Arnold A. The retinoblastoma gene in human pituitary tumors. J Clin Endocrinol Metab. 1993;77:644–646. doi: 10.1210/jcem.77.3.7690360. [DOI] [PubMed] [Google Scholar]
- Cuny T, Pertuit M, Sahnoun-Fathallah M, Daly A, Occhi G, Odou MF, Tabarin A, Nunes ML, Delemer B, Rohmer V, Desailloud R, Kerlan V, Chabre O, Sadoul JL, Cogne M, Caron P, Cortet-Rudelli C, Lienhardt A, Raingeard I, Guedj AM, Brue T, Beckers A, Weryha G, Enjalbert A, Barlier A. Genetic analysis in young patients with sporadic pituitary macroadenomas: besides AIP don't forget MEN1 genetic analysis. Eur J Endocrinol. 2013;168:533–541. doi: 10.1530/EJE-12-0763. [DOI] [PubMed] [Google Scholar]
- da Rocha ST, Edwards CA, Ito M, Ogata T, Ferguson-Smith AC. Genomic imprinting at the mammalian Dlk1-Dio3 domain. Trends Genet. 2008;24:306–316. doi: 10.1016/j.tig.2008.03.011. [DOI] [PubMed] [Google Scholar]
- Dahia PL, Aguiar RC, Honegger J, Fahlbush R, Jordan S, Lowe DG, Lu X, Clayton RN, Besser GM, Grossman AB. Mutation and expression analysis of the p27/kip1 gene in corticotrophin-secreting tumours. Oncogene. 1998;16:69–76. doi: 10.1038/sj.onc.1201516. [DOI] [PubMed] [Google Scholar]
- Daly AF, Vanbellinghen JF, Khoo SK, Jaffrain-Rea ML, Naves LA, Guitelman MA, Murat A, Emy P, Gimenez-Roqueplo AP, Tamburrano G, Raverot G, Barlier A, De Herder W, Penfornis A, Ciccarelli E, Estour B, Lecomte P, Gatta B, Chabre O, Sabate MI, Bertagna X, Garcia Basavilbaso N, Stalldecker G, Colao A, Ferolla P, Wemeau JL, Caron P, Sadoul JL, Oneto A, Archambeaud F, Calender A, Sinilnikova O, Montanana CF, Cavagnini F, Hana V, Solano A, Delettieres D, Luccio-Camelo DC, Basso A, Rohmer V, Brue T, Bours V, Teh BT, Beckers A. Aryl hydrocarbon receptor-interacting protein gene mutations in familial isolated pituitary adenomas: analysis in 73 families. J Clin Endocrinol Metab. 2007;92:1891–1896. doi: 10.1210/jc.2006-2513. [DOI] [PubMed] [Google Scholar]
- Dawson MA, Kouzarides T. Cancer epigenetics: from mechanism to therapy. Cell. 2012;150:12–27. doi: 10.1016/j.cell.2012.06.013. [DOI] [PubMed] [Google Scholar]
- De Craene B, Berx G. Regulatory networks defining EMT during cancer initiation and progression. Nat Rev Cancer. 2013;13:97–110. doi: 10.1038/nrc3447. [DOI] [PubMed] [Google Scholar]
- Debelenko LV, Zhuang Z, Emmert-Buck MR, Chandrasekharappa SC, Manickam P, Guru SC, Marx SJ, Skarulis MC, Spiegel AM, Collins FS, Jensen RT, Liotta LA, Lubensky IA. Allelic deletions on chromosome 11q13 in multiple endocrine neoplasia type 1-associated and sporadic gastrinomas and pancreatic endocrine tumors. Cancer Res. 1997;57:2238–2243. [PubMed] [Google Scholar]
- Denison MS, Nagy SR. Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Annu Rev Pharmacol Toxicol. 2003;43:309–334. doi: 10.1146/annurev.pharmtox.43.100901.135828. [DOI] [PubMed] [Google Scholar]
- Denison MS, Soshilov AA, He G, DeGroot DE, Zhao B. Exactly the same but different: promiscuity and diversity in the molecular mechanisms of action of the aryl hydrocarbon (dioxin) receptor. Toxicol Sci. 2011;124:1–22. doi: 10.1093/toxsci/kfr218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dever DP, Opanashuk LA. The aryl hydrocarbon receptor contributes to the proliferation of human medulloblastoma cells. Mol Pharmacol. 2012;81:669–678. doi: 10.1124/mol.111.077305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Di Fiore R, D'Anneo A, Tesoriere G, Vento R. RB1 in cancer: different mechanisms of RB1 inactivation and alterations of pRb pathway in tumorigenesis. J Cell Physiol. 2013;228:1676–1687. doi: 10.1002/jcp.24329. [DOI] [PubMed] [Google Scholar]
- Dick FA, Rubin SM. Molecular mechanisms underlying RB protein function. Nat Rev Mol Cell Biol. 2013;14:297–306. doi: 10.1038/nrm3567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DiGiovanni R, Serra S, Ezzat S, Asa SL. AIP Mutations are not identified in patients with sporadic pituitary adenomas. Endocr Pathol. 2007;18:76–78. doi: 10.1007/s12022-007-0010-z. [DOI] [PubMed] [Google Scholar]
- Dittmar KA, Jiang P, Park JW, Amirikian K, Wan J, Shen S, Xing Y, Carstens RP. Genome-wide determination of a broad ESRP-regulated posttranscriptional network by high-throughput sequencing. Mol Cell Biol. 2012;32:1468–1482. doi: 10.1128/MCB.06536-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Djos A, Martinsson T, Kogner P, Caren H. The RASSF gene family members RASSF5, RASSF6 and RASSF7 show frequent DNA methylation in neuroblastoma. Mol Cancer. 2012;11:40. doi: 10.1186/1476-4598-11-40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Donninger H, Vos MD, Clark GJ. The RASSF1A tumor suppressor. J Cell Sci. 2007;120:3163–3172. doi: 10.1242/jcs.010389. [DOI] [PubMed] [Google Scholar]
- Dumenco LL, Allay E, Norton K, Gerson SL. The prevention of thymic lymphomas in transgenic mice by human O6-alkylguanine-DNA alkyltransferase. Science. 1993;259:219–222. doi: 10.1126/science.8421782. [DOI] [PubMed] [Google Scholar]
- Elferink CJ, Ge NL, Levine A. Maximal aryl hydrocarbon receptor activity depends on an interaction with the retinoblastoma protein. Mol Pharmacol. 2001;59:664–673. doi: 10.1124/mol.59.4.664. [DOI] [PubMed] [Google Scholar]
- Elston MS, Gill AJ, Conaglen JV, Clarkson A, Cook RJ, Little NS, Robinson BG, Clifton-Bligh RJ, McDonald KL. Nuclear accumulation of e-cadherin correlates with loss of cytoplasmic membrane staining and invasion in pituitary adenomas. J Clin Endocrinol Metab. 2009;94:1436–1442. doi: 10.1210/jc.2008-2075. [DOI] [PubMed] [Google Scholar]
- Evang JA, Berg JP, Casar-Borota O, Lekva T, Kringen MK, Ramm-Pettersen J, Bollerslev J. Reduced levels of E-cadherin correlate with progression of corticotroph pituitary tumours. Clin Endocrinol (Oxf) 2011;75:811–818. doi: 10.1111/j.1365-2265.2011.04109.x. [DOI] [PubMed] [Google Scholar]
- Ezzat S, Asa SL, Couldwell WT, Barr CE, Dodge WE, Vance ML, McCutcheon IE. The prevalence of pituitary adenomas: a systematic review. Cancer. 2004;101:613–619. doi: 10.1002/cncr.20412. [DOI] [PubMed] [Google Scholar]
- Fan W, Richter G, Cereseto A, Beadling C, Smith KA. Cytokine response gene 6 induces p21 and regulates both cell growth and arrest. Oncogene. 1999;18:6573–6582. doi: 10.1038/sj.onc.1203054. [DOI] [PubMed] [Google Scholar]
- Farrell WE, Simpson DJ, Bicknell J, Magnay JL, Kyrodimou E, Thakker RV, Clayton RN. Sequence analysis and transcript expression of the MEN1 gene in sporadic pituitary tumours. Br J Cancer. 1999;80:44–50. doi: 10.1038/sj.bjc.6690319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fealey ME, Scheithauer BW, Horvath E, Erickson D, Kovacs K, McLendon R, Lloyd RV. MGMT immunoexpression in silent subtype 3 pituitary adenomas: possible therapeutic implications. Endocr Pathol. 2010;21:161–165. doi: 10.1007/s12022-010-9120-0. [DOI] [PubMed] [Google Scholar]
- Fero ML, Rivkin M, Tasch M, Porter P, Carow CE, Firpo E, Polyak K, Tsai LH, Broudy V, Perlmutter RM, Kaushansky K, Roberts JM. A syndrome of multiorgan hyperplasia with features of gigantism, tumorigenesis, and female sterility in p27(Kip1)-deficient mice. Cell. 1996;85:733–744. doi: 10.1016/s0092-8674(00)81239-8. [DOI] [PubMed] [Google Scholar]
- Fougner SL, Lekva T, Borota OC, Hald JK, Bollerslev J, Berg JP. The expression of E-cadherin in somatotroph pituitary adenomas is related to tumor size, invasiveness, and somatostatin analog response. J Clin Endocrinol Metab. 2010;95:2334–2342. doi: 10.1210/jc.2009-2197. [DOI] [PubMed] [Google Scholar]
- Franklin DS, Godfrey VL, Lee H, Kovalev GI, Schoonhoven R, Chen-Kiang S, Su L, Xiong Y. CDK inhibitors p18(INK4c) and p27(Kip1) mediate two separate pathways to collaboratively suppress pituitary tumorigenesis. Genes Dev. 1998;12:2899–2911. doi: 10.1101/gad.12.18.2899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Freda PU, Chung WK, Matsuoka N, Walsh JE, Kanibir MN, Kleinman G, Wang Y, Bruce JN, Post KD. Analysis of GNAS mutations in 60 growth hormone secreting pituitary tumors: correlation with clinical and pathological characteristics and surgical outcome based on highly sensitive GH and IGF-I criteria for remission. Pituitary. 2007;10:275–282. doi: 10.1007/s11102-007-0058-2. [DOI] [PubMed] [Google Scholar]
- Friedman E, Sakaguchi K, Bale AE, Falchetti A, Streeten E, Zimering MB, Weinstein LS, McBride WO, Nakamura Y, Brandi ML, et al. Clonality of parathyroid tumors in familial multiple endocrine neoplasia type 1. N Engl J Med. 1989;321:213–218. doi: 10.1056/NEJM198907273210402. [DOI] [PubMed] [Google Scholar]
- Fukino K, Kitamura Y, Sanno N, Teramoto A, Emi M. Analysis of the MEN1 gene in sporadic pituitary adenomas from Japanese patients. Cancer Lett. 1999;144:85–92. doi: 10.1016/s0304-3835(99)00201-3. [DOI] [PubMed] [Google Scholar]
- Fukuda N, Saitoh M, Kobayashi N, Miyazono K. Execution of BMP-4-induced apoptosis by p53-dependent ER dysfunction in myeloma and B-cell hybridoma cells. Oncogene. 2006;25:3509–3517. doi: 10.1038/sj.onc.1209393. [DOI] [PubMed] [Google Scholar]
- Gao SB, Feng ZJ, Xu B, Wu Y, Yin P, Yang Y, Hua X, Jin GH. Suppression of lung adenocarcinoma through menin and polycomb gene-mediated repression of growth factor pleiotrophin. Oncogene. 2009;28:4095–4104. doi: 10.1038/onc.2009.273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gejman R, Batista DL, Zhong Y, Zhou Y, Zhang X, Swearingen B, Stratakis CA, Hedley-Whyte ET, Klibanski A. Selective loss of MEG3 expression and intergenic differentially methylated region hypermethylation in the MEG3/DLK1 locus in human clinically nonfunctioning pituitary adenomas. J Clin Endocrinol Metab. 2008;93:4119–4125. doi: 10.1210/jc.2007-2633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Geli J, Kiss N, Lanner F, Foukakis T, Natalishvili N, Larsson O, Kogner P, Hoog A, Clark GJ, Ekstrom TJ, Backdahl M, Farnebo F, Larsson C. The Ras effectors NORE1A and RASSF1A are frequently inactivated in pheochromocytoma and abdominal paraganglioma. Endocr Relat Cancer. 2007;14:125–134. doi: 10.1677/ERC-06-0031. [DOI] [PubMed] [Google Scholar]
- Geli J, Kogner P, Lanner F, Natalishvili N, Juhlin C, Kiss N, Clark GJ, Ekstrom TJ, Farnebo F, Larsson C. Assessment of NORE1A as a putative tumor suppressor in human neuroblastoma. Int J Cancer. 2008;123:389–394. doi: 10.1002/ijc.23533. [DOI] [PubMed] [Google Scholar]
- Georgitsi M, Raitila A, Karhu A, Tuppurainen K, Makinen MJ, Vierimaa O, Paschke R, Saeger W, van der Luijt RB, Sane T, Robledo M, De Menis E, Weil RJ, Wasik A, Zielinski G, Lucewicz O, Lubinski J, Launonen V, Vahteristo P, Aaltonen LA. Molecular diagnosis of pituitary adenoma predisposition caused by aryl hydrocarbon receptor-interacting protein gene mutations. Proc Natl Acad Sci USA. 2007;104:4101–4105. doi: 10.1073/pnas.0700004104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Georgitsi M, De Menis E, Cannavo S, Makinen MJ, Tuppurainen K, Pauletto P, Curto L, Weil RJ, Paschke R, Zielinski G, Wasik A, Lubinski J, Vahteristo P, Karhu A, Aaltonen LA. Aryl hydrocarbon receptor interacting protein (AIP) gene mutation analysis in children and adolescents with sporadic pituitary adenomas. Clin Endocrinol (Oxf) 2008a;69:621–627. doi: 10.1111/j.1365-2265.2008.03266.x. [DOI] [PubMed] [Google Scholar]
- Georgitsi M, Heliovaara E, Paschke R, Kumar AV, Tischkowitz M, Vierimaa O, Salmela P, Sane T, De Menis E, Cannavo S, Gundogdu S, Lucassen A, Izatt L, Aylwin S, Bano G, Hodgson S, Koch CA, Karhu A, Aaltonen LA. Large genomic deletions in AIP in pituitary adenoma predisposition. J Clin Endocrinol Metab. 2008b;93:4146–4151. doi: 10.1210/jc.2008-1003. [DOI] [PubMed] [Google Scholar]
- Giacomini D, Paez-Pereda M, Theodoropoulou M, Labeur M, Refojo D, Gerez J, Chervin A, Berner S, Losa M, Buchfelder M, Renner U, Stalla GK, Arzt E. Bone morphogenetic protein-4 inhibits corticotroph tumor cells: involvement in the retinoic acid inhibitory action. Endocrinology. 2006;147:247–256. doi: 10.1210/en.2005-0958. [DOI] [PubMed] [Google Scholar]
- Gicquel C, Le Bouc Y, Luton JP, Girard F, Bertagna X. Monoclonality of corticotroph macroadenomas in Cushing's disease. J Clin Endocrinol Metab. 1992;75:472–475. doi: 10.1210/jcem.75.2.1322426. [DOI] [PubMed] [Google Scholar]
- Gicquel C, Leblond-Francillard M, Bertagna X, Louvel A, Chapuis Y, Luton JP, Girard F, Le Bouc Y. Clonal analysis of human adrenocortical carcinomas and secreting adenomas. Clin Endocrinol (Oxf) 1994;40:465–477. doi: 10.1111/j.1365-2265.1994.tb02485.x. [DOI] [PubMed] [Google Scholar]
- Gordon FE, Nutt CL, Cheunsuchon P, Nakayama Y, Provencher KA, Rice KA, Zhou Y, Zhang X, Klibanski A. Increased expression of angiogenic genes in the brains of mouse meg3-null embryos. Endocrinology. 2010;151:2443–2452. doi: 10.1210/en.2009-1151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grombacher T, Eichhorn U, Kaina B. P53 is involved in regulation of the DNA repair gene O6-methylguanine-DNA methyltransferase (MGMT) by DNA damaging agents. Oncogene. 1998;17:845–851. doi: 10.1038/sj.onc.1202000. [DOI] [PubMed] [Google Scholar]
- Guo W, Dong Z, Guo Y, Chen Z, Kuang G, Yang Z. Methylation-mediated repression of GADD45A and GADD45G expression in gastric cardia adenocarcinoma. Int J Cancer. 2013a;133:2043–2053. doi: 10.1002/ijc.28223. [DOI] [PubMed] [Google Scholar]
- Guo W, Zhu T, Dong Z, Cui L, Zhang M, Kuang G. Decreased expression and aberrant methylation of Gadd45G is associated with tumor progression and poor prognosis in esophageal squamous cell carcinoma. Clin Exp Metastasis. 2013b doi: 10.1007/s10585-013-9597-2. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
- Hajra KM, Chen DY, Fearon ER. The SLUG zinc-finger protein represses E-cadherin in breast cancer. Cancer Res. 2002;62:1613–1618. [PubMed] [Google Scholar]
- Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2013;144:646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- Harding B, Lemos MC, Reed AA, Walls GV, Jeyabalan J, Bowl MR, Tateossian H, Sullivan N, Hough T, Fraser WD, Ansorge O, Cheeseman MT, Thakker RV. Multiple endocrine neoplasia type 1 knockout mice develop parathyroid, pancreatic, pituitary and adrenal tumours with hypercalcaemia, hypophosphataemia and hypercorticosteronaemia. Endocr Relat Cancer. 2009;16:1313–1327. doi: 10.1677/ERC-09-0082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harvey M, Vogel H, Lee EY, Bradley A, Donehower LA. Mice deficient in both p53 and Rb develop tumors primarily of endocrine origin. Cancer Res. 1995;55:1146–1151. [PubMed] [Google Scholar]
- Henley SA, Dick FA. The retinoblastoma family of proteins and their regulatory functions in the mammalian cell division cycle. Cell Div. 2012;7:10. doi: 10.1186/1747-1028-7-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herman V, Fagin J, Gonsky R, Kovacs K, Melmed S. Clonal origin of pituitary adenomas. J Clin Endocrinol Metab. 1990;71:1427–1433. doi: 10.1210/jcem-71-6-1427. [DOI] [PubMed] [Google Scholar]
- Herman V, Drazin NZ, Gonsky R, Melmed S. Molecular screening of pituitary adenomas for gene mutations and rearrangements. J Clin Endocrinol Metab. 1993;77:50–55. doi: 10.1210/jcem.77.1.8100831. [DOI] [PubMed] [Google Scholar]
- Hiyama H, Kubo O, Kawamata T, Ishizaki R, Hori T. Expression of cyclin kinase inhibitor p21/WAF1 protein in pituitary adenomas: correlations with endocrine activity, but not cell proliferation. Acta Neurochir (Wien) 2002;144:481–488. doi: 10.1007/s007010200069. [DOI] [PubMed] [Google Scholar]
- Hoffmann A, Spengler D. A new coactivator function for Zac1's C2H2 zinc finger DNA-binding domain in selectively controlling PCAF activity. Mol Cell Biol. 2008;28:6078–6093. doi: 10.1128/MCB.00842-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoffmann A, Ciani E, Boeckardt J, Holsboer F, Journot L, Spengler D. Transcriptional activities of the zinc finger protein Zac are differentially controlled by DNA binding. Mol Cell Biol. 2003;23:988–1003. doi: 10.1128/MCB.23.3.988-1003.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Honda S, Tanaka-Kosugi C, Yamada S, Sano T, Matsumoto T, Itakura M, Yoshimoto K. Human pituitary adenomas infrequently contain inactivation of retinoblastoma 1 gene and activation of cyclin dependent kinase 4 gene. Endocr J. 2003;50:309–318. doi: 10.1507/endocrj.50.309. [DOI] [PubMed] [Google Scholar]
- Hossain MG, Iwata T, Mizusawa N, Qian ZR, Shima SW, Okutsu T, Yamada S, Sano T, Yoshimoto K. Expression of p18(INK4C) is down-regulated in human pituitary adenomas. Endocr Pathol. 2009;20:114–121. doi: 10.1007/s12022-009-9076-0. [DOI] [PubMed] [Google Scholar]
- Hu N, Gutsmann A, Herbert DC, Bradley A, Lee WH, Lee EY. Heterozygous Rb-1 delta 20/+mice are predisposed to tumors of the pituitary gland with a nearly complete penetrance. Oncogene. 1994;9:1021–1027. [PubMed] [Google Scholar]
- Huang SM, Schonthal AH, Stallcup MR. Enhancement of p53-dependent gene activation by the transcriptional coactivator Zac1. Oncogene. 2001;20:2134–2143. doi: 10.1038/sj.onc.1204298. [DOI] [PubMed] [Google Scholar]
- Huang ZY, Wu Y, Hedrick N, Gutmann DH. T-cadherin-mediated cell growth regulation involves G2 phase arrest and requires p21(CIP1/WAF1) expression. Mol Cell Biol. 2003;23:566–578. doi: 10.1128/MCB.23.2.566-578.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang SM, Huang SP, Wang SL, Liu PY. Importin alpha1 is involved in the nuclear localization of Zac1 and the induction of p21WAF1/CIP1 by Zac1. Biochem J. 2007;402:359–366. doi: 10.1042/BJ20061295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Igreja S, Chahal HS, Akker SA, Gueorguiev M, Popovic V, Damjanovic S, Burman P, Wass JA, Quinton R, Grossman AB, Korbonits M. Assessment of p27 (cyclin-dependent kinase inhibitor 1B) and aryl hydrocarbon receptor-interacting protein (AIP) genes in multiple endocrine neoplasia (MEN1) syndrome patients without any detectable MEN1 gene mutations. Clin Endocrinol (Oxf) 2009;70:259–264. doi: 10.1111/j.1365-2265.2008.03379.x. [DOI] [PubMed] [Google Scholar]
- Igreja S, Chahal HS, King P, Bolger GB, Srirangalingam U, Guasti L, Chapple JP, Trivellin G, Gueorguiev M, Guegan K, Stals K, Khoo B, Kumar AV, Ellard S, Grossman AB, Korbonits M, International FC. Characterization of aryl hydrocarbon receptor interacting protein (AIP) mutations in familial isolated pituitary adenoma families. Hum Mutat. 2010;31:950–960. doi: 10.1002/humu.21292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ikeda H, Yoshimoto T, Shida N. Molecular analysis of p21 and p27 genes in human pituitary adenomas. Br J Cancer. 1997;76:1119–1123. doi: 10.1038/bjc.1997.521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Indovina P, Marcelli E, Casini N, Rizzo V, Giordano A. Emerging roles of RB family: new defense mechanisms against tumor progression. J Cell Physiol. 2013;228:525–535. doi: 10.1002/jcp.24170. [DOI] [PubMed] [Google Scholar]
- Ivo D, Corset L, Desbourdes L, Gaudray P, Weber G. Menin controls the concentration of retinoblastoma protein. Cell Cycle. 2011;10:166–168. doi: 10.4161/cc.10.1.14447. [DOI] [PubMed] [Google Scholar]
- Iwakuma T, Sakumi K, Nakatsuru Y, Kawate H, Igarashi H, Shiraishi A, Tsuzuki T, Ishikawa T, Sekiguchi M. High incidence of nitrosamine-induced tumorigenesis in mice lacking DNA repair methyltransferase. Carcinogenesis. 1997;18:1631–1635. doi: 10.1093/carcin/18.8.1631. [DOI] [PubMed] [Google Scholar]
- Iwata T, Yamada S, Mizusawa N, Golam HM, Sano T, Yoshimoto K. The aryl hydrocarbon receptor-interacting protein gene is rarely mutated in sporadic GH-secreting adenomas. Clin Endocrinol (Oxf) 2007;66:499–502. doi: 10.1111/j.1365-2265.2007.02758.x. [DOI] [PubMed] [Google Scholar]
- Jacks T, Fazeli A, Schmitt EM, Bronson RT, Goodell MA, Weinberg RA. Effects of an Rb mutation in the mouse. Nature. 1992;359:295–300. doi: 10.1038/359295a0. [DOI] [PubMed] [Google Scholar]
- Jacoby LB, Hedley-Whyte ET, Pulaski K, Seizinger BR, Martuza RL. Clonal origin of pituitary adenomas. J Neurosurg. 1990;73:731–735. doi: 10.3171/jns.1990.73.5.0731. [DOI] [PubMed] [Google Scholar]
- Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet. 2003;33(Suppl):245–254. doi: 10.1038/ng1089. [DOI] [PubMed] [Google Scholar]
- Jaffrain-Rea ML, Ferretti E, Toniato E, Cannita K, Santoro A, Di Stefano D, Ricevuto E, Maroder M, Tamburrano G, Cantore G, Gulino A, Martinotti S. P16 (INK4a, MTS-1) gene polymorphism and methylation status in human pituitary tumours. Clin Endocrinol (Oxf) 1999;51:317–325. doi: 10.1046/j.1365-2265.1999.00774.x. [DOI] [PubMed] [Google Scholar]
- Jarmalaite S, Laurinaviciene A, Tverkuviene J, Kalinauskaite N, Petroska D, Bohling T, Husgafvel-Pursiainen K. Tumor suppressor gene ZAC/PLAGL1: altered expression and loss of the nonimprinted allele in pheochromocytomas. Cancer Genet. 2011;204:398–404. doi: 10.1016/j.cancergen.2011.07.002. [DOI] [PubMed] [Google Scholar]
- Jeanes A, Gottardi CJ, Yap AS. Cadherins and cancer: how does cadherin dysfunction promote tumor progression? Oncogene. 2008;27:6920–6929. doi: 10.1038/onc.2008.343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jordan S, Lidhar K, Korbonits M, Lowe DG, Grossman AB. Cyclin D and cyclin E expression in normal and adenomatous pituitary. Eur J Endocrinol. 2000;143:R1–6. doi: 10.1530/eje.0.143r001. [DOI] [PubMed] [Google Scholar]
- Kaina B, Christmann M, Naumann S, Roos WP. MGMT: key node in the battle against genotoxicity, carcinogenicity and apoptosis induced by alkylating agents. DNA Repair (Amst) 2007;6:1079–1099. doi: 10.1016/j.dnarep.2007.03.008. [DOI] [PubMed] [Google Scholar]
- Kaji H, Canaff L, Lebrun JJ, Goltzman D, Hendy GN. Inactivation of menin, a Smad3-interacting protein, blocks transforming growth factor type beta signaling. Proc Natl Acad Sci USA. 2001;98:3837–3842. doi: 10.1073/pnas.061358098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalmes M, Hennen J, Clemens J, Blomeke B. Impact of aryl hydrocarbon receptor (AhR) knockdown on cell cycle progression in human HaCaT keratinocytes. Biol Chem. 2011;392:643–651. doi: 10.1515/BC.2011.067. [DOI] [PubMed] [Google Scholar]
- Kamikihara T, Arima T, Kato K, Matsuda T, Kato H, Douchi T, Nagata Y, Nakao M, Wake N. Epigenetic silencing of the imprinted gene ZAC by DNA methylation is an early event in the progression of human ovarian cancer. Int J Cancer. 2005;115:690–700. doi: 10.1002/ijc.20971. [DOI] [PubMed] [Google Scholar]
- Kamiya M, Judson H, Okazaki Y, Kusakabe M, Muramatsu M, Takada S, Takagi N, Arima T, Wake N, Kamimura K, Satomura K, Hermann R, Bonthron DT, Hayashizaki Y. The cell cycle control gene ZAC/PLAGL1 is imprinted – a strong candidate gene for transient neonatal diabetes. Hum Mol Genet. 2000;9:453–460. doi: 10.1093/hmg/9.3.453. [DOI] [PubMed] [Google Scholar]
- Karmodiya K, Krebs AR, Oulad-Abdelghani M, Kimura H, Tora L. H3K9 and H3K14 acetylation co-occur at many gene regulatory elements, while H3K14ac marks a subset of inactive inducible promoters in mouse embryonic stem cells. BMC Genomics. 2012;13:424. doi: 10.1186/1471-2164-13-424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karnik SK, Hughes CM, Gu X, Rozenblatt-Rosen O, McLean GW, Xiong Y, Meyerson M, Kim SK. Menin regulates pancreatic islet growth by promoting histone methylation and expression of genes encoding p27Kip1 and p18INK4c. Proc Natl Acad Sci USA. 2005;102:14659–14664. doi: 10.1073/pnas.0503484102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kawamoto H, Mizoue T, Arita K, Tominaga A, Eguchi K, Kurisu K. Expression of epithelial cadherin and cavernous sinus invasion in human pituitary adenomas. J Neurooncol. 1997;34:105–109. doi: 10.1023/a:1005709014239. [DOI] [PubMed] [Google Scholar]
- Kawashima ST, Usui T, Sano T, Iogawa H, Hagiwara H, Tamanaha T, Tagami T, Naruse M, Hojo M, Takahashi JA, Shimatsu A. P53 gene mutation in an atypical corticotroph adenoma with Cushing's disease. Clin Endocrinol (Oxf) 2009;70:656–657. doi: 10.1111/j.1365-2265.2008.03404.x. [DOI] [PubMed] [Google Scholar]
- Kazlauskas A, Poellinger L, Pongratz I. The immunophilin-like protein XAP2 regulates ubiquitination and subcellular localization of the dioxin receptor. J Biol Chem. 2000;275:41317–41324. doi: 10.1074/jbc.M007765200. [DOI] [PubMed] [Google Scholar]
- Kim YS, Burns AL, Goldsmith PK, Heppner C, Park SY, Chandrasekharappa SC, Collins FS, Spiegel AM, Marx SJ. Stable overexpression of MEN1 suppresses tumorigenicity of RAS. Oncogene. 1999;18:5936–5942. doi: 10.1038/sj.onc.1203005. [DOI] [PubMed] [Google Scholar]
- Kirsch M, Morz M, Pinzer T, Schackert HK, Schackert G. Frequent loss of the CDKN2C (p18INK4c) gene product in pituitary adenomas. Genes Chromosom Cancer. 2009;48:143–154. doi: 10.1002/gcc.20621. [DOI] [PubMed] [Google Scholar]
- Kirschner LS, Carney JA, Pack SD, Taymans SE, Giatzakis C, Cho YS, Cho-Chung YS, Stratakis CA. Mutations of the gene encoding the protein kinase A type I-alpha regulatory subunit in patients with the Carney complex. Nat Genet. 2000;26:89–92. doi: 10.1038/79238. [DOI] [PubMed] [Google Scholar]
- Kiyokawa H, Kineman RD, Manova-Todorova KO, Soares VC, Hoffman ES, Ono M, Khanam D, Hayday AC, Frohman LA, Koff A. Enhanced growth of mice lacking the cyclin-dependent kinase inhibitor function of p27(Kip1) Cell. 1996;85:721–732. doi: 10.1016/s0092-8674(00)81238-6. [DOI] [PubMed] [Google Scholar]
- Knudson AG., Jr Mutation and cancer: statistical study of retinoblastoma. Proc Natl Acad Sci USA. 1971;68:820–823. doi: 10.1073/pnas.68.4.820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kolluri SK, Weiss C, Koff A, Gottlicher M. P27(Kip1) induction and inhibition of proliferation by the intracellular Ah receptor in developing thymus and hepatoma cells. Genes Dev. 1999;13:1742–1753. doi: 10.1101/gad.13.13.1742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Komatsubara K, Tahara S, Umeoka K, Sanno N, Teramoto A, Osamura RY. Immunohistochemical analysis of p27 (Kip1) in human pituitary glands and in various types of pituitary adenomas. Endocr Pathol. 2001;12:181–188. doi: 10.1385/ep:12:2:181. [DOI] [PubMed] [Google Scholar]
- Korbonits M, Chahal HS, Kaltsas G, Jordan S, Urmanova Y, Khalimova Z, Harris PE, Farrell WE, Claret FX, Grossman AB. Expression of phosphorylated p27(Kip1) protein and Jun activation domain-binding protein 1 in human pituitary tumors. J Clin Endocrinol Metab. 2002;87:2635–2643. doi: 10.1210/jcem.87.6.8517. [DOI] [PubMed] [Google Scholar]
- Kruse JP, Gu W. Modes of p53 regulation. Cell. 2009;137:609–622. doi: 10.1016/j.cell.2009.04.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kudo T, Ikeda M, Nishikawa M, Yang Z, Ohno K, Nakagawa K, Hata Y. The RASSF3 candidate tumor suppressor induces apoptosis and G1-S cell-cycle arrest via p53. Cancer Res. 2012;72:2901–2911. doi: 10.1158/0008-5472.CAN-12-0572. [DOI] [PubMed] [Google Scholar]
- Kuroda A, Rauch TA, Todorov I, Ku HT, Al-Abdullah IH, Kandeel F, Mullen Y, Pfeifer GP, Ferreri K. Insulin gene expression is regulated by DNA methylation. PLoS One. 2009;4:e6953. doi: 10.1371/journal.pone.0006953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- La P, Silva AC, Hou Z, Wang H, Schnepp RW, Yan N, Shi Y, Hua X. Direct binding of DNA by tumor suppressor menin. J Biol Chem. 2004;279:49045–49054. doi: 10.1074/jbc.M409358200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Labeur M, Paez-Pereda M, Haedo M, Arzt E, Stalla GK. Pituitary tumors: cell type-specific roles for BMP-4. Mol Cell Endocrinol. 2010;326:85–88. doi: 10.1016/j.mce.2010.04.006. [DOI] [PubMed] [Google Scholar]
- Lacerte A, Lee EH, Reynaud R, Canaff L, De Guise C, Devost D, Ali S, Hendy GN, Lebrun JJ. Activin inhibits pituitary prolactin expression and cell growth through Smads, Pit-1 and menin. Mol Endocrinol. 2004;18:1558–1569. doi: 10.1210/me.2003-0470. [DOI] [PubMed] [Google Scholar]
- Landis CA, Masters SB, Spada A, Pace AM, Bourne HR, Vallar L. GTPase inhibiting mutations activate the alpha chain of Gs and stimulate adenylyl cyclase in human pituitary tumours. Nature. 1989;340:692–696. doi: 10.1038/340692a0. [DOI] [PubMed] [Google Scholar]
- LaPres JJ, Glover E, Dunham EE, Bunger MK, Bradfield CA. ARA9 modifies agonist signaling through an increase in cytosolic aryl hydrocarbon receptor. J Biol Chem. 2000;275:6153–6159. doi: 10.1074/jbc.275.9.6153. [DOI] [PubMed] [Google Scholar]
- Larsson C, Skogseid B, Oberg K, Nakamura Y, Nordenskjold M. Multiple endocrine neoplasia type 1 gene maps to chromosome 11 and is lost in insulinoma. Nature. 1988;332:85–87. doi: 10.1038/332085a0. [DOI] [PubMed] [Google Scholar]
- Lau Q, Scheithauer B, Kovacs K, Horvath E, Syro LV, Lloyd R. MGMT immunoexpression in aggressive pituitary adenoma and carcinoma. Pituitary. 2010;13:367–379. doi: 10.1007/s11102-010-0249-0. [DOI] [PubMed] [Google Scholar]
- Lee CK, Lee JH, Lee MG, Jeong SI, Ha TK, Kang MJ, Ryu BK, Hwangbo Y, Shim JJ, Jang JY, Lee KY, Kim HJ, Chi SG. Epigenetic inactivation of the NORE1 gene correlates with malignant progression of colorectal tumors. BMC Cancer. 2010;10:577. doi: 10.1186/1471-2407-10-577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lekva T, Berg JP, Fougner SL, Olstad OK, Ueland T, Bollerslev J. Gene expression profiling identifies ESRP1 as a potential regulator of epithelial mesenchymal transition in somatotroph adenomas from a large cohort of patients with acromegaly. J Clin Endocrinol Metab. 2012;97:E1506–1514. doi: 10.1210/jc.2012-1760. [DOI] [PubMed] [Google Scholar]
- Lekva T, Berg JP, Lyle R, Heck A, Ringstad G, Olstad OK, Michelsen AE, Casar-Borota O, Bollerslev J, Ueland T. Epithelial splicing regulator protein 1 and alternative splicing in somatotroph adenomas. Endocrinology. 2013;154:3331–3343. doi: 10.1210/en.2013-1051. [DOI] [PubMed] [Google Scholar]
- Lemeta S, Jarmalaite S, Pylkkanen L, Bohling T, Husgafvel-Pursiainen K. Preferential loss of the nonimprinted allele for the ZAC1 tumor suppressor gene in human capillary hemangioblastoma. J Neuropathol Exp Neurol. 2007;66:860–867. doi: 10.1097/nen.0b013e318149ee64. [DOI] [PubMed] [Google Scholar]
- Lemos MC, Thakker RV. Multiple endocrine neoplasia type 1 (MEN1): analysis of 1336 mutations reported in the first decade following identification of the gene. Hum Mutat. 2008;29:22–32. doi: 10.1002/humu.20605. [DOI] [PubMed] [Google Scholar]
- Leonhardt H, Page AW, Weier HU, Bestor TH. A targeting sequence directs DNA methyltransferase to sites of DNA replication in mammalian nuclei. Cell. 1992;71:865–873. doi: 10.1016/0092-8674(92)90561-p. [DOI] [PubMed] [Google Scholar]
- Leontieva OV, Ionov Y. RNA-binding motif protein 35A is a novel tumor suppressor for colorectal cancer. Cell Cycle. 2009;8:490–497. doi: 10.4161/cc.8.3.7679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leontiou CA, Gueorguiev M, van der Spuy J, Quinton R, Lolli F, Hassan S, Chahal HS, Igreja SC, Jordan S, Rowe J, Stolbrink M, Christian HC, Wray J, Bishop-Bailey D, Berney DM, Wass JA, Popovic V, Ribeiro-Oliveira A, Jr, Gadelha MR, Monson JP, Akker SA, Davis JR, Clayton RN, Yoshimoto K, Iwata T, Matsuno A, Eguchi K, Musat M, Flanagan D, Peters G, Bolger GB, Chapple JP, Frohman LA, Grossman AB, Korbonits M. The role of the aryl hydrocarbon receptor-interacting protein gene in familial and sporadic pituitary adenomas. J Clin Endocrinol Metab. 2008;93:2390–2401. doi: 10.1210/jc.2007-2611. [DOI] [PubMed] [Google Scholar]
- Leung SW, Wloga EH, Castro AF, Nguyen T, Bronson RT, Yamasaki L. A dynamic switch in Rb+/− mediated neuroendocrine tumorigenesis. Oncogene. 2004;23:3296–3307. doi: 10.1038/sj.onc.1207457. [DOI] [PubMed] [Google Scholar]
- Levy A, Hall L, Yeudall WA, Lightman SL. P53 gene mutations in pituitary adenomas: rare events. Clin Endocrinol (Oxf) 1994;41:809–814. doi: 10.1111/j.1365-2265.1994.tb02797.x. [DOI] [PubMed] [Google Scholar]
- Lidhar K, Korbonits M, Jordan S, Khalimova Z, Kaltsas G, Lu X, Clayton RN, Jenkins PJ, Monson JP, Besser GM, Lowe DG, Grossman AB. Low expression of the cell cycle inhibitor p27Kip1 in normal corticotroph cells, corticotroph tumors, and malignant pituitary tumors. J Clin Endocrinol Metab. 1999;84:3823–3830. doi: 10.1210/jcem.84.10.6066. [DOI] [PubMed] [Google Scholar]
- Liebermann DA, Tront JS, Sha X, Mukherjee K, Mohamed-Hadley A, Hoffman B. Gadd45 stress sensors in malignancy and leukemia. Crit Rev Oncog. 2011;16:129–140. doi: 10.1615/critrevoncog.v16.i1-2.120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin BC, Sullivan R, Lee Y, Moran S, Glover E, Bradfield CA. Deletion of the aryl hydrocarbon receptor-associated protein 9 leads to cardiac malformation and embryonic lethality. J Biol Chem. 2007;282:35924–35932. doi: 10.1074/jbc.M705471200. [DOI] [PubMed] [Google Scholar]
- Liu PY, Chan JY, Lin HC, Wang SL, Liu ST, Ho CL, Chang LC, Huang SM. Modulation of the cyclin-dependent kinase inhibitor p21(WAF1/Cip1) gene by Zac1 through the antagonistic regulators p53 and histone deacetylase 1 in HeLa Cells. Mol Cancer Res. 2008;6:1204–1214. doi: 10.1158/1541-7786.MCR-08-0123. [DOI] [PubMed] [Google Scholar]
- Liu PY, Hsieh TY, Liu ST, Chang YL, Lin WS, Wang WM, Huang SM. Zac1, an Sp1-like protein, regulates human p21(WAF1/Cip1) gene expression in HeLa cells. Exp Cell Res. 2011;317:2925–2937. doi: 10.1016/j.yexcr.2011.09.018. [DOI] [PubMed] [Google Scholar]
- Loffler KA, Biondi CA, Gartside M, Waring P, Stark M, Serewko-Auret MM, Muller HK, Hayward NK, Kay GF. Broad tumor spectrum in a mouse model of multiple endocrine neoplasia type 1. Int J Cancer. 2007;120:259–267. doi: 10.1002/ijc.22288. [DOI] [PubMed] [Google Scholar]
- Lombardo Y, Scopelliti A, Cammareri P, Todaro M, Iovino F, Ricci-Vitiani L, Gulotta G, Dieli F, de Maria R, Stassi G. Bone morphogenetic protein 4 induces differentiation of colorectal cancer stem cells and increases their response to chemotherapy in mice. Gastroenterology. 2011;140:297–309. doi: 10.1053/j.gastro.2010.10.005. [DOI] [PubMed] [Google Scholar]
- Lowe SW, Cepero E, Evan G. Intrinsic tumour suppression. Nature. 2004;432:307–315. doi: 10.1038/nature03098. [DOI] [PubMed] [Google Scholar]
- Lubensky IA, Debelenko LV, Zhuang Z, Emmert-Buck MR, Dong Q, Chandrasekharappa S, Guru SC, Manickam P, Olufemi SE, Marx SJ, Spiegel AM, Collins FS, Liotta LA. Allelic deletions on chromosome 11q13 in multiple tumors from individual MEN1 patients. Cancer Res. 1996;56:5272–5278. [PubMed] [Google Scholar]
- Lyons J, Landis CA, Harsh G, Vallar L, Grunewald K, Feichtinger H, Duh QY, Clark OH, Kawasaki E, Bourne HR, et al. Two G protein oncogenes in human endocrine tumors. Science. 1990;249:655–659. doi: 10.1126/science.2116665. [DOI] [PubMed] [Google Scholar]
- Ma Q, Whitlock JP., Jr A novel cytoplasmic protein that interacts with the Ah receptor, contains tetratricopeptide repeat motifs, and augments the transcriptional response to 2,3,7,8-tetrachlorodibenzo-p-dioxin. J Biol Chem. 1997;272:8878–8884. [PubMed] [Google Scholar]
- Ma W, Ikeda H, Yoshimoto T. Clinicopathologic study of 123 cases of prolactin-secreting pituitary adenomas with special reference to multihormone production and clonality of the adenomas. Cancer. 2002;95:258–266. doi: 10.1002/cncr.10676. [DOI] [PubMed] [Google Scholar]
- Macheiner D, Heller G, Kappel S, Bichler C, Stattner S, Ziegler B, Kandioler D, Wrba F, Schulte-Hermann R, Zochbauer-Muller S, Grasl-Kraupp B. NORE1B, a candidate tumor suppressor, is epigenetically silenced in human hepatocellular carcinoma. J Hepatol. 2006;45:81–89. doi: 10.1016/j.jhep.2005.12.017. [DOI] [PubMed] [Google Scholar]
- Macleod KF. The role of the RB tumour suppressor pathway in oxidative stress responses in the haematopoietic system. Nat Rev Cancer. 2008;8:769–781. doi: 10.1038/nrc2504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Madak-Erdogan Z, Katzenellenbogen BS. Aryl hydrocarbon receptor modulation of estrogen receptor alpha-mediated gene regulation by a multimeric chromatin complex involving the two receptors and the coregulator RIP140. Toxicol Sci. 2012;125:401–411. doi: 10.1093/toxsci/kfr300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mallette FA, Calabrese V, Ilangumaran S, Ferbeyre G. SOCS1, a novel interaction partner of p53 controlling oncogene-induced senescence. Aging (Albany NY) 2010;2:445–452. doi: 10.18632/aging.100163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marlowe JL, Knudsen ES, Schwemberger S, Puga A. The aryl hydrocarbon receptor displaces p300 from E2F-dependent promoters and represses S phase-specific gene expression. J Biol Chem. 2004;279:29013–29022. doi: 10.1074/jbc.M404315200. [DOI] [PubMed] [Google Scholar]
- Marlowe JL, Fan Y, Chang X, Peng L, Knudsen ES, Xia Y, Puga A. The aryl hydrocarbon receptor binds to E2F1 and inhibits E2F1-induced apoptosis. Mol Biol Cell. 2008;19:3263–3271. doi: 10.1091/mbc.E08-04-0359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McCormack AI, McDonald KL, Gill AJ, Clark SJ, Burt MG, Campbell KA, Braund WJ, Little NS, Cook RJ, Grossman AB, Robinson BG, Clifton-Bligh RJ. Low O6-methylguanine-DNA methyltransferase (MGMT) expression and response to temozolomide in aggressive pituitary tumours. Clin Endocrinol (Oxf) 2009;71:226–233. doi: 10.1111/j.1365-2265.2008.03487.x. [DOI] [PubMed] [Google Scholar]
- Meng W, Takeichi M. Adherens junction: molecular architecture and regulation. Cold Spring Harb Perspect Biol. 2009;1:a002899. doi: 10.1101/cshperspect.a002899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mezzomo LC, Gonzales PH, Pesce FG, Kretzmann Filho N, Ferreira NP, Oliveira MC, Kohek MB. Expression of cell growth negative regulators MEG3 and GADD45gamma is lost in most sporadic human pituitary adenomas. Pituitary. 2012;15:420–427. doi: 10.1007/s11102-011-0340-1. [DOI] [PubMed] [Google Scholar]
- Michaelis KA, Knox AJ, Xu M, Kiseljak-Vassiliades K, Edwards MG, Geraci M, Kleinschmidt-DeMasters BK, Lillehei KO, Wierman ME. Identification of growth arrest and DNA-damage-inducible gene beta (GADD45beta) as a novel tumor suppressor in pituitary gonadotrope tumors. Endocrinology. 2011;152:3603–3613. doi: 10.1210/en.2011-0109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Milne TA, Briggs SD, Brock HW, Martin ME, Gibbs D, Allis CD, Hess JL. MLL targets SET domain methyltransferase activity to Hox gene promoters. Mol Cell. 2002;10:1107–1117. doi: 10.1016/s1097-2765(02)00741-4. [DOI] [PubMed] [Google Scholar]
- Milne TA, Dou Y, Martin ME, Brock HW, Roeder RG, Hess JL. MLL associates specifically with a subset of transcriptionally active target genes. Proc Natl Acad Sci USA. 2005a;102:14765–14770. doi: 10.1073/pnas.0503630102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Milne TA, Hughes CM, Lloyd R, Yang Z, Rozenblatt-Rosen O, Dou Y, Schnepp RW, Krankel C, Livolsi VA, Gibbs D, Hua X, Roeder RG, Meyerson M, Hess JL. Menin and MLL cooperatively regulate expression of cyclin-dependent kinase inhibitors. Proc Natl Acad Sci USA. 2005b;102:749–754. doi: 10.1073/pnas.0408836102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miyazono K, Maeda S, Imamura T. BMP receptor signaling: transcriptional targets, regulation of signals, and signaling cross-talk. Cytokine Growth Factor Rev. 2005;16:251–263. doi: 10.1016/j.cytogfr.2005.01.009. [DOI] [PubMed] [Google Scholar]
- Morgan RM, Hernandez-Ramirez LC, Trivellin G, Zhou L, Roe SM, Korbonits M, Prodromou C. Structure of the TPR domain of AIP: lack of client protein interaction with the C-terminal alpha-7 helix of the TPR domain of AIP is sufficient for pituitary adenoma predisposition. PLoS One. 2012;7:e53339. doi: 10.1371/journal.pone.0053339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mortusewicz O, Schermelleh L, Walter J, Cardoso MC, Leonhardt H. Recruitment of DNA methyltransferase I to DNA repair sites. Proc Natl Acad Sci USA. 2005;102:8905–8909. doi: 10.1073/pnas.0501034102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moshnikova A, Frye J, Shay JW, Minna JD, Khokhlatchev AV. The growth and tumor suppressor NORE1A is a cytoskeletal protein that suppresses growth by inhibition of the ERK pathway. J Biol Chem. 2006;281:8143–8152. doi: 10.1074/jbc.M511837200. [DOI] [PubMed] [Google Scholar]
- Moskalev AA, Smit-McBride Z, Shaposhnikov MV, Plyusnina EN, Zhavoronkov A, Budovsky A, Tacutu R, Fraifeld VE. Gadd45 proteins: relevance to aging, longevity and age-related pathologies. Ageing Res Rev. 2012;11:51–66. doi: 10.1016/j.arr.2011.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nakayama K, Ishida N, Shirane M, Inomata A, Inoue T, Shishido N, Horii I, Loh DY. Mice lacking p27(Kip1) display increased body size, multiple organ hyperplasia, retinal dysplasia, and pituitary tumors. Cell. 1996;85:707–720. doi: 10.1016/s0092-8674(00)81237-4. [DOI] [PubMed] [Google Scholar]
- Nevins JR. The Rb/E2F pathway and cancer. Hum Mol Genet. 2001;10:699–703. doi: 10.1093/hmg/10.7.699. [DOI] [PubMed] [Google Scholar]
- Nikitin A, Lee WH. Early loss of the retinoblastoma gene is associated with impaired growth inhibitory innervation during melanotroph carcinogenesis in Rb+/− mice. Genes Dev. 1996;10:1870–1879. doi: 10.1101/gad.10.15.1870. [DOI] [PubMed] [Google Scholar]
- Nukaya M, Lin BC, Glover E, Moran SM, Kennedy GD, Bradfield CA. The aryl hydrocarbon receptor-interacting protein (AIP) is required for dioxininduced hepatotoxicity but not for the induction of the Cyp1a1 and Cyp1a2 genes. J Biol Chem. 2010;285:35599–35605. doi: 10.1074/jbc.M110.132043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oberg K, Skogseid B, Eriksson B. Multiple endocrine neoplasia type 1 (MEN-1). Clinical, biochemical and genetical investigations. Acta Oncol. 1989;28:383–387. doi: 10.3109/02841868909111211. [DOI] [PubMed] [Google Scholar]
- O'Brien T, O'Riordan DS, Gharib H, Scheithauer BW, Ebersold MJ, van Heerden JA. Results of treatment of pituitary disease in multiple endocrine neoplasia, type I. Neurosurgery. 1996;39:273–278. doi: 10.1097/00006123-199608000-00008. discussion 278-279. [DOI] [PubMed] [Google Scholar]
- Occhi G, Trivellin G, Ceccato F, De Lazzari P, Giorgi G, Dematte S, Grimaldi F, Castello R, Davi MV, Arnaldi G, Salviati L, Opocher G, Mantero F, Scaroni C. Prevalence of AIP mutations in a large series of sporadic Italian acromegalic patients and evaluation of CDKN1B status in acromegalic patients with multiple endocrine neoplasia. Eur J Endocrinol. 2010;163:369–376. doi: 10.1530/EJE-10-0327. [DOI] [PubMed] [Google Scholar]
- Ogino A, Yoshino A, Katayama Y, Watanabe T, Ota T, Komine C, Yokoyama T, Fukushima T. The p15(INK4b)/p16(INK4a)/RB1 pathway is frequently deregulated in human pituitary adenomas. J Neuropathol Exp Neurol. 2005;64:398–403. doi: 10.1093/jnen/64.5.398. [DOI] [PubMed] [Google Scholar]
- Okano M, Xie S, Li E. Cloning and characterization of a family of novel mammalian DNA (cytosine-5) methyltransferases. Nat Genet. 1998;19:219–220. doi: 10.1038/890. [DOI] [PubMed] [Google Scholar]
- Okano M, Bell DW, Haber DA, Li E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell. 1999;99:247–257. doi: 10.1016/s0092-8674(00)81656-6. [DOI] [PubMed] [Google Scholar]
- Olivier M, Hollstein M, Hainaut P. TP53 mutations in human cancers: origins, consequences, and clinical use. Cold Spring Harb Perspect Biol. 2010;2:a001008. doi: 10.1101/cshperspect.a001008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Osada T, Sakamoto M, Ino Y, Iwamatsu A, Matsuno Y, Muto T, Hirohashi S. E-cadherin is involved in the intrahepatic metastasis of hepatocellular carcinoma. Hepatology. 1996;24:1460–1467. doi: 10.1053/jhep.1996.v24.pm0008938181. [DOI] [PubMed] [Google Scholar]
- Paez-Pereda M, Giacomini D, Refojo D, Nagashima AC, Hopfner U, Grubler Y, Chervin A, Goldberg V, Goya R, Hentges ST, Low MJ, Holsboer F, Stalla GK, Arzt E. Involvement of bone morphogenetic protein 4 (BMP-4) in pituitary prolactinoma pathogenesis through a Smad/estrogen receptor crosstalk. Proc Natl Acad Sci USA. 2003;100:1034–1039. doi: 10.1073/pnas.0237312100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pagotto U, Arzberger T, Ciani E, Lezoualc'h F, Pilon C, Journot L, Spengler D, Stalla GK. Inhibition of Zac1, a new gene differentially expressed in the anterior pituitary, increases cell proliferation. Endocrinology. 1999;140:987–996. doi: 10.1210/endo.140.2.6532. [DOI] [PubMed] [Google Scholar]
- Pagotto U, Arzberger T, Theodoropoulou M, Grubler Y, Pantaloni C, Saeger W, Losa M, Journot L, Stalla GK, Spengler D. The expression of the antiproliferative gene ZAC is lost or highly reduced in nonfunctioning pituitary adenomas. Cancer Res. 2000;60:6794–6799. [PubMed] [Google Scholar]
- Pang PH, Lin YH, Lee YH, Hou HH, Hsu SP, Juan SH. Molecular mechanisms of p21 and p27 induction by 3-methylcholanthrene, an arylhydrocarbon receptor agonist, involved in antiproliferation of human umbilical vascular endothelial cells. J Cell Physiol. 2008;215:161–171. doi: 10.1002/jcp.21299. [DOI] [PubMed] [Google Scholar]
- Park J, Kang SI, Lee SY, Zhang XF, Kim MS, Beers LF, Lim DS, Avruch J, Kim HS, Lee SB. Tumor suppressor ras association domain family 5 (RASSF5/NORE1) mediates death receptor ligand-induced apoptosis. J Biol Chem. 2010;285:35029–35038. doi: 10.1074/jbc.M110.165506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pegg AE, Dolan ME, Moschel RC. Structure, function, and inhibition of O6-alkylguanine-DNA alkyltransferase. Prog Nucleic Acid Res Mol Biol. 1995;51:167–223. doi: 10.1016/s0079-6603(08)60879-x. [DOI] [PubMed] [Google Scholar]
- Pellegata NS, Quintanilla-Martinez L, Siggelkow H, Samson E, Bink K, Hofler H, Fend F, Graw J, Atkinson MJ. Germ-line mutations in p27Kip1 cause a multiple endocrine neoplasia syndrome in rats and humans. Proc Natl Acad Sci USA. 2006;103:15558–15563. doi: 10.1073/pnas.0603877103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peltomaki P. Mutations and epimutations in the origin of cancer. Exp Cell Res. 2012;318:299–310. doi: 10.1016/j.yexcr.2011.12.001. [DOI] [PubMed] [Google Scholar]
- Peng H, Liu H, Zhao S, Wu J, Fan J, Liao J. Silencing of RASSF3 by DNA hypermethylation is associated with tumorigenesis in somatotroph adenomas. PLoS One. 2013;8:e59024. doi: 10.1371/journal.pone.0059024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perl AK, Wilgenbus P, Dahl U, Semb H, Christofori G. A causal role for E-cadherin in the transition from adenoma to carcinoma. Nature. 1998;392:190–193. doi: 10.1038/32433. [DOI] [PubMed] [Google Scholar]
- Perry ME. The regulation of the p53-mediated stress response by MDM2 and MDM4. Cold Spring Harb Perspect Biol. 2010;2:a000968. doi: 10.1101/cshperspect.a000968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Petrulis JR, Hord NG, Perdew GH. Subcellular localization of the aryl hydrocarbon receptor is modulated by the immunophilin homolog hepatitis B virus X-associated protein 2. J Biol Chem. 2000;275:37448–37453. doi: 10.1074/jbc.M006873200. [DOI] [PubMed] [Google Scholar]
- Philippova M, Joshi MB, Kyriakakis E, Pfaff D, Erne P, Resink TJ. A guide and guard: the many faces of T-cadherin. Cell Signal. 2009;21:1035–1044. doi: 10.1016/j.cellsig.2009.01.035. [DOI] [PubMed] [Google Scholar]
- Philippova M, Joshi MB, Pfaff D, Kyriakakis E, Maslova K, Erne P, Resink TJ. T-cadherin attenuates insulin-dependent signalling, eNOS activation, and angiogenesis in vascular endothelial cells. Cardiovasc Res. 2012;93:498–507. doi: 10.1093/cvr/cvs004. [DOI] [PubMed] [Google Scholar]
- Poncin J, Stevenaert A, Beckers A. Somatic MEN1 gene mutation does not contribute significantly to sporadic pituitary tumorigenesis. Eur J Endocrinol. 1999;140:573–576. doi: 10.1530/eje.0.1400573. [DOI] [PubMed] [Google Scholar]
- Prezant TR, Levine J, Melmed S. Molecular characterization of the men1 tumor suppressor gene in sporadic pituitary tumors. J Clin Endocrinol Metab. 1998;83:1388–1391. doi: 10.1210/jcem.83.4.4859. [DOI] [PubMed] [Google Scholar]
- Puga A, Barnes SJ, Dalton TP, Chang C, Knudsen ES, Maier MA. Aromatic hydrocarbon receptor interaction with the retinoblastoma protein potentiates repression of E2F-dependent transcription and cell cycle arrest. J Biol Chem. 2000;275:2943–2950. doi: 10.1074/jbc.275.4.2943. [DOI] [PubMed] [Google Scholar]
- Qian ZR, Sano T, Yoshimoto K, Yamada S, Ishizuka A, Mizusawa N, Horiguchi H, Hirokawa M, Asa SL. Inactivation of RASSF1A tumor suppressor gene by aberrant promoter hypermethylation in human pituitary adenomas. Lab Invest. 2005;85:464–473. doi: 10.1038/labinvest.3700248. [DOI] [PubMed] [Google Scholar]
- Qian ZR, Sano T, Yoshimoto K, Asa SL, Yamada S, Mizusawa N, Kudo E. Tumor-specific downregulation and methylation of the CDH13 (H-cadherin) and CDH1 (E-cadherin) genes correlate with aggressiveness of human pituitary adenomas. Mod Pathol. 2007;20:1269–1277. doi: 10.1038/modpathol.3800965. [DOI] [PubMed] [Google Scholar]
- Raitila A, Georgitsi M, Karhu A, Tuppurainen K, Makinen MJ, Birkenkamp-Demtroder K, Salmenkivi K, Orntoft TF, Arola J, Launonen V, Vahteristo P, Aaltonen LA. No evidence of somatic aryl hydrocarbon receptor interacting protein mutations in sporadic endocrine neoplasia. Endocr Relat Cancer. 2007;14:901–906. doi: 10.1677/ERC-07-0025. [DOI] [PubMed] [Google Scholar]
- Raitila A, Lehtonen HJ, Arola J, Heliovaara E, Ahlsten M, Georgitsi M, Jalanko A, Paetau A, Aaltonen LA, Karhu A. Mice with inactivation of aryl hydrocarbon receptor-interacting protein (Aip) display complete penetrance of pituitary adenomas with aberrant ARNT expression. Am J Pathol. 2010;177:1969–1976. doi: 10.2353/ajpath.2010.100138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ratineau C, Bernard C, Poncet G, Blanc M, Josso C, Fontaniere S, Calender A, Chayvialle JA, Zhang CX, Roche C. Reduction of menin expression enhances cell proliferation and is tumorigenic in intestinal epithelial cells. J Biol Chem. 2004;279:24477–24484. doi: 10.1074/jbc.M401835200. [DOI] [PubMed] [Google Scholar]
- Rayess H, Wang MB, Srivatsan ES. Cellular senescence and tumor suppressor gene p16. Int J Cancer. 2012;130:1715–1725. doi: 10.1002/ijc.27316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reinke LM, Xu Y, Cheng C. Snail represses the splicing regulator epithelial splicing regulatory protein 1 to promote epithelial–mesenchymal transition. J Biol Chem. 2012;287:36435–36442. doi: 10.1074/jbc.M112.397125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Romero-Tlalolini MA, Chavez Olmos P, Garrido E. Differential DNA methylation patterns in the CD86 gene controls its constitutive expression in keratinocytes. Biochem Biophys Res Commun. 2013;438:54–60. doi: 10.1016/j.bbrc.2013.07.023. [DOI] [PubMed] [Google Scholar]
- Rubina K, Kalinina N, Potekhina A, Efimenko A, Semina E, Poliakov A, Wilkinson DG, Parfyonova Y, Tkachuk V. T-cadherin suppresses angiogenesis in vivo by inhibiting migration of endothelial cells. Angiogenesis. 2007;10:183–195. doi: 10.1007/s10456-007-9072-2. [DOI] [PubMed] [Google Scholar]
- Ruebel KH, Jin L, Zhang S, Scheithauer BW, Lloyd RV. Inactivation of the p16 gene in human pituitary nonfunctioning tumors by hypermethylation is more common in null cell adenomas. Endocr Pathol. 2001;12:281–289. doi: 10.1385/ep:12:3:281. [DOI] [PubMed] [Google Scholar]
- Ruebel KH, Jin L, Qian X, Scheithauer BW, Kovacs K, Nakamura N, Zhang H, Raz A, Lloyd RV. Effects of DNA methylation on galectin-3 expression in pituitary tumors. Cancer Res. 2005;65:1136–1140. doi: 10.1158/0008-5472.CAN-04-3578. [DOI] [PubMed] [Google Scholar]
- Saeger W, Schreiber S, Ludecke DK. Cyclins D1 and D3 and topoisomerase II alpha in inactive pituitary adenomas. Endocr Pathol. 2001;12:39–47. doi: 10.1385/ep:12:1:39. [DOI] [PubMed] [Google Scholar]
- Salehi F, Scheithauer BW, Kros JM, Lau Q, Fealey M, Erickson D, Kovacs K, Horvath E, Lloyd RV. MGMT promoter methylation and immunoexpression in aggressive pituitary adenomas and carcinomas. J Neurooncol. 2011;104:647–657. doi: 10.1007/s11060-011-0532-6. [DOI] [PubMed] [Google Scholar]
- Schmidt MC, Henke RT, Stangl AP, Meyer-Puttlitz B, Stoffel-Wagner B, Schramm J, von Deimling A. Analysis of the MEN1 gene in sporadic pituitary adenomas. J Pathol. 1999;188:168–173. doi: 10.1002/(SICI)1096-9896(199906)188:2<168::AID-PATH342>3.0.CO;2-4. [DOI] [PubMed] [Google Scholar]
- Schnepp RW, Hou Z, Wang H, Petersen C, Silva A, Masai H, Hua X. Functional interaction between tumor suppressor menin and activator of S-phase kinase. Cancer Res. 2004;64:6791–6796. doi: 10.1158/0008-5472.CAN-04-0724. [DOI] [PubMed] [Google Scholar]
- Schnepp RW, Chen YX, Wang H, Cash T, Silva A, Diehl JA, Brown E, Hua X. Mutation of tumor suppressor gene Men1 acutely enhances proliferation of pancreatic islet cells. Cancer Res. 2006;66:5707–5715. doi: 10.1158/0008-5472.CAN-05-4518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seemann N, Kuhn D, Wrocklage C, Keyvani K, Hackl W, Buchfelder M, Fahlbusch R, Paulus W. CDKN2A/p16 inactivation is related to pituitary adenoma type and size. J Pathol. 2001;193:491–497. doi: 10.1002/path.833. [DOI] [PubMed] [Google Scholar]
- Sharma S, Salehi F, Scheithauer BW, Rotondo F, Syro LV, Kovacs K. Role of MGMT in tumor development, progression, diagnosis, treatment and prognosis. Anticancer Res. 2009;29:3759–3768. [PubMed] [Google Scholar]
- Sherr CJ, Roberts JM. CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev. 1999;13:1501–1512. doi: 10.1101/gad.13.12.1501. [DOI] [PubMed] [Google Scholar]
- Shi K, Parekh VI, Roy S, Desai SS, Agarwal SK. The embryonic transcription factor Hlxb9 is a menin interacting partner that controls pancreatic beta-cell proliferation and the expression of insulin regulators. Endocr Relat Cancer. 2013;20:111–122. doi: 10.1530/ERC-12-0077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shirai YT, Ehata S, Yashiro M, Yanagihara K, Hirakawa K, Miyazono K. Bone morphogenetic protein-2 and -4 play tumor suppressive roles in human diffuse-type gastric carcinoma. Am J Pathol. 2011;179:2920–2930. doi: 10.1016/j.ajpath.2011.08.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shivakumar L, Minna J, Sakamaki T, Pestell R, White MA. The RASSF1A tumor suppressor blocks cell cycle progression and inhibits cyclin D1 accumulation. Mol Cell Biol. 2002;22:4309–4318. doi: 10.1128/MCB.22.12.4309-4318.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simpson DJ, Bicknell JE, McNicol AM, Clayton RN, Farrell WE. Hypermethylation of the p16/CDKN2A/MTSI gene and loss of protein expression is associated with nonfunctional pituitary adenomas but not somatotrophinomas. Genes Chromosom Cancer. 1999;24:328–336. [PubMed] [Google Scholar]
- Simpson DJ, Hibberts NA, McNicol AM, Clayton RN, Farrell WE. Loss of pRb expression in pituitary adenomas is associated with methylation of the RB1 CpG Island. Cancer Res. 2000;60:1211–1216. [PubMed] [Google Scholar]
- Simpson DJ, Frost SJ, Bicknell JE, Broome JC, McNicol AM, Clayton RN, Farrell WE. Aberrant expression of G(1)/S regulators is a frequent event in sporadic pituitary adenomas. Carcinogenesis. 2001;22:1149–1154. doi: 10.1093/carcin/22.8.1149. [DOI] [PubMed] [Google Scholar]
- Simpson DJ, McNicol AM, Murray DC, Bahar A, Turner HE, Wass JA, Esiri MM, Clayton RN, Farrell WE. Molecular pathology shows p16 methylation in nonadenomatous pituitaries from patients with Cushing's disease. Clin Cancer Res. 2004;10:1780–1788. doi: 10.1158/1078-0432.ccr-1127-3. [DOI] [PubMed] [Google Scholar]
- Song MS, Song SJ, Ayad NG, Chang JS, Lee JH, Hong HK, Lee H, Choi N, Kim J, Kim H, Kim JW, Choi EJ, Kirschner MW, Lim DS. The tumour suppressor RASSF1A regulates mitosis by inhibiting the APC-Cdc20 complex. Nat Cell Biol. 2004;6:129–137. doi: 10.1038/ncb1091. [DOI] [PubMed] [Google Scholar]
- Song MS, Song SJ, Kim SY, Oh HJ, Lim DS. The tumour suppressor RASSF1A promotes MDM2 self-ubiquitination by disrupting the MDM2-DAXX-HAUSP complex. EMBO J. 2008;27:1863–1874. doi: 10.1038/emboj.2008.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soto E, Yanagisawa M, Marlow LA, Copland JA, Perez EA, Anastasiadis PZ. P120 catenin induces opposing effects on tumor cell growth depending on E-cadherin expression. J Cell Biol. 2008;183:737–749. doi: 10.1083/jcb.200805113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soufir N, Avril MF, Chompret A, Demenais F, Bombled J, Spatz A, Stoppa-Lyonnet D, Benard J, Bressac-de Paillerets B. Prevalence of p16 and CDK4 germline mutations in 48 melanoma-prone families in France. The French Familial Melanoma Study Group. Hum Mol Genet. 1998;7:209–216. doi: 10.1093/hmg/7.2.209. [DOI] [PubMed] [Google Scholar]
- Soussi T. TP53 mutations in human cancer: database reassessment and prospects for the next decade. Adv Cancer Res. 2011;110:107–139. doi: 10.1016/B978-0-12-386469-7.00005-0. [DOI] [PubMed] [Google Scholar]
- Spengler D, Villalba M, Hoffmann A, Pantaloni C, Houssami S, Bockaert J, Journot L. Regulation of apoptosis and cell cycle arrest by Zac1, a novel zinc finger protein expressed in the pituitary gland and the brain. EMBO J. 1997;16:2814–2825. doi: 10.1093/emboj/16.10.2814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sperka T, Wang J, Rudolph KL. DNA damage checkpoints in stem cells, ageing and cancer. Nat Rev Mol Cell Biol. 2013;13:579–590. doi: 10.1038/nrm3420. [DOI] [PubMed] [Google Scholar]
- St Croix B, Sheehan C, Rak JW, Florenes VA, Slingerland JM, Kerbel RS. E-Cadherin-dependent growth suppression is mediated by the cyclin-dependent kinase inhibitor p27(KIP1) J Cell Biol. 1998;142:557–571. doi: 10.1083/jcb.142.2.557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stalberg P, Grimfjard P, Santesson M, Zhou Y, Lindberg D, Gobl A, Oberg K, Westin G, Rastad J, Wang S, Skogseid B. Transfection of the multiple endocrine neoplasia type 1 gene to a human endocrine pancreatic tumor cell line inhibits cell growth and affects expression of JunD, delta-like protein 1/preadipocyte factor-1, proliferating cell nuclear antigen, and QM/Jif-1. J Clin Endocrinol Metab. 2004;89:2326–2337. doi: 10.1210/jc.2003-031228. [DOI] [PubMed] [Google Scholar]
- Su D, Peng X, Zhu S, Huang Y, Dong Z, Zhang Y, Zhang J, Liang Q, Lu J, Huang B. Role of p38 MAPK pathway in BMP4-mediated Smaddependent premature senescence in lung cancer cells. Biochem J. 2011;433:333–343. doi: 10.1042/BJ20100404. [DOI] [PubMed] [Google Scholar]
- Suetake I, Shinozaki F, Miyagawa J, Takeshima H, Tajima S. DNMT3L stimulates the DNA methylation activity of Dnmt3a and Dnmt3b through a direct interaction. J Biol Chem. 2004;279:27816–27823. doi: 10.1074/jbc.M400181200. [DOI] [PubMed] [Google Scholar]
- Suliman M, Royds J, Cullen D, Timperley W, Powell T, Battersby R, Jones TH. Mdm2 and the p53 pathway in human pituitary adenomas. Clin Endocrinol (Oxf) 2001;54:317–325. doi: 10.1046/j.1365-2265.2001.01195.x. [DOI] [PubMed] [Google Scholar]
- Takeda M, Otsuka F, Suzuki J, Kishida M, Ogura T, Tamiya T, Makino H. Involvement of activin/BMP system in development of human pituitary gonadotropinomas and nonfunctioning adenomas. Biochem Biophys Res Commun. 2003;306:812–818. doi: 10.1016/s0006-291x(03)01052-0. [DOI] [PubMed] [Google Scholar]
- Takeuchi S, Koeffler HP, Hinton DR, Miyoshi I, Melmed S, Shimon I. Mutation and expression analysis of the cyclin-dependent kinase inhibitor gene p27/Kip1 in pituitary tumors. J Endocrinol. 1998;157:337–341. doi: 10.1677/joe.0.1570337. [DOI] [PubMed] [Google Scholar]
- Tamura RE, de Vasconcellos JF, Sarkar D, Libermann TA, Fisher PB, Zerbini LF. GADD45 proteins: central players in tumorigenesis. Curr Mol Med. 2012;12:634–651. doi: 10.2174/156652412800619978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tanaka C, Kimura T, Yang P, Moritani M, Yamaoka T, Yamada S, Sano T, Yoshimoto K, Itakura M. Analysis of loss of heterozygosity on chromosome 11 and infrequent inactivation of the MEN1 gene in sporadic pituitary adenomas. J Clin Endocrinol Metab. 1998;83:2631–2634. doi: 10.1210/jcem.83.8.4888. [DOI] [PubMed] [Google Scholar]
- Tanizaki Y, Jin L, Scheithauer BW, Kovacs K, Roncaroli F, Lloyd RV. P53 gene mutations in pituitary carcinomas. Endocr Pathol. 2007;18:217–222. doi: 10.1007/s12022-007-9006-y. [DOI] [PubMed] [Google Scholar]
- Tateno T, Zhu X, Asa SL, Ezzat S. Chromatin remodeling and histone modifications in pituitary tumors. Mol Cell Endocrinol. 2010;326:66–70. doi: 10.1016/j.mce.2009.12.028. [DOI] [PubMed] [Google Scholar]
- Teodoro JG, Evans SK, Green MR. Inhibition of tumor angiogenesis by p53: a new role for the guardian of the genome. J Mol Med. 2007;85:1175–1186. doi: 10.1007/s00109-007-0221-2. [DOI] [PubMed] [Google Scholar]
- Thakker RV, Bouloux P, Wooding C, Chotai K, Broad PM, Spurr NK, Besser GM, O'Riordan JL. Association of parathyroid tumors in multiple endocrine neoplasia type 1 with loss of alleles on chromosome 11. N Engl J Med. 1989;321:218–224. doi: 10.1056/NEJM198907273210403. [DOI] [PubMed] [Google Scholar]
- Thaler S, Hahnel PS, Schad A, Dammann R, Schuler M. RASSF1A mediates p21Cip1/Waf1-dependent cell cycle arrest and senescence through modulation of the Raf-MEK-ERK pathway and inhibition of Akt. Cancer Res. 2009;69:1748–1757. doi: 10.1158/0008-5472.CAN-08-1377. [DOI] [PubMed] [Google Scholar]
- Thaler S, Schmidt M, Schad A, Sleeman JP. RASSF1A inhibits estrogen receptor alpha expression and estrogen-independent signalling: implications for breast cancer development. Oncogene. 2012;31:4912–4922. doi: 10.1038/onc.2011.658. [DOI] [PubMed] [Google Scholar]
- Theodoropoulou M, Zhang J, Laupheimer S, Paez-Pereda M, Erneux C, Florio T, Pagotto U, Stalla GK. Octreotide, a somatostatin analogue, mediates its antiproliferative action in pituitary tumor cells by altering phosphatidylinositol 3-kinase signaling and inducing Zac1 expression. Cancer Res. 2006;66:1576–1582. doi: 10.1158/0008-5472.CAN-05-1189. [DOI] [PubMed] [Google Scholar]
- Tichomirowa MA, Barlier A, Daly AF, Jaffrain-Rea ML, Ronchi C, Yaneva M, Urban JD, Petrossians P, Elenkova A, Tabarin A, Desailloud R, Maiter D, Schurmeyer T, Cozzi R, Theodoropoulou M, Sievers C, Bernabeu I, Naves LA, Chabre O, Montanana CF, Hana V, Halaby G, Delemer B, Aizpun JI, Sonnet E, Longas AF, Hagelstein MT, Caron P, Stalla GK, Bours V, Zacharieva S, Spada A, Brue T, Beckers A. High prevalence of AIP gene mutations following focused screening in young patients with sporadic pituitary macroadenomas. Eur J Endocrinol. 2011;165:509–515. doi: 10.1530/EJE-11-0304. [DOI] [PubMed] [Google Scholar]
- Tiwari N, Gheldof A, Tatari M, Christofori G. EMT as the ultimate survival mechanism of cancer cells. Semin Cancer Biol. 2012;22:194–207. doi: 10.1016/j.semcancer.2012.02.013. [DOI] [PubMed] [Google Scholar]
- Trivellin G, Korbonits M. AIP and its interacting partners. J Endocrinol. 2011;210:137–155. doi: 10.1530/JOE-11-0054. [DOI] [PubMed] [Google Scholar]
- Trump D, Farren B, Wooding C, Pang JT, Besser GM, Buchanan KD, Edwards CR, Heath DA, Jackson CE, Jansen S, Lips K, Monson JP, O'Halloran D, Sampson J, Shalet SM, Wheeler MH, Zink A, Thakker RV. Clinical studies of multiple endocrine neoplasia type 1 (MEN1) QJM. 1996;89:653–669. doi: 10.1093/qjmed/89.9.653. [DOI] [PubMed] [Google Scholar]
- Tryndyak VP, Beland FA, Pogribny IP. E-cadherin transcriptional downregulation by epigenetic and microRNA-200 family alterations is related to mesenchymal and drug-resistant phenotypes in human breast cancer cells. Int J Cancer. 2010;126:2575–2583. doi: 10.1002/ijc.24972. [DOI] [PubMed] [Google Scholar]
- Vairapandi M, Balliet AG, Fornace AJ, Jr, Hoffman B, Liebermann DA. The differentiation primary response gene MyD118, related to GADD45, encodes for a nuclear protein which interacts with PCNA and p21WAF1/CIP1. Oncogene. 1996;12:2579–2594. [PubMed] [Google Scholar]
- Vairapandi M, Balliet AG, Hoffman B, Liebermann DA. GADD45b and GADD45g are cdc2/cyclinB1 kinase inhibitors with a role in S and G2/M cell cycle checkpoints induced by genotoxic stress. J Cell Physiol. 2002;192:327–338. doi: 10.1002/jcp.10140. [DOI] [PubMed] [Google Scholar]
- Valleley EM, Cordery SF, Carr IM, MacLennan KA, Bonthron DT. Loss of expression of ZAC/PLAGL1 in diffuse large B-cell lymphoma is independent of promoter hypermethylation. Genes Chromosom Cancer. 2010;49:480–486. doi: 10.1002/gcc.20758. [DOI] [PubMed] [Google Scholar]
- van der Weyden L, Adams DJ. The Ras-association domain family (RASSF) members and their role in human tumourigenesis. Biochim Biophys Acta. 2007;1776:58–85. doi: 10.1016/j.bbcan.2007.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Roy F, Berx G. The cell-cell adhesion molecule E-cadherin. Cell Mol Life Sci. 2008;65:3756–3788. doi: 10.1007/s00018-008-8281-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vax VV, Bibi R, Diaz-Cano S, Gueorguiev M, Kola B, Borboli N, Bressac-de Paillerets B, Walker GJ, Dedov II, Grossman AB, Korbonits M. Activating point mutations in cyclin-dependent kinase 4 are not seen in sporadic pituitary adenomas, insulinomas or Leydig cell tumours. J Endocrinol. 2003;178:301–310. doi: 10.1677/joe.0.1780301. [DOI] [PubMed] [Google Scholar]
- Verges B, Boureille F, Goudet P, Murat A, Beckers A, Sassolas G, Cougard P, Chambe B, Montvernay C, Calender A. Pituitary disease in MEN type 1 (MEN1): data from the France–Belgium MEN1 multicenter study. J Clin Endocrinol Metab. 2002;87:457–465. doi: 10.1210/jcem.87.2.8145. [DOI] [PubMed] [Google Scholar]
- Vierimaa O, Georgitsi M, Lehtonen R, Vahteristo P, Kokko A, Raitila A, Tuppurainen K, Ebeling TM, Salmela PI, Paschke R, Gundogdu S, De Menis E, Makinen MJ, Launonen V, Karhu A, Aaltonen LA. Pituitary adenoma predisposition caused by germline mutations in the AIP gene. Science. 2006;312:1228–1230. doi: 10.1126/science.1126100. [DOI] [PubMed] [Google Scholar]
- Vleminckx K, Vakaet L, Jr, Mareel M, Fiers W, van Roy F. Genetic manipulation of E-cadherin expression by epithelial tumor cells reveals an invasion suppressor role. Cell. 1991;66:107–119. doi: 10.1016/0092-8674(91)90143-m. [DOI] [PubMed] [Google Scholar]
- Vousden KH, Prives C. Blinded by the light: the growing complexity of p53. Cell. 2009;137:413–431. doi: 10.1016/j.cell.2009.04.037. [DOI] [PubMed] [Google Scholar]
- Wade M, Wang YV, Wahl GM. The p53 orchestra: Mdm2 and Mdmx set the tone. Trends Cell Biol. 2010;20:299–309. doi: 10.1016/j.tcb.2010.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walls GV, Lemos MC, Javid M, Bazan-Peregrino M, Jeyabalan J, Reed AA, Harding B, Tyler DJ, Stuckey DJ, Piret S, Christie PT, Ansorge O, Clarke K, Seymour L, Thakker RV. MEN1 gene replacement therapy reduces proliferation rates in a mouse model of pituitary adenomas. Cancer Res. 2012;72:5060–5068. doi: 10.1158/0008-5472.CAN-12-1821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang SP, Wang WL, Chang YL, Wu CT, Chao YC, Kao SH, Yuan A, Lin CW, Yang SC, Chan WK, Li KC, Hong TM, Yang PC. P53 controls cancer cell invasion by inducing the MDM2-mediated degradation of Slug. Nat Cell Biol. 2009;11:694–704. doi: 10.1038/ncb1875. [DOI] [PubMed] [Google Scholar]
- Wang WM, Liu ST, Huang SM, Lin WS, Chen SG, Chang YL. Zac1 functional interactions mediate AP-1 transcriptional activity. Biochim Biophys Acta. 2011;1813:2050–2060. doi: 10.1016/j.bbamcr.2011.08.005. [DOI] [PubMed] [Google Scholar]
- Warfel NA, El-Deiry WS. P21WAF1 and tumourigenesis: 20 years after. Curr Opin Oncol. 2013;25:52–58. doi: 10.1097/CCO.0b013e32835b639e. [DOI] [PubMed] [Google Scholar]
- Warzecha CC, Sato TK, Nabet B, Hogenesch JB, Carstens RP. ESRP1 and ESRP2 are epithelial cell-type-specific regulators of FGFR2 splicing. Mol Cell. 2009;33:591–601. doi: 10.1016/j.molcel.2009.01.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Warzecha CC, Jiang P, Amirikian K, Dittmar KA, Lu H, Shen S, Guo W, Xing Y, Carstens RP. An ESRP-regulated splicing programme is abrogated during the epithelial–mesenchymal transition. EMBO J. 2010;29:3286–3300. doi: 10.1038/emboj.2010.195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wenbin C, Asai A, Teramoto A, Sanno N, Kirino T. Mutations of the MEN1 tumor suppressor gene in sporadic pituitary tumors. Cancer Lett. 1999;142:43–47. doi: 10.1016/s0304-3835(99)00111-1. [DOI] [PubMed] [Google Scholar]
- Whitson RJ, Lucia MS, Lambert JR. Growth differentiation factor-15 (GDF-15) suppresses in vitro angiogenesis through a novel interaction with connective tissue growth factor (CCN2) J Cell Biochem. 2013;114:1424–1433. doi: 10.1002/jcb.24484. [DOI] [PubMed] [Google Scholar]
- Williams BO, Remington L, Albert DM, Mukai S, Bronson RT, Jacks T. Cooperative tumorigenic effects of germline mutations in Rb and p53. Nat Genet. 1994;7:480–484. doi: 10.1038/ng0894-480. [DOI] [PubMed] [Google Scholar]
- Woloschak M, Yu A, Xiao J, Post KD. Frequent loss of the P16INK4a gene product in human pituitary tumors. Cancer Res. 1996;56:2493–2496. [PubMed] [Google Scholar]
- Woloschak M, Yu A, Post KD. Frequent inactivation of the p16 gene in human pituitary tumors by gene methylation. Mol Carcinog. 1997;19:221–224. doi: 10.1002/(sici)1098-2744(199708)19:4<221::aid-mc1>3.0.co;2-f. [DOI] [PubMed] [Google Scholar]
- Wong PS, Li W, Vogel CF, Matsumura F. Characterization of MCF mammary epithelial cells overexpressing the Arylhydrocarbon receptor (AhR) BMC Cancer. 2009;9:234. doi: 10.1186/1471-2407-9-234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu T, Zhang X, Huang X, Yang Y, Hua X. Regulation of cyclin B2 expression and cell cycle G2/m transition by menin. J Biol Chem. 2010;285:18291–18300. doi: 10.1074/jbc.M110.106575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu B, Sano T, Yoshimoto K, Yamada S. Downregulation of E-cadherin and its undercoat proteins in pituitary growth hormone cell adenomas with prominent fibrous bodies. Endocr Pathol. 2002;13:341–351. doi: 10.1385/ep:13:4:341. [DOI] [PubMed] [Google Scholar]
- Yacqub-Usman K, Duong CV, Clayton RN, Farrell WE. Epigenomic silencing of the BMP-4 gene in pituitary adenomas: a potential target for epidrug-induced re-expression. Endocrinology. 2012a;153:3603–3612. doi: 10.1210/en.2012-1231. [DOI] [PubMed] [Google Scholar]
- Yacqub-Usman K, Richardson A, Duong CV, Clayton RN, Farrell WE. The pituitary tumour epigenome: aberrations and prospects for targeted therapy. Nat Rev Endocrinol. 2012b;8:486–494. doi: 10.1038/nrendo.2012.54. [DOI] [PubMed] [Google Scholar]
- Yin XF, Chen J, Mao W, Wang YH, Chen MH. Downregulation of aryl hydrocarbon receptor expression decreases gastric cancer cell growth and invasion. Oncol Rep. 2013;30:364–370. doi: 10.3892/or.2013.2410. [DOI] [PubMed] [Google Scholar]
- Yokoyama A, Cleary ML. Menin critically links MLL proteins with LEDGF on cancer-associated target genes. Cancer Cell. 2008;14:36–46. doi: 10.1016/j.ccr.2008.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yokoyama A, Wang Z, Wysocka J, Sanyal M, Aufiero DJ, Kitabayashi I, Herr W, Cleary ML. Leukemia proto-oncoprotein MLL forms a SET1-like histone methyltransferase complex with menin to regulate Hox gene expression. Mol Cell Biol. 2004;24:5639–5649. doi: 10.1128/MCB.24.13.5639-5649.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yoshino A, Katayama Y, Ogino A, Watanabe T, Yachi K, Ohta T, Komine C, Yokoyama T, Fukushima T. Promoter hypermethylation profile of cell cycle regulator genes in pituitary adenomas. J Neurooncol. 2007;83:153–162. doi: 10.1007/s11060-006-9316-9. [DOI] [PubMed] [Google Scholar]
- You JS, Jones PA. Cancer genetics and epigenetics: two sides of the same coin? Cancer Cell. 2012;22:9–20. doi: 10.1016/j.ccr.2012.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu R, Bonert V, Saporta I, Raffel LJ, Melmed S. Aryl hydrocarbon receptor interacting protein variants in sporadic pituitary adenomas. J Clin Endocrinol Metab. 2006;91:5126–5129. doi: 10.1210/jc.2006-1731. [DOI] [PubMed] [Google Scholar]
- Zahedi A, Booth GL, Smyth HS, Farrell WE, Clayton RN, Asa SL, Ezzat S. Distinct clonal composition of primary and metastatic adrencorticotrophic hormone-producing pituitary carcinoma. Clin Endocrinol (Oxf) 2001;55:549–556. doi: 10.1046/j.1365-2265.2001.01322.x. [DOI] [PubMed] [Google Scholar]
- Zhang X, Sun H, Danila DC, Johnson SR, Zhou Y, Swearingen B, Klibanski A. Loss of expression of GADD45 gamma, a growth inhibitory gene, in human pituitary adenomas: implications for tumorigenesis. J Clin Endocrinol Metab. 2002;87:1262–1267. doi: 10.1210/jcem.87.3.8315. [DOI] [PubMed] [Google Scholar]
- Zhang X, Zhou Y, Mehta KR, Danila DC, Scolavino S, Johnson SR, Klibanski A. A pituitary-derived MEG3 isoform functions as a growth suppressor in tumor cells. J Clin Endocrinol Metab. 2003;88:5119–5126. doi: 10.1210/jc.2003-030222. [DOI] [PubMed] [Google Scholar]
- Zhang X, Rice K, Wang Y, Chen W, Zhong Y, Nakayama Y, Zhou Y, Klibanski A. Maternally expressed gene 3 (MEG3) noncoding ribonucleic acid: isoform structure, expression, and functions. Endocrinology. 2010;151:939–947. doi: 10.1210/en.2009-0657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang J, Li H, Yu JP, Wang SE, Ren XB. Role of SOCS1 in tumor progression and therapeutic application. Int J Cancer. 2012;130:1971–1980. doi: 10.1002/ijc.27318. [DOI] [PubMed] [Google Scholar]
- Zhao J, Dahle D, Zhou Y, Zhang X, Klibanski A. Hypermethylation of the promoter region is associated with the loss of MEG3 gene expression in human pituitary tumors. J Clin Endocrinol Metab. 2005;90:2179–2186. doi: 10.1210/jc.2004-1848. [DOI] [PubMed] [Google Scholar]
- Zhao J, Zhang X, Zhou Y, Ansell PJ, Klibanski A. Cyclic AMP stimulates MEG3 gene expression in cells through a cAMP-response element (CRE) in the MEG3 proximal promoter region. Int J Biochem Cell Biol. 2006;38:1808–1820. doi: 10.1016/j.biocel.2006.05.004. [DOI] [PubMed] [Google Scholar]
- Zhou Y, Zhong Y, Wang Y, Zhang X, Batista DL, Gejman R, Ansell PJ, Zhao J, Weng C, Klibanski A. Activation of p53 by MEG3 non-coding RNA. J Biol Chem. 2007;282:24731–24742. doi: 10.1074/jbc.M702029200. [DOI] [PubMed] [Google Scholar]
- Zhou Y, Cheunsuchon P, Nakayama Y, Lawlor MW, Zhong Y, Rice KA, Zhang L, Zhang X, Gordon FE, Lidov HG, Bronson RT, Klibanski A. Activation of paternally expressed genes and perinatal death caused by deletion of the Gtl2 gene. Development. 2010;137:2643–2652. doi: 10.1242/dev.045724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou Y, Zhang X, Klibanski A. MEG3 noncoding RNA: a tumor suppressor. J Mol Endocrinol. 2012;48:R45–53. doi: 10.1530/JME-12-0008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu X, Asa SL, Ezzat S. Ikaros is regulated through multiple histone modifications and deoxyribonucleic acid methylation in the pituitary. Mol Endocrinol. 2007a;21:1205–1215. doi: 10.1210/me.2007-0053. [DOI] [PubMed] [Google Scholar]
- Zhu X, Lee K, Asa SL, Ezzat S. Epigenetic silencing through DNA and histone methylation of fibroblast growth factor receptor 2 in neoplastic pituitary cells. Am J Pathol. 2007b;170:1618–1628. doi: 10.2353/ajpath.2007.061111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu X, Mao X, Hurren R, Schimmer AD, Ezzat S, Asa SL. Deoxyribonucleic acid methyltransferase 3B promotes epigenetic silencing through histone 3 chromatin modifications in pituitary cells. J Clin Endocrinol Metab. 2008;93:3610–3617. doi: 10.1210/jc.2008-0578. [DOI] [PubMed] [Google Scholar]
- Zhuang Z, Ezzat SZ, Vortmeyer AO, Weil R, Oldfield EH, Park WS, Pack S, Huang S, Agarwal SK, Guru SC, Manickam P, Debelenko LV, Kester MB, Olufemi SE, Heppner C, Crabtree JS, Burns AL, Spiegel AM, Marx SJ, Chandrasekharappa SC, Collins FS, Emmert-Buck MR, Liotta LA, Asa SL, Lubensky IA. Mutations of the MEN1 tumor suppressor gene in pituitary tumors. Cancer Res. 1997;57:5446–5451. [PubMed] [Google Scholar]
- Zuo L, Weger J, Yang Q, Goldstein AM, Tucker MA, Walker GJ, Hayward N, Dracopoli NC. Germline mutations in the p16INK4a binding domain of CDK4 in familial melanoma. Nat Genet. 1996;12:97–99. doi: 10.1038/ng0196-97. [DOI] [PubMed] [Google Scholar]
