role of the aryl hydrocarbon receptor-interacting protein in

15
681 Review www.expert-reviews.com ISSN 1744-6651 © 2010 Expert Reviews Ltd 10.1586/EEM.10.42 Overview of pituitary adenoma Pituitary adenomas are benign, monoclonal neoplasms of the anterior pituitary gland, accounting for approximately 10–15% of intracranial neoplasms [1,2] . A meta-analysis by Ezzat et al. in 2004 showed that pituitary adenomas occur with a frequency of 14.4% (range: 1–35%) and 22.5% (range: 1–40%) in pooled autopsy and radiological series, respectively [3] . This study indicated that cur- rent prevalence figures are significantly skewed by under-diagnosis of preclinical adenomas. A recent study on symptomatic pituitary adeno- mas in Liege, Belgium, provides an indication of the incidence of clinically apparent pitu- itary adenomas, showing a prevalence of one in 1064, a figure three- to five-times higher than earlier estimates [4] . This figure has been verified in a more recent study in Banbury, UK, which estimated a similar prevalence of one in 1288 individuals [5] . Two thirds of pituitary adenomas produce an excess of endogenous hormones, while a third are clinically nonfunctioning adenomas presenting owing to symptoms of local expansion and loss of endocrine function [6,7] . Pituitary adenomas are largely sporadic, with approximately 5% of pituitary adenomas being familial in origin [8] . Approximately 3% of pituitary adenomas arise due to mutations in the multiple endocrine neoplasia type 1 (MEN1) gene or owing to the Carney complex (CNC) [9,10] . The MEN1 gene is located on chromosome 11q13 and acts as a tumor-suppressor gene, encoding the nuclear protein menin [10] . Mutations typically result in a truncated menin protein. Approximately 40% of those possessing a mutated MEN1 gene develop pituitary adenomas [11] , although almost all carrier subjects develop at least one feature of the disease by the age of 50 years [12,13] . More recently, a MEN1-like syndrome has been reported relating to mutations in the Joshua W Cain 1 , Dragana Miljic 2 , Vera Popovic 2 and Márta Korbonits †1 1 Department of Endocrinology, Barts and the London School of Medicine, Queen Mary University of London, EC1M 6BQ, UK 2 Institute of Endocrinology, School of Medicine, University Belgrade Belgrade, Serbia Author for correspondence: Tel.: +44 207 882 6238 Fax: +44 207 882 6197 [email protected] Pituitary adenomas are typically sporadic benign tumors. However, approximately 5% of cases have been found to be familial in origin. Of these, approximately 40% occur in the absence of multiple endocrine neoplasia type 1 or Carney complex and have been termed ‘familial isolated pituitary adenoma’ (FIPA). Recently, germline mutations in the aryl hydrocarbon receptor- interacting protein ( AIP ) gene have been described in 15–20% of these families, identifying an autosomal dominant condition with incomplete penetrance termed ‘pituitary adenoma predisposition’. Pituitary adenoma predisposition cohorts show a marked disposition to develop large, aggressive somatotroph, somatolactotroph or lactotroph adenomas, typically presenting at a young age. AIP mutation families have a distinct clinical phenotype compared with AIP mutation-negative FIPA families. Current evidence suggests that AIP is a tumor-suppressor gene. AIP has been demonstrated to interact with a number of cellular proteins, including several nuclear receptors, heat-shock protein 90 and survivin, although the mechanism of the tumor-suppressor effect is unknown. This article summarizes available data regarding the role of AIP in pituitary tumorigenesis and the clinical features of FIPA. KEYWORDS: AhR • AIP • aryl hydrocarbon receptor • aryl hydrocarbon receptor-interacting protein • familial isolated pituitary adenoma • FIPA • PAP • pituitary adenoma • pituitary adenoma predisposition • tumor-suppressor gene Role of the aryl hydrocarbon receptor-interacting protein in familial isolated pituitary adenoma Expert Rev. Endocrinol. Metab. 5(5), 681–695 (2010) For reprint orders, please contact [email protected]

Upload: others

Post on 12-Sep-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Role of the aryl hydrocarbon receptor-interacting protein in

681

Review

www.expert-reviews.com ISSN 1744-6651© 2010 Expert Reviews Ltd10.1586/EEM.10.42

Overview of pituitary adenomaPituitary adenomas are benign, monoclonal neoplasms of the anterior pituitary gland, accounting for approximately 10–15% of intracranial neoplasms [1,2]. A meta-ana lysis by Ezzat et al. in 2004 showed that pituitary adenomas occur with a frequency of 14.4% (range: 1–35%) and 22.5% (range: 1–40%) in pooled autopsy and radiological series, respectively [3]. This study indicated that cur-rent prevalence figures are significantly skewed by under-diagnosis of preclinical adenomas. A recent study on symptomatic pituitary adeno-mas in Liege, Belgium, provides an indication of the incidence of clinically apparent pitu-itary adenomas, showing a prevalence of one in 1064, a figure three- to five-times higher than earlier estimates [4]. This figure has been verified in a more recent study in Banbury, UK, which estimated a similar prevalence of one in 1288 individuals [5].

Two thirds of pituitary adenomas produce an excess of endogenous hormones, while a third are clinically nonfunctioning adenomas presenting owing to symptoms of local expansion and loss of endocrine function [6,7]. Pituitary adenomas are largely sporadic, with approximately 5% of pituitary adenomas being familial in origin [8]. Approximately 3% of pituitary adenomas arise due to mutations in the multiple endocrine neoplasia type 1 (MEN1) gene or owing to the Carney complex (CNC) [9,10]. The MEN1 gene is located on chromosome 11q13 and acts as a tumor-suppressor gene, encoding the nuclear protein menin [10]. Mutations typically result in a truncated menin protein. Approximately 40% of those possessing a mutated MEN1 gene develop pituitary adenomas [11], although almost all carrier subjects develop at least one feature of the disease by the age of 50 years [12,13]. More recently, a MEN1-like syndrome has been reported relating to mutations in the

Joshua W Cain1, Dragana Miljic2, Vera Popovic2 and Márta Korbonits†1

1Department of Endocrinology, Barts and the London School of Medicine, Queen Mary University of London, EC1M 6BQ, UK 2Institute of Endocrinology, School of Medicine, University Belgrade Belgrade, Serbia †Author for correspondence:Tel.: +44 207 882 6238 Fax: +44 207 882 6197 [email protected]

Pituitary adenomas are typically sporadic benign tumors. However, approximately 5% of cases have been found to be familial in origin. Of these, approximately 40% occur in the absence of multiple endocrine neoplasia type 1 or Carney complex and have been termed ‘familial isolated pituitary adenoma’ (FIPA). Recently, germline mutations in the aryl hydrocarbon receptor-interacting protein (AIP) gene have been described in 15–20% of these families, identifying an autosomal dominant condition with incomplete penetrance termed ‘pituitary adenoma predisposition’. Pituitary adenoma predisposition cohorts show a marked disposition to develop large, aggressive somatotroph, somatolactotroph or lactotroph adenomas, typically presenting at a young age. AIP mutation families have a distinct clinical phenotype compared with AIP mutation-negative FIPA families. Current evidence suggests that AIP is a tumor-suppressor gene. AIP has been demonstrated to interact with a number of cellular proteins, including several nuclear receptors, heat-shock protein 90 and survivin, although the mechanism of the tumor-suppressor effect is unknown. This article summarizes available data regarding the role of AIP in pituitary tumorigenesis and the clinical features of FIPA.

Keywords: AhR • AIP • aryl hydrocarbon receptor • aryl hydrocarbon receptor-interacting protein • familial isolated pituitary adenoma • FIPA • PAP • pituitary adenoma • pituitary adenoma predisposition • tumor-suppressor gene

Role of the aryl hydrocarbon receptor-interacting protein in familial isolated pituitary adenomaExpert Rev. Endocrinol. Metab. 5(5), 681–695 (2010)

For reprint orders, please contact [email protected]

Page 2: Role of the aryl hydrocarbon receptor-interacting protein in

682

Review

Expert Rev. Endocrinol. Metab. 5(5), (2010)

Cain, Miljic, Popovic & Korbonits

cyclin-dependent kinase inhibitor 1B (CDKN1B) gene at chro-mosome 12p13 [14]; however, this syndrome has only been found in five families to date [14–17]. CNC has a significantly lower prevalence than MEN1, with only 500 cases having been identi-fied to date [18]. Approximately 60% of CNCs arise owing to a mutation in the protein kinase A 1A regulatory subunit gene (PRKAR1A) located on chromosome 17q22–24 or owing to a currently unknown gene at the chromosome 2p16 locus [19].

Pituitary adenoma patients with an established family history of pituitary adenomas are classified as having familial isolated pituitary adenoma (FIPA) in the absence of features of either MEN1 or CNC syndrome [20,21]. In 1995, Benlian et al. estab-lished that there must be a novel genetic cause for low-penetrance familial pituitary adenomas in a study assessing a FIPA family with MEN1-negative somatotropinomas. The study concluded that there was a low-penetrance causative gene close to the MEN1 locus [22]. Frohman’s and Yamada’s research group, among others, has also observed a loss of heterozygosity (LOH) at the MEN1 locus, and similarly suggested that the causative gene is distinct from MEN1 [2,23–27]. Finally, in 2006, Vierimaa et al. identified the aryl hydrocarbon receptor-interacting protein (AIP) gene at chromosome 11q13, near the MEN1 locus, as being a causative gene in some FIPA patients, and suggested the term ‘pituitary adenoma predisposition’ (PAP) to describe patients harboring AIP mutations [28]. Further studies have indicated that the AIP gene is responsible for approximately 15–20% of families with pituitary adenomas observed in FIPA cohorts, and this figure rises to 40% if FIPA data are selected only for families with two or more members with somatotropinoma (termed isolated familial somato tropinoma [24]) [8,29]. The large proportion of AIP mutation-negative FIPA families suggests that there may be more causative genes associated with FIPA yet to be identified.

Identification of the AIP geneIn 2006, two families with pituitary adenomas in Northern Finland were selected for study by Vierimaa et al. [28]. Using high-stringency criteria (considering only those with acromegaly or gigantism), a genome-wide single-nucleotide polymorphism study on 16 patients was performed to identify the gene locus. Linkage ana lysis identified linkage at chromosome 11q12–11q13, which also includes the MEN1 gene locus; however, no MEN1 mutations were detected in the cohort. Further genotyping with 36 markers in both families revealed a logarithm of the odds score of 7.1 with high-stringency criteria at chromosome 11q13, with both families sharing the linked haplotype, segregating perfectly with acromegaly. This evidence supported previously published data indicating that this locus contained a gene responsible for familial acromegaly [2,22–27].

Expression profiling on leukocyte-derived RNA was performed and the linked region was mapped with 172 probe sets. Based on these data, two candidate genes were chosen for investigation, AIP and galectin-12 (LGALS12). The team found a nonsense mutation, p.Q14X, in exon 1 of the AIP gene that segregated with disease status (although two young female patients with prolac-tinoma turned out to be phenocopies because no AIP mutation

was identified). The p.Q14X mutation was absent in 209 local blood donor controls. Interestingly, genotyping of multiple AIP-carrier families in recent publications has shown this mutation to be unique to the two Finnish families, and a small number of Finnish sporadic pituitary adenoma patients, suggesting it to be a founder mutation [30,31].

To date, 49 variants have been identified throughout the AIP gene (Table 1 & Figure 1). These mutations have included deletions, insertions, segmental duplications, nonsense and missense muta-tions [24,26–28,31–52]. Splice-site and promoter mutations have also been described (Figure 2) [28,32,33]. The most commonly occurring mutations are at the p.R304 locus where both members of a CpG site have been found to be mutated, resulting in either a non-sense (c.910C>T, p.R304X) or a missense (c.911G>A, p.R304Q) mutation [31,33,37,51]. These mutations have now been reported in multiple unrelated families and in sporadic cases, indicating a potential mutational hot-spot [29,52]. Approximately 10% of families investigated by DNA sequencing will harbor larger AIP gene deletions [31,37,51]. This indicates that genetic identification in suspected families should not be limited to the sequencing of exons, intron junctions and promoter regions, but should include a technique such as the multiplex ligation-dependent probe ampli-fication (MLPA) technique, a technique specifically designed for identification of larger gene deletions that are frequently missed by conventional sequencing [37,51,53]. Some of the variants identi-fied and listed in Table 1 & Figure 1 may be rare polymorphisms and not disease-causing mutations. Data based on ana lysis of the general population, LOH and functional studies suggest that the p.R16H [32,36,38,48,54], p.V49M [35] and p.A299V [36,51] variants may be rare single-nucleotide polymorphisms.

Analysis of AIP status in family pedigrees has indicated that PAP is a heterozygous autosomal dominant disease with variable penetrance [55]. The observed LOH at the AIP locus in affected individuals suggests the role of AIP to be a tumor-suppressor gene, with loss of the wild type allele causing PAP in concordance with the Knudson’s two-hit hypothesis [15]. All known affected indi-viduals are heterozygous for the AIP mutation, with knock-out studies thus far determining homozygote mice being nonviable owing to fatal congenital cardiovascular abnormalities occurring during embryonic development [56]. Penetrance is suggested to be incomplete at approximately 30% [39,51]; however, this figure varies widely between families [8,33].

Characteristics of AIPAIP, also called the aryl hydrocarbon receptor-associated pro-tein 9 (ARA9) [57] was initially identified via interaction with the hepatitis B virus X protein [58] and termed the ‘immuno philin-like X-associated protein 2’ (XAP2) [59]. The AIP gene consists of six exons, coding a 330 amino acid, 37-kDa protein that exhibits FK506-binding protein 12 (FKBP12) homology in the amino ter-minus [60] and contains three protein–protein interaction-mediating tetratricopeptide repeats (TPR) and an a-helix in the C-terminal region (Figure 3) [61]. Despite the FKBP12 homology domain, AIP does not exhibit the properties of an immunophilin [60], while AIP displays properties of other TPR-domain peptides such as

Page 3: Role of the aryl hydrocarbon receptor-interacting protein in

www.expert-reviews.com 683

ReviewRole of the AIP gene in familial isolated pituitary adenoma

HOP, CHIP, Cyp40, FKBP51, FKBP52 and PP5, in terms of binding numerous partners including the heat-shock protein (Hsp) 90 [57].

AIP acts as a regulator of aryl hydro-carbon receptor (AhR) activity, mediating the response to xenobiotic compounds [59]. Formation of an AIP/AhR/Hsp90-dimer/p23 complex (Figure 4) results in sta-bilization of AhR within the cytoplasm, preventing unliganded nucleocytoplas-mic shuttling and protecting AhR from ubiquitin-proteasome pathway degrada-tion [62–64]. Structural ana lysis of AIP has demonstrated that the last five amino acids at the C-terminus are vital for association with AhR. Studies have shown that any mutation leading to a truncated protein, or disruption in the terminal five amino acids, results in failure to associate with AhR [59,61,65–67] and loss of function [33]. Studies have also shown that the third TPR domain is required in the binding of both Hsp90 and AhR [59,65,66]. Although it is not known if AhR is the relevant interacting factor for the tumorigenesis role of AIP, the two terminal TPR domains and the final a-helix are probably important to binding several, if not all, known partners of AIP. Therefore, it is not surprising that of the known AIP mutations, 71% cause disrup-tion to the C-terminus of the AIP protein (Figure 1).

AIP also interacts with a number of other compounds within the cell, including PPARa [68], the glucocorticoid receptor [69], translocase of the outer membrane of mito-chondria 20 (TOMM20) [70], b-thyroid receptor (THRb1) [71], survivin [72], phospho diesterase (PDE) 4A5 and 2A [73,74], RET, a proto-oncogene receptor tyrosine kinase [75], the subtype 13 a-subunit of the G protein family (Ga13

) [60], the Epstein-Barr

virus-encoded nuclear protein (EBNA-3) [76] and Hsc70, a Hsp70 family member [70], shown to preferentially bind AIP over Hsp90 in the absence of AhR [70,77].

The aryl hydrocarbon receptorAhR is a ligand-activated member of the basic helix-loop-helix/Per-Arnt-Sim (bHLH/PAS) family of transcription fac-tors [78] that forms a tetrameric complex with AIP, a 90-kD Hsp90 dimer [67] and the co-chaperone p23 (Figure 4) [79]. AhR

Table 1. Mutations identified within the aryl hydrocarbon receptor-interacting protein gene.

AIP mutation Mutation type Ref.

Transcript Protein level

c.40C>T p.Q14X Nonsense [28,36,46]

c.64C>T p.R22X Nonsense [31]

c.70G>T p.E24X Nonsense [24,33]

c.241C>T p.R81X Nonsense [26,33,49,50]

c.424C>T p.Q142X Nonsense [32]

c.490C>T p.Q164X Nonsense [40]

c.601A>T p.K201X Nonsense [38]

c.649C>T p.Q217X Nonsense [32]

c.715C>T p.Q239X Nonsense [32]

c.721A>T p.K241X Nonsense [41]

c.804A>C p.Y268X Nonsense [43,44]

c.910C>T p.R304X Nonsense [28,32,33,38,40]

c.47G>A p.R16H Missense [32]

c.145G>A p.V49M Missense [35]

c.308A>G p.K103R Missense [41]

c.713G>A p.C238Y Missense [24,33]

c.721A>G p.K241E Missense [32]

c.769A>G p.I257V Missense [47]

c.811C>T p.R271W Missense [32,52]

c.896C>T p.A299V Missense [36]

c.911G>A p.R304Q Missense [33,36,38]

c.74_81delins7 p.L25PfsX130 fs [51]

c.286_287delGT p.V96PfsX32 fs [27,35]

p.P114fs fs [41]

c.404delA p.H135LfsX20 fs [38]

c.500delC p.P167HfsX3 fs [131]

c.517_521delGAAGA p.E174fsX47 fs [32,39]

c.542delT p.L181fsX13 fs [36]

c.662dupC p.E222X fs [51]

c.824_825insA p.H275QfsX12 fs [36]

c.854_857delAGGC p.Q285fsX16 fs [32]

c.919insC p.Q307RfsX103 fs [41]

Novel fs mutations in Ex2 fs [48]

c.100–1G>C IVS2–1G>C Splice site [36]

c.249G>T p.G83AfsX15 Splice site [51]

AIP: Aryl hydrocarbon receptor-interacting protein; del: Deletion; dup: Duplication; Ex2: Exon 2; fs: Frameshift; ins: Insertion.

Page 4: Role of the aryl hydrocarbon receptor-interacting protein in

Expert Rev. Endocrinol. Metab. 5(5), (2010)684

Review Cain, Miljic, Popovic & Korbonits

also interacts with the E3 ubiquitin ligase C-terminal of Hsp70 interacting protein (CHIP) [80], a more recently identified bind-ing partner believed to mediate ubiquitation of Hsp90 binding proteins [81], indicating a potential role in AhR regulation [82].

AhR mediates the cellular response to xenobiotic compounds such as dioxin, also known as 2,3,7,8-tetrachloro dibenzo-p-dioxin [57,58], and other hydrophobic environmental pollutants such as polycyclic aromatic hydrocarbons (PAH) – a group of over 200 compounds characterized structurally by the presence of two or more fused benzene rings [83]. PAHs are formed as a result of the incomplete combustion of fossil fuels [84–86] and have been found in cigarette smoke and charbroiled or smoked foods [87,88]. Dioxins and PAHs are known to exhibit mutagenic and carcino-genic effects [83]. AhR, when activated by PAH, is known to associate with RelB, driving the production of proinflammatory mediators such as IL-8 [89].

Prior to activation, dormant AhR is localized in the cytoplasm held in a stabilizing AhR/AIP/Hsp90/p23 complex; however, upon interaction with xenobiotic compounds, the cytoplasmic AhR/AIP/Hsp90 complex translocates to the nucleus where AhR undergoes a conformational change, detaching AhR from the protein complex [63,90–92]. The role of p23 in translo cation of AhR is yet to be identified. Within the nucleus, AhR forms a transcriptionally active dimeric complex with the AhR nuclear translator (ARNT), following which the AhR/ARNT complex

binds to DNA at the xenobiotic response element, also known as the dioxin response element, sites within the promoter region of target genes, inducing transcription of xenobiotic-metabolizing enzymes [60,93,94]. Gene targets include the cytochrome p450 (CYP) drug-metabolizing enzyme fam-ily (CYP1A1, CYP1A2 and CYP1B1), glutathionine-S-transferase, NADPH/quinine oxidoreductase and aldehyde dehydrogenase 3 [60,94,95].

However, there is evidence that AhR undergoes nucleocytoplasmic shuttling in the absence of exogenous xenobiotic compounds [64,96–98], with more recent studies suggesting that AhR undergoes nuclear translocation in the presence of cAMP [82,99]. This may be relevant because AIP has been shown to bind some phos-phodiesterases (PDEs) – a group of cAMP-degrading enzymes [73,74]. cAMP appears to interact with AhR as part of an endog-enous system that has been suggested to be important for the embryonic develop-ment of the liver and vascular system and maturation of the immune system [100–102].

Current data regarding the role of AIP in AhR regulation are conflicting, with some studies indicating that AIP increases AhR levels in the cytoplasm, increasing the tran-

scriptional activity of AhR [64,67], while other studies have shown that AIP has an inhibitory effect on AhR-induced transcription [62,103]. Additionally, there is also controversy regarding whether AhR itself is tumorigenic or inhibits cell proliferation. Numerous studies point to a tumorigenic effect [83,104–108], while others to a tumor-suppressor effect [108–111]. A unifying explanation to these controversial effects, although not compatible with all studies [108], was suggested by Peng et al. [112]. In the absence of ligand, AhR promotes cell cycle progression via binding cyclin-depen-dent kinase 4 [113], while AhR activation results in interactions with the retinoblastoma protein that lead to perturbation of the cell cycle, G0/G1 arrest, diminished capacity for DNA replica-tion and inhibition of cell proliferation. It is hypothesized [114] that AhR may function as a tumor-suppressor gene that becomes silenced in the process of tumor formation. AhR silencing may be associated with cancer progression, as shown in experiments using liver [112] or breast cell lines [115].

Role of AIP in tumorigenesisThe exact mechanism of tumorigenesis in the absence of AIP is not fully understood. Recent studies have indicated that AIP slows cell proliferation in pituitary cells when overexpressed [33] and a lack of AIP induces cell proliferation [116], reinforcing the idea of AIP being a tumor-suppressor gene, as previously hypoth-esized [24,28]. This provides an indication of the role of AIP in

Table 1. Mutations identified within the aryl hydrocarbon receptor-interacting protein gene (cont.).

AIP mutation Mutation type Ref.

Transcript Protein level

c.468 and 15C>T IVS3+15C>T Splice site [47]

c.469–2A>G IVS3–2A>G Splice site [38]

c.469–1G>A IVS3–1G>A Splice site [28]

c.807C>T p.F269= Splice site [33,34]

c.-270_269CG>AA and c.-220G>A

Promoter [33]

c.66_71delAGGAGA p.G23_E24del In-frame deletion [36]

c.138_161del24 p.G47_R54del In-frame deletion [32]

c.742_744delTAC p.Y248del In-frame deletion [45]

c.880_891delCTGGACCCAGCCandc.878_879AG>GT

p.L294_A297delandp.E293G

In-frame deletion [36]

c.805_825dup p.F269_H275dup In-frame insertion [26,33,137]

c.100–1025_279+357 (Ex2del) p.A34_K93 Large genomic del [37]

c.1104–109_279+578 (Ex1_Ex2del)

Large genomic del [37]

Whole gene del Large genomic del [51]

c.1-?_993+?del- Whole protein del Large genomic del [51]

AIP: Aryl hydrocarbon receptor-interacting protein; del: Deletion; dup: Duplication; Ex2: Exon 2; fs: Frameshift; ins: Insertion.

Page 5: Role of the aryl hydrocarbon receptor-interacting protein in

www.expert-reviews.com 685

Review

Missense mutation In-frame deletion

In-frame insertion

C-teminus conserved protein:

C-terminus truncated protein: Nonsense mutation

Large deletions

Splice site

Promoter mutation

Frameshift mutation

Whole gene deletion

c.-270_269CG>AA and c.-220G>A

Novel frameshift p.P114fs p.Q142X c.1-?_993+?del- p. F269_H275dup

p.V96PfsX32

IVS3+15G>T p.E222X p.Q239X p.H275QfsX12

p.G23_E24del p.G83AfsX15 IVS3-1G>A p.K241Xp.L294_A297del

andp.E293G

p.G47_R54del p.Q217X

p.P167HfsX3

p.R304X

p.Q307fsX103p.L25PfsX130

p.E24X

p.R22X

p.R16H

p.Q14X

p.V49M

p.R81X

IVS2-1G>C

Ex2del

Ex1_Ex2del

p.K103R

IVS3-2A>G

p.H135LfsX20

p.K201X

p.C238Y

p.K241E

p.L181fsX13

p.E174fsX47

p.Q164X p.Y248del

p.I257V

p.R304Q

p.A299V

p.Q285fsX16

p.R271W

p.F269=

p.Y268X

Exon 2 Exon 3 Exon 4 Exon 5 Exon 6Exon 1

Role of the AIP gene in familial isolated pituitary adenoma

pituitary cell proliferation. This evidence suggests that loss-of-function mutations in the AIP gene may result in dysregulated cell proliferation that is likely to lead to neoplasia.

Studies regarding AIP–AhR interaction and the tumorigenic potential of AhR are often conflicting [117–119]. Several pathways have been suggested to be involved in AhR-related tumorigenesis, including those of JunD and cyclin A [105], E2F1 [120], p53 deg-radation [104] and the AhR repressor [106], as well as the telomere-independent senescence response [107]. A recent study by Park et al. produced evidence that AhR can cause DNA damage without the formation of an AhR/ARNT complex and without interaction with the xenobiotic response element region of DNA [83]. The study found that translocation of AhR when associated with PAHs can go on to directly damage DNA by concentrating the PAHs within the nucleus, leading to oxidative DNA strand breaks. The role of AhR in pituitary tumorigenesis is unclear, but it has been observed that AhR expression is reduced in AIP mutant pitui-tary adenomas [121]. The expression of ARNT is reduced in AIP mutation-positive adenomas compared with AIP-negative sporadic cases [116], while the expression of AhR repressor is increased in sporadic growth hormone (GH)-secreting tumors [122].

AIP is known to interact with the PDEs. There are more than 50 isoforms of PDE, and binding of AIP varies between differ-ent isoforms [73]. AIP is only known to bind to two PDE iso-forms, 4A5 and 2A [73,74], via the TPR region of the protein [74]. PDEs moderate cellular cAMP levels via hydrolysis of cAMP to AMP [73].

Phosphodiesterase 4A5 was the first PDE identified as a binding partner of AIP, binding via interaction with the TPR domains and reducing PDE4A5 function. Reduced function indirectly raises cAMP levels [73,123]. Given that AIP inhibits the enzymatic function of PDE4A5, this would suggest that loss of AIP function leads to reduced cAMP levels. Somatotroph cell cAMP levels are vital for regulating cell replication and hormone synthesis [124] and increased cAMP could result in hypertrophy and hyperplasia of the pituitary, which could lead to the development of pituitary neoplasia [125,126]. These data do not support the tumorigenic role of AIP in somatotroph cells via this pathway.

de Oliveira et al. identified PDE2A as a binding partner of AIP, associating with the TPR domains of AIP via a GAF-B domain present in the PDE2A structure [74]. They suggested

Figure 1. Representation of each known mutation identified to date within the aryl hydrocarbon receptor-interacting protein gene.

Page 6: Role of the aryl hydrocarbon receptor-interacting protein in

Expert Rev. Endocrinol. Metab. 5(5), (2010)686

Review

Splice site

Nonsense

Large genomic deletion

In-frame insertion

In-framedeletion

FrameshiftPromoter

Missense

12.50%

25.00%

25.00%

18.75%

2.08%

2.08%

8.33%

8.33%

Cain, Miljic, Popovic & Korbonits

that the association of PDE2A with AIP results in PDE2A being brought into close proximity to cytoplasmic AhR proteins, local-ized within the AIP/AhR/Hsp90/p23 complex. The catalytic activity of PDE2A is not affected by association with AIP; there-fore, the complex reduces local cAMP levels, reducing cAMP interaction with AhR [74]. It has been recognized that cAMP can cause translocation of AhR into the nucleus, independent of exogenous compounds [99]; therefore, the local reduction in cAMP in the vicinity of AhR provides a possible mechanism of action for AIP as a tumor-suppressor gene [82].

Another protein found to be associated with AIP is sur-vivin. Survivin is a member of the inhibitor of apoptosis fam-ily of proteins that prevent apoptosis via association with and inactivation of caspases. This inactivation prevents caspase-driven apoptosis [72,127]. Survivin forms a complex with AIP and Hsp90 resulting in stabilization of survivin by preventing ubiquitination [72,128]. Knock-out studies have shown that the loss of AIP results in decreased survivin levels, resulting in apoptosis [72]. These data do not support the tumor-suppressor role of AIP in this context. However, another binding partner of AIP, RET, can change the AIP–survivin interaction [75]. RET has an interesting dual role – it produces survival signals in the presence of its ligand glial cell line-derived neurotropic factor. However, in the absence of glial cell line-derived neurotropic factor, it is proapoptopic. Activating RET mutations result in MEN2 or medullary thyroid carcinoma syndrome, while inactivating mutations cause Hirschprung disease. Binding of AIP to RET results in disassociation of AIP from survivin, resulting in increased ubiquitination of survivin, reducing cell

survival signals [75]. This proposed inter-action supports the hypothesis that AIP is a tumor-suppressor gene.

AIP has been found to be widely dis-tributed in the tissues of the body, hav-ing been isolated in tissues of the brain, colon, heart, kidney, leukocytes, liver, lung, ovary, pancreas, pituitary, pla-centa, prostate, skeletal muscle, small intestine, spleen, testes and thymus [33,58]. Considering that AIP is thought to be a tumor-suppressor gene, it could be expected that AIP mutation-positive patients are at an increased risk of devel-oping neoplastic changes in any tissue expressing AIP. Indeed, a few AIP muta-tion-positive families have been described by various types of other tumors [33], but these tumors are not uniform in their tis-sue of origin, and given the high preva-lence of tumors in general, not enough data are available to confirm the hypoth-esis. In terms of AIP mutations in other types of tumors, no somatic AIP muta-tions were identified in 373 patients with colorectal cancer, 82 patients with breast

cancer, 44 patients with prostate cancer [129] and 75 endocrine tumors (four paragangliomas, four pancreatic endocrine tumors, eight parathyroid, 26 thyroid, 19 adrenal, 16 neuroendocrine tumors and two mixed tumors) [46].

Comparison of the clinical characteristics of FIPA cohorts with & without AIP mutationsStudies into FIPA families have consistently found a dramatically different clinical picture in families harboring AIP mutations compared with both AIP mutation-negative FIPA families and those with sporadic pituitary adenoma.

Age at diagnosisThe mean age of diagnosis in our cohort of 64 families was found to be 33.2 ± 15.6 years, with AIP mutation-positive patients presenting 16.6 years earlier, on average, than those without AIP mutations (23.5 ± 11.0 years vs 39.8 ± 14.8 years; p < 0.0001).These data agree with the current literature, showing the mean age of diagnosis in those harboring AIP mutations to be, on average, 13–15 years earlier than in AIP mutation-negative patients [8,28,33].

The 2006 study by Daly et al. found that the age of diagnosis within FIPA families appears to decrease from one generation to another (50.5 ± 14.2 years vs 29.0 ± 16.3 years; p < 0.001) [20]. A similar trend can be observed in our cohort. We believe this phenomena is most probably not due to genetic anticipa-tion but to an increased awareness of symptoms due to the family history [8,20]. When selecting for families with more than one recorded generation, our data show that tumors in

Figure 2. A pie chart demonstrating the number and proportion of each known mutation type identified to date within the aryl hydrocarbon receptor interacting protein gene.

Page 7: Role of the aryl hydrocarbon receptor-interacting protein in

www.expert-reviews.com 687

Review

N-terminus

C-terminusα-helix

TPR3

TPR2

TPR1

Role of the AIP gene in familial isolated pituitary adenoma

the second generation are diagnosed, on average, 17.7 years earlier than those in the first generation (41.3 ± 17.1 years vs 23.6 ± 10.4 years; p < 0.0001).

Pituitary tumor typesThe most common adenoma types in sporadic cohorts are prolac-tinomas (30–45%), somatotropinomas (20%), adrenocorticotro-pin hormone-secreting adenomas (10–12%), clinically nonfunc-tioning adenomas (20–25%), somatolactotropinomas, secreting both prolactin and GH (7%) and thyrotropinomas (1–2%) [1,6]. Within the unselected FIPA cohort of Beckers’ group, adenoma presentation is similar to that of the sporadic onset patients, with 41% presenting with prolactinomas, 30% somatotropinomas, 7% somatolactotropinomas and the remaining 23% compris-ing of adrenocorticotropin hormone-secreting adenomas, clini-cally nonfunctioning and thyrotropinomas [15]. Conversely, AIP mutation-positive families in this cohort exhibit 87.5% soma-totropinomas and 9.4% prolactinomas [130], with nonfunctioning and corticotropic pituitary adenomas being rare [8]. However, in our cohort of 64 FIPA families, we observed a predisposition to somatotropinomas, primarily in AIP mutation-positive patients but also in AIP-negative families (Table 2).

Gender differenceInterestingly, our data suggest that there is a gender difference in prevalence between pituitary adenomas in AIP mutation -positive and -negative FIPA families, with approximately two out of three (65.7%, 69 male vs 33 female; n = 105) AIP mutation-posi-tive patients being male, compared with 57.0% in unselected

FIPA patients (114 male vs 86 female) or 47.5% in AIP negative FIPA patients. While our data are supported by earlier reports regarding male predominance (71%) in AIP mutation posi-tive patients [130], we do not find female preponderance [15] in AIP-negative families.

Owing to the interesting link between gender and tumorigen-esis in AIP-positive families, we decided to look into the mode of inheritance in AIP mutation-positive families and unselected FIPA cohorts to determine if there was any difference in mater-nal and paternal transmission between the groups. We gathered inheritance information on 127 individuals, 82 of whom pos-sessed an AIP mutation. We determined that in unselected FIPA cases, maternal to paternal transmission was 52:75, respectively, indicating that transmission was 40.9% maternal and 59.1% paternal, whereas within AIP mutation-positive families, the maternal to paternal transmission was 27:55, respectively, giving a 32.9% maternal versus 67.1% paternal transmission, indicat-ing that both FIPA and PAP transmission via the father is more common. This trend may suggest a link to potential genomic imprinting, although if this were the case, transmission would likely be more skewed to paternal transmission. Another potential explanation for this may relate to a differing effect of pituitary adenomas on fertility in males and females. We believe that dis-rupted pituitary function results in abnormal luteinizing hor-mone/follicle-stimulating hormone regulation, which will lead to loss of ovulation in females. However, in males, reduction of luteinizing hormone and follicle-stimulating hormone might result in lower testosterone levels without complete loss of sperma-togenesis; therefore, some level of fertility is maintained. Because AIP mutation-positive patients are affected at a younger age, this effect would be more pronounced in this population.

PenetranceOur data were analyzed to determine whether there is a dif-ference in penetrance between AIP mutation-positive and -negative FIPA cohorts. Our 44 AIP mutation-negative families contained 96 affected individuals and a calculated number of carriers of 293, giving an estimated penetrance of 32.7%. Our ana lysis of the 20 AIP mutation-positive families, containing 66 affected individuals and 200 carriers, demonstrated a simi-lar penetrance of 33.0%. While this figure agrees with current

Figure 3. The three tetratricopeptide repeats regions of the aryl hydrocarbon recptor-interacting protein and the terminal α-helix.TPR: Tetratricopeptide repeats.

p23 Hsp90 dimer AIP AhR

Figure 4. The AIP/AhR/Hsp90/p23 complex.AhR: Aryl hydrocarbon receptor; AIP: Aryl hydrocarbon receptor-interacting protein; Hsp: Heat-shock protein.

Page 8: Role of the aryl hydrocarbon receptor-interacting protein in

Expert Rev. Endocrinol. Metab. 5(5), (2010)688

Review

210 cmat age of 21 years

175 cmat age of 10 years

Cain, Miljic, Popovic & Korbonits

estimates placing the penetrance in AIP mutation-positive fami-lies between 15–30% [39,51], it does not indicate a significant difference in penetrance between AIP mutation-negative and -positive families with FIPA. However, approaching the same questions in an alternative way, there is a noted difference in the number of individuals with disease in FIPA families with and without AIP mutations [51]. Analysis of 64 families, 20 AIP mutation-positive and 44 AIP mutation-negative, has shown that AIP-positive families have, on average, 3.2 ± 1.8 compared with 2.2 ± 0.5 members (p = 0.0006) in AIP mutation-negative fami-lies. This appears to indicate a difference in penetrance between AIP mutation-positive and -negative FIPA cohorts. However, we need to be aware that while AIP mutations often cause early-onset GH-secreting adenomas making the clinical diagnosis relatively straight forward, the more variable tumor type and later onset in AIP mutation-negative families could confound recognition of the disease and therefore penetrance calculations.

The factors influencing penetrance are dif-ficult to determine and the possibility of a second locus has been suggested, although not confirmed [131].

We also looked into the penetrance for specific mutation types within AIP muta-tion-positive families. A total of 16 out of 20 of our AIP mutation-positive fami-lies posessed a truncating mutation, with 56 affected individuals and 177 carriers, giving a penetrance of 32.7%. Interestingly, our four AIP mutation-positive families with nontruncating mutations included

ten affected individuals and 23 carriers, giving a penetrance of 43.5%. We believe that the number of families available for these calculations excludes the possibility to provide reliable estimates here, but does suggest an interesting point for further investiga-tion witha larger sample size.

The AIP-positive patients typically develop aggressive soma-totroph macroadenomas [32,33,130]. AIP-positive patients also typically have a young age at diagnosis (<25–30 years) [15], whereas with sporadic pituitary adenomas, occurrence in the young is rare. In our cohort, 32 patients presented younger than 18 years – of these, 78.1% (n = 25) were AIP mutation positive. The predisposition to develop adenomas at a young age results in 80% (16 out of 20) of families harboring AIP mutations experiencing childhood-onset adenoma, while only 11.4% (5 out of 44) of AIP mutation-negative FIPA families have a childhood-onset case. It is therefore important to con-sider an AIP mutation in all patients with early-onset pituitary

adenoma or gigantism. A large number of families recruited into our cohort were initially identified via early-onset disease (Figure 5). We believe that through wide-spread awareness of the high prevalence of AIP mutation in early-onset pituitary ade-noma, more families would be investigated for potential mutations, allowing better management of other carriers who are at risk of developing disease.

The term ‘acromegaly’ was first used by Pierre Marie in 1886, describing a condi-tion characterized by hypertrophy of the hands, feet and face, although many other physicians had previously described the same condition [132,133]. Initially, acromeg-aly and gigantism were considered separate clinical entities; later, gigantism was con-sidered a congenital disease and acromegaly an acquired disease [134]. With the increas-ing recognition of AIP mutations in famil-ial and sporadic giants, the original idea that gigantism can be a result of a genetic predisposition is, in some cases, not too far from the truth.

Table 2. Pituitary adenoma types in our cohort of 64 families.

Adenoma type All FIPA (%) AIP mutation-positive (%)

AIP mutation-negative (%)

Somatotropinoma 65 (n = 104) 76.6 (n = 49) 57.3 (n = 55)

Lactotroph adenoma 19.4 (n = 31) 10.9 (n = 7) 25.0 (n = 24)

Somatolactotropinoma 4.4 (n = 7) 7.8 (n = 5) 2.1 (n = 2)

Clinically nonfunctioning pituitary adenoma

10.6 (n = 17) 4.7 (n = 3) 14.6 (n = 14)

Corticotropinoma 0.6 (n = 1) 0.0 (n = 0) 1.0 9 (n = 1)

AIP: Aryl hydrocarbon receptor-interacting protein; FIPA: Familial isolated pituitary adenoma.

Figure 5. Family tree and photograph of two aryl hydrocarbon receptor-interacting protein mutation positive siblings with gigantism. The female patient was diagnosed with a growth hormone-secreting macroadenoma at the age of 10 years (175 cm, >>99 percentile), was treated with two surgeries and radiotherapy and currently is cured of acromegaly and on full pituitary hormone replacement. Her brother was diagnosed age 21 years (210 cm) with a growth hormone-secreting macroadenoma, and after surgery and radiotherapy, he has residual disease with poor response to somatostatin analogs.

Page 9: Role of the aryl hydrocarbon receptor-interacting protein in

www.expert-reviews.com 689

ReviewRole of the AIP gene in familial isolated pituitary adenoma

Prevalence of AIP mutations identified in sporadic patientsRecent cohort studies agree that AIP muta-tions can be identified in approximately 3–4% of unselected sporadic pituitary adenoma cases [36,38]. These mutations are germline mutations and no somatic muta-tions have been found in pituitary adeno-mas to date [31,33,75]. Evidence suggests that presentation of seemingly sporadic AIP mutation-positive pituitary adenoma is simi-lar to that in the familial setting. Patients typically present at a young age (majority are <25 years), most commonly with somatotro-pinoma. Therefore, it is important to con-sider an AIP mutation in all sporadic patients presenting early with somatotropinoma. However, in our experience [33], the major-ity of childhood-onset sporadic acromegaly cases do not carry AIP mutations, such as the patient shown in Figure 6.

We analyzed all the AIP mutation-positive sporadic patients published in the literature to date, together with two AIP mutation-positive sporadic patients from our cohort (Table 3) [Korbonits M, Unpublished

Data]. Analysis of the data reveals a mean age of diagnosis of 24.4 ± 9.2 years, with 59.3% of patients being male (16 out of 27) and 40.7% of patients being female (11 out of 27). Of the AIP mutation-positive spo-radic patients, 75% (21 out of 28) of the group possessed somatotropinomas. These trends fit with those observed in FIPA cohorts harboring an AIP mutation, sug-gesting that the presentation of AIP muta-tion-positive patients is similar in both the familial and sporadic settings.

Interestingly, eight of the 28 patients pos-sessed the p.Q14X mutation, all of which originated in Finland, suggesting that these cases could be part of the founder mutation cohort identified in that region.

Role of immunohistochemistry in identification of AIP mutation-positive tumor tissuesAIP is preferentially expressed in somatotrophs and lactotrophs. This corresponds with the observation that AIP mutation-positive patients preferentially develop somatotropinomas and lactotropi-nomas. Currently, suspected AIP mutation-positive patients are diagnosed via conventional sequencing and MLPA. The potential role of immunohistochemistry in detection of reduced or absent AIP expression in excised pituitary adenomas has been studied and found not to be a reliable tool to predict the presence of AIP mutations [33,36,121]. In somatotroph adenomas where AIP staining

is not detected, an AIP mutation is likely, but positive AIP stain-ing does not rule out AIP mutations. This suggests that there is probably little role for immunohistochemistry in screening and the identification of AIP-mutant patients.

Clinical management of PAP familiesStudies regarding MEN1 have indicated that changes in the levels of IGF-1 and prolactin can be detected up to 10 years before a pitui-tary adenoma becomes clinically apparent [135]. Additionally, MRI scanning can often identify small adenomas before overt clinical

Table 3. AIP mutation-positive sporadic patients identified in the literature to date.

Gender Condition Age (years)

Mutation Ref.

Female Acromegaly 24 p.Q14X [1]

Male Acromegaly 42 p.Q14X [1]

Female Acromegaly 26 IVS3–1G>A [1]

Female Acromegaly 22 p.Q14X [1]

Female Acromegaly 20 p.Q14X [1]

Male Acromegaly 36 p.Q14X [2]

Female Acromegaly 41 p.Q14X [2]

Male Acromegaly 20 IVS2–1G>C [2]

Male Gigantism 8 p.H275QfsX12 [2]

Unknown Cushing’s syndrome 26 p.R304Q [2]

Female Somatotropinoma 16 p.R304X [2]

Male Gigantism 28 p.V49M [35]

Male Acromegaly 24 p.R22X [4]

Male Gigantism 15 p.H135LfsX20 [12]

Male Acromegaly 40 IVS3–2A>G [12]

Female Acromegaly 27 p.K201X [12]

Male Acromegaly 24 p.K201X [12]

Female Acromegaly 37 p.R304Q [12]

Male Gigantism 17 p.R304X [12]

Male Prolactinoma 35 p.Q14X [3]

Female Somatolactotropinoma 35 p.Q14X [3]

Male Gigantism 19 p.Y248del [27]

Female Acromegaly 19 IVS3+15C>T [47]

Male Gigantism 15 fs in exon 2 [48]

Male Acromegaly 20 p.G23_E24del [2]

Male Acromegaly 18 p.L181fsX13 [2]

Male Gigantism 15 p.R271W [Korbonits M, Unpublished Data]

Female Gigantism 15 p.R304X [Korbonits M, Unpublished Data]

del: Deletion; fs: frameshift.

Page 10: Role of the aryl hydrocarbon receptor-interacting protein in

Expert Rev. Endocrinol. Metab. 5(5), (2010)690

Review Cain, Miljic, Popovic & Korbonits

symptoms occur. If preclinical changes in blood bio chemistry were to be observed in AIP mutation-positive families this would allow early identification of pituitary adenomas in identified AIP mutation-positive families, via regular blood investigations. We suggest that patients with an AIP mutation should undergo base-line MRI and yearly screening for abnormal pituitary function, as is currently indicated in MEN1-positive patients [136].

The management of PAP families is a challenging task. Patients usually have large invasive adenomas and are often not responsive to somatostatin analogs [33]. Long-term follow-up of patients and asymptomatic carriers is vital for early prevention of disease. AIP mutation screening should be considered for all familial pituitary adenoma cases where MEN1 and CNC is unlikely and the genetic screening should involve both DNA sequencing and MLPA. This service is now available from an accredited NHS Laboratory in the UK. Owing to the young age of onset of pituitary adenoma in AIP mutation-positive families, predictive genetic testing of the children of the affected or carrier members should be considered at an early age, and based on the youngest known manifested case with AIP

mutation, screening by the age of 4 years could be considered. Identified carriers should undergo yearly clinical and biochemical assessments for abnormal pituitary function.

Expert commentaryFamilial isolated pituitary adenomas are increasingly recognized and awareness needs to be raised to their existence in addition to the classical MEN1 and CNC syndromes. A total of 15–20% of FIPA families harbor a mutation in the AIP gene. It is not known how AIP, a molecular co-chaperone, affects pituitary tumorigenesis, but over expression reduces, while inhibition increases cell pro-liferation in experimental settings. Thorough genetic ana lysis, including tests for large gene deletions, and possible assessment of functional status of a sequence change, helps to appropriately classify families.

Phenotypically, AIP mutation-positive and -negative families are quite distinct as a group, although individual families need to be studied carefully. The disease has a low penetrance, raising issues about clinical screening and genetic counseling.

Five-year viewAnalyzing and reporting families with FIPA will raise awareness of the possibility of a familial link in patients with pituitary ade-noma. Physicians will be able to refer these families to specialists early, allowing screening, early diagnosis and better management of a condition, which, if left untreated, could lead to considerable morbidity. Identification of the mechanism of how AIP is involved in pituitary tumorigenesis will help to design novel strategies to fight the disease. We envisage that other genes will be identified in the next few years that are responsible for the AIP mutation-negative cases in FIPA cohorts. We hope that advancements will also allow understanding of the causes of the incomplete pen-etrance observed in FIPA cohorts to the benefit of counseling carrier subjects.

Figure 6. Patient (17 years of age, 190 cm) with growth hormone-secreting macroadenoma. Following two pituitary surgeries and stereotactic radiotherapy, the patient is on somatostatin analog therapy. No family history of pituitary adenomas and no AIP mutation have been identified.

Key issues

• Pituitary adenomas have a prevalence of approximately one in 1100, 2% of which are classified as familial isolated pituitary adenoma (FIPA). Patients with a family history of isolated pituitary adenomas most often have growth hormone- or prolactin-secreting adenomas.

• A total of 15–20% of FIPA cases harbor an autosomal dominant mutation in the aryl hydrocarbon receptor-interacting protein (AIP) gene causing pituitary disease with incomplete penetrance (approximately 30%).

• AIP mutation-positive patients have distinct clinical characteristics. They present, on average, 16 years younger than AIP-negative FIPA patients, and have a predisposition to developing somatotropinoma, often leading to gigantism. These tumors tend to respond poorly to somatostatin analogs.

• Occasionally, seemingly sporadic pituitary patients are also found to harbor AIP mutations. The characteristic clinical features of AIP mutation-positive patients appear to be conserved through the sporadic and familial settings and allow a key indication of potential AIP mutation carriers.

• AIP is thought to be a tumor-suppressor gene, with loss of the normal allele resulting in tumor formation. It is unclear how AIP exerts its tumor-suppressor function. The roles of several interacting partners have been considered.

• Currently, research is ongoing to identify the gene or genes causing AIP mutation-negative FIPA.

• It is important to raise the awareness of the possible familial nature of pituitary tumors.

Page 11: Role of the aryl hydrocarbon receptor-interacting protein in

www.expert-reviews.com 691

ReviewRole of the AIP gene in familial isolated pituitary adenoma

ReferencesPapers of special note have been highlighted as:• of interest•• of considerable interest

1 Heaney AP, Melmed S. Molecular targets in pituitary tumours. Nat. Rev. Cancer 4, 285–295 (2004).

2 Frohman LA, Eguchi K. Familial acromegaly. Growth Horm. IGF Res. 14(Suppl. A), S90–S96 (2004).

3 Ezzat S, Asa SL, Couldwell WT et al. The prevalence of pituitary adenomas: a systematic review. Cancer 101, 613–619 (2004).

4 Daly AF, Rixhon M, Adam C, Dempegioti A, Tichomirowa MA, Beckers A. High prevalence of pituitary adenomas: a cross-sectional study in the province of Liege, Belgium. J. Clin. Endocrinol. Metab. 91, 4769–4775 (2006).

5 Fernandez A, Karavitaki N, Wass JA. Prevalence of pituitary adenomas: a community-based, cross-sectional study in Banbury (Oxfordshire, UK). Clin. Endocrinol. (Oxf). 72(3), 377–382 (2010).

6 Arafah BM, Nasrallah MP. Pituitary tumors: pathophysiology, clinical manifestations and management. Endocr. Relat. Cancer 8, 287–305 (2001).

7 Melmed S. Mechanisms for pituitary tumorigenesis: the plastic pituitary. J. Clin. Invest. 112, 1603–1618 (2003).

8 Beckers A, Daly AF. The clinical, pathological, and genetic features of familial isolated pituitary adenomas. Eur. J. Endocrinol. 157, 371–382 (2007).

9 Bertherat J. Carney complex (CNC). Orphanet. J. Rare Dis. 1, 21–26 (2006).

10 Chandrasekharappa SC, Guru SC, Manickam P et al. Positional cloning of the gene for multiple endocrine neoplasia-type 1. Science 276, 404–407 (1997).

11 Verges B, Boureille F, Goudet P et al. Pituitary disease in MEN type 1 (MEN1): data from the France-Belgium MEN1 multicenter study. J. Clin. Endocrinol. Metab. 87, 457–465 (2002).

12 Thakker RV, Bouloux P, Wooding C et al. Association of parathyroid tumors in multiple endocrine neoplasia type 1

with loss of alleles on chromosome 11. N. Engl. J. Med. 321, 218–224 (1989).

13 Turner JJ, Christie PT, Pearce SH, Turnpenny PD, Thakker RV. Diagnostic challenges due to phenocopies: lessons from Multiple Endocrine Neoplasia type 1 (MEN1). Hum. Mutat. 31, E1089–E1101 (2010).

14 Pellegata NS, Quintanilla-Martinez L, Siggelkow H et al. Germ-line mutations in p27Kip1 cause a multiple endocrine neoplasia syndrome in rats and humans. Proc. Natl Acad. Sci. USA 103, 15558–15563 (2006).

15 Daly AF, Tichomirowa MA, Beckers A. Genetic, molecular and clinical features of familial isolated pituitary adenomas. Horm. Res. 71(Suppl. 2), 116–122 (2009).

16 Agarwal SK, Mateo CM, Marx SJ. Rare germline mutations in cyclin-dependent kinase inhibitor genes in multiple endocrine neoplasia type 1 and related states. J. Clin. Endocrinol. Metab. 94, 1826–1834 (2009).

17 Georgitsi M, Raitila A, Karhu A et al. Germline CDKN1B/p27Kip1 mutation in multiple endocrine neoplasia. J. Clin. Endocrinol. Metab. 92, 3321–3325 (2007).

18 Boikos SA, Stratakis CA. Carney complex: the first 20 years. Curr. Opin Oncol. 19, 24–29 (2007).

19 Casey M, Mah C, Merliss AD et al. Identification of a novel genetic locus for familial cardiac myxomas and Carney complex. Circulation 98, 2560–2566 (1998).

20 Daly AF, Jaffrain-Rea ML, Ciccarelli A et al. Clinical characterization of familial isolated pituitary adenomas. J. Clin. Endocrinol. Metab. 91, 3316–3323 (2006).

•• Thispaperdescribedtheclinicalcharacteristicsoffamilialisolatedpituitaryadenoma(FIPA)cohortsincomparisontotheestablishedclinicalcharacteristicsofsporadicpituitaryadenoma,concludingthatFIPAhasdistinctclinicalcharacteristicsthatarenottypicalofsporadiccohorts.

21 Daly AF, Jaffrain-Rea ML, Beckers A. Clinical and genetic features of familial pituitary adenomas. Horm. Metab. Res. 37, 347–354 (2005).

22 Benlian P, Giraud S, Lahlou N et al. Familial acromegaly: a specific clinical entity – further evidence from the genetic study of a three-generation family. Eur. J. Endocrinol. 133, 451–456 (1995).

23 Tanaka C, Yoshimoto K, Yamada S et al. Absence of germ-line mutations of the multiple endocrine neoplasia type 1 (MEN1) gene in familial pituitary adenoma in contrast to MEN1 in Japanese. J. Clin. Endocrinol. Metab. 83, 960–965 (1998).

24 Gadelha MR, Prezant TR, Une KN et al. Loss of heterozygosity on chromosome 11q13 in two families with acromegaly/gigantism is independent of mutations of the multiple endocrine neoplasia type I gene. J. Clin. Endocrinol. Metab. 84, 249–256 (1999).

25 Ackermann F, Krohn K, Windgassen M, Buchfelder M, Fahlbusch R, Paschke R. Acromegaly in a family without a mutation in the menin gene. Exp. Clin. Endocrinol. Diabetes 107, 93–96 (1999).

26 Soares BS, Eguchi K, Frohman LA. Tumor deletion mapping on chromosome 11q13 in eight families with isolated familial somatotropinoma and in 15 sporadic somatotropinomas. J. Clin. Endocrinol. Metab. 90, 6580–6587 (2005).

27 Yamada S, Yoshimoto K, Sano T et al. Inactivation of the tumor suppressor gene on 11q13 in brothers with familial acrogigantism without multiple endocrine neoplasia type 1. J. Clin. Endocrinol. Metab. 82, 239–242 (1997).

28 Vierimaa O, Georgitsi M, Lehtonen R et al. Pituitary adenoma predisposition caused by germline mutations in the AIP gene. Science 312, 1228–1230 (2006).

•• ThisseminalpaperidentifiedAIPasthecausitivegeneforsomepatientswithFIPA,introducingtheterm‘pituitaryadenomapredisposition’(PAP)torefertothesepatients.

29 Chahal HS, Chapple JP, Frohman LA, Grossman AB, Korbonits M. Clinical, genetic and molecular characterisation of patients with familial isolated pituitary adenomas. Trends Endocrinol. Metab. 21(7), 419–427 (2010).

Financial & competing interests disclosureJoshua W Cain was supported by the Wolfson foundation and the Association of Physicians of Great Britain and Ireland. Our laboratories’ FIPA project is supported by the Cancer Research Committee of St Bartholomew’s Hospital (London, UK). The FIPA project in Serbia was supported by a grant from 

the Ministry of Science (145019). The authors have no other relevant affili-ations or financial involvement with any organization or entity with a finan-cial  interest  in or financial conflict with the  subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

Page 12: Role of the aryl hydrocarbon receptor-interacting protein in

Expert Rev. Endocrinol. Metab. 5(5), (2010)692

Review Cain, Miljic, Popovic & Korbonits

30 Yu R, Bonert V, Saporta I, Raffel LJ, Melmed S. Aryl hydrocarbon receptor interacting protein variants in sporadic pituitary adenomas. J. Clin. Endocrinol. Metab. 91, 5126–5129 (2006).

31 Barlier A, Vanbellinghen JF, Daly AF et al. Mutations in the aryl hydrocarbon receptor interacting protein gene are not highly prevalent among subjects with sporadic pituitary adenomas. J. Clin. Endocrinol. Metab. 92, 1952–1955 (2007).

32 Daly AF, Vanbellinghen JF, Khoo SK et al. Aryl hydrocarbon receptor-interacting protein gene mutations in familial isolated pituitary adenomas: ana lysis in 73 families. J. Clin. Endocrinol. Metab. 92, 1891–1896 (2007).

33 Leontiou CA, Gueorguiev M, van der Spuy J et al. The role of the aryl hydrocarbon receptor-interacting protein gene in familial and sporadic pituitary adenomas. J. Clin. Endocrinol. Metab. 93, 2390–2401 (2008).

34 McCarthy MI, Noonan K, Wass JA, Monson JP. Familial acromegaly: studies in three families. Clin. Endocrinol. (Oxf). 32, 719–728 (1990).

35 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). 66, 499–502 (2007).

36 Georgitsi M, Raitila A, Karhu A et al. Molecular diagnosis of pituitary adenoma predisposition caused by aryl hydrocarbon receptor-interacting protein gene mutations. Proc. Natl Acad. Sci. USA 104, 4101–4105 (2007).

37 Georgitsi M, Heliovaara E, Paschke R et al. Large genomic deletions in AIP in pituitary adenoma predisposition. J. Clin. Endocrinol. Metab. 93, 4146–4151 (2008).

38 Cazabat L, Libe R, Perlemoine K et al. Germline inactivating mutations of the aryl hydrocarbon receptor-interacting protein gene in a large cohort of sporadic acromegaly: mutations are found in a subset of young patients with macroadenomas. Eur. J. Endocrinol. 157, 1–8 (2007).

39 Naves LA, Daly AF, Vanbellinghen JF et al. Variable pathological and clinical features of a large Brazilian family harboring a mutation in the aryl hydrocarbon receptor-interacting protein gene. Eur. J. Endocrinol. 157, 383–391 (2007).

40 Chahal HS, Igreja SC, Gueorguiev M et al. The clinical and genetic characteristics of patients with familial isolated pituitary

adenomas. Hormones, 11th International Workshop of MEN meeting, Delphi. 7(Suppl. 2), O8 (2008).

41 Stratakis CA, Tichomirowa MA, Boikos S et al. The role of germline AIP, MEN1, PRKAR1A, CDKN1B and CDKN2C mutations in causing pituitary adenomas in a large cohort of children, adolescents, and patients with genetic syndromes. Clin. Genet. DOI: 10.1111/j.1399-0004.2010.01406.x (2010) (Epub ahead of print).

42 Matsuno A, Teramoto A, Yamada S et al. Gigantism in sibling unrelated to multiple endocrine neoplasia: case report. Neurosurgery 35, 952–955 (1994).

43 Jorge BH, Agarwal SK, Lando VS et al. Study of the multiple endocrine neoplasia type 1, growth hormone-releasing hormone receptor, Gs a, and Gi2 a genes in isolated familial acromegaly. J. Clin. Endocrinol. Metab. 86, 542–544 (2001).

44 Toledo RA, Lourenco DM Jr, Liberman B et al. Germline mutation in the aryl hydrocarbon receptor interacting protein gene in familial somatotropinoma. J. Clin. Endocrinol. Metab. 92, 1934–1937 (2007).

45 Georgitsi M, De ME, Cannavo S et al. Aryl hydrocarbon receptor interacting protein (AIP) gene mutation ana lysis in children and adolescents with sporadic pituitary adenomas. Clin. Endocrinol. (Oxf). 69, 621–627 (2008).

46 Raitila A, Georgitsi M, Karhu A et al. No evidence of somatic aryl hydrocarbon receptor interacting protein mutations in sporadic endocrine neoplasia. Endocr. Relat. Cancer 14, 901–906 (2007).

47 Montanana CF, Daly AF, Riego-Suarez R et al. AIP mutations in familial and sporadic pituitary adenomas: local experience and review of the literature. Endocrinol. Nutr. 56 (7), 369–377 (2009).

48 Yaneva M, Daly AF, Tichomirowa MA et al. Aryl hydrocarbon receptor interacting protein gene mutations in bulgarian FIPA and young sporadic pituitary adenoma patients. Proceedings of the 90th Annual Meet of the Endocrine Society. (2008) (Abstract P3-520).

49 Luccio-Camelo DC, Une KN, Ferreira RE et al. A meiotic recombination in a new isolated familial somatotropinoma kindred. Eur. J. Endocrinol. 150, 643–648 (2004).

50 Toledo RA, Mendonca BB, Longuini VC et al. Familial somatotropinoma and adrenocortical carcinoma due to a novel germline mutation and loss of the wild-type allele of the AIP gene. Proceedings of the 90th Annual Meet of the Endocrine Society. (2008) (Abstract P1-488).

51 Igreja SC, Chahal HS, King P et al. Characterization of aryl hydrocarbon receptor interacting protein (AIP) mutations in familial isolated pituitary adenoma families. Hum. Mutat. 31(8), 950–960 (2010).

52 Jennings JE, Georgitsi M, Holdaway I et al. Aggressive pituitary adenomas occurring in young patients in a large Polynesian kindred with a germline R271W mutation in the AIP gene. Eur. J. Endocrinol. 161, 799–804 (2009).

53 Kikuchi M, Ohkura N, Yamaguchi K, Obara T, Tsukada T. Gene dose mapping delineated boundaries of a large germline deletion responsible for multiple endocrine neoplasia type 1. Cancer Lett. 208, 81–88 (2004).

54 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 116, 625–628 (2008).

55 Soares BS, Frohman LA. Isolated familial somatotropinoma. Pituitary 7, 95–101 (2004).

56 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. Bio. Chem. 282, 35924–35932 (2007).

57 Carver LA, Bradfield CA. Ligand-dependent interaction of the aryl hydrocarbon receptor with a novel immunophilin homolog in vivo. J. Bio. Chem. 272, 11452–11456 (1997).

58 Kuzhandaivelu N, Cong YS, Inouye C, Yang WM, Seto E. XAP2, a novel hepatitis B virus X-associated protein that inhibits X transactivation. Nucleic Acids Res. 24, 4741–4750 (1996).

59 Petrulis JR, Perdew GH. The role of chaperone proteins in the aryl hydrocarbon receptor core complex. Chem. Biol. Interact. 141, 25–40 (2002).

60 Nakata A, Urano D, Fujii-Kuriyama Y, Mizuno N, Tago K, Itoh H. G-protein signalling negatively regulates the stability of aryl hydrocarbon receptor. EMBO Rep. 10, 622–628 (2009).

61 Carver LA, LaPres JJ, Jain S, Dunham EE, Bradfield CA. Characterization of the Ah receptor-associated protein, ARA9. J. Bio. Chem. 273, 33580–33587 (1998).

Page 13: Role of the aryl hydrocarbon receptor-interacting protein in

www.expert-reviews.com 693

ReviewRole of the AIP gene in familial isolated pituitary adenoma

62 Pollenz RS, Dougherty EJ. Redefining the role of the endogenous XAP2 and C-terminal hsp70-interacting protein on the endogenous Ah receptors expressed in mouse and rat cell lines. J. Bio. Chem. 280, 33346–33356 (2005).

63 Kazlauskas A, Poellinger L, Pongratz I. The immunophilin-like protein XAP2 regulates ubiquitination and subcellular localization of the dioxin receptor. J. Bio. Chem. 275, 41317–41324 (2000).

64 LaPres JJ, Glover E, Dunham EE, Bunger MK, Bradfield CA. ARA9 modifies agonist signaling through an increase in cytosolic aryl hydrocarbon receptor. J. Bio. Chem. 275, 6153–6159 (2000).

65 Bell DR, Poland A. Binding of aryl hydrocarbon receptor (AhR) to AhR-interacting protein. The role of hsp90. J. Bio. Chem. 275, 36407–36414 (2000).

66 Meyer BK, Petrulis JR, Perdew GH. Aryl hydrocarbon (Ah) receptor levels are selectively modulated by hsp90-associated immunophilin homolog XAP2. Cell Stress Chaperones 5, 243–254 (2000).

67 Meyer BK, Perdew GH. Characterization of the AhR-hsp90-XAP2 core complex and the role of the immunophilin-related protein XAP2 in AhR stabilization. Biochemistry 38, 8907–8917 (1999).

68 Sumanasekera WK, Tien ES, Turpey R, Vanden Heuvel JP, Perdew GH. Evidence that peroxisome proliferator-activated receptor alpha is complexed with the 90-kDa heat shock protein and the hepatitis virus B X-associated protein 2. J. Bio. Chem. 278, 4467–4473 (2003).

69 Laenger A, Lang-Rollin I, Kozany C et al. XAP2 inhibits glucocorticoid receptor activity in mammalian cells. FEBS Lett. 583, 1493–1498 (2009).

70 Yano M, Terada K, Mori M. AIP is a mitochondrial import mediator that binds to both import receptor Tom20 and preproteins. J. Cell. Biol. 163, 45–56 (2003).

71 Froidevaux MS, Berg P, Seugnet I et al. The co-chaperone XAP2 is required for activation of hypothalamic thyrotropin-releasing hormone transcription in vivo. EMBO Rep. 7, 1035–1039 (2006).

72 Kang BH, Altieri DC. Regulation of survivin stability by the aryl hydrocarbon receptor-interacting protein. J. Bio. Chem. 281, 24721–24727 (2006).

73 Bolger GB, Peden AH, Steele MR et al. Attenuation of the activity of the cAMP-specific phosphodiesterase PDE4A5 by interaction with the immunophilin XAP2. J. Bio. Chem. 278, 33351–33363 (2003).

74 de Oliveira SK, Hoffmeister M, Gambaryan S, Muller-Esterl W, Guimaraes JA, Smolenski AP. Phosphodiesterase 2A forms a complex with the co-chaperone XAP2 and regulates nuclear translocation of the aryl hydrocarbon receptor. J. Biol. Chem. 282, 13656–13663 (2007).

• FirstidentifiedPDE2Aasabindingpartnerofarylhydrocarbonreceptor-interactingprotein(AIP),suggestingthatAIP–PDE2AinteractionslowerlocalcAMPlevels,decreasingunligandednucleartranslocationofAhR,providingapotentialexplanationofthemechanismbehindthetumor-supressorroleofAIP.

75 Vargiolu M, Fusco D, Kurelac I et al. The tyrosine kinase receptor RET interacts in vivo with aryl hydrocarbon receptor-interacting protein to alter survivin availability. J. Clin. Endocrinol. Metab. 94, 2571–2578 (2009).

76 Kashuba E, Kashuba V, Pokrovskaja K, Klein G, Szekely L. Epstein-Barr virus encoded nuclear protein EBNA-3 binds XAP-2, a protein associated with hepatitis B virus X antigen. Oncogene 19, 1801–1806 (2000).

77 Meyer BK, Pray-Grant MG, Vanden Heuvel JP, Perdew GH. Hepatitis B virus X-associated protein 2 is a subunit of the unliganded aryl hydrocarbon receptor core complex and exhibits transcriptional enhancer activity. Mol. Cell. Biol. 18, 978–988 (1998).

78 Gu YZ, Hogenesch JB, Bradfield CA. The PAS superfamily: sensors of environmental and developmental signals. Ann. Rev. Pharmacol. Toxicol. 40, 519–561 (2000).

79 Kazlauskas A, Poellinger L, Pongratz I. Evidence that the co-chaperone p23 regulates ligand responsiveness of the dioxin (Aryl hydrocarbon) receptor. J. Bio. Chem. 274, 13519–13524 (1999).

80 Morales JL, Perdew GH. Carboxyl terminus of hsc70-interacting protein (CHIP) can remodel mature aryl hydrocarbon receptor (AhR) complexes and mediate ubiquitination of both the AhR and the 90 kDa heat-shock protein (hsp90) in vitro. Biochemistry 46, 610–621 (2007).

81 Pearl LH, Prodromou C, Workman P. The Hsp90 molecular chaperone: an open and shut case for treatment.  Biochem. J. 410, 439–453 (2008).

82 de Oliveira SK, Smolenski A. Phosphodiesterases link the aryl hydrocarbon receptor complex to cyclic nucleotide signaling.  Biochem. Pharmacol. 77, 723–733 (2009).

83 Park JH, Mangal D, Frey AJ, Harvey RG, Blair IA, Penning TM. Aryl hydrocarbon receptor facilitates DNA strand breaks and 8-oxo-2 -́deoxyguanosine formation by the aldo-keto reductase product benzo[a]pyrene-7,8-dione. J. Bio. Chem. 284, 29725–29734 (2009).

84 Grimmer G, Bohnke H. Polycyclic aromatic hydrocarbon profile ana lysis of high-protein foods, oils, and fats by gas chromatography. J. Assoc. Off. Anal. Chem. 58, 725–733 (1975).

85 Dipple A. Formation, metabolism, and mechanism of action of polycyclic aromatic hydrocarbons. Cancer Res. 43, 2422S-2425S (1983).

86 Rothman N, Poirier MC, Baser ME et al. Formation of polycyclic aromatic hydrocarbon-DNA adducts in peripheral white blood cells during consumption of charcoal-broiled beef. Carcinogenesis 11, 1241–1243 (1990).

87 Hecht SS. Tobacco smoke carcinogens and lung cancer. J. Natl Cancer Inst. 91, 1194–1210 (1999).

88 Pfeifer GP, Denissenko MF, Olivier M, Tretyakova N, Hecht SS, Hainaut P. Tobacco smoke carcinogens, DNA damage and p53 mutations in smoking-associated cancers. Oncogene 21, 7435–7451 (2002).

89 Vogel CF, Sciullo E, Li W, Wong P, Lazennec G, Matsumura F. RelB, a new partner of aryl hydrocarbon receptor-mediated transcription. Mol. Endocrinol. 21, 2941–2955 (2007).

90 Ramadoss P, Petrulis JR, Hollingshead BD, Kusnadi A, Perdew GH. Divergent roles of hepatitis B virus X-associated protein 2 (XAP2) in human versus mouse Ah receptor complexes. Biochemistry 43, 700–709 (2004).

91 Petrulis JR, Kusnadi A, Ramadoss P, Hollingshead B, Perdew GH. The hsp90 co-chaperone XAP2 alters importin beta recognition of the bipartite nuclear localization signal of the Ah receptor and represses transcriptional activity. J. Bio. Chem. 278, 2677–2685 (2003).

92 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. Bio. Chem. 275, 37448–37453 (2000).

93 Reyes H, Reisz-Porszasz S, Hankinson O. Identification of the Ah receptor nuclear translocator protein (Arnt) as a component of the DNA binding form of the Ah receptor. Science 256, 1193–1195 (1992).

Page 14: Role of the aryl hydrocarbon receptor-interacting protein in

Expert Rev. Endocrinol. Metab. 5(5), (2010)694

Review Cain, Miljic, Popovic & Korbonits

94 Hankinson O. The aryl hydrocarbon receptor complex. Ann. Rev. Pharmacol. Toxicol. 35, 307–340 (1995).

95 Fujii-Kuriyama Y, Mimura J. Molecular mechanisms of AhR functions in the regulation of cytochrome P450 genes. Biochem. Biophys. Res. Commun. 338, 311–317 (2005).

96 Ikuta T, Tachibana T, Watanabe J, Yoshida M, Yoneda Y, Kawajiri K. Nucleocytoplasmic shuttling of the aryl hydrocarbon receptor. J. Biochem. 127, 503–509 (2000).

97 Kazlauskas A, Sundstrom S, Poellinger L, Pongratz I. The hsp90 chaperone complex regulates intracellular localization of the dioxin receptor. Mol. Cell. Biol. 21, 2594–2607 (2001).

98 Richter CA, Tillitt DE, Hannink M. Regulation of subcellular localization of the aryl hydrocarbon receptor (AhR). Arch. Biochem. Biophys. 389, 207–217 (2001).

99 Oesch-Bartlomowicz B, Huelster A, Wiss O et al. Aryl hydrocarbon receptor activation by cAMP vs. dioxin: divergent signaling pathways. Proc. Natl Acad. Sci. USA 102, 9218–9223 (2005).

• FirstidentifiedcAMPasadirectinteractingpartnerofthearylhydrocarbonreceptor(AhR),resultinginnucleartranslocationofAhRindependentofligand.ThisfindingresultedinincreasedinterestinthephosphodiesterasesandtheirinteractionswiththeAIPasapotentialtumor-supressormechanismofAIP.

100 Fernandez-Salguero P, Pineau T, Hilbert DM et al. Immune system impairment and hepatic fibrosis in mice lacking the dioxin-binding Ah receptor. Science 268, 722–726 (1995).

101 Schmidt JV, Su GH, Reddy JK, Simon MC, Bradfield CA. Characterization of a murine Ahr null allele: involvement of the Ah receptor in hepatic growth and development. Proc. Natl Acad. Sci. USA 93, 6731–6736 (1996).

102 Lahvis GP, Lindell SL, Thomas RS et al. Portosystemic shunting and persistent fetal vascular structures in aryl hydrocarbon receptor-deficient mice. Proc. Natl Acad. Sci. USA 97, 10442–10447 (2000).

103 Hollingshead BD, Petrulis JR, Perdew GH. The aryl hydrocarbon (Ah) receptor transcriptional regulator hepatitis B virus X-associated protein 2 antagonizes p23 binding to Ah receptor-Hsp90

complexes and is dispensable for receptor function. J. Bio. Chem. 279, 45652–45661 (2004).

104 Paajarvi G, Viluksela M, Pohjanvirta R, Stenius U, Hogberg J. TCDD activates Mdm2 and attenuates the p53 response to DNA damaging agents. Carcinogenesis 26, 201–208 (2005).

105 Weiss C, Faust D, Schreck I et al. TCDD deregulates contact inhibition in rat liver oval cells via Ah receptor, JunD and cyclin A. Oncogene 27, 2198–2207 (2008).

106 Zudaire E, Cuesta N, Murty V et al. The aryl hydrocarbon receptor repressor is a putative tumor suppressor gene in multiple human cancers. J. Clin. Invest. 118, 640–650 (2008).

107 Ray S, Swanson HI. Activation of the aryl hydrocarbon receptor by TCDD inhibits senescence: a tumor promoting event?  Biochem. Pharmacol. 77, 681–688 (2009).

108 Ishida M, Mikami S, Kikuchi E et al. Activation of the aryl hydrocarbon receptor pathway enhances cancer cell invasion by upregulating the MMP expression and is associated with poor prognosis in upper urinary tract urothelial cancer. Carcinogenesis 31, 287–295 (2010).

109 Huang G, Elferink CJ. Multiple mechanisms are involved in Ah receptor-mediated cell cycle arrest. Mol. Pharmacol. 67, 88–96 (2005).

110 Marlowe JL, Puga A. Aryl hydrocarbon receptor, cell cycle regulation, toxicity, and tumorigenesis. J. Cell. Biochem. 96, 1174–1184 (2005).

111 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. Bio. Chem. 275, 2943–2950 (2000).

112 Peng L, Mayhew CN, Schnekenburger M, Knudsen ES, Puga A. Repression of Ah receptor and induction of transforming growth factor-beta genes in DEN-induced mouse liver tumors. Toxicology 246, 242–247 (2008).

113 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. 77, 195–201 (2010).

114 Fan Y, Boivin GP, Knudsen ES, Nebert DW, Xia Y, Puga A. The aryl hydrocarbon receptor functions as a tumor suppressor of liver carcinogenesis. Cancer Res. 70, 212–220 (2010).

115 Hall JM, Barhoover MA, Kazmin D, McDonnell DP, Greenlee WF, Thomas RS. Activation of the aryl-hydrocarbon receptor inhibits invasive and metastatic features of human breast cancer cells and promotes breast cancer cell differentiation. Mol. Endocrinol. 24, 359–369 (2010).

116 Heliovaara E, Raitila A, Launonen V et al. The expression of AIP-related molecules in elucidation of cellular pathways in pituitary adenomas. Am. J. Pathol. 175, 2501–2507 (2009).

117 Wyde ME, Braen AP, Hejtmancik M et al. Oral and dermal exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induces cutaneous papillomas and squamous cell carcinomas in female hemizygous Tg.AC transgenic mice. Toxicol. Sci. 82, 34–45 (2004).

118 Hebert CD, Harris MW, Elwell MR, Birnbaum LS. Relative toxicity and tumor-promoting ability of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), 2,3,4,7,8-pentachloro-dibenzofuran (PCDF), and 1,2,3,4,7,8-hexachlorodibenzofuran (HCDF) in hairless mice. Toxicol. Appl. Pharmacol. 102, 362–377 (1990).

119 Poland A, Palen D, Glover E. Tumour promotion by TCDD in skin of HRS/J hairless mice. Nature 300, 271–273 (1982).

120 Marlowe JL, Fan Y, Chang X et al. The aryl hydrocarbon receptor binds to E2F1 and inhibits E2F1-induced apoptosis. Mol. Biol. Cell. 19, 3263–3271 (2008).

121 Jaffrain-Rea ML, Angelini M, Gargano D et al. Expression of aryl hydrocarbon receptor (AHR) and AHR-interacting protein in pituitary adenomas: pathological and clinical implications. Endocr. Relat. Cancer 16, 1029–1043 (2009).

122 Trivellin G, Occhi G, Albiger N et al. AHRR is overexpressed in somatotropinomas. Presented at: XXXIV Giornate Endocrinologiche Pisane. Pisa, Italy, 10–12 June 2010.

123 McPhee I, Pooley L, Lobban M, Bolger G, Houslay MD. Identification, characterization and regional distribution in brain of RPDE-6 (RNPDE4A5), a novel splice variant of the PDE4A cyclic AMP phosphodiesterase family.  Biochem. J. 310(Pt 3), 965–974 (1995).

Page 15: Role of the aryl hydrocarbon receptor-interacting protein in

www.expert-reviews.com 695

ReviewRole of the AIP gene in familial isolated pituitary adenoma

124 Landis CA, Masters SB, Spada A, Pace AM, Bourne HR, Vallar L. GTPase inhibiting mutations activate the a chain of Gs and stimulate adenylyl cyclase in human pituitary tumours. Nature 340, 692–696 (1989).

125 Horvath E, Kovacs K. Pathology of acromegaly. Neuroendocrinology 83, 161–165 (2006).

126 Melmed S. Aryl hydrocarbon receptor interacting protein and pituitary tumorigenesis: another interesting protein. J. Clin. Endocrinol. Metab. 92, 1617–1619 (2007).

127 Salvesen GS, Duckett CS. IAP proteins: blocking the road to death’s door. Nat. Rev. Mol. Cell Biol. 3, 401–410 (2002).

128 Fortugno P, Beltrami E, Plescia J et al. Regulation of survivin function by Hsp90. Proc. Natl Acad. Sci. USA 100, 13791–13796 (2003).

129 Georgitsi M, Karhu A, Winqvist R et al. Mutation ana lysis of aryl hydrocarbon receptor interacting protein (AIP)

gene in colorectal, breast, and prostate cancers. Br. J. Cancer 96, 352–356 (2007).

130 Cazabat L, Guillaud-Bataille M, Bertherat J, Raffin-Sanson ML. Mutations of the gene for the aryl hydrocarbon receptor-interacting protein in pituitary adenomas. Horm. Res.  71, 132–141 (2009).

131 Khoo SK, Pendek R, Nickolov R et al. Genome-wide scan identifies novel modifier loci of acromegalic phenotypes for isolated familial somatotropinoma. Endocr. Relat. Cancer 16, 1057–1063 (2009).

132 Marrie P. Sur deux cas d’acromegalie. Hypertrophe singuliere no congénitale des extrémités supérieures, inférieures et cephalique. Révue Medicale Française 6, 197–333 (1886).

133 de Herder WW. Acromegaly and gigantism in the medical literature. Case descriptions in the era before and the early

years after the initial publication of Pierre Marie (1886). Pituitary 12, 236–244 (2009).

134 Langer K. Über Wachstum des menschlichen Skelets, mit Bezug auf den Riesen. Denkschr der Kais Akad der Wissensch zu Wien Math-naturw Kl. 31 (1872).

135 Karhu A, Aaltonen LA. Susceptibility to pituitary neoplasia related to MEN-1, CDKN1B and AIP mutations: an update. Hum. Mol. Genet. 16(Spec. No. 1), R73–R79 (2007).

136 Brandi ML, Gagel RF, Angeli A et al. Guidelines for diagnosis and therapy of MEN type 1 and type 2. J. Clin. Endocrinol. Metab. 86, 5658–5671 (2001).

137 Prescott RWG, Spruce BA, Kendall-Taylor P, Hall K, Hall R. Acromegaly and gigantism presenting in two brothers. Presented at: 1st Joint Meeting British Endocrine Society London, UK, 25–27 May 1982.