Expression of pituitary developmental signalling molecules and TFs
1. Introduction
Pituitary adenomas (PA) are frequent and typically benign endocrine neoplasia, which clinical prevalence is estimated around 1/1000 inhabitants [1]. The vast majority are sporadic. PA are endowed with significant clinical morbidity related to hormonal hypersecretion, neurological symptoms due to intracranial mass effects or invasion of the surrounding structures and/or secondary hypopituitarism. Their evolution is quite variable, ranging from indolent tumours with an extremely slow growing potential, to recurrent, aggressive, and exceptionally malignant tumours. Their current clinical management is based on pharmacological treatment, mainly dopamine-agonists (DA) and somatostatin analogues (SSA), surgery and radiotherapy [2]. Despite considerable progress in the management of PA, a significant subset of patients are not satisfactorily controlled. Long-term uncontrolled pituitary hormone hypersecretion, leading to potential severe systemic diseases, and tumour recurrence or aggressiveness still represent a difficult clinical challenge. Understanding the mechanisms involved in the pathogenesis of PA is essential for the development of new therapeutic strategies. In this chapter, we will summarize current concepts in pituitary tumorigenesis and focus our attention on the most recent insights and new perspectives in this field.
2. Pituitary tumours
Primary pituitary tumours in human are mainly represented by PA arising from endocrine cells in the anterior lobe and craniopharyngiomas. These latter are divided into adamantinomatous, which derive from the Rathke’s pouch, and papillary craniopharyngiomas. Additional tumour types deriving from non-endocrine cells of the anterior pituitary and the neurophypohysis can be found [3], which pathogenesis is poorly known and will not be considered in this review.
2.1. Classification
PA may be classified according to their macroscopic characteristics into micro- (< 1 cm) or macro-adenomas (≥ 1 cm), and enclosed or invasive adenomas. Invasion of the surrounding structures (cavernous sinuses, bone, sphenoidal sinus) is generally defined according to neuroradiological imaging – especially magnetic resonance imaging -, although intra-operative findings may introduce some correction to the pre-operative radiological classification or reveal macroscopic dural invasion. Of note, microscopic evidence of dural invasion is rarely present on surgical samples. The functional classification of PA is based on their hormone-secreting potential, which may be associated with bio-clinical evidence of hormone hypersecretion or recognized by immunohistochemistry (IHC) for pituitary hormones and/or by specific ultrastructural features. From an epidemiological point of view, prolactinomas are by far the most frequent (50-60%), followed by clinically non-functioning PA (NFPA) (20-30%), somatotrophinomas (10-15%), corticotrophinomas (5-10%) and thyreotrophinomas (1-2%) [1,2,4]. Recruitment bias are frequently encountered in pathological series, since most prolactinomas are treated by DA only. Clinico-pathological correlations in PA have been recently reviewed [4]. In the large majority of PA associated with bio-clinical evidence of hormone secretion, except functional hyperprolactinemia, pathological examination will confirm the diagnosis of PRL, GH, ACTH or TSH-secreting tumours and potentially identify bi- or multi-hormonal secretion. This is especially true for GH-secreting PA, which may also secrete PRL, less frequently glycoprotein hormones, or both (multihormonal). TSH-secreting adenomas are rare and frequently multihormonal. Ultrastructural studies of secreting PA may disclose a “densely granulated” (DG) or “sparsely granulated” (SG) pattern, which may reflect significant differences in hormone secretion and tumour behaviour. This has been well studied in somatotrophinomas, where IHC for cytokeratin can be used to disclose the typical “dot-like” staining pattern of SG adenomas. Although a continuum exists between the SG and DG types, pure SG are typically more aggressive than DG somatotrophinomas [5]. Pathological examination of NFPA may show negative immunostaining for all pituitary hormones (the “null cell” or endocrine inactive histotype), positive immunostaining for FSH and/or LH (gonadotrophinomas) or reveal silent secretion of other pituitary hormones, in particular ACTH and GH (“silent” secreting PA). Cell lineage may also be identified by the expression of specific transcription factors (see “pituitary ontogenesis”) [4]. It is generally accepted that most NFPA derive from the gonadotroph lineage, since data obtained from primary cultures or molecular analysis of these tumours frequently reveal a silent expression of gonadotropins or their subunits, including α−subunit only. Also, transgenic mice overexpressing SV40 under the control of the βFSH promoter develop gonadotroph hyperplasia and PA with reduced gonadotropin immunoreactivity and ultrastructural characteristics similar to human null cell PA [6]. Silent corticotroph adenomas are commonly aggressive and different subtypes have been described. Pituitary carcinomas are strictly defined by the presence of extra-pituitary dissemination (see “pituitary carcinomas”). Therefore, no diagnosis of pituitary carcinoma can be made on a surgical pituitary sample. Considerable efforts have been made to recognize the aggressive potential of PA according to pathological criteria. However, mitoses are generally rare and the percentage of cells immunopositive for the Ki67 antigen (which is expressed throughout the cell cycle and detected with the MIB1 monoclonal antibody) is currently considered as the best marker of cell proliferation. A Ki67 index ≥3% is commonly associated with invasiveness [4], although it can be reduced by pre-operative pharmacological treatment in secreting PA [7]. Immunostaining for p53 is also frequently associated with invasiveness and is typically present in carcinomas [4]. For these reasons, the 2004 WHO conference has proposed to define as “atypical adenomas” a subset of invasive PA characterized by Ki67 labelling ≥ 3% and extensive p53 nuclear staining [8]. However, many criticisms remain and search for reliable markers of aggressiveness or malignancy is going on. Gene expression profiling comparing non-invasive adenomas with their invasive counterpart [9, 10] or with pituitary carcinomas [11] represents a promising approach.
2.2. Origin
The pathogenesis of PA is multifactorial. Traditionally, two theories have been proposed: the primary pituitary origin, and the hypothalamic origin of PA or, more generally speaking, the concept that PA may derive from abnormal pituitary regulation. Acromegaly or Cushing’s disease resulting from ectopic secretion of GHRH or CRH by neuroendocrine tumours, respectively, or estrogen-induced prolactinomas, represent the main evidence for the second theory. However, the chief lesion in such conditions is hyperplasia, with PA developing in a very minority of cases, whereas hyperplasia is exceptionally observed surrounding the tumoral tissue in human PA. Although hyperplasia may be difficult to identify (reticulin staining is not routinely proposed), there is accumulating evidence supporting the primary pituitary origin of PA. Indeed, PA are essentially monoclonal in origin, and a number of genetic abnormalities have been identified these tumours, which include somatic events and inherited predisposition. The relatively low rate of recurrences following complete surgical removal of enclosed PA, especially microadenomas, also favours a primary pituitary hypothesis. However, polyclonality can also be observed on PA, and different clones may develop in a synchronous or delayed pattern, possibly accounting for tumour progression or regrowth [12]. A unifying view is that pituitary tumorigenesis is a multistep and multifactorial process, which includes early initiating genetic events, growth promotion by extracellular factors (including the extracellular matrix, growth factors, cytokines, neuropeptides and peripheral hormones) and additional genetic events contributing to a further progression of the tumour in terms of invasiveness, recurrences and, exceptionally, metastasis [13,14].
3. Pituitary developmental pathways and their potential alterations in pituitary tumours
Because the expanding knowledge about the molecular mechanisms involved in pituitary ontogenesis is providing new clues in the understanding of pituitary tumorigenesis, relevant findings in this field will be summarized.
3.1. Ontogenesis of the pituitary gland
The anterior pituitary lobe (AP) derives from an invagination of the oral ectoderm forming the Rathke’s pouch (RP), which cells proliferate and subsequently undergo progressive terminal differentiation into the 5 adult pituitary cell types (corticotrophs; somatotrophs, lactotrophs, gonadotrophs and thyreotrophs), whereas the posterior lobe (or neurohypophysis) derives from a specialized region of the neuroectoderm, the infundibulum. The intermediate lobe also arises from the RP and contains melanotrophs in rodents but is virtually absent in humans. The molecular mechanisms of pituitary ontogenesis have been mainly studied in the mouse and, in addition to genetic models which have contributed to elucidate the role of single proteins [6], analysis of transcriptomes obtained from cDNA libraries in the developing embryo represents a promising tool to identify new genes involved in this process [15,16]. Complex interactions between signalling molecules (in particular opposite signals coming from the diencephalon and the ventral ectoderm) and pituitary transcriptions factors (TFs) are involved and tightly regulated in a spatially and temporally organised manner. Extensive reviews are available on this topic [17,18]. Genetic defects in pituitary TFs are responsible for inherited abnormalities in pituitary development, spanning from syndromic diseases due to defects in early factors (
|
|
|
|
|
|||
BMPs (BMP 2 and 4) |
VD | BMP4: essential for the invagination of Rathke’s pouch; BMP2 |
BMP4: overexpressed in prolactinomas, underexpressed in corticotrophinomas |
FGFs (FGF 8 and 10) |
VD/RP | FGF10 : essential for cell survival FGF8: involved in the maintenance of RP cells proliferation opposing gradients with BMP2 |
NA |
WNT4 and 5A/ β-catenin |
VD/RP | Involved in pituitary development and in the induction of |
Wnt4 : expressed in GH/PRL/TSH-secreting PA |
Shh/ |
VD; oral ectoderm except RP | Involved in early proliferation and cell type determination as well as in the control of adult pituitary cell function and proliferation | Shh: underexpressed in PA, including corticotrophinomas |
Notch 2 and 3 | VD/RP | Required for early lineage commitment and the terminal differentiation of distinct cell lineages | Notch3: overexpressed in NFPA |
Sox2 | VD/RP | Required for pituitary development and the maintenance or proliferation of pituitary progenitor cells | Expressed in adamantinomatous craniopharyngiomas |
|
|||
Hesx1 | VD/RP | Involved in early pituitary development | Expressed in all PA phenotypes |
Otx2 | RP | Involved in RP development, in particular in the expression of |
NA |
Lhx3-4 | RP | Required for cell survival and prevention of apoptosis | NA |
Isl1 | RP | Required for the proliferation and differentiation of pituitary progenitors | NA |
Six 1, 3 and 6 | RP | Six1 regulates cell proliferation; Six6 is required for pituitary development and cell proliferation; Six3 interacts with Hesx1 | NA |
Pitx 1 and 2 | RP | Pitx1 e Pitx2 activate the early transcription of the Pitx1 also binds the |
Pitx1: expressed in all PA phenotypes Pitx2: overexpressed in NFPA |
FOXL2 | AP | A transcriptional activator of |
Overexpressed in NFPA |
|
|||
NeuroD1 | RP | A transcriptional activator of |
Expressed in corticotrophinomas and a subset of NFPA |
Tpit | POMC precursor | A transcriptional activator of |
Expressed in secreting and silent corticotrophinomas |
|
|||
Prop1 | RP | A transcriptional activator of |
Expressed in all PA phenotypes |
Gata2 | RP | Interacts and cooperates with |
Expressed essentially in gonadotroph and thyreotroph PA |
Pit-1 | Pit-1 lineages | Essential for the terminal differentiation and expansion of GH/PRL/TSH-secreting cells; an enhancer of |
Expressed in GH/PRL/TSH-secreting PA (including NFPA with silent secretion) |
CREB | Pit-1 lineages | Required in somatotrophs | Expressed in all somatotrophinomas |
|
|||
Gata2 | RP/pituitary | A promoter of |
See above |
SF1 | Gonadotrope | A promoter of |
Expressed in the majority of gonadotroph/NFPA, not in GH/PRL-secreting PA |
Egr1 | Gonadotrope | Required for |
NA |
Table 1.
3.2. Developmental pathways in pituitary adenomas
Three master developmental pathways – Wnt, Hedgehog and Notch – have been involved in several human diseases, in particular in cancer. We will attempt to summarize current knowledge about their role in pituitary development and tumorigenesis, which is likely to expand significantly in the next years.
3.2.1. The Wnt/beta-catenin pathway
The canonical Wnt/β-catenin signalling pathway plays a role in the specification of pituitary progenitor/stem cells and β-catenin modulates the transcriptional activity of several pituitary TFs, in particular Pitx2, PROP-1 (thereby promoting the activation of
3.2.2. Sonic hedgehog
Sonic hedgehog (Shh) is involved in pituitary ontogenesis and regulates pituitary hormone release. Shh signalling is mediated by Patched receptors (Ptc1, Ptc2) and the Gli family of TFs. During ontogenesis, Shh is expressed by the ventral diencephalon and by the oral ectoderm immediately adjacent to RP. There is recent evidence that Shh signalling is of particular importance before the RP detachs from the oral ectoderm, and that Gli2 is responsible for the proliferation of early pituitary progenitors and for the diencephalic induction of FGF8 and BMP4 [32]. This is consistent with reports of human inactivating Gli2 germline mutations associated with severe pituitary developmental defects and different degrees of craniofacial abnormalities [33]. In the adult pituitary Shh/Ptc2/Gli1 signalling is active in corticotrophs and stimulates POMC transcription and ACTH release; Gli1 is necessary for CRF signalling [34]. Contrasting with the corticotroph specificity of Shh, Ptc receptors are also expressed in other pituitary secreting cells, although in a phenotype-specific manner [34]. Shh immunostaining was found low or absent in a large series of PA, including corticotrophinomas, whereas the expression of Ptch receptors was retained [35]. Shh was also found to stimulate ACTH, GH and PRL secretion by normal and tumorous pituitary cells
3.2.3. Notch/Hes1
Notch signalling regulates progenitor cell differentiation during embryogenesis. Notch is a cell-to-cell signalling network composed by transmembrane ligands (in mammals Delta-like 1,3,4 and Jagged 1,2), the transmembrane Notch receptors (Notch 1-4), and a transcription factor (in mammals RBPJ), which is activated by the intracellular fragment of Notch after ligand-induced Notch cleavage has occurred. Notch target genes encode beta-helix loop-helix (bHLH) TFs, in particular the
3.3. Pituitary stem cells
The pituitary gland is characterized by a high degree of plasticity, which is involved in pituitary function changes through life and adaptation to physiological and pathological variations in peripheral hormone feed-back. Several conditions require a re-organization of pituitary cell composition, with an expansion of a specific cell pool (
4. Genetics of pituitary tumours
PA are triggered by a variety of genetic abnormalities [13,14,47]. The vast majority occur at a somatic level, but inherited predisposition is being increasingly recognized.
4.1. Inherited predisposition to pituitary tumours
Although inherited predisposition to PAs is often recognized in a familial setting, a sporadic presentation may occur. In addition to their implications for familial screening, genes involved in hereditary tumours may provide important clues in the comprehension of the molecular basis of tumorigenesis. We have recently reviewed this topic and proposed an algorithm for genetic screening in patients with PA [48].
4.1.1. Multiple Endocrine Neoplasia type 1 (MEN1)
MEN1 is an autosomal dominant condition defined by the presence, in a single subject or within a single family, of two or more hyperplastic and/or adenomatous lesions of the parathyroid glands (~90%), the gastro-entero-pancreatic (GEP) tract (30-80%) and/or the anterior pituitary (~40%). The clinical characteristics of MEN1-related PA have been recently reviewed elsewhere [48,49]. Briefly, their phenotypic distribution is unremarkable when compared to sporadic PA, with a predominance of prolactinomas (~60%), but they are more frequently invasive and resistant to pharmacological treatment. Pediatric onset is not uncommon, especially for PRL- and ACTH-secreting PA, and carcinomas have been reported. Peri-tumoral hyperplasia is rare, but multiple and plurihormonal adenomas are more frequent than in sporadic PA [50]. Since the identification of the
4.1.2. Carney Complex (CNC)
The "complex of myxomas, spotty skin pigmentation, and endocrine overactivity” is a rare and heterogeneous condition, characterized by the association of endocrine overactivity and tumors - Primary Pigmented Nodular Adrenocortical Disease (PPNAD), acromegaly, thyroid and gonadal tumors – with cardiac myxomas, schwannomas and skin pigmented lesions [57]. Primary pituitary presentation is rare and pituitary abnormalities are mainly represented by an hyperplasia of GH/PRL-secreting cells, which frequently translates into mild hyperprolactinemia and/or subclinical alterations of GH/IGF1 secretion. Early onset GH/IGF1 hypersecretion may induce gigantism, but acromegaly and somatotrophinomas develop in a minority of patients (15%). Up to 70% of CNC patients present in a familial setting, with an autosomal dominant transmission. Germline heterozygote mutations in the
4.1.3. McCune Albright Syndrome (MAS)
MAS is a rare sporadic disease due to post-zygotic activating mutations in the α
4.1.4. MEN-4 and other CDKI-related disorders
Inactivating mutations in the
4.1.5. The pituitary adenoma-paraganglioma/pheochromocytoma association: A new syndrome?
Succinate dehydrogenase (SDH) is a mitochondrial enzyme composed of four functionally different subunits A,B,C,D. Mutations in genes encoding the SDH B,C and D subunits predispose to pheochromocytomas/ paragangliomas and additional tumours, including thyroid cancer. Recently, a germline mutation in the
4.1.6. Familial Isolated Pituitary Adenomas (FIPA) and the Aryl hydrocarbon receptor Interacting Protein (AIP) gene
Familial Isolated Pituitary Adenomas (FIPA) are defined by the familial presentation of PA in the absence of syndromic features. In 2006, 64 European FIPA kindreds were reported, including patients affected by prolactinomas (55%), somatotrophinomas (~30%), non-secreting PA (~15%) and corticotrophinomas (<5%) [69]. The prevalence of FIPA among PA was estimated about 2-3% and kindreds were almost equally divided into homogeneous and heterogeneous, as defined by the familial association of PA with a single or multiple phenotypes, respectively. Familial homogeneous somatotrophinomas, previously reported as “Isolated Familial Somatotrophinomas”, accounted for ~20% of the whole series. In the same year, the
4.2. Somatic events in pituitary tumours
Somatic events in pituitary tumours include genetic and epigenetic changes. Intragenic mutations are less frequently encountered than in other solid tumours. Indeed, oncogene activation is mainly triggered by the overexpression of genes involved in extracellular signalling and cell cycle progression, with a few gain-of-function mutations and occasional rearrangements being reported. Inactivation of tumour suppressor genes is very common and occurs through epigenetic changes more frequently than through loss-of-function mutations and/or allelic deletions. MicroRNAs (miRs) have also recently emerged as important regulators of gene/protein expression in pituitary tumours.
4.2.1. Chromosome and DNA alterations in PA
Several chromosome abnormalities have been reported in PA, although on limited series of tumours, and include aneuploidy and evidence for intrachromosomal gain or loss of DNA. For example, trisomies involving chromosomes 5, 8 and 12 appear to be very frequent in prolactinomas [82], whereas trisomies involving chromosomes 7, 9 and 20 were reported in NFPA [83]. Conversely, monosomy of chromosome 11 was observed in an aggressive MEN1-related prolactinoma [84] and LOH with single or multiple allelic deletions at different loci have been reported, which also appear to be more frequent in invasive PA. For instance, the frequency of LOH in 11q13, 13q12-14 and/or 10q26 was found to increase from <10% in low grade to nearly 75% in high grade PA, according to a modified Hardy’s classification [85]. As compared to other solid tumours, classical somatic oncogenic mutations are relatively rare in PA [reviewed in 13,47]. The most frequent oncogenic mutation is represented by activating missense mutations in the
4.2.2. Epigenetics in PA
Epigenetic changes are mainly characterized by DNA methylation and histone modifications, leading to chromatin remodelling and regulation of gene expression through a modulation of DNA accessibility to TFs. Epigenetic changes may coexist with genetic events (
4.2.3. The emerging role of microRNAs and other non-coding RNAs
MicroRNAs (miRs) are small non-coding RNA molecules (18-25 nucleotides) involved in the post-transcriptional regulation of mRNAs stability. More than 600 miRs have been reported in the human genome and up to 30% of genes are believed to be regulated by miRs. Briefly, miRs derive from the intracellular processing of precursors called pri- and pre-miRNAs, and mature miRs bind to the 3’ untranslated sequence of mRNA target molecules, leading to the formation of miRs duplexes. MiRs duplexes are incorporated into protein containing RNA-induced silencing complexes (RISC) and, according to their degree of complementarity with miRs, target mRNAs can be cleaved (perfect complementarity) or undergo partial degradation and translational inhibition (imperfect complementarity, which is the most common figure). MiRs have been involved in the control of pituitary development [100] and function [101]. Alterations in miRs expression profile, including loss or gain of expression, have been increasingly involved in pituitary tumorigenesis. Since the first report on miR15a and miR16-1 downregulation in GH- and PRL-secreting macroadenomas [102], more than 100 dysregulated miRs have been identified in PA, with phenotype-specific expression profiles being reported in GH- and/or PRL-secreting, ACTH-secreting or NFPA [103-112]. Although in most cases their biological significance is not fully understood, several miRs have been involved in the control of pituitary cell cell proliferation, differentiation, apoptosis, cell adhesion and metabolism, and a subset has been linked to a more aggressive behavior or even to malignant transformation [103, 104, 110]. Target mRNAs are also being increasingly recognized. For instance,
|
|
|
|
|
|
miR-128a | 2q21.3 | Overexpression | NFPA | Target gene: |
109 |
Down-regulation | All PA GH |
Target gene: |
103 111 |
||
miR-26a/b | 3p21 (a) 2q35 (b) |
Overexpression | NFPA (miR-26a) GH (miR-26a/b) |
Potential target genes: Target gene (miR26b): |
103 111 |
Down-regulation | GH/PRL/NFPA (miR 26a) |
Target genes: |
112 | ||
miR-191 | 3p21 | Overexpression | All PA | Cell proliferation | 103 |
miR-145 | 5q32 | Down-regulation | GH | Target genes: |
106 |
miR-30a/b/c/d | 6q13 (c); 8q24.2 (b) |
Overexpression | ACTH | - | 103 |
miR-320 | 8p21.3 | Overexpression | GH | - | 107 |
miR-24-1 | 9q22.1 | Down-regulation | All PA | Predicted target genes: |
103 |
Let7-a | 9q22.32 | Down-regulation | All PA | Target genes: Potential target genes: |
103, 104, 112 |
miR-126 | 9q34.3 | Down-regulation | GH | Amplification of PI3K signalling; |
106 |
miR-107 | 10q23 | Overexpression | GH/ NFPA | Target gene: |
105, 111 |
miR-326 | 11q13.4 | Down-regulation | GH, some PRL NFPA |
Target genes: |
107 |
miR-141 | 12p13 | Down-regulation | ACTH | Tumor growth and tumor local invasion | 103 |
miR-16-1 | 13q14 | Down-regulation | All PA | Target genes: |
102- 104, 112 |
miR-15a | 13q14 | Down-regulation | All PA, in particular GH/PRL | Target genes: |
102, 103, 112 |
miR-20a | 13q31.3 | Overexpression | NFPA | Target gene: |
109 |
miR-493 | 14q32.2 | Overexpression | ACTH (carcinoma) | Target genes: |
110 |
miR-381 | 14q32.31 | Down-regulation | GH | Target gene: |
106 |
miR-140 | 16q22.1 | Overexpression | NFPA (macroadenomas) | Tumor growth | 103 |
miR-212 | 17p13.3 | Overexpression | All PA | Target gene: |
103 |
miR-132 | 17p13.3 | Down-regulation | All PA | - | 103 |
miR-152 | 17q21 | Overexpression | All PA | Cell proliferation | 103 |
miR-122 | 18q21.31 | Overexpression | ACTH (carcinoma) | - | 110 |
miR-23a | 19p13.13 | Overexpression | GH/PRL | - | 103 |
miR-24-2 | 19p13.13 | Overexpression | GH/PRL | See miR-24-1 | 103 |
Down-regulation | ACTH/ NFPA | See miR-24-1 | 103 | ||
miR-155 | 21q21.3 | Overexpression | GH/PRL/NFPA | Target gene: |
109 |
miR-098 |
Xp11.2 | Down-regulation | All PA | Predicted target genes involved in cell progression, cytoskeleton and vesicle organization | 103 |
Table 2.
Differential expression of microRNAs in pituitary tumours
*a transcriptional repressor of PTEN
5. Alterations in neuropeptide signalling in pituitary tumours
Hypothalamic peptides play an essential role in normal pituitary cells and their biological effects are mediated by G-protein coupled receptors (GPCRs) [19]. In addition to their dynamic control on pituitary hormone secretion and release, they may exert trophic effects on target cells - such as GHRH and CRH on somatotrophs and corticotrophs, respectively – or limit their proliferation – such as dopamine in lactotrophs. The expression of neuropeptide receptors is generally conserved in pituitary tumours and represents the molecular basis for their pharmacological treatment with DA and SSA [2]. However, abnormal expression of these receptors may be involved in paradoxical responses or in pharmacological resistance. Neuropeptides may also be produced by the pituitary gland or ectopically by neuroendocrine tumours, and their ectopic secretion may lead to pituitary hyperplasia and/or adenoma. Finally, abnormal intracellular signalling may occur in PA, contribute to pituitary tumorigenesis and potentially influence the response to pharmacological treatment. An extensive review of such processes would be beyond the scope of this work, so we will focus on the most relevant and recent findings in this field.
5.1. Abnormalities in the cAMP-PKA pathway
The cAMP/PKA pathway is essential in pituitary cells, especially in somatotrophs and in corticotrophs. Briefly, the α-subunit of the stimulatory G protein (Gsα), encoded by the
5.2. Neuropeptides, neuropeptide receptors and their implications in pituitary tumour pathogenesis and treatment
Abnormal hypothalamic neuropeptide signalling has long been involved in the pathogenesis of PA. GHRH stimulates somatotroph proliferation and causes somatotroph hyperplasia in mice, with PA developing in old transgenic GHRH animals. Hypothalamic acromegaly has been exceptionally reported in patients with gangliocytomas [118] and ectopic secretion of GHRH by neuroendocrine tumours has been well characterized. In a retrospective analysis of 21 patients with ectopic acromegaly [119], most patients showed radiological evidence of pituitary enlargement, but 5 had normal pituitary imaging and 6 had suspected PA, respectively. Out of the four cases who underwent pituitary surgery, all had somatotroph hyperplasia and 2 had concomitant GH/PRL-secreting PA. Noteworthy, a MEN1 context was identified in 8/11 cases. Ectopic secretion of CRH by neuroendocrine tumours may cause ACTH-dependent hypercortisolism, but this is an exceptional condition and ectopic secretion of ACTH and other POMC-derived peptides is by far most frequent. TRH stimulates thyrotrophs and lactotrophs, mainly through the IP3/calcium pathway, and patients with long-standing, severe, primary hypothyroidism may develop pituitary thyrotroph and lactotroph hyperplasia due to an altered feedback on TRH/TSH secretion. Similar mechanisms are involved in gonadotroph hyperplasia secondary to long-standing hypogonadism (of note, pituitary hyperplasia is not a feature of menopause). However, true “feed-back” PA are rare and are likely to require additional somatic events. Abnormal/ectopic, expression of neuropeptide receptors may also occur in PA and account for some “paradoxical” responses observed
6. Dysregulated cell growth and survival in pituitary tumours
Alterations in cell cycle control and imbalance between cell proliferation and apoptosis are general mechanisms in tumours. Such alterations may be driven by abnormal extracellular signalling and/or abnormal intracellular responses to extracellular signals, including constitutive activation of proliferative pathways and/or escape to normal regulatory signals.
6.1. Extracellular signalling in PA
In addition to neuropeptides, pituitary cells depend on extracellular signalling from the extracellular matrix, a variety of growth factors (GFs) and peripheral hormones secreted by target glands. Extracellular signalling molecules are involved in a complex network of paracrine and autocrine pathways, which are tightly regulated in the developing and adult pituitary [125]. Cell-to-cell signalling regulates pituitary hormone secretion, cell differentiation, growth, survival and plasticity, as well as angiogenesis. Abnormal signalling may therefore participate in pituitary tumorigenesis.
6.1.1. Growth factors and cytokines
The pituitary gland is an abundant source of GFs, in particular the FGF, EGF and VEGF families, and cytokines (the TGFβ family, interleukins and chemokines) [19,125]. Due to the abundant literature in this field, we will present an overview of the best studied GFs and related pathways in PA. FGFs play an important role in pituitary development. FGFs include >20 members and FGF signalling is mediated by 4 FGF receptors (FGFR1-4), with different isoforms (cell-bound, secreted, truncated) generated by alternative splicing. FGFs and FGFRs are involved in proliferative and anti-proliferative signals. The
6.1.2. Steroid hormones
Steroid hormones may play an important role in pituitary tumorigenesis. The best characterized model is represented by estrogen-induced lactotroph hyperplasia and prolactinomas. Estrogen-induced lactotroph hyperplasia occurs physiologically during pregnancy, but sustained estrogen exposure may lead to prolactinomas in some strains of rats. Although evidence for estrogen-induced prolactinomas in humans is very poor, prolactinoma growth and/or intra-tumoral hemorrhage may occur during pregnancy, especially in macroprolactinomas [144]. Among PA, prolactinomas express the higher concentration of ER [145] and estrogens can increase PA cell proliferation in primary culture [146]. Importantly, ERα and ERβ play distinct roles and an imbalance between these two isoforms has been reported in PA. Indeed, nuclear overexpression of ERα has been linked to tumour aggressiveness in prolactinomas and NFPA, whereas invasive NFPA were found to express lower ERβ [147]. Ablation of ERβ in mice was also associated with the development of invasive gonadotrophinomas in females [148]. Estrogens exert multiple effects on pituitary cells, especially on lactotrophs. They enhance the transcription of the
6.1.3. Adhesion molecules
Adhesion molecules are important to maintain cell-to-cell contact and a normal cell morphology and tissue architecture. Similarly to other epithelial tumours, PA may develop some degree of epithelial-to-mesenchymal transition, which is associated with loss of cell adherence and tumour aggressiveness. The N- and C-cadherins are involved in pituitary development and in the organization of the adult pituitary into a functional network involving FCS cells [154]. E-cadherin has been largely recognized as a TSG in the pituitary. Reduced E-cadherin expression was first associated with an aggressive behavior in prolactinomas [155]. Subsequently, downregulation of the E-cadherin gene (
6.2. Abnormalities in cell cycle control
The main positive regulators of the cell cycle are the cyclin dependent kinases (CDKs), which are activated by specific associations with cyclins (A, B, D, E). The progression through the different phases of the cell cycle, in particular the G1/S and G2/M transitions, are stimulated by cyclin/CDK complexes and suppressed by their inhibitors, CKIs. The latter are divided into the INK4 family (p16Ink4a, p15Ink4b, p18Ink4c), which negatively regulates the G1/S transition through a direct interaction with the CDK4/6 containing complexes, and the so-called “universal” Cip/Kip family (p21Cip1, p27Kip1, p57Kip2), which is involved at various phases of the cell cycle by interacting with different cyclin/CDK complexes. Down-regulation of cell cycle inhibitors (

Figure 1.
Cell cycle dysregulation in pituitary tumours
6.2.1. Abnormalities in cell cycle progression
Increased expression of D-type cyclins is a key event in the exit from the quiescent G0 state under mitogenic stimulation by GFs. They activate CDK4 and CDK6, which phosphorylate pRB and therefore indirectly induce the transcription of S-phase genes by releasing TFs of the E2F family from their interaction with pRB. Transition from late G1 to the S phase is also driven by Cyclin E/ CDK2 complexes. Overexpression of cyclins D1 and D3 has been reported in PA of different histotypes [160]. In particular, Cyclin D1 is overexpressed in ~70% of NFPA and 40% of somatotrophinomas, especially in invasive PA, with allelic imbalance suggesting gene amplification in 25% of the cases [161]. Increased GFs and Wnt/β-catenin signalling is also likely to contribute to
6.2.2. The universal Cip/kip CDKI family
Both p27Kip1 and p21Cip1/Waf1 have been involved in pituitary tumorigenesis. The p27Kip1 protein is normally localized in the nucleus of quiescent cells and undergoes rapid degradation upon mitogenic stimulation. Phosphorylation of p27Kip1 is involved in its degradation through the ubiquitin/proteasome pathway. Loss of nuclear p27 Kip1 due to a reduced expression or an abnormal cytoplasmic localization has a negative prognostic value in a variety of human neoplasia.
6.2.3. PTTG
Although PTTG1 – the most abundant and widely studied member of the PTTG family, commonly referred to as PTTG - was first identified in the rat pituitary GH4 cells, it has important oncogenic properties in a number of additional endocrine and non-endocrine neoplasia and cells overexpressing PTTG are tumorigenic
6.2.4. HMGA proteins
The HMGA family of nuclear proteins - HMGA1
6.3. Abnormal proliferative pathways in pituitary tumours
The Raf/MEK/ERK and PI3K/Akt/mTor pathways are typically activated by GFs but crosstalks with neuropeptide/GPCRs signalling pathways are being increasingly recognized [186]. Crosstalks between the Raf/MEK/ERK and PI3K/Akt/mTor pathways result in the modulation of ERK1/2 activity, which is involved in the regulation of cell growth and differentiation, depending on the cellular context, and represents an important proliferative pathway in cancer. Secondary activation of mTOR signalling represents an important link between cell proliferation and metabolism [187]. These pathways are essential in oncology, many drugs have been designed to target their effector molecules at different steps [188] and similar strategies may be of interest in selected PA [189,190].
6.3.1. The Ras/Raf/MEK/ERK pathway
The Ras/Raf/MEK/ERK pathway originates at the cell membrane with receptors for GFs or cytokines, which activate the GTPAse Ras protein family through the coupling complex Shc/Grb2/SOS. The active, GTP bound, form of Ras recruits Raf proteins, which in turn activate a cascade of phosphorylations on cytoplasmic MAP kinases (MEKs and ERKs). A large number of cytoplasmic and nuclear proteins have been recognized as ERK1/2 targets, including the ribosomal S6 kinase (which in turns phosphorylates CREB), TFs (
6.3.2. The PI3K/Akt/mTOR pathway
The PI3K/AKT/mTOR pathway also initiates at the cell membrane in response to a variety of GFs and hormones, including insulin. PI3K phosphorylates phosphoinositides, resulting in the production of phosphoinositide 3-phosphate which in turn regulates the activity and intracellular localization of a number of target proteins, among which the best characterized is Akt (also known as Protein Kinase B/PKB). PI3K is negatively regulated by the tumour suppressor PTEN. The activation of Akt/PKB results in a cascade of phosphorylations, including mTOR, GSK3β, crosstalks with the Raf/MEK/ERK pathway at different levels, and has been involved in cell proliferation and motility in a number of cancers. Moreover, repression of the tumour suppressor Zac1 and activation of β-catenin through GSK3β are indirect effects of PI3K/Akt activation. The mTOR pathway is also activated by nutrients, cellular energy levels (ATP, O2) and stress conditions; it is a major regulator of ribosomal biogenesis and protein synthesis, in particular through the activation of the ribosomal S6 kinase p70S6K and 4EBP1, which enhances the translation of
6.4. Angiogenesis and hypoxia-pathways
Although angiogenesis is involved in the progression of many solid tumours, its pathogenetic role in PA is still not well defined. The normal pituitary gland is already highly vascularised and different studies on microvessel density (MVD) in PA have provided evidence for a reduced vascularity as compared to the normal tissue. On the other hand, increased vascularity can occur, as reported in estrogen-induced prolactinoma models and in some human pituitary tumours [142]. Despite conflicting results about potential factors associated with an increased vascularity (
6.5. Apoptosis in pituitary tumours
As in other tissues, apoptosis is a physiological event during pituitary ontogenesis [212]. It is also believed to contribute to pituitary plasticity and may occur in PA, either spontaneously or in response to pharmacological treatment. Apoptosis is generally low in normal pituitaries and in PA, but is increased in pituitary carcinomas [213]. Apoptotic cells in PA can be detected on routine examination on the basis of their morphological changes, though they can be missed even by experienced pathologists, so that specific assays are more suitable for the definition of apoptotic indexes [214]. The ISEL and TUNEL assays, which are based on the visualization of DNA breaks, can be used on paraffin-embedded sections, but should be combined with morphological criteria to minimize artefacts [214]. Immunohistochemical detection of proteins involved in the apoptotic process such as activated caspase 3, cleaved cytokeratins or annexin-5 are also useful. As a general rule, apoptosis can be triggered by extra-cellular signalling by Fas ligand (FasL) or TNF interacting with “death receptors” or by endogenous signalling following mitochondrial or DNA damage, which is able to induce apoptosis in a p53-dependent manner. No significant relationship between p53 expression and apoptosis have been reported in PA, with the exception of nuclear p53 in corticotrophinomas [214]. An important determinant of apoptosis is the cellular expression of the Bcl2 family of proteins, which contains pro-apoptotic (
6.6. Senescence
Senescence is an alternative tumour-suppressive cell faith to apoptosis in benign neoplasia. It is characterised by an irreversible arrest in cell proliferation and accompanied by an increase in cell cycle inhibitors such as p53, p19ARF, p21Cip1 and p16Ink4a. Because PA remain typically benign, even in the presence of invasive features, it has been proposed that a senescence buffer in PA cells exerts a protective effect against malignancy. This could in particular explain the very low prevalence of GH-secreting carcinomas and NFPA. Indeed, beta-galactosidase, a marker of senescence, was recently found overexpressed in somatotrophinomas and NFPA [176]. Interestingly, this could be explained by different molecular mechanisms. Somatotrophinomas show intranuclear p21 accumulation (possibly induced by aneuploidy and/or p53), which is able to restrain cell proliferation [176,231]. In gonadotroph PA, which express low nuclear p21Cip1, high levels of clusterin have been proposed to restrain cell proliferation by triggering the CKIs p15Ink4b/p16Ink4a/p27Kip1 [232]. However, in both cases overexpression of PTTG and DNA damage are present [231,232]. Because senescence may also be activated by oncogenes, as originally described for Ras, this may apply to PTTG in pituitary tumours. Oncogene-induced senescence (OIS) is a protective mechanism against cancer which may also involve cytokines. Due to its role in pituitary development and its frequent expression in PA, IL6 is an attractive candidate for OIS in PA [131].
7. Pituitary carcinomas
Pituitary carcinomas represent about 0.2% of symptomatic primary pituitary tumours and are defined exclusively by the presence of metastases. Their prevalence may be somewhat underestimated, since metastases can be discovered post-mortem and the number of reported cases has been significantly increasing during the last 15 years [233-235]. The current interest in pituitary carcinomas certainly reflects the recent improvements in their diagnosis and therapeutic management. Their clinical characteristics have been reviewed in details elsewhere [233-236]. Briefly, most pituitary carcinomas are secreting (>80%), with malignant prolactinomas and corticotrophinomas being the most frequently encountered. They usually present as recurrent invasive macroadenomas, with an increasing degree of pharmacological resistance. Noteworthy, silent ACTH-secreting PA may become functional as malignant transformation occurs. Metastases may develop in the central nervous system (with intracranial and/or spinal localizations) or present as systemic secondary tumours, in particular in the bones, lungs or liver. Therefore, malignant transformation is a late event, which typically complicates the evolution of an aggressive PA, although exceptions have been reported. Yet, there is no specific biological marker of pituitary carcinoma and no reliable prognostic marker of potential malignant transformation in PA. The primary pituitary tumour often displays a high mitotic index and extensive p53 immunostaining, the apoptotic index is typically higher than in PA, but none of these features is invariably present and no threshold value of Ki67 or p53 immunopositivity can be defined. Several molecular abnormalities are encountered more frequently in pituitary carcinomas, such as chromosome gains,
8. Conclusion
Pituitary tumours are very heterogeneous and their pathogenesis is multifactorial. During the last two decades, increasing knowledge about factors involved in pituitary ontogenesis, physiology and genetics have provided significant new information concerning dysregulated pathways in pituitary tumorigenesis. The rapid development of new methodological approaches allowing to explore hundreds of genes and proteins simultaneously (genomics/epigenomics/proteomics) has become an essential tool to unravel new players in pituitary tumourigenesis, although data obtained with such screening methods need to be validated on large series of pituitary tumours and integrated with functional studies. Molecular signatures of functional PA phenotypes are emerging and may provide significant information in terms of pathogenesis, prognosis and treatment. The identification of reliable markers of aggressiveness remains a priority for a better understanding and management of secreting PA resistant to conventional pharmacological treatment, NFPA and pituitary carcinomas.
Glossary
αGSU= α-glycoprotein subunit ACP: adamantinomatous craniopharyngioma ACTH: adrenocoticotropic hormone AHR: aryl hydrocarbon receptor AIP: aryl hydrocarbon receptor interacting protein AP: anterior pituitary BAG1: Bcl2-associated athanogene 1 Bcl2: B-cell lymphoma 2 BMP: bone morphogenetic protein CDK: cyclin dependent kinase CKI: cyclin dependent kinase inhibitor CNC: Carney complex CREB: cAMP response element-binding protein CRH: corticotrophin-releasing hormone CTNNB1: cadherin-associated protein β1 (β-catenin) D2R: D2 dopamine receptor DA: dopamine agonist DAPK1: death-associated protein kinase 1 EGF: epidermal growth factor ER: estrogen receptor Erk: extracellular-signal-regulated kinase FasL: Fas ligand FCS: folliculostellate cells FGF: fibroblast growth factor FIPA: familial isolated pituitary adenomas FISH: Fluorescence in situ hybridization FSH: follicle-stimulating hormone GADD45: growth arrest and DNA damage gene |
GF: growth factor GH: growth hormone GHRH: growth-hormone-releasing hormone GNAS1: α-subunit of the stimulatory G protein GnRH: gonadotropin-releasing hormone GPCR: G-protein coupled receptor GSK-3β: glycogen synthase kinase 3β HDAC: hystone deacetylase HIFα: hypoxia-inducible factor α HMGA: high mobility group AT-hook protein IGF1: insulin-like growth factor 1 IHC: immunohystochemistry IP3: inositol triphosphate ISEL: immunogold electron microscopy in situ end-labeling LOH: loss of heterozigosity MAGE-A3: melanoma-associated antigen 3 MAPK: mitogen-activated protein kinase MAS: McCune Albright syndrome MEG3: maternally expressed gene 3 MEN1: multiple endocrine neoplasia type 1 MEN4: multiple endocrine neoplasia type 4 MGMT: methylguanine methyltransferase miR: microRNA MMP: matrix metalloproteinase mTor: mammalian target of rapamycin MVD: microvessel density NFPA: non-functioning pituitary adenoma PA: pituitary adenoma |
PAP: pituitary adenoma predisposition PDE: phosphodiesterase PI3K: phosphatidylinositide 3-kinases PIK3CA: phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit α PKA: protein kinase A PKC: protein kinase C PKRAR1A: regulatory subunit type 1A of cAMP-dependent protein kinase POMC: proopiomelanocortin PPAR: peroxisome proliferator-activated receptor PROP-1: Prophet of Pit1 PRL: prolactin PTAG: pituitary tumor derived apoptosis gene PTTG: pituitary tumour trasforming gene RIα: regulatory subunit type 1A RP : Rathke’s pouch Shh: Sonic Hedgehog SSA: somatostatin analogues SSTR: somatostatin receptor TGF: trasforming growth factor TF: transcriptions factor TNF: tumor necrosis factor TPR: tetratricopeptide repeats TRH: TSH-releasing hormone TSG: tumor suppressor gene TSH: thyroid-stimulating hormone TUNEL: terminal deoxynucleotidyl transferase VEGF: vascular endothelial growth factor WHO: world health organization |
References
- 1.
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. The Journal of Clinical Endocrinology & Metabolism 2006;91(12) 4769-75. - 2.
Arafah BM, Nasrallah MP. Pituitary tumors: pathophysiology, clinical manifestations and management. Endocrine Related Cancer 2001;8(4) 287-305. - 3.
Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW, Kleihues P. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathologica 2007;114(2) 97-109. - 4.
Mete O, Asa SL. Clinicopathological correlations in pituitary adenomas. Brain Pathology 2012;22(4) 443-53. - 5.
Obari A, Sano T, Ohyama K, Kudo E, Qian ZR, Yoneda A, Rayhan N, Mustafizur Rahman M, Yamada S. Clinicopathological features of growth hormone-producing pituitary adenomas: difference among various types defined by cytokeratin distribution pattern including a transitional form. Endocrine Pathology 2008;19(2) 82-91. - 6.
Asa SL. Transgenic and knockout mouse models clarify pituitary development, function and disease. Brain Pathology 2001;11(3) 371-84. - 7.
Jaffrain-Rea ML, Di Stefano D, Minniti G, Esposito V, Bultrini A, Ferretti E, Santoro A, Faticanti Scucchi L, Gulino A, Cantore G. A critical reappraisal of MIB-1 labelling index significance in a large series of pituitary tumours: secreting versus non-secreting adenomas. Endocrine Related Cancer 2002;9(2) 103-13. - 8.
De Lellis RA., Lloyd RV., Heitz PU., Eng C. World Health Organization classification of tumours: tumours of endocrine organs. Lyon: IARC press; 2004. - 9.
Raverot G, Wierinckx A, Dantony E, Auger C, Chapas G, Villeneuve L, Brue T, Figarella-Branger D, Roy P, Jouanneau E, Jan M, Lachuer J, Trouillas J; HYPOPRONOS. Prognostic factors in prolactin pituitary tumors: clinical, histological, and molecular data from a series of 94 patients with a long postoperative follow-up. The Journal of Clinical Endocrinology & Metabolism 2010;95(4) 1708-16. - 10.
Galland F, Lacroix L, Saulnier P, Dessen P, Meduri G, Bernier M, Gaillard S, Guibourdenche J, Fournier T, Evain-Brion D, Bidart JM, Chanson P. Differential gene expression profiles of invasive and non-invasive non-functioning pituitary adenomas based on microarray analysis. Endocrine Related Cancer 2010;17(2) 361-71. - 11.
Ruebel KH, Leontovich AA, Jin L, Stilling GA, Zhang H, Qian X, Nakamura N, Scheithauer BW, Kovacs K, Lloyd RV. Patterns of gene expression in pituitary carcinomas and adenomas analyzed by high-density oligonucleotide arrays, reverse transcriptase-quantitative PCR, and protein expression. Endocrine 2006;29(3) 435-44. - 12.
Clayton RN, Farrell WE. Pituitary tumour clonality revisited. Frontiers of Hormone Research 2004;32 186-204. - 13.
Dworakowska D, Grossman AB. The pathophysiology of pituitary adenomas. Best Practice & Research: Clinical Endocrinology & Metabolism 2009;23(5) 525-41. - 14.
Melmed S. Pathogenesis of pituitary tumors. Nature Reviews Endocrinology 2011;7(5) 257-66. - 15.
Brinkmeier ML, Davis SW, Carninci P, MacDonald JW, Kawai J, Ghosh D, Hayashizaki Y, Lyons RH, Camper SA. Discovery of transcriptional regulators and signaling pathways in the developing pituitary gland by bioinformatic and genomic approaches. Genomics 2009;93(5) 449-60. - 16.
Davis SW, Castinetti F, Carvalho LR, Ellsworth BS, Potok MA, Lyons RH, Brinkmeier ML, Raetzman LT, Carninci P, Mortensen AH, Hayashizaki Y, Arnhold IJ, Mendonça BB, Brue T, Camper SA. Molecular mechanisms of pituitary organogenesis: in search of novel regulatory genes. Molecular and Cellular Endocrinology 2010;323(1) 4-19. - 17.
Kelberman D, Rizzoti K, Lovell-Badge R, Robinson IC, Dattani MT. Genetic regulation of pituitary gland development in human and mouse. Endocrine Reviews 2009;30(7) 790-829. - 18.
De Moraes DC, Vaisman M, Conceição FL, Ortiga-Carvalho TM. Pituitary development: a complex, temporal regulated process dependent on specific transcriptional factors. Journal of Endocrinology 2012;215(2) 239-45. - 19.
Perez-Castro C, Renner U, Haedo MR, Stalla GK, Arzt E. Cellular and molecular specificity of pituitary gland physiology. Physiological Reviews 2012;92(1) 1-38. - 20.
Fratticci A, Grieco FA, Spilioti C, Giangaspero F, Ventura L, Esposito V, Piccirilli M, Santoro A, Gulino A, Cantore G, Alesse E, Jaffrain-Rea ML. Differential expression of neurogenins and NeuroD1 in human pituitary tumours. Journal of Endocrinology 2007;194(3) 475-84. - 21.
Mantovani G, Asteria C, Pellegrini C, Bosari S, Alberti L, Bondioni S, Peverelli E, Spada A, Beck-Peccoz P. HESX1 expression in human normal pituitaries and pituitary adenomas. Molecular and Cellular Endocrinology 2006;247(1-2) 135-9. - 22.
Pellegrini-Bouiller I, Manrique C, Gunz G, Grino M, Zamora AJ, Figarella-Branger D, Grisoli F, Jaquet P, Enjalbert A. Expression of the members of the Ptx family of transcription factors in human pituitary adenomas. The Journal of Clinical Endocrinology & Metabolism 1999;84(6) 2212-20. - 23.
Nakamura S, Ohtsuru A, Takamura N, Kitange G, Tokunaga Y, Yasunaga A, Shibata S, Yamashita S. Prop-1 gene expression in human pituitary tumors. The Journal of Clinical Endocrinology & Metabolism 1999;84(7) 2581-4. - 24.
Egashira N, Minematsu T, Miyai S, Takekoshi S, Camper SA, Osamura RY. Pituitary changes in Prop1 transgenic mice: hormone producing tumors and signet-ring type gonadotropes. Acta Histochemica and Cytochemica 2008;41(3) 47-57. - 25.
Egashira N, Takekoshi S, Takei M, Teramoto A, Osamura RY. Expression of FOXL2 in human normal pituitaries and pituitary adenomas. Modern Pathology 2011;24(6) 765-73. - 26.
Gueorguiev M, Grossman AB. Pituitary gland and beta-catenin signaling: from ontogeny to oncogenesis. Pituitary 2009;12(3) 245-55. - 27.
Sekine S, Shibata T, Kokubu A, Morishita Y, Noguchi M, Nakanishi Y, Sakamoto M, Hirohashi S. Craniopharyngiomas of adamantinomatous type harbor beta-catenin gene mutations. American Journal of Pathology 2002;161(6) 1997-2001. - 28.
Buslei R, Nolde M, Hofmann B, Meissner S, Eyupoglu IY, Siebzehnrübl F, Hahnen E, Kreutzer J, Fahlbusch R. Common mutations of beta-catenin in adamantinomatous craniopharyngiomas but not in other tumours originating from the sellar region. Acta Neuropathologica 2005;109(6) 589-97. - 29.
Oikonomou E, Barreto DC, Soares B, De Marco L, Buchfelder M, Adams EF. Beta-catenin mutations in craniopharyngiomas and pituitary adenomas. Journal of Neuro-Oncology 2005;73(3) 205-9. - 30.
Gaston-Massuet C, Andoniadou CL, Signore M, Jayakody SA, Charolidi N, Kyeyune R, Vernay B, Jacques TS, Taketo MM, Le Tissier P, Dattani MT, Martinez-Barbera JP. Increased Wingless (Wnt) signaling in pituitary progenitor/stem cells gives rise to pituitary tumors in mice and humans. Proceedings of the National Academy of Sciences of the United States of America 2011;108(28) 11482-7. - 31.
Elston MS, Clifton-Bligh RJ. Identification of Wnt family inhibitors: a pituitary tumor directed whole genome approach. Molecular and Cellular Endocrinology 2010;326(1-2) 48-54. - 32.
Wang Y, Martin JF, Bai CB. Direct and indirect requirements of Shh/Gli signaling in early pituitary development. Developmental Biology 2010;348(2) 199-209. - 33.
Roessler E, Du YZ, Mullor JL, Casas E, Allen WP, Gillessen-Kaesbach G, Roeder ER, Ming JE, Ruiz i Altaba A, Muenke M. Loss-of-function mutations in the human GLI2 gene are associated with pituitary anomalies and holoprosencephaly-like features. Proceedings of the National Academy of Sciences of the United States of America 2003;100(23) 13424-9. - 34.
Vila G, Papazoglou M, Stalla J, Theodoropoulou M, Stalla GK, Holsboer F, Paez-Pereda M. Sonic hedgehog regulates CRH signal transduction in the adult pituitary. FASEB Journal 2005;19(2) 281-3. - 35.
Vila G, Theodoropoulou M, Stalla J, Tonn JC, Losa M, Renner U, Stalla GK, Paez-Pereda M. Expression and function of sonic hedgehog pathway components in pituitary adenomas: evidence for a direct role in hormone secretion and cell proliferation. The Journal of Clinical Endocrinology & Metabolism 2005;90(12) 6687-94. - 36.
Thurston G, Kitajewski J. VEGF and Delta-Notch: interacting signalling pathways in tumour angiogenesis. British Journal of Cancer 2008;99(8) 1204-9. - 37.
Raetzman LT, Ross SA, Cook S, Dunwoodie SL, Camper SA, Thomas PQ. Developmental regulation of Notch signaling genes in the embryonic pituitary: Prop1 deficiency affects Notch2 expression. Developmental Biology 2004;265(2) 329-40. - 38.
Monahan P, Rybak S, Raetzman LT. The notch target gene HES1 regulates cell cycle inhibitor expression in the developing pituitary. Endocrinology 2009;150(9) 4386-94. - 39.
Miao Z, Miao Y, Lin Y, Lu X. Overexpression of the Notch3 receptor in non-functioning pituitary tumours. Journal of Clinical Neuroscience 2012;19(1) 107-10. - 40.
Evans CO, Moreno CS, Zhan X, McCabe MT, Vertino PM, Desiderio DM, Oyesiku NM. Molecular pathogenesis of human prolactinomas identified by gene expression profiling, RT-qPCR, and proteomic analyses. Pituitary 2008;11(3) 231-45. - 41.
Jiang Z, Gui S, Zhang Y. Analysis of differential gene expression in plurihormonal pituitary adenomas using bead-based fiber-optic arrays. Journal of Neuro-Oncology 2012;108(3) 341-8. - 42.
Rizzoti K. Adult pituitary progenitors/stem cells: from in vitro characterization to in vivo function. European Journal of Neuroscience 2010;32 2053–2062. - 43.
Florio T. Adult pituitary stem cells: from pituitary plasticity to adenoma development. Neuroendocrinology 2011;94(4) 265-77. - 44.
Hosoyama T, Nishijo K, Garcia MM, Schaffer BS, Ohshima-Hosoyama S, Prajapati SI, Davis MD, Grant WF, Scheithauer BW, Marks DL, Rubin BP, Keller C. A postnatal Pax7 progenitor gives rise to pituitary adenomas. Genes Cancer 2010;1(4) 388-402. - 45.
Xu Q, Yuan X, Tunici P, Liu G, Fan X, Xu M, Hu J, Hwang JY, Farkas DL, Black KL, Yu JS. Isolation of tumour stem-like cells from benign tumours. British Journal of Cancer 2009; 101(2) 303-11. - 46.
Garcia-Lavandeira M, Saez C, Diaz-Rodriguez E, Perez-Romero S, Senra A, Dieguez C, Japon MA, Alvarez CV. Craniopharyngiomas express embryonic stem cell markers (SOX2, OCT4, KLF4, and SOX9) as pituitary stem cells but do not coexpress RET/GFRA3 receptors. The Journal of Clinical Endocrinology & Metabolism 2012;97(1) E80-7. - 47.
46.Vandeva S, Jaffrain-Rea ML, Daly AF, Tichomirowa M, Zacharieva S, Beckers A. The genetics of pituitary adenomas. Best Practice & Research: Clinical Endocrinology & Metabolism 2010;24(3) 461-76. - 48.
Jaffrain-Rea ML, Daly AF, Angelini M, Petriossians P, Bours V, Beckers A. Genetic susceptibility in pituitary adenomas: from pathogenesis to clinical implications. Expert Review of Endocrinology & Metabolism 2011;(2) 195-214. - 49.
Agarwal SK, Ozawa A, Mateo CM, Marx SJ. The MEN1 gene and pituitary tumours. Hormone Research 2009;71 Suppl 2 131-8. - 50.
Trouillas J, Labat-Moleur F, Sturm N, Kujas M, Heymann MF, Figarella-Branger D, Patey M, Mazucca M, Decullier E, Vergès B, Chabre O, Calender A, Groupe d'études des Tumeurs Endocrines. Pituitary tumors and hyperplasia in multiple endocrine neoplasia type 1 syndrome (MEN1): a case-control study in a series of 77 patients versus 2509 non-MEN1 patients. The American Journal of Surgical Pathology 2008;32(4) 534-43. - 51.
Thakker RV. Multiple endocrine neoplasia type 1 (MEN1). Best Practice & Research: Clinical Endocrinology & Metabolism 2010;24(3) 355-70. - 52.
Lips CJ, Dreijerink KM, Höppener JW. Variable clinical expression in patients with a germline MEN1 disease gene mutation: clues to a genotype-phenotype correlation. Clinics 2012;67 Suppl 1 49-56. - 53.
Ellard S, Hattersley AT, Brewer CM, Vaidya B. Detection of a MEN1 gene mutation depends on clinical features and supports current referral criteria for diagnostic molecular genetic testing. Clinical Endocrinology 2005;62(2) 169-75. - 54.
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 Research 2012;72(19) 5060-8. - 55.
Dreijerink KM, Lips CJ, Timmers HT. Multiple endocrine neoplasia type 1: a chromatin writer's block. Journal of Internal Medicine 2009;266(1) 53-9. - 56.
Theodoropoulou M, Cavallari I, Barzon L, D'Agostino DM, Ferro T, Arzberger T, Grübler Y, Schaaf L, Losa M, Fallo F, Ciminale V, Stalla GK, Pagotto U. Differential expression of menin in sporadic pituitary adenomas. Endocrine Related Cancer 2004;11(2) 333-44. - 57.
Rothenbuhler A, Stratakis CA. Clinical and molecular genetics of Carney complex. Best Practice & Research: Clinical Endocrinology & Metabolism 2010;24(3) 389-99. - 58.
Bertherat J, Horvath A, Groussin L, Grabar S, Boikos S, Cazabat L, Libe R, René-Corail F, Stergiopoulos S, Bourdeau I, Bei T, Clauser E, Calender A, Kirschner LS, Bertagna X, Carney JA, Stratakis CA. Mutations in regulatory subunit type 1A of cyclic adenosine 5'-monophosphate-dependent protein kinase (PRKAR1A): phenotype analysis in 353 patients and 80 different genotypes. The Journal of Clinical Endocrinology & Metabolism 2009;94(6) 2085-91. - 59.
Yin Z, Williams-Simons L, Parlow AF, Asa S, Kirschner LS. Pituitary-specific knockout of the Carney complex gene Prkar1a leads to pituitary tumorigenesis. Molecular Endocrinology 2008;22(2) 380-7. - 60.
Veugelers M, Bressan M, McDermott DA, Weremowicz S, Morton CC, Mabry CC, Lefaivre JF, Zunamon A, Destree A, Chaudron JM, Basson CT. Mutation of perinatal myosin heavy chain associated with a Carney complex variant. The New England Journal of Medicine 2004;351(5) 460-9. - 61.
Libé R, Horvath A, Vezzosi D, Fratticci A, Coste J, Perlemoine K, Ragazzon B, Guillaud-Bataille M, Groussin L, Clauser E, Raffin-Sanson ML, Siegel J, Moran J, Drori-Herishanu L, Faucz FR, Lodish M, Nesterova M, Bertagna X, Bertherat J, Stratakis CA. Frequent phosphodiesterase 11A gene (PDE11A) defects in patients with Carney complex (CNC) caused by PRKAR1A mutations: PDE11A may contribute to adrenal and testicular tumors in CNC as a modifier of the phenotype. The Journal of Clinical Endocrinology & Metabolism 2011;96(1) E208-14. - 62.
Dumitrescu CE, Collins MT. McCune-Albright syndrome. Orphanet Journal of Rare Diseases 2008;3 12. - 63.
Lumbroso S, Paris F, Sultan C; Activating Gsalpha mutations: analysis of 113 patients with signs of McCune-Albright syndrome. A European collaborative study. The Journal of Clinical Endocrinology & Metabolism 2004;89(5) 2107-13. - 64.
Pellegata NS, Quintanilla-Martinez L, Siggelkow H, Samson E, Bink K, Höfler H, Fend F, Graw J, Atkinson MJ. Germ-line mutations in p27Kip1 cause a multiple endocrine neoplasia syndrome in rats and humans. Proceedings of the National Academy of Sciences of the United States of America 2006;103(50) 19213. - 65.
Pellegata NS. MENX and MEN4. Clinics (Sao Paulo) 2012;67 Suppl 1 13-8. - 66.
Agarwal SK, Mateo CM, Marx SJ. Rare germline mutations in cyclin-dependent kinase inhibitor genes in multiple endocrine neoplasia type 1 and related states. The Journal of Clinical Endocrinology & Metabolism 2009;94(5) 1826-34. - 67.
Ozawa A, Agarwal SK, Mateo CM, Burns AL, Rice TS, Kennedy PA, Quigley CM, Simonds WF, Weinstein LS, Chandrasekharappa SC, Collins FS, Spiegel AM, Marx SJ. The parathyroid/pituitary variant of multiple endocrine neoplasia type 1 usually has causes other than p27Kip1 mutations. The Journal of Clinical Endocrinology & Metabolism 2007;92(5) 1948-51. - 68.
Xekouki P, Pacak K, Almeida M, Wassif CA, Rustin P, Nesterova M, de la Luz Sierra M, Matro J, Ball E, Azevedo M, Horvath A, Lyssikatos C, Quezado M, Patronas N, Ferrando B, Pasini B, Lytras A, Tolis G, Stratakis CA. Succinate dehydrogenase (SDH) D subunit (SDHD) inactivation in a growth-hormone-producing pituitary tumor: a new association for SDH? The Journal of Clinical Endocrinology & Metabolism 2012;97(3) E357-66. - 69.
Daly AF, Jaffrain-Rea ML, Ciccarelli A, Valdes-Socin H, Rohmer V, Tamburrano G, Borson-Chazot C, Estour B, Ciccarelli E, Brue T, Ferolla P, Emy P, Colao A, De Menis E, Lecomte P, Penfornis F, Delemer B, Bertherat J, Wémeau JL, De Herder W, Archambeaud F, Stevenaert A, Calender A, Murat A, Cavagnini F, Beckers A. Clinical characterization of familial isolated pituitary adenomas. The Journal of Clinical Endocrinology & Metabolism 2006;91(9) 3316-23. - 70.
Vierimaa O, Georgitsi M, Lehtonen R, Vahteristo P, Kokko A, Raitila A, Tuppurainen K, Ebeling TM, Salmela PI, Paschke R, Gündogdu S, De Menis E, Mäkinen MJ, Launonen V, Karhu A, Aaltonen LA. Pituitary adenoma predisposition caused by germline mutations in the AIP gene. Science 2006;312(5777) 1228-30. - 71.
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, Sabaté MI, Bertagna X, Garcia Basavilbaso N, Stalldecker G, Colao A, Ferolla P, Wémeau JL, Caron P, Sadoul JL, Oneto A, Archambeaud F, Calender A, Sinilnikova O, Montañana 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. The Journal of Clinical Endocrinology & Metabolism 2007;92(5) 1891-6. - 72.
Trivellin G, Korbonits M. AIP and its interacting partners. Journal of Endocrinology 2011;210(2) 137-55. - 73.
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, Schürmeyer T, Cozzi R, Theodoropoulou M, Sievers C, Bernabeu I, Naves LA, Chabre O, Montañana CF, Hana V, Halaby G, Delemer B, Aizpún JI, Sonnet E, Longás 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. European Journal of Endocrinology 2011;165(4) 509-15. - 74.
Cazabat L, Bouligand J, Chanson P. AIP mutation in pituitary adenomas. New England Journal of Medicine 2011;364(20) 1974-5. - 75.
Daly AF, Tichomirowa MA, Petrossians P, Heliövaara E, Jaffrain-Rea ML, BarlierA, Naves LA, Ebeling T, Karhu A, Raappana A, Cazabat L, De Menis E, Montañana CF, Raverot G, Weil RJ, Sane T, Maiter D, Neggers S, Yaneva M, Tabarin A, Verrua E, Eloranta E, Murat A, Vierimaa O, Salmela PI, Emy P, Toledo RA, Sabaté MI, Villa C, Popelier M, Salvatori R, Jennings J, Longás AF, Labarta Aizpún JI, Georgitsi M, Paschke R, Ronchi C, Valimaki M, Saloranta C, De Herder W, Cozzi R, Guitelman M, Magri F, Lagonigro MS, Halaby G, Corman V, Hagelstein MT, Vanbellinghen JF, Barra GB, Gimenez-Roqueplo AP, Cameron FJ, Borson-Chazot F, Holdaway I, Toledo SP, Stalla GK, Spada A, Zacharieva S, Bertherat J, Brue T, Bours V, Chanson P, Aaltonen LA, Beckers A. Clinical characteristics and therapeutic responses in patients with germ-line AIP mutations and pituitary adenomas: an international collaborative study. The Journal of Clinical Endocrinology & Metabolism 2010;95(11) E373-83. - 76.
Raitila A, Lehtonen HJ, Arola J, Heliövaara 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. American Journal of Pathology 2010;177(4) 1969-76. - 77.
Khoo SK, Pendek R, Nickolov R, Luccio-Camelo DC, Newton TL, Massie A, Petillo D, Menon J, Cameron D, Teh BT, Chan SP. Genome-wide scan identifies novel modifier loci of acromegalic phenotypes for isolated familial somatotropinoma. Endocrine Related Cancer 2009;16(3) 1057-63. - 78.
Villa C, Lagonigro MS, Magri F, Koziak M, Jaffrain-Rea ML, Brauner R, Bouligand J, Junier MP, Di Rocco F, Sainte-Rose C, Beckers A, Roux FX, Daly AF, Chiovato L. Hyperplasia-adenoma sequence in pituitary tumorigenesis related to aryl hydrocarbon receptor interacting protein gene mutation. Endocrine Related Cancer 2011;18(3) 347-56. - 79.
Kasuki Jomori de Pinho L, Vieira Neto L, Armondi Wildemberg LE, Gasparetto EL, Marcondes J, de Almeida Nunes B, Takiya CM, Gadelha MR. Low aryl hydrocarbon receptor-interacting protein expression is a better marker of invasiveness in somatotropinomas than Ki-67 and p53. Neuroendocrinology 2011;94(1) 39-48. - 80.
Chahal HS, Trivellin G, Leontiou CA, Alband N, Fowkes RC, Tahir A, Igreja SC, Chapple JP, Jordan S, Lupp A, Schulz S, Ansorge O, Karavitaki N, Carlsen E, Wass JA, Grossman AB, Korbonits M. Somatostatin analogs modulate AIP in somatotroph adenomas: the role of the ZAC1 pathway. The Journal of Clinical Endocrinology & Metabolism 2012;97(8) E1411-20. - 81.
Kasuki L, Vieira Neto L, Wildemberg LE, Colli LM, de Castro M, Takiya CM, Gadelha MR. AIP expression in sporadic somatotropinomas is a predictor of the response to octreotide LAR therapy independent of SSTR2 expression. Endocrine Related Cancer 2012;19(3) L25-9. - 82.
Finelli P, Giardino D, Rizzi N, Buiatiotis S, Virduci T, Franzin A, Losa M, Larizza L. Non-random trisomies of chromosomes 5, 8 and 12 in the prolactinoma sub-type of pituitary adenomas: conventional cytogenetics and interphase FISH study. International Journal of Cancer 2000;86(3) 344-50. - 83.
Larsen JB, Schrøder HD, Sørensen AG, Bjerre P, Heim S. Simple numerical chromosome aberrations characterize pituitary adenomas. Cancer Genetics and Cytogenetics 1999;114(2) 144-9. - 84.
Kontogeorgos G, Kapranos N, Tzavara I, Thalassinos N, Rologis D. Monosomy of chromosome 11 in pituitary adenoma in a patient with familial multiple endocrine neoplasia type 1. Clinical Endocrinology 2001;54(1) 117-20. - 85.
Bates AS, Farrell WE, Bicknell EJ, McNicol AM, Talbot AJ, Broome JC, Perrett CW, Thakker RV, Clayton RN. Allelic deletion in pituitary adenomas reflects aggressive biological activity and has potential value as a prognostic marker. The Journal of Clinical Endocrinology & Metabolism 1997;82(3) 818-24. - 86.
Lania A, Spada A. G-protein and signalling in pituitary tumours. Hormone Research 2009;71 Suppl 2 95-100. - 87.
De Martino I, Fedele M, Palmieri D, Visone R, Cappabianca P, Wierinckx A, Trouillas J, Fusco A. B-RAF mutations are a rare event in pituitary adenomas. Journal of Endocrinological Investigation 2007;30(1) RC1-3. - 88.
Lin Y, Jiang X, Shen Y, Li M, Ma H, Xing M, Lu Y. Frequent mutations and amplifications of the PIK3CA gene in pituitary tumors. Endocrine Related Cancer 2009;16(1) 301-10. - 89.
Tanizaki Y, Jin L, Scheithauer BW, Kovacs K, Roncaroli F, Lloyd RV. P53 gene mutations in pituitary carcinomas. Endocrine Pathology 2007;18(4) 217-22. - 90.
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 Research 2000;60(5) 1211-6. - 91.
Wierinckx A, Raverot G, Nazaret N, Jouanneau E, Auger C, Lachuer J, Trouillas J. Proliferation markers of human pituitary tumors: contribution of a genome-wide transcriptome approach. Molecular and Cellular Endocrinology 2010; 326(1-2) 30-9. - 92.
Tong Y, Zheng Y, Zhou J, Oyesiku NM, Koeffler HP, Melmed S. Genomic characterization of human and rat prolactinomas. Endocrinology 2012;153(8) 3679-91. - 93.
Xu M, Knox AJ, Michaelis KA, Kiseljak-Vassiliades K, Kleinschmidt-DeMasters BK, Lillehei KO, Wierman ME. Reprimo (RPRM) is a novel tumor suppressor in pituitary tumors and regulates survival, proliferation, and tumorigenicity. Endocrinology 2012;153(7) 2963-73. - 94.
Jenuwein T, Allis CD. Translating the histone code. Science 2001;293(5532) 1074-80. - 95.
Dudley KJ, Revill K, Clayton RN, Farrell WE. Pituitary tumours: all silent on the epigenetics front. Journal of Molecular Endocrinology 2009;42(6) 461-8. - 96.
Ezzat S. Epigenetic control in pituitary tumors. Endocrine Journal 2008;55(6) 951-7. - 97.
Duong CV, Emes RD, Wessely F, Yacqub-Usman K, Clayton RN, Farrell WE. Quantitative, genome-wide analysis of the DNA methylome in sporadic pituitary adenomas. Endocrine-Related Cancer 2012;19(6) 805-16. - 98.
Yacqub-Usman K, Richardson A, Duong CV, Clayton RN, Farrell WE. The pituitary tumour epigenome: aberrations and prospects for targeted therapy. Nature Reviews Endocrinology 2012;8(8) 486-94. - 99.
Wu X, Hua X. Menin, histone H3 methyltransferases, and regulation of cell proliferation: current knowledge and perspective. Current Molecular Medicine 2008; 8(8) 805-15. - 100.
Zhang Z, Florez S, Gutierrez-Hartmann A, Martin JF, Amendt BA. MicroRNAs regulate pituitary development, and microRNA 26b specifically targets lymphoid enhancer factor 1 (Lef-1), which modulates pituitary transcription factor 1 (Pit-1) expression. The Journal of Biological Chemistry 2010;285(45) 34718-28. - 101.
Yuen T, Ruf F, Chu T, Sealfon SC. Microtranscriptome regulation by gonadotropin-releasing hormone. Molecular and Cellular Endocrinology 2009;302(1) 12-7. - 102.
Bottoni A, Piccin D, Tagliati F, Luchin A, Zatelli MC, degli Uberti EC. miR-15a and miR-16-1 down-regulation in pituitary adenomas. Journal of Cellular Physiology 2005;204(1) 280-5. - 103.
Bottoni A, Zatelli MC, Ferracin M, Tagliati F, Piccin D, Vignali C, Calin GA, Negrini M, Croce CM, Degli Uberti EC. Identification of differentially expressed microRNAs by microarray: a possible role for microRNA genes in pituitary adenomas. Journal of Cellular Physiology 2007;210(2) 370-7. - 104.
Sivapragasam M, Rotondo F, Lloyd RV, Scheithauer BW, Cusimano M, Syro LV, Kovacs K. MicroRNAs in the human pituitary. Endocrine Pathology 2011;22(3) 134-43. - 105.
Trivellin G, Butz H, Delhove J, Igreja S, Chahal HS, Zivkovic V, McKay T, Patócs A, Grossman AB, Korbonits M. MicroRNA miR-107 is overexpressed in pituitary adenomas and inhibits the expression of aryl hydrocarbon receptor-interacting protein in vitro. American Journal of Physiology – Endocrinology and Metabolism 2012;303(6) E708-19. - 106.
Mao ZG, He DS, Zhou J, Yao B, Xiao WW, Chen CH, Zhu YH, Wang HJ. Differential expression of microRNAs in GH-secreting pituitary adenomas. Diagnostic Pathology 2010;5 79. - 107.
D'Angelo D, Palmieri D, Mussnich P, Roche M, Wierinckx A, Raverot G, Fedele M, Croce CM, Trouillas J, Fusco A. Altered microRNA expression profile in human pituitary GH adenomas: down-regulation of miRNA targeting HMGA1, HMGA2, and E2F1. The Journal of Clinical Endocrinology & Metabolism 2012;97(7) E1128-38. - 108.
Wang C, Su Z, Sanai N, Xue X, Lu L, Chen Y, Wu J, Zheng W, Zhuge Q, Wu ZB. microRNA expression profile and differentially-expressed genes in prolactinomas following bromocriptine treatment. Oncology Reports 2012;27(5) 1312-20. - 109.
Butz H, Likó I, Czirják S, Igaz P, Khan MM, Zivkovic V, Bálint K, Korbonits M, Rácz K, Patócs A. Down-regulation of Wee1 kinase by a specific subset of microRNA in human sporadic pituitary adenomas. The Journal of Clinical Endocrinology & Metabolism 2010;95(10) E181-91. - 110.
Stilling G, Sun Z, Zhang S, Jin L, Righi A, Kovācs G, Korbonits M, Scheithauer BW, Kovacs K, Lloyd RV. MicroRNA expression in ACTH-producing pituitary tumors: up-regulation of microRNA-122 and -493 in pituitary carcinomas. Endocrine 2010;38(1) 67-75. - 111.
Palumbo T, Faucz FR, Azevedo M, Xekouki P, Iliopoulos D, Stratakis CA. Functional screen analysis reveals miR-26b and miR-128 as central regulators of pituitary somatomammotrophic tumor growth through activation of the PTEN-AKT pathway. Oncogene 2012 [Epub ahead of print]. - 112.
Palmieri D, D'Angelo D, Valentino T, De Martino I, Ferraro A, Wierinckx A, Fedele M, Trouillas J, Fusco A. Downregulation of HMGA-targeting microRNAs has a critical role in human pituitary tumorigenesis. Oncogene 2012;31(34) 3857-65. - 113.
Zhou Y, Zhang X, Klibanski A. MEG3 noncoding RNA: a tumor suppressor. Journal of Molecular Endocrinology 2012; 48(3) R45-53. - 114.
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. American Journal of Pathology 2011;179(4) 2120-30. - 115.
Pertuit M, Barlier A, Enjalbert A, Gérard C. Signalling pathway alterations in pituitary adenomas: involvement of Gsalpha, cAMP and mitogen-activated protein kinases. Journal of Neuroendocrinology 2009;21(11) 869-77. - 116.
Lania AG, Mantovani G, Ferrero S, Pellegrini C, Bondioni S, Peverelli E, Braidotti P, Locatelli M, Zavanone ML, Ferrante E, Bosari S, Beck-Peccoz P, Spada A. Proliferation of transformed somatotroph cells related to low or absent expression of protein kinase a regulatory subunit 1A protein. Cancer Research 2004;64(24) 9193-8. - 117.
Peverelli E, Ermetici F, Filopanti M, Elli FM, Ronchi CL, Mantovani G, Ferrero S, Bosari S, Beck-Peccoz P, Lania A, Spada A. Analysis of genetic variants of phosphodiesterase 11A in acromegalic patients. European Journal of Endocrinology 2009;161(5) 687-94. - 118.
Asa SL, Scheithauer BW, Bilbao JM, Horvath E, Ryan N, Kovacs K, Randall RV, Laws ER Jr, Singer W, Linfoot JA, Thorner MO, Vale W. A case for hypothalamic acromegaly: a clinicopathological study of six patients with hypothalamic gangliocytomas producing growth hormone-releasing factor. The Journal of Clinical Endocrinology & Metabolism 1984;58 796-803. - 119.
Garby L, Caron P, Claustrat F, Chanson P, Tabarin A, Rohmer V, Arnault G, Bonnet F, Chabre O, Christin-Maitre S, du-Boullay H, Murat A, Nakib I, Sadoul JL, Sassolas G, Claustrat B, Raverot G, Borson-Chazot F; GTE Group. Clinical characteristics and outcome of acromegaly induced by ectopic secretion of growth hormone-releasing hormone (GHRH): a French nationwide series of 21 cases. The Journal of Clinical Endocrinology & Metabolism 2012;97(6) 2093-104. - 120.
Senovilla L, Núñez L, de Campos JM, de Luis DA, Romero E, Sánchez A, García-Sancho J, Villalobos C. Multifunctional cells in human pituitary adenomas: implications for paradoxical secretion and tumorigenesis. The Journal of Clinical Endocrinology & Metabolism 2004;89(9) 4545-52. - 121.
Molitch ME. Pharmacologic resistance in prolactinoma patients. Pituitary 2005;8(1) 43-52. - 122.
Peverelli E, Mantovani G, Vitali E, Elli FM, Olgiati L, Ferrero S, Laws ER, Della Mina P, Villa A, Beck-Peccoz P, Spada A, Lania AG. Filamin-A is essential for dopamine D2 receptor expression and signaling in tumorous lactotrophs. The Journal of Clinical Endocrinology & Metabolism 2012;97(3) 967-77. - 123.
Lania A, Mantovani G, Spada A. Genetic abnormalities of somatostatin receptors in pituitary tumors. Molecular and Cellular Endocrinology 2008;286(1-2) 180-6. - 124.
Durán-Prado M, Saveanu A, Luque RM, Gahete MD, Gracia-Navarro F, Jaquet P, Dufour H, Malagón MM, Culler MD, Barlier A, Castaño JP. A potential inhibitory role for the new truncated variant of somatostatin receptor 5, sst5TMD4, in pituitary adenomas poorly responsive to somatostatin analogs. The Journal of Clinical Endocrinology & Metabolism 2010;95(5) 2497-502. - 125.
Denef C. Paracrinicity: the story of 30 years of cellular pituitary crosstalk. Journal of Neuroendocrinology 2008;20(1) 1-70. - 126.
Perumal P, Vrontakis ME. Transgenic mice over-expressing galanin exhibit pituitary adenomas and increased secretion of galanin, prolactin and growth hormone. Journal of Endocrinology 2003;179(2) 145-54. - 127.
Shimon I, Hinton DR, Weiss MH, Melmed S. Prolactinomas express human heparin-binding secretory transforming gene (hst) protein product: marker of tumour invasiveness. Clinical Endocrinology 1998;48(1) 23-9. - 128.
Ezzat S, Zheng L, Zhu XF, Wu GE, Asa SL. Targeted expression of a human pituitary tumor-derived isoform of FGF receptor-4 recapitulates pituitary tumorigenesis. The Journal of Clinical Investigation 2002;109(1) 69-78. - 129.
Mukdsi JH, De Paul AL, Petiti JP, Gutiérrez S, Aoki A, Torres AI. Pattern of FGF-2 isoform expression correlated with its biological action in experimental prolactinomas. Acta Neuropathologica 2006;112(4) 491-501. - 130.
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. Amercian Journal of Pathology 2007;170(5) 1618-28. - 131.
Haedo MR, Gerez J, Fuertes M, Giacomini D, Páez-Pereda M, Labeur M, Renner U, Stalla GK, Arzt E. Regulation of pituitary function by cytokines. Hormone Research 2009;72(5) 266-74. - 132.
Lebrun JJ. Activin, TGF-beta and menin in pituitary tumorigenesis. Advances in Experimental Medicine and Biology 2009;668 69-78. - 133.
Danila DC, Zhang X, Zhou Y, Haidar JN, Klibanski A. Overexpression of wild-type activin receptor alk4-1 restores activin antiproliferative effects in human pituitary tumor cells. The Journal of Clinical Endocrinology & Metabolism 2002;87(10) 4741-6. - 134.
Giacomini D, Acuña M, Gerez J, Nagashima AC, Silberstein S, Páez-Pereda M, Labeur M, Theodoropoulou M, Renner U, Stalla GK, Arzt E. Pituitary action of cytokines: focus on BMP-4 and gp130 family. Neuroendocrinology 2007;85(2) 94-100. - 135.
Barbieri F, Bajetto A, Stumm R, Pattarozzi A, Porcile C, Zona G, Dorcaratto A, Ravetti JL, Minuto F, Spaziante R, Schettini G, Ferone D, Florio T. Overexpression of stromal cell-derived factor 1 and its receptor CXCR4 induces autocrine/paracrine cell proliferation in human pituitary adenomas. Clinical Cancer Research 2008;14(16) 5022-32. - 136.
Cooper O, Vlotides G, Fukuoka H, Greene MI, Melmed S. Expression and function of ErbB receptors and ligands in the pituitary. Endocrine Related Cancer 2011;18(6) R197-211. - 137.
Theodoropoulou M, Arzberger T, Gruebler Y, Jaffrain-Rea ML, Schlegel J, Schaaf L, Petrangeli E, Losa M, Stalla GK, Pagotto U. Expression of epidermal growth factor receptor in neoplastic pituitary cells: evidence for a role in corticotropinoma cells. Journal of Endocrinology 2004;183(2) 385-94. - 138.
Fukuoka H, Cooper O, Ben-Shlomo A, Mamelak A, Ren SG, Bruyette D, Melmed S. EGFR as a therapeutic target for human, canine, and mouse ACTH-secreting pituitary adenomas. The Journal of Clinical Endocrinology 2011;121(12) 4712-21. - 139.
Vlotides G, Siegel E, Donangelo I, Gutman S, Ren SG, Melmed S. Rat prolactinoma cell growth regulation by epidermal growth factor receptor ligands. Cancer Research 2008;68(15) 6377-86. - 140.
Assimakopoulou M, Zolota V, Chondrogianni C, Gatzounis G, Varakis J. p75 and TrkC neurotrophin receptors demonstrate a different immunoreactivity profile in comparison to TrkA and TrkB receptors in human normal pituitary gland and adenomas. Neuroendocrinology 2008;88(2) 127-34. - 141.
Missale C. Nerve growth factor, D2 receptor isoforms, and pituitary tumors. Endocrine 2012;42(3) 466-7. - 142.
Lloyd RV, Vidal S, Horvath E, Kovacs K, Scheithauer B. Angiogenesis in normal and neoplastic pituitary tissues. Microscopy Research and Technique 2003;60(2) 244-50. - 143.
Sánchez-Ortiga R, Sánchez-Tejada L, Moreno-Perez O, Riesgo P, Niveiro M, Picó Alfonso AM. Over-expression of vascular endothelial growth factor in pituitary adenomas is associated with extrasellar growth and recurrence. Pituitary 2012 [Epub ahead of print]. - 144.
Molitch ME. Prolactinoma in pregnancy. Best Practice & Research Clinical Endocrinology & Metabolism 2011;25(6) 885-96. - 145.
Jaffrain-Rea ML, Petrangeli E, Ortolani F, Fraioli B, Lise A, Esposito V, Spagnoli LG, Tamburrano G, Frati L, Gulino A. Cellular receptors for sex steroids in human pituitary adenomas. Journal of Endocrinology 1996; 151(2) 175-84. - 146.
Caronti B, Palladini G, Bevilacqua MG, Petrangeli E, Fraioli B, Cantore G, Tamburrano G, Carapella CM, Jaffrain-Rea ML. Effects of 17 beta-estradiol, progesterone and tamoxifen on in vitro proliferation of human pituitary adenomas: correlation with specific cellular receptors. Tumour Biology 1993;14(1) 59-68. - 147.
Zhou W, Song Y, Xu H, Zhou K, Zhang W, Chen J, Qin M, Yi H, Gustafsson JA, Yang H, Fan X. In nonfunctional pituitary adenomas, estrogen receptors and slug contribute to development of invasiveness. The Journal of Clinical Endocrinology & Metabolism 2011;96(8) E1237-45. - 148.
Fan X, Gabbi C, Kim HJ, Cheng G, Andersson LC, Warner M, Gustafsson JA. Gonadotropin-positive pituitary tumors accompanied by ovarian tumors in aging female ERbeta-/- mice. Proceedings of the National Academy of Sciences of the United States of America 2010;107(14) 6453-8. - 149.
Heaney AP, Fernando M, Melmed S. Functional role of estrogen in pituitary tumor pathogenesis. The Journal of Clinical Investigation 2002;109(2) 277-83. - 150.
Sarkar DK. Genesis of prolactinomas: studies using estrogen-treated animals. Frontiers of Hormone Research 2006;35 32-49. - 151.
Zaldivar V, Magri ML, Zárate S, Jaita G, Eijo G, Radl D, Ferraris J, Pisera D, Seilicovich A. Estradiol increases the expression of TNF-α and TNF receptor 1 in lactotropes. Neuroendocrinology 2011;93(2) 106-13. - 152.
Zárate S, Jaita G, Ferraris J, Eijo G, Magri ML, Pisera D, Seilicovich A. Estrogens induce expression of membrane-associated estrogen receptor α isoforms in lactotropes. PLoS One 2012;7(7) E41299. - 153.
Dworakowska D, Grossman AB. The molecular pathogenesis of corticotroph tumours. European Journal of Clinical Investigation 2012;42(6) 665-76. - 154.
Chauvet N, El-Yandouzi T, Mathieu MN, Schlernitzauer A, Galibert E, Lafont C, Le Tissier P, Robinson IC, Mollard P, Coutry N. Characterization of adherens junction protein expression and localization in pituitary cell networks. Journal of Endocrinology 2009;202(3) 375-87. - 155.
Qian ZR, Li CC, Yamasaki H, Mizusawa N, Yoshimoto K, Yamada S, Tashiro T, Horiguchi H, Wakatsuki S, Hirokawa M, Sano T. Role of E-cadherin, alpha-, beta-, and gamma-catenins, and p120 (cell adhesion molecules) in prolactinoma behavior. Modern Pathology 2002;15(12) 1357-65. - 156.
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. The Journal of Clinical Endocrinology & Metabolism 2009;94(4) 1436-42. - 157.
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. The Journal of Clinical Endocrinology & Metabolism 2010;95(5) 2334-42. - 158.
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. Clinical Endocrinology 2011;75(6) 811-8. - 159.
Trouillas J, Daniel L, Guigard MP, Tong S, Gouvernet J, Jouanneau E, Jan M, Perrin G, Fischer G, Tabarin A, Rougon G, Figarella-Branger D. Polysialylated neural cell adhesion molecules expressed in human pituitary tumors and related to extrasellar invasion. Journal of Neurosurgery 2003; 98(5) 1084-93. - 160.
Quereda V, Malumbres M. Cell cycle control of pituitary development and disease. Journal of Molecular Endocrinology 2009;42(2) 75-86. - 161.
Hibberts NA, Simpson DJ, Bicknell JE, Broome JC, Hoban PR, Clayton RN, Farrell WE. Analysis of cyclin D1 (CCND1) allelic imbalance and overexpression in sporadic human pituitary tumors. Clinical Cancer Research 1999;5(8) 2133-9. - 162.
Roussel-Gervais A, Bilodeau S, Vallette S, Berthelet F, Lacroix A, Figarella-Branger D, Brue T, Drouin J. Cooperation between cyclin E and p27(Kip1) in pituitary tumorigenesis. Molecular Endocrinology 2010;24(9) 1835-45. - 163.
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(8) 1149-54. - 164.
Kirsch M, Mörz M, Pinzer T, Schackert HK, Schackert G. Frequent loss of the CDKN2C (p18INK4c) gene product in pituitary adenomas. Genes Chromosomes Cancer. 2009;48(2) 143-54. - 165.
Foijer F, te Riele H. Check, double check: the G2 barrier to cancer. Cell Cycle 2006;5(8) 831-6. - 166.
Wierinckx A, Auger C, Devauchelle P, Reynaud A, Chevallier P, Jan M, Perrin G, Fèvre-Montange M, Rey C, Figarella-Branger D, Raverot G, Belin MF, Lachuer J, Trouillas J. A diagnostic marker set for invasion, proliferation, and aggressiveness of prolactin pituitary tumors. Endocrine Related Cancer 2007;14(3) 887-900. - 167.
De Martino I, Visone R, Wierinckx A, Palmieri D, Ferraro A, Cappabianca P, Chiappetta G, Forzati F, Lombardi G, Colao A, Trouillas J, Fedele M, Fusco A. HMGA proteins up-regulate CCNB2 gene in mouse and human pituitary adenomas. Cancer Research 2009;69(5) 1844-50. - 168.
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. The Journal of Clinical Endocrinology & Metabolism 2002;87(3) 1262-7. - 169.
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(10) 3603-13. - 170.
St Croix B, Sheehan C, Rak JW, Flørenes VA, Slingerland JM, Kerbel RS. E-Cadherin-dependent growth suppression is mediated by the cyclin-dependent kinase inhibitor p27(KIP1). Journal of Cellular Biology 1998;142(2) 557-71. - 171.
Hubina E, Nanzer AM, Hanson MR, Ciccarelli E, Losa M, Gaia D, Papotti M, Terreni MR, Khalaf S, Jordan S, Czirják S, Hanzély Z, Nagy GM, Góth MI, Grossman AB, Korbonits M. Somatostatin analogues stimulate p27 expression and inhibit the MAP kinase pathway in pituitary tumours. European Journal of Endocrinology 2006;155(2) 371-9. - 172.
Abbass T, Dutta A. p21 in cancer: intricate networks and multiple activities. Nature Review in Cancer 2009; 9(6) 400-14. - 173.
Chesnokova V, Zonis S, Kovacs K, Ben-Shlomo A, Wawrowsky K, Bannykh S, Melmed S. p21(Cip1) restrains pituitary tumor growth. Proceedings of the National Academy of Sciences of the United States of America 2008;105(45) 17498-503. - 174.
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 Neurochirurgica 2002;144(5) 481-8. - 175.
Neto AG, McCutcheon IE, Vang R, Spencer ML, Zhang W, Fuller GN. Elevated expression of p21 (WAF1/Cip1) in hormonally active pituitary adenomas. Annals of Diagnostic Pathology 2005;9(1) 6-10. - 176.
Alexandraki KI, Munayem Khan M, Chahal HS, Dalantaeva NS, Trivellin G, Berney DM, Caron P, Popovic V, Pfeifer M, Jordan S, Korbonits M, Grossman AB. Oncogene-induced senescence in pituitary adenomas and carcinomas. Hormones 2012;11(3) 297-307. - 177.
Vlotides G, Eigler T, Melmed S. Pituitary tumor-transforming gene: physiology and implications for tumorigenesis. Endocrine Reviews 2007;28(2) 165-86. - 178.
Tong Y, Eigler T. Transcriptional targets for pituitary tumor-transforming gene-1. Journal of Molecular Endocrinology 2009;43(5) 179-85. - 179.
Salehi F, Kovacs K, Scheithauer BW, Lloyd RV, Cusimano M. Pituitary tumor-transforming gene in endocrine and other neoplasms: a review and update. Endocrine Related Cancer 2008;15(3) 721-43. - 180.
Abbud RA, Takumi I, Barker EM, Ren SG, Chen DY, Wawrowsky K, Melmed S. Early multipotential pituitary focal hyperplasia in the alpha-subunit of glycoprotein hormone-driven pituitary tumor-transforming gene transgenic mice. Molecular Endocrinology 2005;19(5) 1383-91. - 181.
Cleynen I, Van de Ven WJ. The HMGA proteins: a myriad of functions (Review). International Journal of Oncology 2008;32(2) 289-305. - 182.
Fedele M, Fusco A. Role of the high mobility group A proteins in the regulation of pituitary cell cycle. Journal of Molecular Endocrinology 2010;44(6) 309-18. - 183.
Palmieri D, Valentino T, De Martino I, Esposito F, Cappabianca P, Wierinckx A, Vitiello M, Lombardi G, Colao A, Trouillas J, Pierantoni GM, Fusco A, Fedele M. PIT1 upregulation by HMGA proteins has a role in pituitary tumorigenesis. Endocrine Related Cancer 2012;19(2) 123-35. - 184.
Qian ZR, Asa SL, Siomi H, Siomi MC, Yoshimoto K, Yamada S, Wang EL, Rahman MM, Inoue H, Itakura M, Kudo E, Sano T. Overexpression of HMGA2 relates to reduction of the let-7 and its relationship to clinicopathological features in pituitary adenomas. Modern Pathology 2009;22(3) 431-41. - 185.
Wang EL, Qian ZR, Rahman MM, Yoshimoto K, Yamada S, Kudo E, Sano T. Increased expression of HMGA1 correlates with tumour invasiveness and proliferation in human pituitary adenomas. Histopathology 2010;56(4) 501-9. - 186.
May LT, Hill SJ. ERK phosphorylation: spatial and temporal regulation by G protein-coupled receptors. The International Journal of Biochemistry & Cell Biology 2008;40(10) 2013-7. - 187.
Pópulo H, Lopes JM, Soares P. The mTOR Signalling Pathway in Human Cancer. International Journal of Molecular Sciences 2012;13(2) 1886-918. - 188.
McCubrey JA, Steelman LS, Franklin RA, Abrams SL, Chappell WH, Wong EW, Lehmann BD, Terrian DM, Basecke J, Stivala F, Libra M, Evangelisti C, Martelli AM. Targeting the RAF/MEK/ERK, PI3K/AKT and p53 pathways in hematopoietic drug resistance. Advances in Enzyme Regulation 2007;47 64-103. - 189.
Suojun Z, Feng W, Dongsheng G, Ting L. Targeting Raf/MEK/ERK pathway in pituitary adenomas. European Journal of Cancer 2012;48(3) 389-95. - 190.
Cakir M, Grossman AB. Targeting MAPK (Ras/ERK) and PI3K/Akt pathways in pituitary tumorigenesis. Expert Opinion on Therapeutic Targets 2009;13(9) 1121-34. - 191.
Ewing I, Pedder-Smith S, Franchi G, Ruscica M, Emery M, Vax V, Garcia E, Czirják S, Hanzély Z, Kola B, Korbonits M, Grossman AB. A mutation and expression analysis of the oncogene BRAF in pituitary adenomas. Clinical Endocrinology 2007;66(3) 348-52. - 192.
Dworakowska D, Wlodek E, Leontiou CA, Igreja S, Cakir M, Teng M, Prodromou N, Góth MI, Grozinsky-Glasberg S, Gueorguiev M, Kola B, Korbonits M, Grossman AB. Activation of RAF/MEK/ERK and PI3K/AKT/mTOR pathways in pituitary adenomas and their effects on downstream effectors. Endocrine Related Cancer 2009;16(4) 1329-38. - 193.
Kovalovsky D, Refojo D, Liberman AC, Hochbaum D, Pereda MP, Coso OA, Stalla GK, Holsboer F, Arzt E. Activation and induction of NUR77/NURR1 in corticotrophs by CRH/cAMP: involvement of calcium, protein kinase A, and MAPK pathways. Molecular Endocrinology 2002;16(7) 1638-51. - 194.
Bliss SP, Navratil AM, Xie J, Roberson MS. GnRH signaling, the gonadotrope and endocrine control of fertility. Frontiers in Neuroendocrinology 2010;31(3) 322-40. - 195.
Pertuit M, Romano D, Zeiller C, Barlier A, Enjalbert A, Gerard C. The gsp oncogene disrupts Ras/ERK-dependent prolactin gene regulation in gsp inducible somatotroph cell line. Endocrinology 2011;152(4) 1234-43. - 196.
Musat M, Korbonits M, Kola B, Borboli N, Hanson MR, Nanzer AM, Grigson J, Jordan S, Morris DG, Gueorguiev M, Coculescu M, Basu S, Grossman AB. Enhanced protein kinase B/Akt signalling in pituitary tumours. Endocrine Related Cancer 2005;12(2) 423-33. - 197.
Lu C, Willingham MC, Furuya F, Cheng SY. Activation of phosphatidylinositol 3-kinase signaling promotes aberrant pituitary growth in a mouse model of thyroid-stimulating hormone-secreting pituitary tumors. Endocrinology 2008;149(7) 3339-45. - 198.
Theodoropoulou M, Stalla GK, Spengler D. ZAC1 target genes and pituitary tumorigenesis. Molecular and Cellular Endocrinology 2010;326(1-2) 60-5. - 199.
Pizarro CB, Oliveira MC, Pereira-Lima JF, Leães CG, Kramer CK, Schuch T, Barbosa-Coutinho LM, Ferreira NP. Evaluation of angiogenesis in 77 pituitary adenomas using endoglin as a marker. Neuropathology 2009;29(1) 40-4. - 200.
Recouvreux MV, Camilletti MA, Rifkin DB, Becu-Villalobos D, Díaz-Torga G. Thrombospondin-1 (TSP-1) analogs ABT-510 and ABT-898 inhibit prolactinoma growth and recover active pituitary transforming growth factor-β1 (TGF-β1). Endocrinology 2012;153(8) 3861-71. - 201.
Raica M, Coculescu M, Cimpean AM, Ribatti D. Endocrine gland derived-VEGF is down-regulated in human pituitary adenoma. Anticancer Research 2010;30(10) 3981-6. - 202.
Vidal S, Horvath E, Kovacs K, Kuroki T, Lloyd RV, Scheithauer BW. Expression of hypoxia-inducible factor-1alpha (HIF-1alpha) in pituitary tumours. Histology and Histopathology 2003;18(3) 679-86. - 203.
Yoshida D, Kim K, Noha M, Teramoto A. Anti-apoptotic action by hypoxia inducible factor 1-alpha in human pituitary adenoma cell line, HP-75 in hypoxic condition. Journal of Neuro-oncology 2006;78(3) 217-25. - 204.
Shan B, Gerez J, Haedo M, Fuertes M, Theodoropoulou M, Buchfelder M, Losa M, Stalla GK, Arzt E, Renner U. RSUME is implicated in HIF-1-induced VEGF-A production in pituitary tumour cells. Endocrine Related Cancer 2012;19(1) 13-27. - 205.
Jin Kim Y, Hyun Kim C, Hwan Cheong J, Min Kim J. Relationship between expression of vascular endothelial growth factor and intratumoral hemorrhage in human pituitary adenomas. Tumori 2011;97(5) 639-46. - 206.
Xiao Z, Liu Q, Mao F, Wu J, Lei T. TNF-α-induced VEGF and MMP-9 expression promotes hemorrhagic transformation in pituitary adenomas. International Journal of Molecular Sciences 2011;12(6) 4165-79. - 207.
Xiao Z, Liu Q, Zhao B, Wu J, Lei T. Hypoxia induces hemorrhagic transformation in pituitary adenomas via the HIF-1α signaling pathway. Oncology Reports 2011;26(6) 1457-64. - 208.
Rotondo F, Sharma S, Scheithauer BW, Horvath E, Syro LV, Cusimano M, Nassiri F, Yousef GM, Kovacs K. Endoglin and CD-34 immunoreactivity in the assessment of microvessel density in normal pituitary and adenoma subtypes. Neoplasma 2010;57(6) 590-3. - 209.
Miyajima K, Takekoshi S, Itoh J, Kakimoto K, Miyakoshi T, Osamura RY. Inhibitory effects of anti-VEGF antibody on the growth and angiogenesis of estrogen-induced pituitary prolactinoma in Fischer 344 Rats: animal model of VEGF-targeted therapy for human endocrine tumors. Acta Histochemica Cytochemica 2010;43(2) 33-44. - 210.
Luque GM, Perez-Millán MI, Ornstein AM, Cristina C, Becu-Villalobos D. Inhibitory effects of antivascular endothelial growth factor strategies in experimental dopamine-resistant prolactinomas. Journal of Pharmacology and Experimental Therapeutics 2011;337(3) 766-74. - 211.
Ortiz LD, Syro LV, Scheithauer BW, Ersen A, Uribe H, Fadul CE, Rotondo F, Horvath E, Kovacs K. Anti-VEGF therapy in pituitary carcinoma. Pituitary. 2012;15(3) 445-9. - 212.
Saraga-Babic M, Bazina M, Vukojevic K, Bocina I, Stefanovic V. Involvement of pro-apoptotic and anti-apoptotic factors in the early development of the human pituitary gland. Histology and Histopathology 2008;23(10) 1259-68. - 213.
Kulig E, Jin L, Qian X, Horvath E, Kovacs K, Stefaneanu L, Scheithauer BW, Lloyd RV. Apoptosis in nontumorous and neoplastic human pituitaries: expression of the Bcl-2 family of proteins. American Journal of Pathology 1999;154(3) 767-74. - 214.
Kapranos N, Kontogeorgos G, Horvath E, Kovacs K. Morphology, molecular regulation and significance of apoptosis in pituitary adenomas. Frontiers of Hormone Research 2004;32 217-34. - 215.
Sambaziotis D, Kapranos N, Kontogeorgos G. Correlation of bcl-2 and bax with apoptosis in human pituitary adenomas. Pituitary 2003;6(3) 127-33. - 216.
Jaita G, Zárate S, Ferrari L, Radl D, Ferraris J, Eijo G, Zaldivar V, Pisera D, Seilicovich A. Gonadal steroids modulate Fas-induced apoptosis of lactotropes and somatotropes. Endocrine 2011;39(1) 21-7. - 217.
Chen L, Zhuang G, Li W, Liu Y, Zhang J, Tian X. RGD-FasL induces apoptosis of pituitary adenoma cells. Cellular & Molecular Immunology 2008;5(1) 61-8. - 218.
Candolfi M, Jaita G, Pisera D, Ferrari L, Barcia C, Liu C, Yu J, Liu G, Castro MG, Seilicovich A. Adenoviral vectors encoding tumor necrosis factor-alpha and FasL induce apoptosis of normal and tumoral anterior pituitary cells. Journal of Endocrinology 2006;189(3) 681-90. - 219.
Cañibano C, Rodriguez NL, Saez C, Tovar S, Garcia-Lavandeira M, Borrello MG, Vidal A, Costantini F, Japon M, Dieguez C, Alvarez CV. The dependence receptor Ret induces apoptosis in somatotrophs through a Pit-1/p53 pathway, preventing tumor growth. EMBO Journal 2007;26(8) 2015-28. - 220.
Diaz-Rodriguez E, García-Lavandeira M, Perez-Romero S, Senra A, Cañibano C, Palmero I, Borrello MG, Dieguez C, Alvarez CV. Direct promoter induction of p19Arf by Pit-1 explains the dependence receptor RET/Pit-1/p53-induced apoptosis in the pituitary somatotroph cells. Oncogene 2012;31(23) 2824-35. - 221.
Japón MA, Urbano AG, Sáez C, Segura DI, Cerro AL, Diéguez C, Alvarez CV. Glial-derived neurotropic factor and RET gene expression in normal human anterior pituitary cell types and in pituitary tumors. The Journal of Clinical Endocrinology & Metabolism 2002;87(4) 1879-84. - 222.
Kang BH, Xia F, Pop R, Dohi T, Socolovsky M, Altieri DC. Developmental control of apoptosis by the immunophilin aryl hydrocarbon receptor-interacting protein(AIP) involves mitochondrial import of the survivin protein. The Journal of Biological Chemistry 2011;286(19) 16758-67. - 223.
Vargiolu M, Fusco D, Kurelac I, Dirnberger D, Baumeister R, Morra I, Melcarne A, Rimondini R, Romeo G, Bonora E. The tyrosine kinase receptor RET interacts in vivo with aryl hydrocarbon receptor-interacting protein to alter survivin availability. The Journal of Clinical Endocrinology & Metabolism 2009;94(7) 2571-8. - 224.
Wasko R, Waligorska-Stachura J, Jankowska A, Warchol JB, Liebert W, Sowinski J. Coexpression of survivin and PCNA in pituitary tumors and normal pituitary. Neuro Endocrinology Letters 2009;30(4) 477-81. - 225.
Formosa R, Gruppetta M, Falzon S, Santillo G, DeGaetano J, Xuereb-Anastasi A, Vassallo J. Expression and clinical significance of Wnt players and survivin in pituitary tumours. Endocrine Pathology 2012;23(2) 123-31. - 226.
Tagliati F, Gentilin E, Buratto M, Molè D, degli Uberti EC, Zatelli MC. Magmas, a gene newly identified as overexpressed in human and mouse ACTH-secreting pituitary adenomas, protects pituitary cells from apoptotic stimuli. Endocrinology 2010;151(10) 4635-42. - 227.
Bahar A, Simpson DJ, Cutty SJ, Bicknell JE, Hoban PR, Holley S, Mourtada-Maarabouni M, Williams GT, Clayton RN, Farrell WE. Isolation and characterization of a novel pituitary tumor apoptosis gene. Molecular Endocrinology 2004;18(7) 1827-39. - 228.
Acunzo J, Roche C, Defilles C, Thirion S, Quentien MH, Figarella-Branger D, Graillon T, Dufour H, Brue T, Pellegrini I, Enjalbert A, Barlier A. Inactivation of PITX2 transcription factor induced apoptosis of gonadotroph tumoral cells. Endocrinology 2011;152(10) 3884-92. - 229.
Radl DB, Ferraris J, Boti V, Seilicovich A, Sarkar DK, Pisera D. Dopamine-induced apoptosis of lactotropes is mediated by the short isoform of D2 receptor. PLoS One 2011;6(3) e18097. - 230.
Grozinsky-Glasberg S, Shimon I, Korbonits M, Grossman AB. Somatostatin analogues in the control of neuroendocrine tumours: efficacy and mechanisms. Endocrine Related Cancer 2008;15(3) 701-20. - 231.
223. Chesnokova V, Melmed S. Pituitary senescence: the evolving role of Pttg. Molecular and Cellular Endocrinology 2010;326(1-2) 55-9. - 232.
Chesnokova V, Zonis S, Zhou C, Ben-Shlomo A, Wawrowsky K, Toledano Y, Tong Y, Kovacs K, Scheithauer B, Melmed S. Lineage-specific restraint of pituitary gonadotroph cell adenoma growth. PLoS One 2011;6(3) e17924. - 233.
Ragel BT, Couldwell WT. Pituitary carcinoma: a review of the literature. Neurosurgical Focus 2004;16(4) E7. - 234.
Kaltsas GA, Nomikos P, Kontogeorgos G, Buchfelder M, Grossman AB. Clinical review: Diagnosis and management of pituitary carcinomas. The Journal of Clinical Endocrinology & Metabolism 2005;90(5) 3089-99. - 235.
Colao A, Ochoa AS, Auriemma RS, Faggiano A, Pivonello R, Lombardi G. Pituitary carcinomas. Frontiers of Hormone Research 2010;38 94-108. - 236.
van der Klaauw AA, Kienitz T, Strasburger CJ, Smit JW, Romijn JA. Malignant pituitary corticotroph adenomas: report of two cases and a comprehensive review of the literature. Pituitary 2009;12(1) 57-69. - 237.
Ruebel KH, Leontovich AA, Jin L, Stilling GA, Zhang H, Qian X, Nakamura N, Scheithauer BW, Kovacs K, Lloyd RV. Patterns of gene expression in pituitary carcinomas and adenomas analyzed by high-density oligonucleotide arrays, reverse transcriptase-quantitative PCR, and protein expression. Endocrine 2006;29(3) 435-44. - 238.
Sav A, Rotondo F, Syro LV, Scheithauer BW, Kovacs K. Biomarkers of pituitary neoplasms. Anticancer Research 2012;32(11) 4639-54. - 239.
Syro LV, Ortiz LD, Scheithauer BW, Lloyd R, Lau Q, Gonzalez R, Uribe H, Cusimano M, Kovacs K, Horvath E. Treatment of pituitary neoplasms with temozolomide: a review. Cancer 2011;117(3) 454-62. - 240.
Raverot G, Sturm N, de Fraipont F, Muller M, Salenave S, Caron P, Chabre O, Chanson P, Cortet-Rudelli C, Assaker R, Dufour H, Gaillard S, François P, Jouanneau E, Passagia JG, Bernier M, Cornélius A, Figarella-Branger D, Trouillas J, Borson-Chazot F, Brue T. Temozolomide treatment in aggressive pituitary tumors and pituitary carcinomas: a French multicenter experience. The Journal of Clinical Endocrinology & Metabolism 2010;95(10) 4592-9. - 241.
Raverot G, Castinetti F, Jouanneau E, Morange I, Figarella-Branger D, Dufour H, Trouillas J, Brue T. Pituitary carcinomas and aggressive pituitary tumours: merits and pitfalls of temozolomide treatment. Clinical Endocrinology 2012;76(6) 769-75. - 242.
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. Journal of Neuro-oncology 2011;104(3) 647-57.