SlideShare ist ein Scribd-Unternehmen logo
1 von 12
Downloaden Sie, um offline zu lesen
INDEX www.yassermetwally.com




                                                            INTRODUCTION
                                                        




INTRODUCTION

Pituitary tumors exhibit a spectrum of biology, with variable growth and hormonal behaviors. They therefore provide an
opportunity to examine pathogenetic mechanisms that underlie the neoplastic process. These include alterations in hormone
regulation, growth-factor stimulation, cell-cycle control and cell-stromal interactions that result from genetic mutations or
epigenetic disruption of gene expression. Mouse models have validated the roles of these alterations, which can be targets for the
development of therapies that can manage these lesions. These therapies are increasingly recognized as critical for quality of life.

The pituitary, a small bean-shaped gland at the base of the brain, maintains multiple homeostatic functions including metabolism,
growth and reproduction. Most pituitary tumors are adenomas that develop in the adenohypophysis (the anterior lobe of the
pituitary gland), which is composed of six epithelial cell types that secrete peptide hormones.

Pituitary adenomas exhibit a wide range of hormonal and proliferative behaviors.[1] They may be small, slowly growing and
hormonally in active, and might only be detected as radiographic quot;incidentalomasquot; or at postmortem examination. If, however,
they produce hormones in excess, they can cause mood disorders, sexual dysfunction, infertility, obesity and disfigurement,
hypertension, diabetes mellitus and accelerated heart disease. If untreated, hormone-excess syndromes can be lethal.

Some pituitary adenomas grow rapidly, producing symptoms of an intracranial mass, loss of normal anterior pituitary hormone
production, and visual-field disturbances due to stretching of the overlying optic chiasm. They can invade downward into
paranasal sinuses, laterally into the cavernous sinuses (thereby disrupting coordinated eye movement) and upwards into the
brain. They can cause death by invasion of the brain.

Studies using X-chromosome inactivation proved that pituitary adenomas are monoclonal neoplasms.[2] Results that suggest
polyclonality are attributed to contamination by normal tissue; the small adenomas associated with Cushing's syndrome are
particularly subject to this phenomenon.

In contrast to their high frequency of local invasion, only rarely do pituitary tumors metastasize to distant locations in the central
nervous system, lymph nodes and liver. This pattern of spread is the criterion of malignancy, allowing a diagnosis of pituitary
carcinoma. Many tumors classified as benign are nevertheless associated with significant morbidity and mortality.

In this review we will focus on our understanding of the pathogenetic mechanisms underlying the biologic and morphologic
features of human pituitary tumors. These include alterations in hormonal regulation and hormone receptors, dysregulated
growth factors and alterations in their receptors, abnormalities in signaling proteins that transduce the signals of these stimuli,
and changes in cell-cycle regulators (Figure 1). In addition, the neoplastic process is associated with altered cell-stromal
interactions that have a role in the morphogenesis of pituitary tumors (Figure 2). These changes are summarized below.



                                                                                      Figure 1. Maintaining the delicate balance in
                                                                                      pituitary endocrine cell survival. The
                                                                                      contribution from some of the characterized
                                                                                      factors in anterior pituitary cell growth is
                                                                                      shown. Many hormones bind G-protein-
                                                                                      coupled receptors, which wind back and forth
                                                                                      through the cell membrane seven times and
                                                                                      can exert either stimulatory or inhibitory
                                                                                      effects. Growth-factor receptors can similarly
                                                                                      be stimulatory or inhibitory, and expression
                                                                                      levels of, or mutations in, either the receptors
                                                                                      or their ligands can affect signaling. The
                                                                                      relative contributions of cell-cycle control
                                                                                      proteins involved in pituitary cells are
                                                                                      becoming       better    defined.     Estradiol,
                                                                                      endogenous T3 and glucocorticoids act
                                                                                      through dedicated nuclear steroid hormone
                                                                                      receptors to mediate negative feedback on
                                                                                      respective pituitary stimulatory signals.



CRH, corticotropin-releasing hormone; D2R, dopamine receptor 2; E2, estradiol; EGF, epidermal growth factor; FGFs,
fibroblast growth factors; GC, glucocortiocoids; GH, growth hormone; GHRH, GH-releasing hormone; Gi, inhibitory G protein;
GnRH, gonadotropin-releasing hormone; Gs, stimulatory G protein; PRL, prolactin; ptd-FGFR-4, pituitary-tumor-derived
fibroblast growth factor receptor 4; PTTG, pituitary tumor-transforming gene; Ras, intracellular GTP-binding protein that is
implicated in cell-signaling pathways; Rb1, retinoblastoma 1; SST, somatostatin; TGF, transforming growth factor; TRH, TSH-
releasing hormone.

       Epidemiology and Classification of Pituitary Adenomas
   


There are limited data on the prevalence of pituitary tumors. Although classically considered to be rare, systematic assessments
from autopsy and radiologic studies have shown a prevalence of 17% of the population.[3] The sex incidence is equal. These
tumors are rare in childhood and the incidence increases with age.[1]

Prolactin-producing adenomas are the most common type.[1] One-third are unassociated with hypersecretory syndromes; the
majority of these produce but do not secrete the gonadotropins follicle-stimulating hormone and/or luteinizing hormone. Growth-
hormone (GH)-producing or adrenocorticotropic hormone (ACTH)-producing adenomas each account for 10-15% of pituitary
adenomas. Adenomas producing thyrotropin (TSH) are rare.

Hormonal activity classifies pituitary adenomas: ACTH-producing adenomas are associated with Cushing's or Nelson's
syndromes; GH-producing adenomas are associated with acromegaly and/or gigantism; prolactin-producing adenomas cause
hyperprolactinemia; TSH-producing tumors are associated with thyroid dysfunction; there are rare, clinically detectable,
gonadotroph adenomas; and there are quot;endocrinologically inactivequot; adenomas. Anatomic and/or radiographic classification is
based on tumor size and degree of invasion, information that is critical for patient management and, when surgery is indicated,
guides the surgical approach. Immunohistochemistry allows identification of hormone content that usually correlates with the
clinical presentation ( Table 1 ). Ultrastructural classification by electron microscopy allows identification of sub cellular variants
that now can be characterized using immunohistochemical markers, such as keratins;[1] these variants respond to therapy
differently, with variable prognosis and should, therefore, be considered in patient management. Transcription factors that
regulate hormone expression are used to classify cytogenesis even in hormone-negative adenomas;[1] tumors derived from
different cell lines behave differently, making tumor cytogenesis important for clinical care. It remains to be established whether
other characteristics, such as proliferation markers, growth factors and/or their receptors, or oncogene products, will prove to be
more reliable predictors of invasive growth, recurrence, or metastasis.

Table 1. Pituitary Cells, Hormones, Tumors and Hormone-excess Syndromes




       Alterations in Hormone Regulation
   


The development of the adenohypophysis is dependent on hormones for expansion of differentiated cell populations.
Adenohypophysiotropic hormones that are produced by the hypothalamus and target organs are involved in complex regulatory
feedback mechanisms, which are tightly controlled to maintain homeostasis.[4] Pathology external to the pituitary itself Results in
hyperplasia or atrophy of the relevant adenohypophysial cell population.[1,2,4] For this reason, hormonal stimulation or impaired
feedback inhibition has been thought to underlie the pathogenesis of pituitary tumors.

The data suggest, however, that although hormones may promote cell proliferation, hormonal stimulation is not implicated in the
neoplastic transformation that underlies the development of human pituitary tumors.

GH-releasing hormone (GHRH) stimulates somatotroph proliferation and causes somatotroph hyperplasia in mice ( Table 2 );[2]
in patients with endocrine carcinomas that secrete GHRH ectopically, there is somatotroph hyperplasia but tumors do not
develop in this situation.[2] During pregnancy, estrogen causes lactotroph hyperplasia; conversely, antiestrogen activity induces
lactotroph apoptosis and also suppresses tumor formation.[5] Some lactotroph adenomas grow during gestation;[6] however,
although estrogen was implicated in the pathogenesis of a lactotroph adenoma in male-to-female transsexuals,[7] the clinical use
of estrogen in contraceptive agents or in postmenopausal hormone replacement has not been associated with an increased risk of
pituitary tumor development.[8] TSH-releasing hormone (TRH) stimulates thyrotrophs and lactotrophs, and patients with
primary hypothyroidism develop pituitary thyrotroph and lactotroph hyperplasia.[1] Excess of corticotropin-releasing hormone
causes pituitary corticotroph hyperplasia and Cushing's syndrome in humans with ectopic hormone production by tumors, but
tumor development has not been reported in this setting.[9]
Table 2. Summary of Known Genetic Alterations in
                                                                             Pituitary Tumors




Loss of hormonal inhibition might also be involved in hormonal dysregulation. Dopamine maintains tonic inhibition of
lactotrophs. In prolactinomas, neovascularization that bypassed the hypothalamus was observed, which implicated the presence
of diminished hypo thalamic inhibition, and allowed the tumor cells to escape local inhibition by dopamine.[10] Although
dopamine-receptor-deficient mice develop massive lactotroph hyperplasia,[11] no mutations in the DRD2 gene have been
identified in human pituitary adenomas and most lactotroph tumors respond to dopamine agonists.

Hypothalamic somatostatin inhibits GH secretion, and somatostatin receptors are expressed on somatotroph adenomas.[12,13] A
rare, germline, inactivating mutation in somatostatin receptor 5 was reported in a patient with acromegaly,[14] and expression of
somatostatin is reported to be lower in invasive, GH-secreting tumors than in normal pituitary tissue;[15-18] however, there is no
evidence that loss of somatostatin inhibition is etiologically important in somatotroph adenoma development. GH secretion is also
inhibited by insulin-like growth factor 1 (IGF1). Patients with GH-producing adenomas have elevated levels of circulating IGF1
that would be expected to suppress adenoma cells, but no alterations in IGF1 response have been identified that explain
tumorigenesis.

Patients with long-standing primary hypogonadism develop pituitary gonadotroph adenomas.[1] Similarly, those with primary
hypothyroidism develop thyrotroph hyperplasia that can progress to adenoma if untreated.[1] Primary adrenal failure, in the
setting of prolonged glucocorticoid deficiency, leads to corticotroph hyperplasia and adenoma,[1] suggesting that loss of
glucocorticoid feedback might cause Cushing's disease. A novel germ-line mutation was implicated as the cause of pituitary
Cushing's disease in a small number of patients.[19] Similarly, rare glucocorticoid-receptor mutations that diminished
glucocorticoid inhibition were found in corticotroph tumors.[20,21] The majority of tumors, however, do not exhibit alterations
that would explain dysregulated hormonal inhibition.

Old transgenic mice that overexpress GHRH develop pituitary adenomas, and dopamine-receptor-deficient mice ultimately
develop invasive lactotroph adenomas,[2] but the delayed neoplasia observed in both settings, as in humans with primary failure
of the thyroid, adrenals or gonads, implicates additional genetic events in neoplastic transformation. Indeed, it is distinctly
unusual to find hyperplasia underlying the vast majority of sporadic human pituitary adenomas, suggesting that hormonal
stimulation is not a primary etiologic mechanism. Sustained exposure to hormonal stimulation does not, moreover, always lead to
adenoma formation in humans.[22] Although they might promote proliferation of transformed pituitary cells, it seems that
hormones do not cause pituitary tumors. The role of promotion of established tumors by hormones is supported by the finding of
exaggerated intrapituitary GHRH expression in aggressive somatotroph adenomas.[23]
Alterations of Hormone Receptors
   



Activating mutations of the GHRH receptor might explain transformation of somatotrophs, but this possibility has been excluded.
[24] Instead, a truncated, alternatively spliced form of the receptor with limited signaling properties was identified in GH-
producing pituitary adenomas.[25] There are no reports of activating mutations of the corticotropin-releasing hormone receptor
in corticotroph adenomas.

Recently, the concept of GH and prolactin autoregulation in the pituitary was proven. Prolactin regulates its own secretion at
both hypothalamic and pituitary levels.[26] Prolactin-receptor (PRLR)-deficient mice develop earlier lactotroph hyperplasia, and
adenomas that are larger and more invasive, than the hyperplasia seen in dopamine D2 receptor-deficient mice,[27] proving the
importance of this short autoregulatory loop. Prolactin-producing adenomas have higher levels of PRLR messenger RNA
(mRNA) than other types of adenomas. Dopamine-agonist therapy decreases the levels of PRLR mRNA in human prolactin
adenomas.[28] There is no evidence, however, of disrupted pro lactin autoregulation in lactotroph adenomas. In contrast to the
situation with prolactin, where mouse models indicate autoregulation but there is no human pathology implicating this process,
GH has a more subtle autoregulatory role in mice and a pathogenetic significance in humans. Disruption of GH autoregulation in
mice Results in subtle somatotroph enlargement and hyperplasia.[29] GH-receptor antagonism in human somatotrophs leads to
formation of fibrous bodies–abnormal aggregates of keratin filaments characteristic of a subset of human somatotroph adenomas,
the quot;sparsely granulatedquot; variant.[1] Consistent with this finding, somatic mutations of the GH-receptor were found in these
tumors (Asa SL et al., unpublished data), implicating disruption of GH autoregulation in the development of this subtype of
tumor.

The TRH receptor is expressed in thyrotroph adenomas.[30] In some adenomas, the TRH receptor mRNA is alternatively spliced;
deletion of exon 3 Results in a truncated product that does not bind its ligand efficiently. Elevated levels of similar truncated
products in lactotroph adenomas[31] might explain paradoxical in vivo responses to TRH administration.

Thyroid-hormone receptors (TRs) that bind DNA regulatory elements mediate thyroid hormone regulation. TRa mRNA mis-
sense mutations, and an alternatively spliced TRß2 variant with a 135-base-pair deletion in the ligand-binding domain have been
described in TSH-producing tumors.[32]

These data indicate that abnormal hormone receptor signaling can contribute to dysfunctional responses to feedback inhibition;
however, apart from one subtype of GH-secreting adenomas, hormone receptor alterations do not seem to be common events that
could be implicated in the etiology and pathogenesis of the majority of pituitary adenomas.

       Dysregulated Growth Factors and Their Receptors
   



The pituitary is the target for and site of synthesis of several growth factors that modulate hormone production and regulate
pituitary cell growth.[2] Some growth factors are critical for normal pituitary development. Alterations in their expression or
action might have important roles in the growth of tumors.

The epidermal growth factor (EGF) family and its receptors have been implicated in tumorigenesis in a number of neoplasms.
The pituitary gland expresses the ligands transforming growth factor (TGF)-a and EGF, which alter pituitary hormone secretion
and induce cell proliferation.[2] In fact, TGF-a has been implicated as the mediator of estrogen-induced lactotroph proliferation,
since TGF-a antisense expression specifically inhibits this response.[33] Overexpression of TGF-a, under the control of the
prolactin promoter, Results in lactotroph adenomas in transgenic mice,[34] indicating a role for this growth factor in pituitary
tumorigenesis. The common receptor of EGF and TGF-a, EGFR, is one of a family of four highly homologous receptors
possessing tyrosine kinase activity, along with ErbB2/HER2/neu/p185, ErbB3/HER3, and ErbB4/HER4. EGFR expression
correlates with pituitary tumor aggressiveness–especially among GH-producing tumors.[35] In contrast to other human
neoplasms, however, ERBB2 is not mutated or amplified in human pituitary tumors.[2]

The TGF-ß family is represented in the pituitary by inhibin A (a-ßA heterodimers), inhibin B (a-ßB), activin (ßA-ßB), activin A
(ßA-ßA) and activin B (ßB-ßB). Inhibin subunits are expressed by pituitary gonadotroph adenomas and activin stimulates
hormone secretion by these tumors.[2] The effects of activin are modulated by follistatin and activin receptors. Follistatin binds
activins and downregulates their activity. Activin receptors are expressed in gonadotroph adenomas and follistatin expression is
reduced or diminished,[36] implicating enhanced activin signaling as a pathogenetic mechanism. Additionally, a truncated activin
receptor type-IB isoform that fails to transduce activin-induced growth-arrest signaling was described exclusively in tumors, but
not normal human pituitary cells,[37] underscoring the importance of the activin/follistatin balance in modulating pituitary cell
growth.

The fibroblast growth factors (FGFs) and their receptors (FGFRs) regulate development, growth, and angiogenesis. There are
currently 23 known FGF ligands. Basic (b)FGF was originally described in the bovine pituitary folliculostellate cell, and regulates
pituitary hormones; bFGF is expressed by human pituitary adenomas and elevated circulating FGF is found in patients with
pituitary adenomas.[2] FGF-4 sequences are present in transforming DNA from human pro lactin-secreting tumors,[38] and
FGF-4 facilitates lactotroph proliferation.[39] FGF-8 is necessary for pituitary development,[40] and expression of a soluble
dominant-negative FGFR Results in pituitary dysgenesis.[41] The product of the endogenous FGF antisense gene (NUDT6)
inhibits pituitary cell proliferation.[42] A truncated kinase-containing variant of FGFR-4 with an alternative initiation site[43,44]
was isolated from human pituitary tumors.[45] This pituitary-tumor-derived (ptd)-FGFR-4 isoform (ptd-FGFR-4) causes
transformation in vitro and in vivo and, when expressed in transgenic mice, recapitulates invasive pituitary adenoma formation.
[46]

These studies provide convincing evidence for the important role of growth factors, and in particular, the FGF system, in
pituitary development and tumor formation.

       Abnormal Signaling Proteins
   



Altered signaling by proteins involved in signal transduction of hormones and growth factors might be implicated in pituitary
tumorigenesis.

G-proteins transduce signals from cell-surface ligand-receptor complexes to downstream effectors. The family of stimulatory G
proteins (Gs) transduces GHRH signals. Point mutations in Gsa codon 201 (Arg201Cys) and codon 227 (Gln227Arg) activate
adenylyl cyclase by interfering with GTP hydrolysis, Gsa in a constitutively active state.[45] These G-protein oncogenes (GSPs)
were first described in a subset of pituitary somatotroph adenomas and hormone-secreting tumors of other endocrine glands.
[45,47] GSP mutations are more frequently detected in the maternal allele, which is consistent with monoallelic imprinting of this
gene. There is no correlation between GSP mutations and patient age, sex, tumor size, or circulating GH levels;[48] however, this
mutation correlates with the densely granulated phenotype of somatotroph tumors,[49] elevated levels of phosphorylated cyclic
AMP (cAMP) response element binding protein,[50] and higher circulating levels of free glycoprotein-subunit.[51] These features
might be predictive of GH responsiveness to somatostatin-analog therapy.[52] Inactivating mutations in the a-subunit of GIP2, a
protein coupled to the inhibitory G-protein Gi2a, have been identified.[53] No mutations have been identified in the stimulatory
Gaq or the highly similar Ga11.[54]

Ras proteins are also involved in transducing signals from ligand–receptor complexes at the membrane to downstream effectors.
Ras mutations are common in nonendocrine malignancies, most frequently involving the GTP-binding domain (codons 12/13) or
the GTPase domain (codon 61).[55] Mutation in HRas causing a Gly12Val substitution is distinctly uncommon in pituitary
tumors.[56] Initially described in a highly aggressive prolactinoma, Ras mutations are largely restricted to the rare pituitary
carcinomas.[57]

Protein kinase A is a tetrameric holoenzyme involved in cAMP binding. Adequate levels of the cAMP-dependent protein kinase
type 1 regulatory subunit (the product of PRKAR1A) are critical for intracellular protection against unrestrained catalytic
activity. Consistent with this model, germline mutations in PRKAR1A are associated with Carney's complex,[58] an autosomal-
dominant disorder characterized by development of pituitary and other endocrine tumors.

       Deregulated Cell Cycle Control Elements
   



The autosomal-dominant syndrome, multiple endocrine neoplasia type I (MEN1), to which pituitary tumors are integral, is caused
by germline mutations of the MEN1 tumor-suppressor gene on chromosome 11q13, with loss of heterozygosity (LOH) inactivating
the normal allele in tumors. LOH at this region, observed in 10–30% of sporadic endocrine tumors, suggested that MEN1 could
be implicated in sporadic pituitary adenomas. MEN1 is not, however, responsible for sporadic pituitary adenomas, nor is it
altered in patients with inherited pituitary tumors that are not associated with the MEN1 syndrome. Reduced expression of menin
in sporadic adenomas would indicate that menin functions as a tumor suppressor, but MEN1 mRNA expression is not
downregulated in sporadic pituitary tumors.[2]

Retinoblastoma 1 (Rb1), a tumor-suppressor gene on chromosome 13q, is associated with pituitary intermediate-lobe tumors in
Rb1-deficient mice.[59] Mutations of Rb1 have not been identified in human pituitary adenomas, but the Rb1 promoter is
methylated, resulting in gene silencing.[2] There is LOH at13q in tumors with unmethylated Rb1, suggesting an independent
tumor-suppressor gene at that locus.[2]

Cyclins, cyclin-dependent kinases (CDKs) and CDK inhibitors (CDKIs) regulate cell-cycle progression. CDK activity is
modulated by the CDKIs p27kip1, p57kip2, p16ink4A, p15ink4B, p18ink4C, and p19ink4D. Although CDKN2A (which encodes
p16ink4A) is not mutated in pituitary adenomas, it is frequently silenced by promoter methylation.[60] The CDKI p27kip1 is
important in the pathogenesis and prognosis of several human malignancies. Mice that are Cdkn1b-null do not express p27kip1,
and develop multiorgan neoplasia, including pituitary tumors.[61] Although the CDKN1B gene is not mutated in human pituitary
tumors, expression of its product p27kip1 is reduced in corticotroph adenomas and recurrent human pituitary adenomas.[2]
Protein stability is critical in maintaining p27 protein expression, suggesting epigenetic mechanisms of p27 alteration that can be
targeted for therapy.[62] Mice lacking both p18ink4C and p27kip1 succumb to rapidly lethal pituitary adenomas;[61] however,
there are no reports of p18 alterations in human pituitary tumors.
The growth ? arrest and DNA-damage-inducible gene GADD45G negatively regulates cell growth. Expression of GADD45G
mRNA is significantly diminished in clinically nonfunctioning and hormonally active GH-secreting or prolactin-secreting
pituitary tumors,[63] because of gene silencing through CpG island promoter methylation.[64]

Maternally expressed gene 3 (MEG3) is a human homolog of the mouse MATERNALLY IMPRINTED Gtl2 gene. The MEG3a
isoform, containing an extended exon 5 sequence, inhibits cell growth in vitro. As with GADD45?, MEG3a expression is
diminished in human pituitary tumors associated with 5' promoter hypermethylation.[65]

A number of other cell-cycle regulators have received attention as candidate causes of pituitary tumors. Pituitary tumor
transforming gene 1 (PTTG1) is a member of the securin family that is involved in sister chromatid separation. As expected of a
gene with this function it is upregulated in proliferating cells.[2] Pituitary tumor apoptosis gene (PTAG; this name has not yet
been approved) is a novel antiapoptotic gene on chromosome 22 identified in pituitary tumors but with no known significance.[66]
The tumor suppressor p53, which is mutated or lost in many other forms of cancer, is expressed in pituitary tumors; however,
there is no evidence of TP53 gene mutation or LOH in pituitary tumors.[2] Upregulated expression of this gene probably
represents a compensatory mechanism, so that there might be some prognostic value of p53 staining;[67] however, this is
controversial as there is no consistent correlation between p53 immunoreactivity and tumor invasion or recurrence.[68]

       Dysadhesion and Abnormal Stromal Interactions
   


Progression to neoplasia involves changes in cell-cell and cell-matrix adhesion. Indeed, loss of the reticulin network is the
morphological hallmark of the transition from hyperplasia to adenoma that is commonly used as a diagnostic feature in the
pituitary.[1]

Changes in cell adhesion have been correlated with the abnormal expression of ptd-FGFR-4 in cell lines, transgenic mice and
human pituitary adenomas that result in loss of affinity for extracellular matrix.[69] This loss is due to disruption of a
multiprotein complex involving FGFR-4, N-cadherin (a cell adhesion molecule) and neural cell adhesion molecule 1 (NCAM-1).
Expression of ptd-FGFR-4 Results in diminished and ectopic cytoplasmic expression of N-cadherin, associated with invasive
growth.[69] Uncoupling of N-cadherin from the cytoskeleton destabilizes ß-catenin,[69] and reduced ß-catenin expression in
human pituitary adenomas correlates with tumor invasiveness.[70] Similarly, expression of the polysialated form of NCAM-1
correlates with tumor growth and invasiveness;[71] polysialation Results in steric inhibition of membrane-membrane apposition
and cell adhesiveness and, therefore, probably disrupts the multiprotein complex as does ptd-FGFR-4 (Figure 2). Estrogen, a
known stimulus of pituitary cell proliferation, reduces membranous N-cadherin protein expression to barely detectable levels,
whereas antiestrogen treatment reverses this effect.[72]




Figure 2. The balance between adhesive and dysadhesive forces in pituitary cells. In this model, the membrane location of neural
cell adhesion molecule 1, fibroblast growth factor receptor 4 and N-cadherin is critical for the formation of a proadhesive
multiprotein complex. As shown on the right, wild-type fibroblast growth factor receptor 4 resides predominantly in the cell
membrane and interacts heavily with neural cell adhesion molecule 1 to support stability of N-cadherin and ß-catenin and
maintain actin cytoskeletal structure. By contrast, as shown on the left, receptor isoforms such as pituitary-tumorderived
fibroblast growth factor receptor 4 are located in the cytoplasm, and can displace fibroblast growth factor receptor 4 from neural
cell adhesion molecule 1, thereby disrupting cell-cell and cell-matrix adhesion, which are mediated by N-cadherin and ß-catenin.
A similar form of steric hindrance with this multiprotein proadhesive complex can be achieved by polysialated neural cell
adhesion molecule 1.

FGFR-4, fibroblast growth factor receptor 4; NCAM-1; neural cell adhesion molecule 1; PSA, polysialated; ptd-FGFR-4,
pituitary-tumor-derived fibroblast growth factor receptor 4.

       Implications For Therapy
   


The treatment of pituitary tumors involves medical, surgical, and radiotherapeutic approaches. Pharmacotherapy represents the
cornerstone for treatment of several hormonally active adenomas. Prolactin-secreting tumors are generally treated with
dopamine agonists. Somatostatin analogs are widely adopted to restrain hormone secretion by GH-producing pituitary tumors;
the varying response to somatostatin analogs is now understood to be reflective of the differing pathogenetic mechanisms
underlying the various morphologic tumor types that cause acromegaly. Some of the currently available agents suppress hormone
excess, some result in reduction of tumor size, but they do not achieve a tumoricidal effect. Comparison of somatostatin-sensitive
and somatostatin-resistant tumors, by use of genomic and proteomic approaches, will undoubtedly pave the way for the
development of effective tumoricidal drug treatments; moreover, dopamine and somatostatin analogs are not effective in the
treatment of tumors arising from gonadotrophs or corticotrophs, creating a need for more-effective agents.

Since Rb1 inactivation underlies corticotroph tumor development in mice, this provides a model for the evaluation of gene
therapy approaches in the management of pituitary tumors. Spontaneous pituitary tumors in immunocompetent Rb1-deficient
mice were treated with a recombinant adenovirus carrying Rb1-complementary DNA. Intratumoral Rb1 gene transfer decreased
tumor cell proliferation, re-established innervation by growth-regulatory dopaminergic neurons, and resulted in inhibition of
tumor growth and enhanced animal survival,[73] providing proof of principle.

Deficient or defective delivery of dopamine has been implicated in lactotroph adenomas. This system has been investigated by
transfection of human lactotroph adenomas in vitro with an adenoviral vector that expresses tyrosine hydroxylase. Introduction
of this rate-limiting enzyme in dopamine biosynthesis enhanced dopamine synthesis and diminished prolactin secretion.[74]
Similarly, the pro-opiomelanocortin promoter has also been used to allow selective expression of the toxic herpes simplex virus
thymidine kinase, in mouse pituitary corticotroph cell lines. Infection in vitro or injection in vivo results in selective gene
expression and cytotoxicity.[5] This hormone promoter might, therefore, be used in selective targeting of gene therapy vectors to
tumors associated with excessive ACTH production and Cushing's disease. These studies provide evidence for the feasibility of
gene therapy and the use of pituitary-specific promoters to confer selectivity in treating pituitary tumors.

Corticotroph adenomas are characterized by loss of the tumor suppressor p27, as a result of protein degradation.[62] The actions
of vitamin D3 in restoring p27 protein levels[62] could have therapeutic significance for these tumors, which as yet have no other
targeted medical therapies.

Also close to the clinical horizon is the application of selective tyrosine kinase inhibitors. These agents should be tested in
therapeutic trials in defined patient populations–especially those in whom surgical or medical therapies have failed. Selective
inhibitors of the FGFR and EGFR pathways might be used to treat pituitary tumors effectively. Tumor-associated EGFR and
FGFR could, moreover, be targeted, by delivering toxins or antiproliferative monoclonal antibodies to tumor cells.

       Conclusions
   



The individual contributions of the components involved in the complex cascade of events leading to human neoplasia are finally
unraveling. The delicate balance maintained by the dominant factors in the pituitary is summarized in Figures 1 and 2. It is
important to emphasize that it is becoming increasingly evident, in many systems, that no single factor can effectively explain the
many facets of the tumorigenic process. It is nevertheless anticipated that specific interruption of several signaling cascades will
afford new approaches to the therapeutic armamentarium, permitting more-effective strategies in the management of pituitary
tumors.



References

  1. Asa SL (1998) Tumors of the Pituitary Gland. In Atlas of Tumor Pathology, Series 3, Fascicle 22. Washington, DC: Armed
     Forces Institute of Pathology

  2. Asa SL and Ezzat S (2002) The pathogenesis of pituitary tumours. Nat Rev Cancer 2: 836–849
3. Ezzat S et al. (2004) The prevalence of pituitary adenomas: a systematic review. Cancer 101: 613–619

 4. Asa SL (1991) The role of hypothalamic hormones in the pathogenesis of pituitary adenomas. Pathol Res Pract 187: 581–583

 5. Lee EJ et al. (2001) Adenovirus-directed expression of dominant negative estrogen receptor induces apoptosis in breast
    cancer cells and regression of tumors in nude mice. Mol Med 7: 773–782

 6. Molitch ME (2003) Pituitary tumors and pregnancy. Growth Horm IGF Res 13 (Suppl A): S38–S44

 7. Kovacs K et al. (1994) Prolactin-producing pituitary adenoma in a male-to-female transsexual patient with protracted
    estrogen administration. A morphologic study. Arch Pathol Lab Med 118: 562–565

 8. Testa G et al. (1998) Two-year treatment with oral contraceptives in hyperprolactinemic patients. Contraception 58: 69–73

 9. Carey RM et al. (1984) Ectopic secretion of corticotropin-releasing factor as a cause of Cushing's syndrome. A clinical,
    morphologic, and biochemical study. N Engl J Med 311: 13–20

10. Schechter J et al. (1988) Morphological evidence for the presence of arteries in human prolactinomas. J Clin Endocrinol
    Metab 67: 713–719

11. Kelly MA et al. (1997) Pituitary lactotroph hyperplasia and chronic hyperprolactinemia in dopamine D2 receptor-deficient
    mice. Neuron 19: 103–113

12. Kelijman M et al. (1988) Comparison of the sensitivity of growth hormone secretion to somatostatin in vivo and in vitro in
    acromegaly. J Clin Endocrinol Metab 67: 958–963

13. Reubi JC and Landolt AM (1989) The growth hormone responses to octreotide in acromegaly correlate with adenoma
    somatostatin receptor status. J Clin Endocrinol Metab 68: 844–850

14. Ballare E et al. (2001) Mutation of somatostatin receptor type 5 in an acromegalic patient resistant to somatostatin analog
    treatment. J Clin Endocrinol Metab 86: 3809–3814

15. Peillon F et al. (1988) In vitro secretion of somatostatin (SRIH) by human adenomatous somatotropic cells. Relation with
    somatotropic hormone (GH) release and modulation by thyroliberin (TRH) [French]. CR Acad Sci [III] 306: 161–166

16. Joubert D et al. (1989) Normal and growth hormone (GH)-secreting adenomatous human pituitaries release somatostatin
    and GH-releasing hormone. J Clin Endocrinol Metab 68: 572–577

17. Levy L et al. (1993) Presence and characterization of the somatostatin precursor in normal human pituitaries and in growth
    hormone secreting adenomas. J Clin Endocrinol Metab 76: 85–90

18. Filopanti M et al. (2004) Loss of heterozygosity at the SS receptor type 5 locus in human GH- and TSH-secreting pituitary
    adenomas. J Endocrinol Invest 27: 937–942

19. Karl M et al. (1996) Cushing's disease preceded by generalized glucocorticoid resistance: clinical consequences of a novel
    dominant-negative glucocorticoid receptor mutation. Proc Assoc Am Physicians 108: 296–307

20. Karl M et al. (1996) Nelson's syndrome associated with a somatic frame shift mutation in the glucocorticoid receptor gene. J
    Clin Endocrinol Metab 81: 124–129

21. Ray DW et al. (1994) Human small cell lung cancer cell lines expressing the proopiomelanocortin gene have aberrant
    glucocorticoid receptor function. J Clin Invest: 1625–1630

22. Ezzat S et al. (1994) Somatotroph hyperplasia without pituitary adenoma associated with a long standing growth hormone-
    releasing hormone-producing bronchial carcinoid. J Clin Endocrinol Metab 78: 555–560

23. Thapar K et al. (1997) Overexpression of the growth-hormone-releasing hormone gene in acromegaly-associated pituitary
    tumors. An event associated with neoplastic progression and aggressive behavior. Am J Pathol 151: 769–784

24. Lee EJ et al. (2001) Absence of constitutively activating mutations in the GHRH receptor in GH-producing pituitary
    tumors. J Clin Endocrinol Metab: 3989–3995
25. Hashimoto K et al. (1995) Identification of alternatively spliced messenger ribonucleic acid encoding truncated growth
    hormone-releasing hormone receptor in human pituitary adenomas. J Clin Endocrinol Metab 80: 2933–2939

26. Devost D and Boutin JM (1999) Autoregulation of the rat prolactin gene in lactotrophs. Mol Cell Endocrinol : 99–109

27. Schuff KG et al. (2002) Lack of prolactin receptor signaling in mice results in lactotroph proliferation and prolactinomas by
    dopamine-dependent and -independent mechanisms. J Clin Invest 110: 973–981

28. Jin L et al. (1997) Prolactin receptor messenger ribonucleic acid in normal and neoplastic human pituitary tissues. J Clin
    Endocrinol Metab 82: 963–968

29. Asa SL et al. (2000) Evidence for growth hormone (GH) autoregulation in pituitary somatotrophs in GH antagonist-
    transgenic mice and GH receptor-deficient mice. Am J Pathol 156: 1009–1015

30. Yamada M et al. (1993) Pituitary adenomas of patients with acromegaly express thyrotropin-releasing hormone receptor
    messenger RNA cloning and functional expression of the human thyrotropin-releasing hormone receptor gene. Biochem
    Biophys Res Commun 195: 737–745

31. Yamada M et al. (1997) A novel transcript for the thyrotropin-releasing hormone receptor in human pituitary and pituitary
    tumors. J Clin Endocrinol Metab: 4224–4228

32. Ando S et al. (2001) Aberrant alternative splicing of thyroid hormone receptor in a TSH-secreting pituitary tumor is a
    mechanism for hormone resistance. Mol Endocrinol 15: 1529–1538

33. Oomizu S et al. (2000) Transforming growth factor-a stimulates proliferation of mammotrophs and corticotrophs in the
    mouse pituitary. J Endocrinol 165: 493–501

34. McAndrew J et al. (1995) Targeting of transforming growth factor-a expression to pituitary lactotrophs in transgenic mice
    results in selective lactotroph proliferation and adenomas. Endocrinology 136: 4479–4488

35. LeRiche V et al. (1996) Epidermal growth factor and its receptor (EGF-R) in human pituitary adenomas: EGF-R correlates
    with tumor aggressiveness. J Clin Endocrinol Metab 81: 656–662

36. Penabad JL et al. (1996) Decreased follistatin gene expression in gonadotroph adenomas. J Clin Endocrinol Metab 81:
    3397–3403

37. Zhou Y et al. (2000) Truncated activin type I receptor Alk4 isoforms are dominant negative receptors inhibiting activin
    signaling. Mol Endocrinol 14: 2066–2075

38. Gonsky R et al. (1991) Transforming DNA sequences present in human prolactin-secreting pituitary tumors. Mol
    Endocrinol 5: 1687–1695

39. Shimon I et al. (1996) Heparin-binding secretory transforming gene (hst) facilitates rat lactotrope cell tumorigenesis and
    induces prolactin gene transcription. J Clin Invest 97: 187–195

40. Scully KM and Rosenfeld MG (2002) Pituitary development: regulatory codes in mammalian organogenesis. Science 295:
    2231–2235

41. Celli G et al. (1998) Soluble dominant-negative receptor uncovers essential roles for fibroblast growth factors in multi-organ
    induction and patterning. EMBO J 17: 1642–1655

42. Asa SL et al. (2001) The endogenous fibroblast growth factor-2 antisense gene product regulates pituitary cell growth and
    hormone production. Mol Endocrinol 15: 589–599

43. Yu S et al. (2003) Pituitary tumor AP-2a recognizes a cryptic promoter in intron 4 of fibroblast growth factor receptor 4. J
    Biol Chem 278: 19597–19602

44. Ezzat S et al. (2003) Ikaros isoforms in human pituitary tumors: distinct localization, histone acetylation, and activation of
    the 5' fibroblast growth factor receptor-4 promoter. Am J Pathol 163: 1177–1184

45. Vallar L et al. (1987) Altered Gs and adenylate cyclase activity in human GH-secreting pituitary adenomas. Nature 330:
566–568

46. Ezzat S et al. (2002) Targeted expression of a human pituitary tumor-derived isoform of FGF receptor-4 recapitulates
    pituitary tumorigenesis. J Clin Invest 109: 69–78

47. Lyons J et al. (1990) Two G protein oncogenes in human endocrine tumors. Science 249: 655–659

48. Landis CA et al. (1990) Clinical characteristics of acromegalic patients whose pituitary tumors contain mutant Gs protein. J
    Clin Endocrinol Metab 71: 1416–1420

49. Spada A et al. (1990) Clinical, biochemical and morphological correlates in patients bearing growth hormone-secreting
    pituitary tumors with or without constitutively active adenylyl cyclase. J Clin Endocrinol Metab 71: 1421–1426

50. Bertherat J et al. (1995) The cyclic adenosine 3'5'-monophosphate-responsive factor CREB is constitutively activated in
    human somatotroph adenomas. Mol Endocrinol 9: 777–783

51. Harris PE et al. (1992) Glycoprotein hormone a-subunit production in somatotroph adenomas with and without Gsa
    mutations. J Clin Endocrinol Metab 75: 918–923

52. Bhayana S et al. (2005) The implication of somatotroph adenoma phenotype to somatostatin analogue responsiveness in
    acromegaly. J Clin Endocrinol Metab 90: 6290–6295

53. Williamson EA et al. (1994) Gsa and Gi2a mutations in clinically non-functioning pituitary tumours. Clin Endocrinol (Oxf)
    41: 815–820

54. Oyesiku NM et al. (1997) Pituitary adenomas: screening for Gaq mutations. J Clin Endocrinol Metab: 4184–4188 55

55. Bos JL (1989) Ras oncogenes in human cancer: a review. Cancer Res 49: 4682–4689 56

56. Karga HJ et al. (1992) Ras mutations in human pituitary tumors. J Clin Endocrinol Metab 74: 914–919

57. Cai WY et al. (1994) Ras mutations in human prolactinomas and pituitary carcinomas. J Clin Endocrinol Metab 78: 89–93

58. Kirschner LS et al. (2000) Mutations of the gene encoding the protein kinase A type I-a regulatory subunit in patients with
    the Carney complex. Nat Genet 26: 89–92

59. Jacks T et al. (1992) Effects of an Rb mutation in the mouse. Nature 359: 295–300

60. Woloschak M et al. (1997) Frequent inactivation of the p16 gene in human pituitary tumors by gene methylation. Mol
    Carcinog 19: 221–224

61. Asa SL (2001) Transgenic and knockout mouse models clarify pituitary development, function and disease. Brain Pathol 11:
    371–383

62. Liu W et al. (2002) Vitamin D and its analog EB1089 induce p27 accumulation and diminish association of p27 with Skp2
    independent of PTEN in pituitary corticotroph cells. Brain Pathol 12: 412–419

63. Zhang X et al. (2002) Loss of expression of GADD45?, a growth inhibitory gene, in human pituitary adenomas: implications
    for tumorigenesis. J Clin Endocrinol Metab 87: 1262–1267

64. Bahar A et al. (2004) Loss of expression of the growth inhibitory gene GADD45?, in human pituitary adenomas, is
    associated with CpG island methylation. Oncogene 23: 936–944

65. Zhao J et al. (2005) Hypermethylation of the promoter region is associated with the loss of MEG3 gene expression in human
    pituitary tumors. J Clin Endocrinol Metab 90: 2179–2186

66. Bahar A et al. (2004) Isolation and characterization of a novel pituitary tumor apoptosis gene. Mol Endocrinol: 1827–1839

67. Thapar K et al. (1996) p53 expression in pituitary adenomas and carcinomas: correlation with invasiveness and tumor
    growth fractions. Neurosurgery 38: 765–771
68. Amar AP et al. (1999) Invasive pituitary adenomas: significance of proliferation parameters. Pituitary 2: 117–122

 69. Ezzat S et al. (2004) Pituitary tumor-derived fibroblast growth factor receptor 4 isoform disrupts neural cell-adhesion
     molecule/N-cadherin signaling to diminish cell adhesiveness: a mechanism underlying pituitary neoplasia. Mol Endocrinol
     18: 2543–2552

 70. Qian ZR et al. (2002) Role of E-cadherin, a-, ß-, and ?-catenins, and p120 (cell adhesion molecules) in prolactinoma
     behavior. Mod Pathol 15: 1357–1365

 71. Daniel L et al. (2000) Polysialylated-neural cell adhesion molecule expression in rat pituitary transplantable tumors
     (spontaneous mammotropic transplantable tumor in Wistar-Furth rats) is related to growth rate and malignancy. Cancer
     Res 60: 80–85

 72. Heinrich CA et al. (1999) Negative regulation of Ncadherin- mediated cell–cell adhesion by the estrogen receptor signaling
     pathway in rat pituitary GH3 cells. Endocrine 10: 67–76

 73. Riley DJ et al. (1996)Adenovirus-mediated retinoblastoma gene therapy suppresses spontaneous pituitary melanotroph
     tumors in Rb± mice. Nat Med 2: 1316–1321

 74. Freese A et al. (1996) Transfection of human lactotroph adenoma cells with an adenovirus vector expressing tyrosine
     hydroxylase decreases prolactin release. J Clin Endocrinol Metab 81: 2401–2404



Addendum

        A new version of topic of the month publication is uploaded in my web site every month (it remains  for a month and is changed 
   
       with the monthly update of the neurology bulletin at:.http://neurology.yassermetwally.com)

       To download the current version of topic of the month publication follow the link quot;http://neurology.yassermetwally.com/topic.zipquot;
   
       You can also download the current version of topic of the month publication from within the publication or go to my web site at:
   
       quot;http://yassermetwally.comquot; to download it.
       At the end of each year, all the publications are compiled on a single CD-ROM, please author to know more details.
   
       Screen resolution is better set at 1024*768 pixel screen area for optimum display
   
       For an archive of the previously published topics in downloadable PDF format go to http://yassermetwally.net, then under pages in
   
       the right panel, scroll down and click on the text entry quot;topic of the monthquot;
       In order to view a list of the previously published topics in downloadable PDF format, follow the link:
   
       http://wordpress.com/tag/neurological-topic-of-the-month/



The author: Professor Yasser Metwally, professor of neurology, Ain Shams university, Cairo, Egypt

 www.yassermetwally.com

Weitere ähnliche Inhalte

Was ist angesagt?

Principles of endocrinology
Principles of endocrinologyPrinciples of endocrinology
Principles of endocrinologyNidaPraveen
 
Direct Lineage Reprogramming: Novel Factors involved in Lineage Reprogramming
Direct Lineage Reprogramming: Novel Factors involved in Lineage ReprogrammingDirect Lineage Reprogramming: Novel Factors involved in Lineage Reprogramming
Direct Lineage Reprogramming: Novel Factors involved in Lineage ReprogrammingAhmed Madni
 
Epigenetics
Epigenetics Epigenetics
Epigenetics mpattani
 
Epigenetic role in plant
Epigenetic role in plant Epigenetic role in plant
Epigenetic role in plant harshdeep josan
 
Epigenetics: Introduction and Definition and the mechanism
Epigenetics: Introduction and Definition and the mechanismEpigenetics: Introduction and Definition and the mechanism
Epigenetics: Introduction and Definition and the mechanismMohit Adhikary
 
Cancer epigenetics
Cancer epigeneticsCancer epigenetics
Cancer epigeneticsArad Boustan
 
Epidermal growth factor and its receptor tyrosine kinase
Epidermal growth factor and its receptor tyrosine kinaseEpidermal growth factor and its receptor tyrosine kinase
Epidermal growth factor and its receptor tyrosine kinaseGedion Yilma
 
DNA Promoters presentation
DNA Promoters presentation DNA Promoters presentation
DNA Promoters presentation puff7
 
Maternal Nutrition and Epigenetics
Maternal Nutrition and EpigeneticsMaternal Nutrition and Epigenetics
Maternal Nutrition and EpigeneticsGazal Sharma
 
Epigenetics importance in livestock breeding and production
Epigenetics importance in livestock breeding and productionEpigenetics importance in livestock breeding and production
Epigenetics importance in livestock breeding and productionDr Satheesha G M
 
Mechanism of action of hormones
Mechanism of action of hormonesMechanism of action of hormones
Mechanism of action of hormonesDr.M.Prasad Naidu
 
Seminar on gene regulation
Seminar on gene regulationSeminar on gene regulation
Seminar on gene regulationsaarikapk
 

Was ist angesagt? (20)

Cancer epigenetics
Cancer epigenetics Cancer epigenetics
Cancer epigenetics
 
Principles of endocrinology
Principles of endocrinologyPrinciples of endocrinology
Principles of endocrinology
 
Direct Lineage Reprogramming: Novel Factors involved in Lineage Reprogramming
Direct Lineage Reprogramming: Novel Factors involved in Lineage ReprogrammingDirect Lineage Reprogramming: Novel Factors involved in Lineage Reprogramming
Direct Lineage Reprogramming: Novel Factors involved in Lineage Reprogramming
 
Epigenetics
Epigenetics Epigenetics
Epigenetics
 
Epigenetic
EpigeneticEpigenetic
Epigenetic
 
Epigenetic role in plant
Epigenetic role in plant Epigenetic role in plant
Epigenetic role in plant
 
Epigenetics: Introduction and Definition and the mechanism
Epigenetics: Introduction and Definition and the mechanismEpigenetics: Introduction and Definition and the mechanism
Epigenetics: Introduction and Definition and the mechanism
 
epigenetics of kidney disease
epigenetics of kidney diseaseepigenetics of kidney disease
epigenetics of kidney disease
 
Epigenetics
EpigeneticsEpigenetics
Epigenetics
 
Epigenetic
EpigeneticEpigenetic
Epigenetic
 
Cancer epigenetics
Cancer epigeneticsCancer epigenetics
Cancer epigenetics
 
Epidermal growth factor and its receptor tyrosine kinase
Epidermal growth factor and its receptor tyrosine kinaseEpidermal growth factor and its receptor tyrosine kinase
Epidermal growth factor and its receptor tyrosine kinase
 
DNA Promoters presentation
DNA Promoters presentation DNA Promoters presentation
DNA Promoters presentation
 
Epigenetics
EpigeneticsEpigenetics
Epigenetics
 
Maternal Nutrition and Epigenetics
Maternal Nutrition and EpigeneticsMaternal Nutrition and Epigenetics
Maternal Nutrition and Epigenetics
 
Epigenetics importance in livestock breeding and production
Epigenetics importance in livestock breeding and productionEpigenetics importance in livestock breeding and production
Epigenetics importance in livestock breeding and production
 
Epigenetics
EpigeneticsEpigenetics
Epigenetics
 
Mechanism of action of hormones
Mechanism of action of hormonesMechanism of action of hormones
Mechanism of action of hormones
 
Epigenetics
Epigenetics Epigenetics
Epigenetics
 
Seminar on gene regulation
Seminar on gene regulationSeminar on gene regulation
Seminar on gene regulation
 

Andere mochten auch

Thesis section: The role of neuroimaging in muscle and peripheral nerve disor...
Thesis section: The role of neuroimaging in muscle and peripheral nerve disor...Thesis section: The role of neuroimaging in muscle and peripheral nerve disor...
Thesis section: The role of neuroimaging in muscle and peripheral nerve disor...Professor Yasser Metwally
 
Thesis section...Management of hyperacute stroke
Thesis section...Management of hyperacute strokeThesis section...Management of hyperacute stroke
Thesis section...Management of hyperacute strokeProfessor Yasser Metwally
 
Thesis section...Management of hyperacute stroke
Thesis section...Management of hyperacute strokeThesis section...Management of hyperacute stroke
Thesis section...Management of hyperacute strokeProfessor Yasser Metwally
 
Clinical Features and Molecular Genetics of Hereditary Cerebellar Ataxia
Clinical Features and Molecular Genetics of Hereditary Cerebellar AtaxiaClinical Features and Molecular Genetics of Hereditary Cerebellar Ataxia
Clinical Features and Molecular Genetics of Hereditary Cerebellar AtaxiaProfessor Yasser Metwally
 
Talking psychiatry... Neuroimaging of schizophrenic patients treated with ant...
Talking psychiatry... Neuroimaging of schizophrenic patients treated with ant...Talking psychiatry... Neuroimaging of schizophrenic patients treated with ant...
Talking psychiatry... Neuroimaging of schizophrenic patients treated with ant...Professor Yasser Metwally
 
Short case...Multiple craniocervical anomalies
Short case...Multiple craniocervical anomaliesShort case...Multiple craniocervical anomalies
Short case...Multiple craniocervical anomaliesProfessor Yasser Metwally
 
Case record..Multiple sclerosis presented clinically with bilateral trigemina...
Case record..Multiple sclerosis presented clinically with bilateral trigemina...Case record..Multiple sclerosis presented clinically with bilateral trigemina...
Case record..Multiple sclerosis presented clinically with bilateral trigemina...Professor Yasser Metwally
 
Short case...Cerebellopontine angle meningioma
Short case...Cerebellopontine angle meningiomaShort case...Cerebellopontine angle meningioma
Short case...Cerebellopontine angle meningiomaProfessor Yasser Metwally
 
Case record...Spinal dural arteriovenous fistula with congestive myelopathy
Case record...Spinal dural arteriovenous fistula with congestive myelopathyCase record...Spinal dural arteriovenous fistula with congestive myelopathy
Case record...Spinal dural arteriovenous fistula with congestive myelopathyProfessor Yasser Metwally
 
Topic of the month.... The role of gamma knife in the management of brain met...
Topic of the month.... The role of gamma knife in the management of brain met...Topic of the month.... The role of gamma knife in the management of brain met...
Topic of the month.... The role of gamma knife in the management of brain met...Professor Yasser Metwally
 

Andere mochten auch (20)

Thesis section: The role of neuroimaging in muscle and peripheral nerve disor...
Thesis section: The role of neuroimaging in muscle and peripheral nerve disor...Thesis section: The role of neuroimaging in muscle and peripheral nerve disor...
Thesis section: The role of neuroimaging in muscle and peripheral nerve disor...
 
Thesis section...Management of hyperacute stroke
Thesis section...Management of hyperacute strokeThesis section...Management of hyperacute stroke
Thesis section...Management of hyperacute stroke
 
Case record...Glomus jugulare tumor
Case record...Glomus jugulare tumorCase record...Glomus jugulare tumor
Case record...Glomus jugulare tumor
 
Thesis section...Management of hyperacute stroke
Thesis section...Management of hyperacute strokeThesis section...Management of hyperacute stroke
Thesis section...Management of hyperacute stroke
 
Short case...Progressive supranuclear palsy
Short case...Progressive supranuclear palsyShort case...Progressive supranuclear palsy
Short case...Progressive supranuclear palsy
 
Congenital infection
Congenital infectionCongenital infection
Congenital infection
 
Clinical Features and Molecular Genetics of Hereditary Cerebellar Ataxia
Clinical Features and Molecular Genetics of Hereditary Cerebellar AtaxiaClinical Features and Molecular Genetics of Hereditary Cerebellar Ataxia
Clinical Features and Molecular Genetics of Hereditary Cerebellar Ataxia
 
Women with epilepsy
Women with epilepsyWomen with epilepsy
Women with epilepsy
 
Case record...Occipital lobe cavernoma
Case record...Occipital lobe cavernomaCase record...Occipital lobe cavernoma
Case record...Occipital lobe cavernoma
 
Case record...Intramedullary cavernoma
Case record...Intramedullary cavernomaCase record...Intramedullary cavernoma
Case record...Intramedullary cavernoma
 
Talking psychiatry... Neuroimaging of schizophrenic patients treated with ant...
Talking psychiatry... Neuroimaging of schizophrenic patients treated with ant...Talking psychiatry... Neuroimaging of schizophrenic patients treated with ant...
Talking psychiatry... Neuroimaging of schizophrenic patients treated with ant...
 
Short case...Cortical/cerebellar dysplasia
Short case...Cortical/cerebellar dysplasiaShort case...Cortical/cerebellar dysplasia
Short case...Cortical/cerebellar dysplasia
 
Short case...Multiple craniocervical anomalies
Short case...Multiple craniocervical anomaliesShort case...Multiple craniocervical anomalies
Short case...Multiple craniocervical anomalies
 
Drug resistant epilepsy
Drug resistant epilepsyDrug resistant epilepsy
Drug resistant epilepsy
 
Case record..Multiple sclerosis presented clinically with bilateral trigemina...
Case record..Multiple sclerosis presented clinically with bilateral trigemina...Case record..Multiple sclerosis presented clinically with bilateral trigemina...
Case record..Multiple sclerosis presented clinically with bilateral trigemina...
 
Short case...Cerebellopontine angle meningioma
Short case...Cerebellopontine angle meningiomaShort case...Cerebellopontine angle meningioma
Short case...Cerebellopontine angle meningioma
 
Short case...Focal midbrain glioma
Short case...Focal midbrain gliomaShort case...Focal midbrain glioma
Short case...Focal midbrain glioma
 
Case record...Spinal dural arteriovenous fistula with congestive myelopathy
Case record...Spinal dural arteriovenous fistula with congestive myelopathyCase record...Spinal dural arteriovenous fistula with congestive myelopathy
Case record...Spinal dural arteriovenous fistula with congestive myelopathy
 
Topic of the month.... The role of gamma knife in the management of brain met...
Topic of the month.... The role of gamma knife in the management of brain met...Topic of the month.... The role of gamma knife in the management of brain met...
Topic of the month.... The role of gamma knife in the management of brain met...
 
Short case...Rasmussen's encephalitis
Short case...Rasmussen's encephalitisShort case...Rasmussen's encephalitis
Short case...Rasmussen's encephalitis
 

Ähnlich wie Topic of the month.... Pathogenesis of pituitary adenomas

Growth factors and their receptors
Growth factors and their receptorsGrowth factors and their receptors
Growth factors and their receptorsANIL KUMAR
 
Hormones and related diseases.......pptx
Hormones and related diseases.......pptxHormones and related diseases.......pptx
Hormones and related diseases.......pptxAlyaaKaram1
 
Principles of endocrinal disorders
Principles of endocrinal disordersPrinciples of endocrinal disorders
Principles of endocrinal disordersraj kumar
 
Growth Factors in the Human Body: A Conceptual Update_Crimson Publishers
Growth Factors in the Human Body: A Conceptual Update_Crimson PublishersGrowth Factors in the Human Body: A Conceptual Update_Crimson Publishers
Growth Factors in the Human Body: A Conceptual Update_Crimson PublishersCrimsonpublishersCJMI
 
GENE REGULATION IN PROKARYOTES
 GENE REGULATION IN PROKARYOTES GENE REGULATION IN PROKARYOTES
GENE REGULATION IN PROKARYOTESAshvinkumar Katral
 
An introductiontoendocrinology
An introductiontoendocrinologyAn introductiontoendocrinology
An introductiontoendocrinologyAmour Massoud
 
Introduction to endocrine physiology ( mbbs + bds 2nd yr)
Introduction to endocrine physiology ( mbbs + bds 2nd yr)Introduction to endocrine physiology ( mbbs + bds 2nd yr)
Introduction to endocrine physiology ( mbbs + bds 2nd yr)Prakash Yadav
 
Signalling mechanism in cell growth
Signalling mechanism in cell growthSignalling mechanism in cell growth
Signalling mechanism in cell growthbijitadutta123
 
Drugs Used in Disorders of the Endocrine System, Lectures 1 through 6
Drugs Used in Disorders of the Endocrine System, Lectures 1 through 6Drugs Used in Disorders of the Endocrine System, Lectures 1 through 6
Drugs Used in Disorders of the Endocrine System, Lectures 1 through 6Imhotep Virtual Medical School
 
Oncogenes, proto-oncogenes and tumor suppressor gene
Oncogenes, proto-oncogenes and tumor suppressor geneOncogenes, proto-oncogenes and tumor suppressor gene
Oncogenes, proto-oncogenes and tumor suppressor geneVaishnaviJanjal
 
The Role Of G Protein Coupled Receptorss
The Role Of G Protein Coupled ReceptorssThe Role Of G Protein Coupled Receptorss
The Role Of G Protein Coupled ReceptorssAngela Hays
 

Ähnlich wie Topic of the month.... Pathogenesis of pituitary adenomas (20)

Growth factors and their receptors
Growth factors and their receptorsGrowth factors and their receptors
Growth factors and their receptors
 
Tocolytic (Fartoks)
Tocolytic (Fartoks)Tocolytic (Fartoks)
Tocolytic (Fartoks)
 
Hormones and related diseases.......pptx
Hormones and related diseases.......pptxHormones and related diseases.......pptx
Hormones and related diseases.......pptx
 
Reproductive toxicology
Reproductive toxicologyReproductive toxicology
Reproductive toxicology
 
Principles of endocrinal disorders
Principles of endocrinal disordersPrinciples of endocrinal disorders
Principles of endocrinal disorders
 
Growth Factors in the Human Body: A Conceptual Update_Crimson Publishers
Growth Factors in the Human Body: A Conceptual Update_Crimson PublishersGrowth Factors in the Human Body: A Conceptual Update_Crimson Publishers
Growth Factors in the Human Body: A Conceptual Update_Crimson Publishers
 
GENE REGULATION IN PROKARYOTES
 GENE REGULATION IN PROKARYOTES GENE REGULATION IN PROKARYOTES
GENE REGULATION IN PROKARYOTES
 
An introductiontoendocrinology
An introductiontoendocrinologyAn introductiontoendocrinology
An introductiontoendocrinology
 
Hedgehog pathway
Hedgehog pathwayHedgehog pathway
Hedgehog pathway
 
Epigenetics
EpigeneticsEpigenetics
Epigenetics
 
Introduction to endocrine physiology ( mbbs + bds 2nd yr)
Introduction to endocrine physiology ( mbbs + bds 2nd yr)Introduction to endocrine physiology ( mbbs + bds 2nd yr)
Introduction to endocrine physiology ( mbbs + bds 2nd yr)
 
Proto oncogenes
Proto oncogenesProto oncogenes
Proto oncogenes
 
Signalling mechanism in cell growth
Signalling mechanism in cell growthSignalling mechanism in cell growth
Signalling mechanism in cell growth
 
Signaling molecules
Signaling moleculesSignaling molecules
Signaling molecules
 
Cancer
CancerCancer
Cancer
 
Drugs Used in Disorders of the Endocrine System, Lectures 1 through 6
Drugs Used in Disorders of the Endocrine System, Lectures 1 through 6Drugs Used in Disorders of the Endocrine System, Lectures 1 through 6
Drugs Used in Disorders of the Endocrine System, Lectures 1 through 6
 
Oncogenes, proto-oncogenes and tumor suppressor gene
Oncogenes, proto-oncogenes and tumor suppressor geneOncogenes, proto-oncogenes and tumor suppressor gene
Oncogenes, proto-oncogenes and tumor suppressor gene
 
Prostate BPH- pathophysiology
Prostate  BPH- pathophysiologyProstate  BPH- pathophysiology
Prostate BPH- pathophysiology
 
The Role Of G Protein Coupled Receptorss
The Role Of G Protein Coupled ReceptorssThe Role Of G Protein Coupled Receptorss
The Role Of G Protein Coupled Receptorss
 
1-Hormone.pdf
1-Hormone.pdf1-Hormone.pdf
1-Hormone.pdf
 

Mehr von Professor Yasser Metwally

The Snake, the Scorpion, the turtle in Egypt
The Snake, the Scorpion, the turtle in EgyptThe Snake, the Scorpion, the turtle in Egypt
The Snake, the Scorpion, the turtle in EgyptProfessor Yasser Metwally
 
Radiological pathology of epileptic disorders
Radiological pathology of epileptic disordersRadiological pathology of epileptic disorders
Radiological pathology of epileptic disordersProfessor Yasser Metwally
 
Radiological pathology of cerebrovascular disorders
Radiological pathology of cerebrovascular disordersRadiological pathology of cerebrovascular disorders
Radiological pathology of cerebrovascular disordersProfessor Yasser Metwally
 
Radiological pathology of spontaneous cerebral hemorrhage
Radiological pathology of spontaneous cerebral hemorrhageRadiological pathology of spontaneous cerebral hemorrhage
Radiological pathology of spontaneous cerebral hemorrhageProfessor Yasser Metwally
 
Radiological pathology of cerebral amyloid angiography
Radiological pathology of cerebral amyloid angiographyRadiological pathology of cerebral amyloid angiography
Radiological pathology of cerebral amyloid angiographyProfessor Yasser Metwally
 
Radiological pathology of cerebral microbleeds
Radiological pathology of cerebral microbleedsRadiological pathology of cerebral microbleeds
Radiological pathology of cerebral microbleedsProfessor Yasser Metwally
 
Issues in radiological pathology: Radiological pathology of watershed infarct...
Issues in radiological pathology: Radiological pathology of watershed infarct...Issues in radiological pathology: Radiological pathology of watershed infarct...
Issues in radiological pathology: Radiological pathology of watershed infarct...Professor Yasser Metwally
 
Radiological pathology of cortical laminar necrosis
Radiological pathology of cortical laminar necrosisRadiological pathology of cortical laminar necrosis
Radiological pathology of cortical laminar necrosisProfessor Yasser Metwally
 

Mehr von Professor Yasser Metwally (20)

The Egyptian Zoo in Cairo 2015
The Egyptian Zoo in Cairo 2015The Egyptian Zoo in Cairo 2015
The Egyptian Zoo in Cairo 2015
 
End of the great nile river in Ras Elbar
End of the great nile river in Ras ElbarEnd of the great nile river in Ras Elbar
End of the great nile river in Ras Elbar
 
The Lion and The tiger in Egypt
The Lion and The tiger in EgyptThe Lion and The tiger in Egypt
The Lion and The tiger in Egypt
 
The monkeys in Egypt
The monkeys in EgyptThe monkeys in Egypt
The monkeys in Egypt
 
The Snake, the Scorpion, the turtle in Egypt
The Snake, the Scorpion, the turtle in EgyptThe Snake, the Scorpion, the turtle in Egypt
The Snake, the Scorpion, the turtle in Egypt
 
The Egyptian Parrot
The Egyptian ParrotThe Egyptian Parrot
The Egyptian Parrot
 
The Egyptian Deer
The Egyptian DeerThe Egyptian Deer
The Egyptian Deer
 
The Egyptian Pelican
The Egyptian PelicanThe Egyptian Pelican
The Egyptian Pelican
 
The Flamingo bird in Egypt
The Flamingo bird in EgyptThe Flamingo bird in Egypt
The Flamingo bird in Egypt
 
Egyptian Cats
Egyptian CatsEgyptian Cats
Egyptian Cats
 
Radiological pathology of epileptic disorders
Radiological pathology of epileptic disordersRadiological pathology of epileptic disorders
Radiological pathology of epileptic disorders
 
Radiological pathology of cerebrovascular disorders
Radiological pathology of cerebrovascular disordersRadiological pathology of cerebrovascular disorders
Radiological pathology of cerebrovascular disorders
 
Radiological pathology of spontaneous cerebral hemorrhage
Radiological pathology of spontaneous cerebral hemorrhageRadiological pathology of spontaneous cerebral hemorrhage
Radiological pathology of spontaneous cerebral hemorrhage
 
Radiological pathology of cerebral amyloid angiography
Radiological pathology of cerebral amyloid angiographyRadiological pathology of cerebral amyloid angiography
Radiological pathology of cerebral amyloid angiography
 
Radiological pathology of cerebral microbleeds
Radiological pathology of cerebral microbleedsRadiological pathology of cerebral microbleeds
Radiological pathology of cerebral microbleeds
 
The Egyptian Zoo in Cairo
The Egyptian Zoo in CairoThe Egyptian Zoo in Cairo
The Egyptian Zoo in Cairo
 
Progressive multifocal leukoencephalopathy
Progressive multifocal leukoencephalopathyProgressive multifocal leukoencephalopathy
Progressive multifocal leukoencephalopathy
 
Progressive multifocal leukoencephalopathy
Progressive multifocal leukoencephalopathyProgressive multifocal leukoencephalopathy
Progressive multifocal leukoencephalopathy
 
Issues in radiological pathology: Radiological pathology of watershed infarct...
Issues in radiological pathology: Radiological pathology of watershed infarct...Issues in radiological pathology: Radiological pathology of watershed infarct...
Issues in radiological pathology: Radiological pathology of watershed infarct...
 
Radiological pathology of cortical laminar necrosis
Radiological pathology of cortical laminar necrosisRadiological pathology of cortical laminar necrosis
Radiological pathology of cortical laminar necrosis
 

Kürzlich hochgeladen

Partners Life - Insurer Innovation Award 2024
Partners Life - Insurer Innovation Award 2024Partners Life - Insurer Innovation Award 2024
Partners Life - Insurer Innovation Award 2024The Digital Insurer
 
TrustArc Webinar - Unlock the Power of AI-Driven Data Discovery
TrustArc Webinar - Unlock the Power of AI-Driven Data DiscoveryTrustArc Webinar - Unlock the Power of AI-Driven Data Discovery
TrustArc Webinar - Unlock the Power of AI-Driven Data DiscoveryTrustArc
 
Understanding Discord NSFW Servers A Guide for Responsible Users.pdf
Understanding Discord NSFW Servers A Guide for Responsible Users.pdfUnderstanding Discord NSFW Servers A Guide for Responsible Users.pdf
Understanding Discord NSFW Servers A Guide for Responsible Users.pdfUK Journal
 
Strategies for Unlocking Knowledge Management in Microsoft 365 in the Copilot...
Strategies for Unlocking Knowledge Management in Microsoft 365 in the Copilot...Strategies for Unlocking Knowledge Management in Microsoft 365 in the Copilot...
Strategies for Unlocking Knowledge Management in Microsoft 365 in the Copilot...Drew Madelung
 
Automating Google Workspace (GWS) & more with Apps Script
Automating Google Workspace (GWS) & more with Apps ScriptAutomating Google Workspace (GWS) & more with Apps Script
Automating Google Workspace (GWS) & more with Apps Scriptwesley chun
 
2024: Domino Containers - The Next Step. News from the Domino Container commu...
2024: Domino Containers - The Next Step. News from the Domino Container commu...2024: Domino Containers - The Next Step. News from the Domino Container commu...
2024: Domino Containers - The Next Step. News from the Domino Container commu...Martijn de Jong
 
presentation ICT roal in 21st century education
presentation ICT roal in 21st century educationpresentation ICT roal in 21st century education
presentation ICT roal in 21st century educationjfdjdjcjdnsjd
 
Top 5 Benefits OF Using Muvi Live Paywall For Live Streams
Top 5 Benefits OF Using Muvi Live Paywall For Live StreamsTop 5 Benefits OF Using Muvi Live Paywall For Live Streams
Top 5 Benefits OF Using Muvi Live Paywall For Live StreamsRoshan Dwivedi
 
Workshop - Best of Both Worlds_ Combine KG and Vector search for enhanced R...
Workshop - Best of Both Worlds_ Combine  KG and Vector search for  enhanced R...Workshop - Best of Both Worlds_ Combine  KG and Vector search for  enhanced R...
Workshop - Best of Both Worlds_ Combine KG and Vector search for enhanced R...Neo4j
 
Deploy with confidence: VMware Cloud Foundation 5.1 on next gen Dell PowerEdg...
Deploy with confidence: VMware Cloud Foundation 5.1 on next gen Dell PowerEdg...Deploy with confidence: VMware Cloud Foundation 5.1 on next gen Dell PowerEdg...
Deploy with confidence: VMware Cloud Foundation 5.1 on next gen Dell PowerEdg...Principled Technologies
 
Manulife - Insurer Innovation Award 2024
Manulife - Insurer Innovation Award 2024Manulife - Insurer Innovation Award 2024
Manulife - Insurer Innovation Award 2024The Digital Insurer
 
How to Troubleshoot Apps for the Modern Connected Worker
How to Troubleshoot Apps for the Modern Connected WorkerHow to Troubleshoot Apps for the Modern Connected Worker
How to Troubleshoot Apps for the Modern Connected WorkerThousandEyes
 
Strategies for Landing an Oracle DBA Job as a Fresher
Strategies for Landing an Oracle DBA Job as a FresherStrategies for Landing an Oracle DBA Job as a Fresher
Strategies for Landing an Oracle DBA Job as a FresherRemote DBA Services
 
Top 10 Most Downloaded Games on Play Store in 2024
Top 10 Most Downloaded Games on Play Store in 2024Top 10 Most Downloaded Games on Play Store in 2024
Top 10 Most Downloaded Games on Play Store in 2024SynarionITSolutions
 
GenAI Risks & Security Meetup 01052024.pdf
GenAI Risks & Security Meetup 01052024.pdfGenAI Risks & Security Meetup 01052024.pdf
GenAI Risks & Security Meetup 01052024.pdflior mazor
 
Apidays New York 2024 - Scaling API-first by Ian Reasor and Radu Cotescu, Adobe
Apidays New York 2024 - Scaling API-first by Ian Reasor and Radu Cotescu, AdobeApidays New York 2024 - Scaling API-first by Ian Reasor and Radu Cotescu, Adobe
Apidays New York 2024 - Scaling API-first by Ian Reasor and Radu Cotescu, Adobeapidays
 
Exploring the Future Potential of AI-Enabled Smartphone Processors
Exploring the Future Potential of AI-Enabled Smartphone ProcessorsExploring the Future Potential of AI-Enabled Smartphone Processors
Exploring the Future Potential of AI-Enabled Smartphone Processorsdebabhi2
 
Boost PC performance: How more available memory can improve productivity
Boost PC performance: How more available memory can improve productivityBoost PC performance: How more available memory can improve productivity
Boost PC performance: How more available memory can improve productivityPrincipled Technologies
 
Cloud Frontiers: A Deep Dive into Serverless Spatial Data and FME
Cloud Frontiers:  A Deep Dive into Serverless Spatial Data and FMECloud Frontiers:  A Deep Dive into Serverless Spatial Data and FME
Cloud Frontiers: A Deep Dive into Serverless Spatial Data and FMESafe Software
 

Kürzlich hochgeladen (20)

Partners Life - Insurer Innovation Award 2024
Partners Life - Insurer Innovation Award 2024Partners Life - Insurer Innovation Award 2024
Partners Life - Insurer Innovation Award 2024
 
TrustArc Webinar - Unlock the Power of AI-Driven Data Discovery
TrustArc Webinar - Unlock the Power of AI-Driven Data DiscoveryTrustArc Webinar - Unlock the Power of AI-Driven Data Discovery
TrustArc Webinar - Unlock the Power of AI-Driven Data Discovery
 
Understanding Discord NSFW Servers A Guide for Responsible Users.pdf
Understanding Discord NSFW Servers A Guide for Responsible Users.pdfUnderstanding Discord NSFW Servers A Guide for Responsible Users.pdf
Understanding Discord NSFW Servers A Guide for Responsible Users.pdf
 
Strategies for Unlocking Knowledge Management in Microsoft 365 in the Copilot...
Strategies for Unlocking Knowledge Management in Microsoft 365 in the Copilot...Strategies for Unlocking Knowledge Management in Microsoft 365 in the Copilot...
Strategies for Unlocking Knowledge Management in Microsoft 365 in the Copilot...
 
Automating Google Workspace (GWS) & more with Apps Script
Automating Google Workspace (GWS) & more with Apps ScriptAutomating Google Workspace (GWS) & more with Apps Script
Automating Google Workspace (GWS) & more with Apps Script
 
2024: Domino Containers - The Next Step. News from the Domino Container commu...
2024: Domino Containers - The Next Step. News from the Domino Container commu...2024: Domino Containers - The Next Step. News from the Domino Container commu...
2024: Domino Containers - The Next Step. News from the Domino Container commu...
 
presentation ICT roal in 21st century education
presentation ICT roal in 21st century educationpresentation ICT roal in 21st century education
presentation ICT roal in 21st century education
 
Top 5 Benefits OF Using Muvi Live Paywall For Live Streams
Top 5 Benefits OF Using Muvi Live Paywall For Live StreamsTop 5 Benefits OF Using Muvi Live Paywall For Live Streams
Top 5 Benefits OF Using Muvi Live Paywall For Live Streams
 
Workshop - Best of Both Worlds_ Combine KG and Vector search for enhanced R...
Workshop - Best of Both Worlds_ Combine  KG and Vector search for  enhanced R...Workshop - Best of Both Worlds_ Combine  KG and Vector search for  enhanced R...
Workshop - Best of Both Worlds_ Combine KG and Vector search for enhanced R...
 
+971581248768>> SAFE AND ORIGINAL ABORTION PILLS FOR SALE IN DUBAI AND ABUDHA...
+971581248768>> SAFE AND ORIGINAL ABORTION PILLS FOR SALE IN DUBAI AND ABUDHA...+971581248768>> SAFE AND ORIGINAL ABORTION PILLS FOR SALE IN DUBAI AND ABUDHA...
+971581248768>> SAFE AND ORIGINAL ABORTION PILLS FOR SALE IN DUBAI AND ABUDHA...
 
Deploy with confidence: VMware Cloud Foundation 5.1 on next gen Dell PowerEdg...
Deploy with confidence: VMware Cloud Foundation 5.1 on next gen Dell PowerEdg...Deploy with confidence: VMware Cloud Foundation 5.1 on next gen Dell PowerEdg...
Deploy with confidence: VMware Cloud Foundation 5.1 on next gen Dell PowerEdg...
 
Manulife - Insurer Innovation Award 2024
Manulife - Insurer Innovation Award 2024Manulife - Insurer Innovation Award 2024
Manulife - Insurer Innovation Award 2024
 
How to Troubleshoot Apps for the Modern Connected Worker
How to Troubleshoot Apps for the Modern Connected WorkerHow to Troubleshoot Apps for the Modern Connected Worker
How to Troubleshoot Apps for the Modern Connected Worker
 
Strategies for Landing an Oracle DBA Job as a Fresher
Strategies for Landing an Oracle DBA Job as a FresherStrategies for Landing an Oracle DBA Job as a Fresher
Strategies for Landing an Oracle DBA Job as a Fresher
 
Top 10 Most Downloaded Games on Play Store in 2024
Top 10 Most Downloaded Games on Play Store in 2024Top 10 Most Downloaded Games on Play Store in 2024
Top 10 Most Downloaded Games on Play Store in 2024
 
GenAI Risks & Security Meetup 01052024.pdf
GenAI Risks & Security Meetup 01052024.pdfGenAI Risks & Security Meetup 01052024.pdf
GenAI Risks & Security Meetup 01052024.pdf
 
Apidays New York 2024 - Scaling API-first by Ian Reasor and Radu Cotescu, Adobe
Apidays New York 2024 - Scaling API-first by Ian Reasor and Radu Cotescu, AdobeApidays New York 2024 - Scaling API-first by Ian Reasor and Radu Cotescu, Adobe
Apidays New York 2024 - Scaling API-first by Ian Reasor and Radu Cotescu, Adobe
 
Exploring the Future Potential of AI-Enabled Smartphone Processors
Exploring the Future Potential of AI-Enabled Smartphone ProcessorsExploring the Future Potential of AI-Enabled Smartphone Processors
Exploring the Future Potential of AI-Enabled Smartphone Processors
 
Boost PC performance: How more available memory can improve productivity
Boost PC performance: How more available memory can improve productivityBoost PC performance: How more available memory can improve productivity
Boost PC performance: How more available memory can improve productivity
 
Cloud Frontiers: A Deep Dive into Serverless Spatial Data and FME
Cloud Frontiers:  A Deep Dive into Serverless Spatial Data and FMECloud Frontiers:  A Deep Dive into Serverless Spatial Data and FME
Cloud Frontiers: A Deep Dive into Serverless Spatial Data and FME
 

Topic of the month.... Pathogenesis of pituitary adenomas

  • 1. INDEX www.yassermetwally.com INTRODUCTION  INTRODUCTION Pituitary tumors exhibit a spectrum of biology, with variable growth and hormonal behaviors. They therefore provide an opportunity to examine pathogenetic mechanisms that underlie the neoplastic process. These include alterations in hormone regulation, growth-factor stimulation, cell-cycle control and cell-stromal interactions that result from genetic mutations or epigenetic disruption of gene expression. Mouse models have validated the roles of these alterations, which can be targets for the development of therapies that can manage these lesions. These therapies are increasingly recognized as critical for quality of life. The pituitary, a small bean-shaped gland at the base of the brain, maintains multiple homeostatic functions including metabolism, growth and reproduction. Most pituitary tumors are adenomas that develop in the adenohypophysis (the anterior lobe of the pituitary gland), which is composed of six epithelial cell types that secrete peptide hormones. Pituitary adenomas exhibit a wide range of hormonal and proliferative behaviors.[1] They may be small, slowly growing and hormonally in active, and might only be detected as radiographic quot;incidentalomasquot; or at postmortem examination. If, however, they produce hormones in excess, they can cause mood disorders, sexual dysfunction, infertility, obesity and disfigurement, hypertension, diabetes mellitus and accelerated heart disease. If untreated, hormone-excess syndromes can be lethal. Some pituitary adenomas grow rapidly, producing symptoms of an intracranial mass, loss of normal anterior pituitary hormone production, and visual-field disturbances due to stretching of the overlying optic chiasm. They can invade downward into paranasal sinuses, laterally into the cavernous sinuses (thereby disrupting coordinated eye movement) and upwards into the brain. They can cause death by invasion of the brain. Studies using X-chromosome inactivation proved that pituitary adenomas are monoclonal neoplasms.[2] Results that suggest
  • 2. polyclonality are attributed to contamination by normal tissue; the small adenomas associated with Cushing's syndrome are particularly subject to this phenomenon. In contrast to their high frequency of local invasion, only rarely do pituitary tumors metastasize to distant locations in the central nervous system, lymph nodes and liver. This pattern of spread is the criterion of malignancy, allowing a diagnosis of pituitary carcinoma. Many tumors classified as benign are nevertheless associated with significant morbidity and mortality. In this review we will focus on our understanding of the pathogenetic mechanisms underlying the biologic and morphologic features of human pituitary tumors. These include alterations in hormonal regulation and hormone receptors, dysregulated growth factors and alterations in their receptors, abnormalities in signaling proteins that transduce the signals of these stimuli, and changes in cell-cycle regulators (Figure 1). In addition, the neoplastic process is associated with altered cell-stromal interactions that have a role in the morphogenesis of pituitary tumors (Figure 2). These changes are summarized below. Figure 1. Maintaining the delicate balance in pituitary endocrine cell survival. The contribution from some of the characterized factors in anterior pituitary cell growth is shown. Many hormones bind G-protein- coupled receptors, which wind back and forth through the cell membrane seven times and can exert either stimulatory or inhibitory effects. Growth-factor receptors can similarly be stimulatory or inhibitory, and expression levels of, or mutations in, either the receptors or their ligands can affect signaling. The relative contributions of cell-cycle control proteins involved in pituitary cells are becoming better defined. Estradiol, endogenous T3 and glucocorticoids act through dedicated nuclear steroid hormone receptors to mediate negative feedback on respective pituitary stimulatory signals. CRH, corticotropin-releasing hormone; D2R, dopamine receptor 2; E2, estradiol; EGF, epidermal growth factor; FGFs, fibroblast growth factors; GC, glucocortiocoids; GH, growth hormone; GHRH, GH-releasing hormone; Gi, inhibitory G protein; GnRH, gonadotropin-releasing hormone; Gs, stimulatory G protein; PRL, prolactin; ptd-FGFR-4, pituitary-tumor-derived fibroblast growth factor receptor 4; PTTG, pituitary tumor-transforming gene; Ras, intracellular GTP-binding protein that is implicated in cell-signaling pathways; Rb1, retinoblastoma 1; SST, somatostatin; TGF, transforming growth factor; TRH, TSH- releasing hormone. Epidemiology and Classification of Pituitary Adenomas  There are limited data on the prevalence of pituitary tumors. Although classically considered to be rare, systematic assessments from autopsy and radiologic studies have shown a prevalence of 17% of the population.[3] The sex incidence is equal. These tumors are rare in childhood and the incidence increases with age.[1] Prolactin-producing adenomas are the most common type.[1] One-third are unassociated with hypersecretory syndromes; the majority of these produce but do not secrete the gonadotropins follicle-stimulating hormone and/or luteinizing hormone. Growth- hormone (GH)-producing or adrenocorticotropic hormone (ACTH)-producing adenomas each account for 10-15% of pituitary adenomas. Adenomas producing thyrotropin (TSH) are rare. Hormonal activity classifies pituitary adenomas: ACTH-producing adenomas are associated with Cushing's or Nelson's syndromes; GH-producing adenomas are associated with acromegaly and/or gigantism; prolactin-producing adenomas cause hyperprolactinemia; TSH-producing tumors are associated with thyroid dysfunction; there are rare, clinically detectable, gonadotroph adenomas; and there are quot;endocrinologically inactivequot; adenomas. Anatomic and/or radiographic classification is based on tumor size and degree of invasion, information that is critical for patient management and, when surgery is indicated, guides the surgical approach. Immunohistochemistry allows identification of hormone content that usually correlates with the clinical presentation ( Table 1 ). Ultrastructural classification by electron microscopy allows identification of sub cellular variants
  • 3. that now can be characterized using immunohistochemical markers, such as keratins;[1] these variants respond to therapy differently, with variable prognosis and should, therefore, be considered in patient management. Transcription factors that regulate hormone expression are used to classify cytogenesis even in hormone-negative adenomas;[1] tumors derived from different cell lines behave differently, making tumor cytogenesis important for clinical care. It remains to be established whether other characteristics, such as proliferation markers, growth factors and/or their receptors, or oncogene products, will prove to be more reliable predictors of invasive growth, recurrence, or metastasis. Table 1. Pituitary Cells, Hormones, Tumors and Hormone-excess Syndromes Alterations in Hormone Regulation  The development of the adenohypophysis is dependent on hormones for expansion of differentiated cell populations. Adenohypophysiotropic hormones that are produced by the hypothalamus and target organs are involved in complex regulatory feedback mechanisms, which are tightly controlled to maintain homeostasis.[4] Pathology external to the pituitary itself Results in hyperplasia or atrophy of the relevant adenohypophysial cell population.[1,2,4] For this reason, hormonal stimulation or impaired feedback inhibition has been thought to underlie the pathogenesis of pituitary tumors. The data suggest, however, that although hormones may promote cell proliferation, hormonal stimulation is not implicated in the neoplastic transformation that underlies the development of human pituitary tumors. GH-releasing hormone (GHRH) stimulates somatotroph proliferation and causes somatotroph hyperplasia in mice ( Table 2 );[2] in patients with endocrine carcinomas that secrete GHRH ectopically, there is somatotroph hyperplasia but tumors do not develop in this situation.[2] During pregnancy, estrogen causes lactotroph hyperplasia; conversely, antiestrogen activity induces lactotroph apoptosis and also suppresses tumor formation.[5] Some lactotroph adenomas grow during gestation;[6] however, although estrogen was implicated in the pathogenesis of a lactotroph adenoma in male-to-female transsexuals,[7] the clinical use of estrogen in contraceptive agents or in postmenopausal hormone replacement has not been associated with an increased risk of pituitary tumor development.[8] TSH-releasing hormone (TRH) stimulates thyrotrophs and lactotrophs, and patients with primary hypothyroidism develop pituitary thyrotroph and lactotroph hyperplasia.[1] Excess of corticotropin-releasing hormone causes pituitary corticotroph hyperplasia and Cushing's syndrome in humans with ectopic hormone production by tumors, but tumor development has not been reported in this setting.[9]
  • 4. Table 2. Summary of Known Genetic Alterations in Pituitary Tumors Loss of hormonal inhibition might also be involved in hormonal dysregulation. Dopamine maintains tonic inhibition of lactotrophs. In prolactinomas, neovascularization that bypassed the hypothalamus was observed, which implicated the presence of diminished hypo thalamic inhibition, and allowed the tumor cells to escape local inhibition by dopamine.[10] Although dopamine-receptor-deficient mice develop massive lactotroph hyperplasia,[11] no mutations in the DRD2 gene have been identified in human pituitary adenomas and most lactotroph tumors respond to dopamine agonists. Hypothalamic somatostatin inhibits GH secretion, and somatostatin receptors are expressed on somatotroph adenomas.[12,13] A rare, germline, inactivating mutation in somatostatin receptor 5 was reported in a patient with acromegaly,[14] and expression of somatostatin is reported to be lower in invasive, GH-secreting tumors than in normal pituitary tissue;[15-18] however, there is no evidence that loss of somatostatin inhibition is etiologically important in somatotroph adenoma development. GH secretion is also inhibited by insulin-like growth factor 1 (IGF1). Patients with GH-producing adenomas have elevated levels of circulating IGF1 that would be expected to suppress adenoma cells, but no alterations in IGF1 response have been identified that explain tumorigenesis. Patients with long-standing primary hypogonadism develop pituitary gonadotroph adenomas.[1] Similarly, those with primary hypothyroidism develop thyrotroph hyperplasia that can progress to adenoma if untreated.[1] Primary adrenal failure, in the setting of prolonged glucocorticoid deficiency, leads to corticotroph hyperplasia and adenoma,[1] suggesting that loss of glucocorticoid feedback might cause Cushing's disease. A novel germ-line mutation was implicated as the cause of pituitary Cushing's disease in a small number of patients.[19] Similarly, rare glucocorticoid-receptor mutations that diminished glucocorticoid inhibition were found in corticotroph tumors.[20,21] The majority of tumors, however, do not exhibit alterations that would explain dysregulated hormonal inhibition. Old transgenic mice that overexpress GHRH develop pituitary adenomas, and dopamine-receptor-deficient mice ultimately develop invasive lactotroph adenomas,[2] but the delayed neoplasia observed in both settings, as in humans with primary failure of the thyroid, adrenals or gonads, implicates additional genetic events in neoplastic transformation. Indeed, it is distinctly unusual to find hyperplasia underlying the vast majority of sporadic human pituitary adenomas, suggesting that hormonal stimulation is not a primary etiologic mechanism. Sustained exposure to hormonal stimulation does not, moreover, always lead to adenoma formation in humans.[22] Although they might promote proliferation of transformed pituitary cells, it seems that hormones do not cause pituitary tumors. The role of promotion of established tumors by hormones is supported by the finding of exaggerated intrapituitary GHRH expression in aggressive somatotroph adenomas.[23]
  • 5. Alterations of Hormone Receptors  Activating mutations of the GHRH receptor might explain transformation of somatotrophs, but this possibility has been excluded. [24] Instead, a truncated, alternatively spliced form of the receptor with limited signaling properties was identified in GH- producing pituitary adenomas.[25] There are no reports of activating mutations of the corticotropin-releasing hormone receptor in corticotroph adenomas. Recently, the concept of GH and prolactin autoregulation in the pituitary was proven. Prolactin regulates its own secretion at both hypothalamic and pituitary levels.[26] Prolactin-receptor (PRLR)-deficient mice develop earlier lactotroph hyperplasia, and adenomas that are larger and more invasive, than the hyperplasia seen in dopamine D2 receptor-deficient mice,[27] proving the importance of this short autoregulatory loop. Prolactin-producing adenomas have higher levels of PRLR messenger RNA (mRNA) than other types of adenomas. Dopamine-agonist therapy decreases the levels of PRLR mRNA in human prolactin adenomas.[28] There is no evidence, however, of disrupted pro lactin autoregulation in lactotroph adenomas. In contrast to the situation with prolactin, where mouse models indicate autoregulation but there is no human pathology implicating this process, GH has a more subtle autoregulatory role in mice and a pathogenetic significance in humans. Disruption of GH autoregulation in mice Results in subtle somatotroph enlargement and hyperplasia.[29] GH-receptor antagonism in human somatotrophs leads to formation of fibrous bodies–abnormal aggregates of keratin filaments characteristic of a subset of human somatotroph adenomas, the quot;sparsely granulatedquot; variant.[1] Consistent with this finding, somatic mutations of the GH-receptor were found in these tumors (Asa SL et al., unpublished data), implicating disruption of GH autoregulation in the development of this subtype of tumor. The TRH receptor is expressed in thyrotroph adenomas.[30] In some adenomas, the TRH receptor mRNA is alternatively spliced; deletion of exon 3 Results in a truncated product that does not bind its ligand efficiently. Elevated levels of similar truncated products in lactotroph adenomas[31] might explain paradoxical in vivo responses to TRH administration. Thyroid-hormone receptors (TRs) that bind DNA regulatory elements mediate thyroid hormone regulation. TRa mRNA mis- sense mutations, and an alternatively spliced TRß2 variant with a 135-base-pair deletion in the ligand-binding domain have been described in TSH-producing tumors.[32] These data indicate that abnormal hormone receptor signaling can contribute to dysfunctional responses to feedback inhibition; however, apart from one subtype of GH-secreting adenomas, hormone receptor alterations do not seem to be common events that could be implicated in the etiology and pathogenesis of the majority of pituitary adenomas. Dysregulated Growth Factors and Their Receptors  The pituitary is the target for and site of synthesis of several growth factors that modulate hormone production and regulate pituitary cell growth.[2] Some growth factors are critical for normal pituitary development. Alterations in their expression or action might have important roles in the growth of tumors. The epidermal growth factor (EGF) family and its receptors have been implicated in tumorigenesis in a number of neoplasms. The pituitary gland expresses the ligands transforming growth factor (TGF)-a and EGF, which alter pituitary hormone secretion and induce cell proliferation.[2] In fact, TGF-a has been implicated as the mediator of estrogen-induced lactotroph proliferation, since TGF-a antisense expression specifically inhibits this response.[33] Overexpression of TGF-a, under the control of the prolactin promoter, Results in lactotroph adenomas in transgenic mice,[34] indicating a role for this growth factor in pituitary tumorigenesis. The common receptor of EGF and TGF-a, EGFR, is one of a family of four highly homologous receptors possessing tyrosine kinase activity, along with ErbB2/HER2/neu/p185, ErbB3/HER3, and ErbB4/HER4. EGFR expression correlates with pituitary tumor aggressiveness–especially among GH-producing tumors.[35] In contrast to other human neoplasms, however, ERBB2 is not mutated or amplified in human pituitary tumors.[2] The TGF-ß family is represented in the pituitary by inhibin A (a-ßA heterodimers), inhibin B (a-ßB), activin (ßA-ßB), activin A (ßA-ßA) and activin B (ßB-ßB). Inhibin subunits are expressed by pituitary gonadotroph adenomas and activin stimulates hormone secretion by these tumors.[2] The effects of activin are modulated by follistatin and activin receptors. Follistatin binds activins and downregulates their activity. Activin receptors are expressed in gonadotroph adenomas and follistatin expression is reduced or diminished,[36] implicating enhanced activin signaling as a pathogenetic mechanism. Additionally, a truncated activin receptor type-IB isoform that fails to transduce activin-induced growth-arrest signaling was described exclusively in tumors, but not normal human pituitary cells,[37] underscoring the importance of the activin/follistatin balance in modulating pituitary cell growth. The fibroblast growth factors (FGFs) and their receptors (FGFRs) regulate development, growth, and angiogenesis. There are currently 23 known FGF ligands. Basic (b)FGF was originally described in the bovine pituitary folliculostellate cell, and regulates pituitary hormones; bFGF is expressed by human pituitary adenomas and elevated circulating FGF is found in patients with pituitary adenomas.[2] FGF-4 sequences are present in transforming DNA from human pro lactin-secreting tumors,[38] and
  • 6. FGF-4 facilitates lactotroph proliferation.[39] FGF-8 is necessary for pituitary development,[40] and expression of a soluble dominant-negative FGFR Results in pituitary dysgenesis.[41] The product of the endogenous FGF antisense gene (NUDT6) inhibits pituitary cell proliferation.[42] A truncated kinase-containing variant of FGFR-4 with an alternative initiation site[43,44] was isolated from human pituitary tumors.[45] This pituitary-tumor-derived (ptd)-FGFR-4 isoform (ptd-FGFR-4) causes transformation in vitro and in vivo and, when expressed in transgenic mice, recapitulates invasive pituitary adenoma formation. [46] These studies provide convincing evidence for the important role of growth factors, and in particular, the FGF system, in pituitary development and tumor formation. Abnormal Signaling Proteins  Altered signaling by proteins involved in signal transduction of hormones and growth factors might be implicated in pituitary tumorigenesis. G-proteins transduce signals from cell-surface ligand-receptor complexes to downstream effectors. The family of stimulatory G proteins (Gs) transduces GHRH signals. Point mutations in Gsa codon 201 (Arg201Cys) and codon 227 (Gln227Arg) activate adenylyl cyclase by interfering with GTP hydrolysis, Gsa in a constitutively active state.[45] These G-protein oncogenes (GSPs) were first described in a subset of pituitary somatotroph adenomas and hormone-secreting tumors of other endocrine glands. [45,47] GSP mutations are more frequently detected in the maternal allele, which is consistent with monoallelic imprinting of this gene. There is no correlation between GSP mutations and patient age, sex, tumor size, or circulating GH levels;[48] however, this mutation correlates with the densely granulated phenotype of somatotroph tumors,[49] elevated levels of phosphorylated cyclic AMP (cAMP) response element binding protein,[50] and higher circulating levels of free glycoprotein-subunit.[51] These features might be predictive of GH responsiveness to somatostatin-analog therapy.[52] Inactivating mutations in the a-subunit of GIP2, a protein coupled to the inhibitory G-protein Gi2a, have been identified.[53] No mutations have been identified in the stimulatory Gaq or the highly similar Ga11.[54] Ras proteins are also involved in transducing signals from ligand–receptor complexes at the membrane to downstream effectors. Ras mutations are common in nonendocrine malignancies, most frequently involving the GTP-binding domain (codons 12/13) or the GTPase domain (codon 61).[55] Mutation in HRas causing a Gly12Val substitution is distinctly uncommon in pituitary tumors.[56] Initially described in a highly aggressive prolactinoma, Ras mutations are largely restricted to the rare pituitary carcinomas.[57] Protein kinase A is a tetrameric holoenzyme involved in cAMP binding. Adequate levels of the cAMP-dependent protein kinase type 1 regulatory subunit (the product of PRKAR1A) are critical for intracellular protection against unrestrained catalytic activity. Consistent with this model, germline mutations in PRKAR1A are associated with Carney's complex,[58] an autosomal- dominant disorder characterized by development of pituitary and other endocrine tumors. Deregulated Cell Cycle Control Elements  The autosomal-dominant syndrome, multiple endocrine neoplasia type I (MEN1), to which pituitary tumors are integral, is caused by germline mutations of the MEN1 tumor-suppressor gene on chromosome 11q13, with loss of heterozygosity (LOH) inactivating the normal allele in tumors. LOH at this region, observed in 10–30% of sporadic endocrine tumors, suggested that MEN1 could be implicated in sporadic pituitary adenomas. MEN1 is not, however, responsible for sporadic pituitary adenomas, nor is it altered in patients with inherited pituitary tumors that are not associated with the MEN1 syndrome. Reduced expression of menin in sporadic adenomas would indicate that menin functions as a tumor suppressor, but MEN1 mRNA expression is not downregulated in sporadic pituitary tumors.[2] Retinoblastoma 1 (Rb1), a tumor-suppressor gene on chromosome 13q, is associated with pituitary intermediate-lobe tumors in Rb1-deficient mice.[59] Mutations of Rb1 have not been identified in human pituitary adenomas, but the Rb1 promoter is methylated, resulting in gene silencing.[2] There is LOH at13q in tumors with unmethylated Rb1, suggesting an independent tumor-suppressor gene at that locus.[2] Cyclins, cyclin-dependent kinases (CDKs) and CDK inhibitors (CDKIs) regulate cell-cycle progression. CDK activity is modulated by the CDKIs p27kip1, p57kip2, p16ink4A, p15ink4B, p18ink4C, and p19ink4D. Although CDKN2A (which encodes p16ink4A) is not mutated in pituitary adenomas, it is frequently silenced by promoter methylation.[60] The CDKI p27kip1 is important in the pathogenesis and prognosis of several human malignancies. Mice that are Cdkn1b-null do not express p27kip1, and develop multiorgan neoplasia, including pituitary tumors.[61] Although the CDKN1B gene is not mutated in human pituitary tumors, expression of its product p27kip1 is reduced in corticotroph adenomas and recurrent human pituitary adenomas.[2] Protein stability is critical in maintaining p27 protein expression, suggesting epigenetic mechanisms of p27 alteration that can be targeted for therapy.[62] Mice lacking both p18ink4C and p27kip1 succumb to rapidly lethal pituitary adenomas;[61] however, there are no reports of p18 alterations in human pituitary tumors.
  • 7. The growth ? arrest and DNA-damage-inducible gene GADD45G negatively regulates cell growth. Expression of GADD45G mRNA is significantly diminished in clinically nonfunctioning and hormonally active GH-secreting or prolactin-secreting pituitary tumors,[63] because of gene silencing through CpG island promoter methylation.[64] Maternally expressed gene 3 (MEG3) is a human homolog of the mouse MATERNALLY IMPRINTED Gtl2 gene. The MEG3a isoform, containing an extended exon 5 sequence, inhibits cell growth in vitro. As with GADD45?, MEG3a expression is diminished in human pituitary tumors associated with 5' promoter hypermethylation.[65] A number of other cell-cycle regulators have received attention as candidate causes of pituitary tumors. Pituitary tumor transforming gene 1 (PTTG1) is a member of the securin family that is involved in sister chromatid separation. As expected of a gene with this function it is upregulated in proliferating cells.[2] Pituitary tumor apoptosis gene (PTAG; this name has not yet been approved) is a novel antiapoptotic gene on chromosome 22 identified in pituitary tumors but with no known significance.[66] The tumor suppressor p53, which is mutated or lost in many other forms of cancer, is expressed in pituitary tumors; however, there is no evidence of TP53 gene mutation or LOH in pituitary tumors.[2] Upregulated expression of this gene probably represents a compensatory mechanism, so that there might be some prognostic value of p53 staining;[67] however, this is controversial as there is no consistent correlation between p53 immunoreactivity and tumor invasion or recurrence.[68] Dysadhesion and Abnormal Stromal Interactions  Progression to neoplasia involves changes in cell-cell and cell-matrix adhesion. Indeed, loss of the reticulin network is the morphological hallmark of the transition from hyperplasia to adenoma that is commonly used as a diagnostic feature in the pituitary.[1] Changes in cell adhesion have been correlated with the abnormal expression of ptd-FGFR-4 in cell lines, transgenic mice and human pituitary adenomas that result in loss of affinity for extracellular matrix.[69] This loss is due to disruption of a multiprotein complex involving FGFR-4, N-cadherin (a cell adhesion molecule) and neural cell adhesion molecule 1 (NCAM-1). Expression of ptd-FGFR-4 Results in diminished and ectopic cytoplasmic expression of N-cadherin, associated with invasive growth.[69] Uncoupling of N-cadherin from the cytoskeleton destabilizes ß-catenin,[69] and reduced ß-catenin expression in human pituitary adenomas correlates with tumor invasiveness.[70] Similarly, expression of the polysialated form of NCAM-1 correlates with tumor growth and invasiveness;[71] polysialation Results in steric inhibition of membrane-membrane apposition and cell adhesiveness and, therefore, probably disrupts the multiprotein complex as does ptd-FGFR-4 (Figure 2). Estrogen, a known stimulus of pituitary cell proliferation, reduces membranous N-cadherin protein expression to barely detectable levels, whereas antiestrogen treatment reverses this effect.[72] Figure 2. The balance between adhesive and dysadhesive forces in pituitary cells. In this model, the membrane location of neural cell adhesion molecule 1, fibroblast growth factor receptor 4 and N-cadherin is critical for the formation of a proadhesive multiprotein complex. As shown on the right, wild-type fibroblast growth factor receptor 4 resides predominantly in the cell membrane and interacts heavily with neural cell adhesion molecule 1 to support stability of N-cadherin and ß-catenin and
  • 8. maintain actin cytoskeletal structure. By contrast, as shown on the left, receptor isoforms such as pituitary-tumorderived fibroblast growth factor receptor 4 are located in the cytoplasm, and can displace fibroblast growth factor receptor 4 from neural cell adhesion molecule 1, thereby disrupting cell-cell and cell-matrix adhesion, which are mediated by N-cadherin and ß-catenin. A similar form of steric hindrance with this multiprotein proadhesive complex can be achieved by polysialated neural cell adhesion molecule 1. FGFR-4, fibroblast growth factor receptor 4; NCAM-1; neural cell adhesion molecule 1; PSA, polysialated; ptd-FGFR-4, pituitary-tumor-derived fibroblast growth factor receptor 4. Implications For Therapy  The treatment of pituitary tumors involves medical, surgical, and radiotherapeutic approaches. Pharmacotherapy represents the cornerstone for treatment of several hormonally active adenomas. Prolactin-secreting tumors are generally treated with dopamine agonists. Somatostatin analogs are widely adopted to restrain hormone secretion by GH-producing pituitary tumors; the varying response to somatostatin analogs is now understood to be reflective of the differing pathogenetic mechanisms underlying the various morphologic tumor types that cause acromegaly. Some of the currently available agents suppress hormone excess, some result in reduction of tumor size, but they do not achieve a tumoricidal effect. Comparison of somatostatin-sensitive and somatostatin-resistant tumors, by use of genomic and proteomic approaches, will undoubtedly pave the way for the development of effective tumoricidal drug treatments; moreover, dopamine and somatostatin analogs are not effective in the treatment of tumors arising from gonadotrophs or corticotrophs, creating a need for more-effective agents. Since Rb1 inactivation underlies corticotroph tumor development in mice, this provides a model for the evaluation of gene therapy approaches in the management of pituitary tumors. Spontaneous pituitary tumors in immunocompetent Rb1-deficient mice were treated with a recombinant adenovirus carrying Rb1-complementary DNA. Intratumoral Rb1 gene transfer decreased tumor cell proliferation, re-established innervation by growth-regulatory dopaminergic neurons, and resulted in inhibition of tumor growth and enhanced animal survival,[73] providing proof of principle. Deficient or defective delivery of dopamine has been implicated in lactotroph adenomas. This system has been investigated by transfection of human lactotroph adenomas in vitro with an adenoviral vector that expresses tyrosine hydroxylase. Introduction of this rate-limiting enzyme in dopamine biosynthesis enhanced dopamine synthesis and diminished prolactin secretion.[74] Similarly, the pro-opiomelanocortin promoter has also been used to allow selective expression of the toxic herpes simplex virus thymidine kinase, in mouse pituitary corticotroph cell lines. Infection in vitro or injection in vivo results in selective gene expression and cytotoxicity.[5] This hormone promoter might, therefore, be used in selective targeting of gene therapy vectors to tumors associated with excessive ACTH production and Cushing's disease. These studies provide evidence for the feasibility of gene therapy and the use of pituitary-specific promoters to confer selectivity in treating pituitary tumors. Corticotroph adenomas are characterized by loss of the tumor suppressor p27, as a result of protein degradation.[62] The actions of vitamin D3 in restoring p27 protein levels[62] could have therapeutic significance for these tumors, which as yet have no other targeted medical therapies. Also close to the clinical horizon is the application of selective tyrosine kinase inhibitors. These agents should be tested in therapeutic trials in defined patient populations–especially those in whom surgical or medical therapies have failed. Selective inhibitors of the FGFR and EGFR pathways might be used to treat pituitary tumors effectively. Tumor-associated EGFR and FGFR could, moreover, be targeted, by delivering toxins or antiproliferative monoclonal antibodies to tumor cells. Conclusions  The individual contributions of the components involved in the complex cascade of events leading to human neoplasia are finally unraveling. The delicate balance maintained by the dominant factors in the pituitary is summarized in Figures 1 and 2. It is important to emphasize that it is becoming increasingly evident, in many systems, that no single factor can effectively explain the many facets of the tumorigenic process. It is nevertheless anticipated that specific interruption of several signaling cascades will afford new approaches to the therapeutic armamentarium, permitting more-effective strategies in the management of pituitary tumors. References 1. Asa SL (1998) Tumors of the Pituitary Gland. In Atlas of Tumor Pathology, Series 3, Fascicle 22. Washington, DC: Armed Forces Institute of Pathology 2. Asa SL and Ezzat S (2002) The pathogenesis of pituitary tumours. Nat Rev Cancer 2: 836–849
  • 9. 3. Ezzat S et al. (2004) The prevalence of pituitary adenomas: a systematic review. Cancer 101: 613–619 4. Asa SL (1991) The role of hypothalamic hormones in the pathogenesis of pituitary adenomas. Pathol Res Pract 187: 581–583 5. Lee EJ et al. (2001) Adenovirus-directed expression of dominant negative estrogen receptor induces apoptosis in breast cancer cells and regression of tumors in nude mice. Mol Med 7: 773–782 6. Molitch ME (2003) Pituitary tumors and pregnancy. Growth Horm IGF Res 13 (Suppl A): S38–S44 7. Kovacs K et al. (1994) Prolactin-producing pituitary adenoma in a male-to-female transsexual patient with protracted estrogen administration. A morphologic study. Arch Pathol Lab Med 118: 562–565 8. Testa G et al. (1998) Two-year treatment with oral contraceptives in hyperprolactinemic patients. Contraception 58: 69–73 9. Carey RM et al. (1984) Ectopic secretion of corticotropin-releasing factor as a cause of Cushing's syndrome. A clinical, morphologic, and biochemical study. N Engl J Med 311: 13–20 10. Schechter J et al. (1988) Morphological evidence for the presence of arteries in human prolactinomas. J Clin Endocrinol Metab 67: 713–719 11. Kelly MA et al. (1997) Pituitary lactotroph hyperplasia and chronic hyperprolactinemia in dopamine D2 receptor-deficient mice. Neuron 19: 103–113 12. Kelijman M et al. (1988) Comparison of the sensitivity of growth hormone secretion to somatostatin in vivo and in vitro in acromegaly. J Clin Endocrinol Metab 67: 958–963 13. Reubi JC and Landolt AM (1989) The growth hormone responses to octreotide in acromegaly correlate with adenoma somatostatin receptor status. J Clin Endocrinol Metab 68: 844–850 14. Ballare E et al. (2001) Mutation of somatostatin receptor type 5 in an acromegalic patient resistant to somatostatin analog treatment. J Clin Endocrinol Metab 86: 3809–3814 15. Peillon F et al. (1988) In vitro secretion of somatostatin (SRIH) by human adenomatous somatotropic cells. Relation with somatotropic hormone (GH) release and modulation by thyroliberin (TRH) [French]. CR Acad Sci [III] 306: 161–166 16. Joubert D et al. (1989) Normal and growth hormone (GH)-secreting adenomatous human pituitaries release somatostatin and GH-releasing hormone. J Clin Endocrinol Metab 68: 572–577 17. Levy L et al. (1993) Presence and characterization of the somatostatin precursor in normal human pituitaries and in growth hormone secreting adenomas. J Clin Endocrinol Metab 76: 85–90 18. Filopanti M et al. (2004) Loss of heterozygosity at the SS receptor type 5 locus in human GH- and TSH-secreting pituitary adenomas. J Endocrinol Invest 27: 937–942 19. Karl M et al. (1996) Cushing's disease preceded by generalized glucocorticoid resistance: clinical consequences of a novel dominant-negative glucocorticoid receptor mutation. Proc Assoc Am Physicians 108: 296–307 20. Karl M et al. (1996) Nelson's syndrome associated with a somatic frame shift mutation in the glucocorticoid receptor gene. J Clin Endocrinol Metab 81: 124–129 21. Ray DW et al. (1994) Human small cell lung cancer cell lines expressing the proopiomelanocortin gene have aberrant glucocorticoid receptor function. J Clin Invest: 1625–1630 22. Ezzat S et al. (1994) Somatotroph hyperplasia without pituitary adenoma associated with a long standing growth hormone- releasing hormone-producing bronchial carcinoid. J Clin Endocrinol Metab 78: 555–560 23. Thapar K et al. (1997) Overexpression of the growth-hormone-releasing hormone gene in acromegaly-associated pituitary tumors. An event associated with neoplastic progression and aggressive behavior. Am J Pathol 151: 769–784 24. Lee EJ et al. (2001) Absence of constitutively activating mutations in the GHRH receptor in GH-producing pituitary tumors. J Clin Endocrinol Metab: 3989–3995
  • 10. 25. Hashimoto K et al. (1995) Identification of alternatively spliced messenger ribonucleic acid encoding truncated growth hormone-releasing hormone receptor in human pituitary adenomas. J Clin Endocrinol Metab 80: 2933–2939 26. Devost D and Boutin JM (1999) Autoregulation of the rat prolactin gene in lactotrophs. Mol Cell Endocrinol : 99–109 27. Schuff KG et al. (2002) Lack of prolactin receptor signaling in mice results in lactotroph proliferation and prolactinomas by dopamine-dependent and -independent mechanisms. J Clin Invest 110: 973–981 28. Jin L et al. (1997) Prolactin receptor messenger ribonucleic acid in normal and neoplastic human pituitary tissues. J Clin Endocrinol Metab 82: 963–968 29. Asa SL et al. (2000) Evidence for growth hormone (GH) autoregulation in pituitary somatotrophs in GH antagonist- transgenic mice and GH receptor-deficient mice. Am J Pathol 156: 1009–1015 30. Yamada M et al. (1993) Pituitary adenomas of patients with acromegaly express thyrotropin-releasing hormone receptor messenger RNA cloning and functional expression of the human thyrotropin-releasing hormone receptor gene. Biochem Biophys Res Commun 195: 737–745 31. Yamada M et al. (1997) A novel transcript for the thyrotropin-releasing hormone receptor in human pituitary and pituitary tumors. J Clin Endocrinol Metab: 4224–4228 32. Ando S et al. (2001) Aberrant alternative splicing of thyroid hormone receptor in a TSH-secreting pituitary tumor is a mechanism for hormone resistance. Mol Endocrinol 15: 1529–1538 33. Oomizu S et al. (2000) Transforming growth factor-a stimulates proliferation of mammotrophs and corticotrophs in the mouse pituitary. J Endocrinol 165: 493–501 34. McAndrew J et al. (1995) Targeting of transforming growth factor-a expression to pituitary lactotrophs in transgenic mice results in selective lactotroph proliferation and adenomas. Endocrinology 136: 4479–4488 35. LeRiche V et al. (1996) Epidermal growth factor and its receptor (EGF-R) in human pituitary adenomas: EGF-R correlates with tumor aggressiveness. J Clin Endocrinol Metab 81: 656–662 36. Penabad JL et al. (1996) Decreased follistatin gene expression in gonadotroph adenomas. J Clin Endocrinol Metab 81: 3397–3403 37. Zhou Y et al. (2000) Truncated activin type I receptor Alk4 isoforms are dominant negative receptors inhibiting activin signaling. Mol Endocrinol 14: 2066–2075 38. Gonsky R et al. (1991) Transforming DNA sequences present in human prolactin-secreting pituitary tumors. Mol Endocrinol 5: 1687–1695 39. Shimon I et al. (1996) Heparin-binding secretory transforming gene (hst) facilitates rat lactotrope cell tumorigenesis and induces prolactin gene transcription. J Clin Invest 97: 187–195 40. Scully KM and Rosenfeld MG (2002) Pituitary development: regulatory codes in mammalian organogenesis. Science 295: 2231–2235 41. Celli G et al. (1998) Soluble dominant-negative receptor uncovers essential roles for fibroblast growth factors in multi-organ induction and patterning. EMBO J 17: 1642–1655 42. Asa SL et al. (2001) The endogenous fibroblast growth factor-2 antisense gene product regulates pituitary cell growth and hormone production. Mol Endocrinol 15: 589–599 43. Yu S et al. (2003) Pituitary tumor AP-2a recognizes a cryptic promoter in intron 4 of fibroblast growth factor receptor 4. J Biol Chem 278: 19597–19602 44. Ezzat S et al. (2003) Ikaros isoforms in human pituitary tumors: distinct localization, histone acetylation, and activation of the 5' fibroblast growth factor receptor-4 promoter. Am J Pathol 163: 1177–1184 45. Vallar L et al. (1987) Altered Gs and adenylate cyclase activity in human GH-secreting pituitary adenomas. Nature 330:
  • 11. 566–568 46. Ezzat S et al. (2002) Targeted expression of a human pituitary tumor-derived isoform of FGF receptor-4 recapitulates pituitary tumorigenesis. J Clin Invest 109: 69–78 47. Lyons J et al. (1990) Two G protein oncogenes in human endocrine tumors. Science 249: 655–659 48. Landis CA et al. (1990) Clinical characteristics of acromegalic patients whose pituitary tumors contain mutant Gs protein. J Clin Endocrinol Metab 71: 1416–1420 49. Spada A et al. (1990) Clinical, biochemical and morphological correlates in patients bearing growth hormone-secreting pituitary tumors with or without constitutively active adenylyl cyclase. J Clin Endocrinol Metab 71: 1421–1426 50. Bertherat J et al. (1995) The cyclic adenosine 3'5'-monophosphate-responsive factor CREB is constitutively activated in human somatotroph adenomas. Mol Endocrinol 9: 777–783 51. Harris PE et al. (1992) Glycoprotein hormone a-subunit production in somatotroph adenomas with and without Gsa mutations. J Clin Endocrinol Metab 75: 918–923 52. Bhayana S et al. (2005) The implication of somatotroph adenoma phenotype to somatostatin analogue responsiveness in acromegaly. J Clin Endocrinol Metab 90: 6290–6295 53. Williamson EA et al. (1994) Gsa and Gi2a mutations in clinically non-functioning pituitary tumours. Clin Endocrinol (Oxf) 41: 815–820 54. Oyesiku NM et al. (1997) Pituitary adenomas: screening for Gaq mutations. J Clin Endocrinol Metab: 4184–4188 55 55. Bos JL (1989) Ras oncogenes in human cancer: a review. Cancer Res 49: 4682–4689 56 56. Karga HJ et al. (1992) Ras mutations in human pituitary tumors. J Clin Endocrinol Metab 74: 914–919 57. Cai WY et al. (1994) Ras mutations in human prolactinomas and pituitary carcinomas. J Clin Endocrinol Metab 78: 89–93 58. Kirschner LS et al. (2000) Mutations of the gene encoding the protein kinase A type I-a regulatory subunit in patients with the Carney complex. Nat Genet 26: 89–92 59. Jacks T et al. (1992) Effects of an Rb mutation in the mouse. Nature 359: 295–300 60. Woloschak M et al. (1997) Frequent inactivation of the p16 gene in human pituitary tumors by gene methylation. Mol Carcinog 19: 221–224 61. Asa SL (2001) Transgenic and knockout mouse models clarify pituitary development, function and disease. Brain Pathol 11: 371–383 62. Liu W et al. (2002) Vitamin D and its analog EB1089 induce p27 accumulation and diminish association of p27 with Skp2 independent of PTEN in pituitary corticotroph cells. Brain Pathol 12: 412–419 63. Zhang X et al. (2002) Loss of expression of GADD45?, a growth inhibitory gene, in human pituitary adenomas: implications for tumorigenesis. J Clin Endocrinol Metab 87: 1262–1267 64. Bahar A et al. (2004) Loss of expression of the growth inhibitory gene GADD45?, in human pituitary adenomas, is associated with CpG island methylation. Oncogene 23: 936–944 65. Zhao J et al. (2005) Hypermethylation of the promoter region is associated with the loss of MEG3 gene expression in human pituitary tumors. J Clin Endocrinol Metab 90: 2179–2186 66. Bahar A et al. (2004) Isolation and characterization of a novel pituitary tumor apoptosis gene. Mol Endocrinol: 1827–1839 67. Thapar K et al. (1996) p53 expression in pituitary adenomas and carcinomas: correlation with invasiveness and tumor growth fractions. Neurosurgery 38: 765–771
  • 12. 68. Amar AP et al. (1999) Invasive pituitary adenomas: significance of proliferation parameters. Pituitary 2: 117–122 69. Ezzat S et al. (2004) Pituitary tumor-derived fibroblast growth factor receptor 4 isoform disrupts neural cell-adhesion molecule/N-cadherin signaling to diminish cell adhesiveness: a mechanism underlying pituitary neoplasia. Mol Endocrinol 18: 2543–2552 70. Qian ZR et al. (2002) Role of E-cadherin, a-, ß-, and ?-catenins, and p120 (cell adhesion molecules) in prolactinoma behavior. Mod Pathol 15: 1357–1365 71. Daniel L et al. (2000) Polysialylated-neural cell adhesion molecule expression in rat pituitary transplantable tumors (spontaneous mammotropic transplantable tumor in Wistar-Furth rats) is related to growth rate and malignancy. Cancer Res 60: 80–85 72. Heinrich CA et al. (1999) Negative regulation of Ncadherin- mediated cell–cell adhesion by the estrogen receptor signaling pathway in rat pituitary GH3 cells. Endocrine 10: 67–76 73. Riley DJ et al. (1996)Adenovirus-mediated retinoblastoma gene therapy suppresses spontaneous pituitary melanotroph tumors in Rb± mice. Nat Med 2: 1316–1321 74. Freese A et al. (1996) Transfection of human lactotroph adenoma cells with an adenovirus vector expressing tyrosine hydroxylase decreases prolactin release. J Clin Endocrinol Metab 81: 2401–2404 Addendum  A new version of topic of the month publication is uploaded in my web site every month (it remains  for a month and is changed   with the monthly update of the neurology bulletin at:.http://neurology.yassermetwally.com) To download the current version of topic of the month publication follow the link quot;http://neurology.yassermetwally.com/topic.zipquot;  You can also download the current version of topic of the month publication from within the publication or go to my web site at:  quot;http://yassermetwally.comquot; to download it. At the end of each year, all the publications are compiled on a single CD-ROM, please author to know more details.  Screen resolution is better set at 1024*768 pixel screen area for optimum display  For an archive of the previously published topics in downloadable PDF format go to http://yassermetwally.net, then under pages in  the right panel, scroll down and click on the text entry quot;topic of the monthquot; In order to view a list of the previously published topics in downloadable PDF format, follow the link:  http://wordpress.com/tag/neurological-topic-of-the-month/ The author: Professor Yasser Metwally, professor of neurology, Ain Shams university, Cairo, Egypt  www.yassermetwally.com