ArticlePDF AvailableLiterature Review

Genomics, Transcriptomics, and Epigenetics of Sporadic Pituitary Tumors

Authors:
  • Unidad de Alta Especialidad Hospital de Especialidades-Centro Medico Nacional Siglo XXI
Archives of Medical Research 54 (2023) 102915
REVIEW ARTICLE
Genomics, Transcriptomics, and Epigenetics of Sporadic Pituitary Tumors
Daniel Marrero-Rodríguez, Sandra Vela-Patiño, Florencia Martinez-Mendoza,
Alejandra Valenzuela-Perez, Eduardo Peña-Martínez, Amayrani Cano-Zaragoza, Jacobo Kerbel,
Sergio Andonegui-Elguera, Shimon S. Glick-Betech, Karla X. Hermoso-Mier,
Sophia Mercado-Medrez, Alberto Moscona-Nissan, Keiko Taniguchi-Ponciano, and Moises Mercado
Endocrine Research Unit, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
Received for publication July 30, 2023; accepted November 7, 2023 (ARCMED-D-23-00617).
Pituitary tumors (PT) are highly heterogeneous neoplasms, comprising functioning and
nonfunctioning lesions. Functioning PT include prolactinomas, causing amenorrhea-
galactorrhea in women and sexual dysfunction in men; GH-secreting adenomas caus-
ing acromegaly-gigantism; ACTH-secreting corticotrophinomas causing Cushing disease
(CD); and the rare TSH-secreting thyrotrophinomas that result in central hyperthy-
roidism. Nonfunctioning PT do not result in a hormonal hypersecretion syndrome and
most of them are of gonadotrope differentiation; other non-functioning PT include null
cell adenomas and silent ACTH-, GH- and PRL-adenomas. Less than 5% of PT occur in
a familial or syndromic context whereby germline mutations of specific genes account
for their molecular pathogenesis. In contrast, the more common sporadic PT do not
result from a single molecular abnormality but rather emerge from several oncogenic
events that culminate in an increased proliferation of pituitary cells, and in the case of
functioning tumors, in a non-regulated hormonal hypersecretion. In recent years, im-
portant advances in the understanding of the molecular pathogenesis of PT have been
made, including the genomic, transcriptomic, epigenetic, and proteomic characterization
of these neoplasms. In this review, we summarize the available molecular informa-
tion pertaining the oncogenesis of PT. © 2023 Instituto Mexicano del Seguro Social
(IMSS). Published by Elsevier Inc. All rights reserved.
Key Wo rd s : Genomics, Transcriptomics, Methylome, Pituitary tumors, scRNAseq.
Introduction
Traditionally, medical therapeutics are based on the patho-
physiology and clinical features of a particular disease.
Although the individualization of treatment based on clin-
ical judgment is obviously a hallmark of good medical
practice, the design and development of a particular drug
is seldom aimed at the individual patient. The emergence
of ever more powerful molecular biology techniques over
the past 30 years has led to unprecedented advances in
the understanding of the etiology and pathophysiology of
a growing number of diseases resulting in efficient, indi-
vidually tailored targeted therapies and precision medicine
Address reprint requests to: Keiko Taniguchi-Ponciano, Unidad de
Investigación Médica en Enfermedades Endocrinas, Hospital de Espe-
cialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del
Seguro Social, Av. Cuauhtémoc 330, Col. Doctores, 06720, Mexico City,
Mexico; E-mail: keiko.taniguchi@hotmail.com
( 1 ). Precision medicine is currently being used to diagnose
and treat a wide variety of conditions, including metabolic,
neurological, immune-mediated, and neoplastic disorders,
among others.
Pituitary tumors (PT) constitute a heterogeneous group
of epithelial neoplasms that include both functioning
and nonfunctioning lesions ( 2 ). Functioning PT in-
clude prolactin (PRL) -secreting prolactinomas, causing
amenorrhea-galactorrhea in women and sexual dysfunction
in men; growth hormone (GH) -secreting adenomas caus-
ing acromegaly-gigantism; adrenocorticotropic hormone
(ACTH) -secreting corticotrophinomas that cause Cush-
ing disease (CD); and the rare TSH (thyrotropin)-secreting
thyrotrophinomas that result in central hyperthyroidism
( 2 ). Besides immunostaining for their cognate hormones,
ACTH-secreting adenomas immunostain for the T-box
transcription factor (TBX19) also known as T-Pit, whereas
GH-, PRL- and TSH-secreting tumors do so for POU
0188-4409/$ - see front matter. Copyright © 2023 Instituto Mexicano del Seguro Social (IMSS). Published by Elsevier Inc. All rights reserved.
https://doi.org/10.1016/j.arcmed.2023.102915
2 Marrero-Rodríguez et al. / Archives of Medical Research 54 (2023) 102915
class 1 homeobox 1 (POU1F1), also known as Pit-1 ( 2 ).
Nonfunctioning PT do not result in a hormonal hyper-
secretion syndrome and most of them are of gonadotrope
differentiation, as they immunostain for the common α-
subunit, luteinizing hormone ( β-LH) or follicle-stimulating
hormone ( β-FSH), as well as for the transcription factor
nuclear receptor subfamily 5 group A member 1 (NR5A1)
also known as steroidogenic factor 1, or SF1 . Other non-
functioning PTs include null cell adenomas and silent
ACTH-, GH-, and PRL-adenomas ( 3 ). Less than 5% of PT
occur in a familial or syndromic context, where germline
mutations of specific genes are responsible for their molec-
ular pathogenesis ( 4–6 ). In contrast, the more common
sporadic PTs do not result from a single molecular ab-
normality, but rather from multiple oncogenic events that
culminate in an increased pituitary cell proliferation and,
in the case of functioning tumors, in a non-regulated hor-
monal hypersecretion ( 2 ). In a way, before the terms “pre-
cision” or “personalized” medicine were coined, pituitary
specialists had been approaching patients with PT in an in-
dividualized manner by immunophenotyping these lesions
according to their expression of hormones, transcription
factors and more recently, somatostatin and dopamine re-
ceptors. Although the immunohistochemical evaluation of
PT has allowed us to better classify and diagnose these
lesions and has contributed to more rational therapeutic
decisions, it remains a methodology that is limited by the
subjective interpretation of a pathologist ( 7 ). Over the past
10 years, there have been important advances in the un-
derstanding of the genomics, transcriptomics, proteomics,
and epigenetics of PT. In this review, we summarize and
critically analyze the molecular landscapes of sporadic
PT, focusing on those findings that are likely to con-
tribute to a better diagnosis and treatment of these patients
( Figure 1 ).
The Genomics of Pituitary Tumor s
The genomic characterization of sporadic PT has gen-
erated information on the mutational burden (single nu-
cleotide variants, or SNVs), and the presence of chromoso-
mal gains and losses in specific cytogenetic regions (copy
number variation, or CNVs). Genetic variations in the hu-
man genome include SNVs, but also large chromosomal
aberrations such as unbalanced deletions, duplications and
amplifications of DNA segments ranging from a few to
several hundred base pairs ( 8 , 9 ). Whereas whole genome
sequencing (WGS) comprises the entire genome, including
exons and introns, whole exome sequencing (WES) only
evaluates coding exons and in some cases, the intron-exon
boundaries ( 10 ). The frequency of chromosomal alterations
in PT varies according to the type of secretion: 7% for non-
functioning gonadotrophinomas and null cell adenomas,
1% for silent ACTH-adenomas, 50% for PRL-adenomas
and TSH-adenomas, 30% for corticotrophinomas causing
CD, and 20% for somatotrophinomas and plurihormonal
tumors ( 11 ).
Corticotrope Tumo rs
Activating mutations of the gene encoding the ubiquitin
specific peptidase 8 ( USP8 ) are found in 20–40% of spo-
radic ACTH-secreting PT causing CD ( 12 , 13 ). Patients har-
boring these genetic variants are characteristically younger,
more frequently women and in some, but not all, studies
have been found to develop long-term recurrences more
frequently ( 14 , 15 ). USP8 mediates the deubiquitination of
the epidermal growth factor receptor ( EGFR ) by inhibiting
its interaction with the protein 14-3-3, which in turn pre-
vents its proteosomal degradation, leading to an increased
signaling by the deubiquitinated EGFR , which ultimately
results in increased cellular proliferation and transcription
of the proopiomelanocortin ( POMC ) gene ( 16 ). The gene
encoding another ubiquitin specific peptidase, USP48 , has
also been found to be somatically mutated in up to 23% of
ACTH tumors, which also results in an increased ACTH
synthesis ( 17 ).
Up to 16% of ACTH-secreting PT harbor the BRAF
(B-Raf Proto-Oncogene, Serine/Threonine Kinase) V600E
variant which is considered a driver mutation that induces
MAP2K1 (mitogen-activated protein kinase kinase 1) and
MAPK1/2 (mitogen-activated protein kinase 1/2) phospho-
rylation and thus, activates the transcription factor NR4A1
(nuclear receptor subfamily 4, group A, member 1) which
in turn promotes the transcription of POMC ( 17 ). The
gene encoding ATRX ( ATRX chromatin remodeler) has
been found to be mutated in one-third of corticotrophi-
nomas, particularly in tumors with an aggressive behav-
ior and in carcinomas ( 18 ). ATRX interacts with DAXX
(death domain associated protein) and histone H3.3 in het-
erochromatin remodeling and telomere maintenance and
function, therefore inactivation of ATRX leads to telomere
destabilization ( 18 ). Phosphatidylinositol-4,5-bisphosphate
3-kinase catalytic subunit α(PIK3CA ) gene mutations have
been found in 4% of ACTH-secreting tumors and are asso-
ciated with invasiveness ( 19 ). Finally, up to 33% of ACTH-
secreting tumors harbor tumor protein p53 ( TP53 ) muta-
tions, which appear to correlate with a more aggressive
behavior ( 13 , 20 ).
We recently evaluated through whole exome sequenc-
ing 10 PTs, representing the whole pathological spec-
trum of the corticotrope, and including 4 CD-causing
ACTH adenomas, three silent corticotrope tumors, one
ACTH-secreting macroadenoma in a patient with Nelson’s
syndrome, a Crooke cell macroadenoma and an ACTH-
secreting carcinoma that developed from a microadenoma
over a 15-year period ( 13 ). We found, previously re-
ported recurrent somatic mutations in 6 pathogenically rel-
evant genes: hydroxy- δ-5-steroid dehydrogenase, 3 β- and
steroid delta-isomerase 1 ( HSD3B1 ) in all 10 tumors, TP53
Molecular Biology of Pituitary Tumors 3
Figure 1. Advances in precision medicine for pituitary tumors. Multiomics (genomic, transcriptomic, proteomic, epigenomic, immunomic, and clinical
data) networks can provide knowledge for better sample classification, disease subtyping, biomarker discovery and prognosis prediction in pituitary
tumors. These insights will contribute to a better diagnosis and treatment of these patients.
in all, except one silent ACTH adenoma, cyclin depen-
dent kinase inhibitor 1A (CDKN1A ) in six tumors, EGFR
in six tumors, Aurora kinase A ( AURKA) in four tumors,
and USP8 in two tumors ( 13 ). The ACTH-secreting carci-
noma was the tumor with the highest number of genomic
abnormalities, including SNV and CNV ( 13 ). A phylo-
genetic inference analysis was carried out to establish a
hypothetical sequential transformation from a silent cor-
ticotrope adenoma to a functional tumor and finally, to
an ACTH-secreting carcinoma ( 13 ). We found two dis-
tinct clades, the first one was characterized by expressing
a SNV of the gene encoding the activating transcription
factor 7-interacting protein (rs.3213764) and includes two
of the three silent adenomas and two of the five corti-
cotrophinomas causing CD ( 13 ). These four tumors have
the same SNV profile, so we can assume that they harbor
all the genes that must be altered to allow the transition
from a silent adenoma to a clinically evident tumor ( 13 ).
The second and largest clade includes the Crooke cell ade-
noma, two ACTH-adenomas causing CD, one of the silent
ACTH-adenomas and the ACTH-secreting carcinoma ( 13 ).
All the tumors in this clade harbor the necessary molecular
alterations to evolve from a functioning ACTH adenoma
to a malignant corticotrophinoma and are characterized by
expressing a polymorphism of the gene encoding the DNA
mismatch repair protein (rs.1650697) ( 13 ).
Clinically non-functioning Pituitary Tumor s
Clinically non-functioning PT harbor very few mutations,
in particular, none of the previously mentioned vari-
ants reported in tumors of other lineages are present in
these neoplasms of gonadotrope differentiation ( 21 ). This
is consistent with their low proliferative index, which
4 Marrero-Rodríguez et al. / Archives of Medical Research 54 (2023) 102915
means that mechanisms other than somatic mutations are
likely to be involved in their oncogenesis ( 21 ). Never-
theless, a few genes, including platelet-derived growth
factor D ( PDGFD ), NDRG family member 4 ( NDRG4 ),
and Mitogen-Activated Protein Kinase Kinase Kinase 20
( MAP3K20 ) ( 21 ), as well as isoleucyl-tRNA synthetase
1 ( IARS1 ) and thyroid hormone receptor interactor 12
( TRIP12 ) ( 22 ) have been described to harbor isolated, non-
recurrent mutations. Some silent corticotrope tumors have
been found to harbor the same USP8 somatic mutation,
previously described in ACTH-secreting adenomas causing
CD ( 23 , 24 ). However, neither Perez-Rivas L, et al. ( 25 ),
nor our group ( 13 ) have found such molecular abnormality
in these clinically silent ACTH-producing lesions. Isolated
inactivating mutations of the gene encoding the aryl hydro-
carbon receptor-interacting protein ( AIP ) have been found
in some clinically silent somatotroph tumors ( 26 ).
Somatotrophinomas
Activating somatic mutations of the gene encoding the
GNAS complex locus ( GNAS ) have been reported in up
to 40–50% of GH-secreting tumors in Caucasian ( 27 ) and
Japanese ( 28 ) patients, although the prevalence of this
molecular abnormality is much lower in Mexican (18%)
( 29 ) and Korean ( 30 ) subjects. This mutation disrupts the
GTPase activity of the Gs protein α-subunit leading to con-
stitutive activation of the growth hormone releasing hor-
mone receptor ( TRH-R ) that results in a constant activa-
tion of adenylate cyclase and thus, an increased generation
of intracellular cyclic adenosine monophosphate (cAMP)
which through cAMP response element binding protein
( CREB ) increases the transcription of the genes encoding
GH and POU1F1 and also results in an increased cellular
proliferation ( 31 ). Tumors bearing GNAS mutations char-
acteristically have a densely granulated pattern by elec-
tron microscopy or CAM5.2 immunostaining, are usually
smaller and less invasive and appear to respond favorably
to somatostatin analogs ( 27 , 29 ).
No other consistently recurrent somatic mutations have
been found upon WGS and WES in patients with GH-
secreting tumors. However, several somatic variants of
genes involved in the cAMP pathway such as protein ki-
nase AMP-activated catalytic subunit alpha 2 ( PRKAA2 ),
calcium signaling such as calcium voltage-gated channel
subunit alpha 1H ( CACNA1H ), as well as AT P signaling
such as suv3 like RNA helicase ( SUPV3L1 ) have been ob-
served in some of these patients ( 32 , 33 ).
Thyrotrophinomas and Prolactinomas
TSH-secreting tumors or thyrotrophinomas are extremely
rare and present clinically with central hyperthyroidism.
Whole exome sequencing studies have identified candi-
date driver mutations in genes such as spermine oxidase
( SMOX) and synaptotagmin like 3 ( SYTL3 ) which are in-
volved in oxidative damage and the Rab signaling pathway,
respectively. No other altered genes were found in these
tumors ( 34 ).
The molecular evaluation of prolactinomas is hampered
by the fact that most of these neoplasms are pharmaco-
logically treated with dopamine agonists and therefore,
tissue availability is very limited. Recently, Li C, et al.
carried out whole genome sequencing of 21 macropro-
lactinomas and found a recurrent somatic missense mu-
tation in the gene encoding splicing factor 3B subunit 1
( SF3B1 ), that changes Arginine for Histidine at position
625 ( SF3B1 R625H ) in two of these tumors ( 35 ). These
authors validated this finding in 206 prolactinomas by dig-
ital PCR analysis and found that 43 of them (20.8%) har-
bored the SF3B1 R625H mutant ( 35 ). None of the 120 non-
prolactinoma PTs harbored this molecular abnormality, and
no germline mutation was found in 13 blood DNA sam-
ples ( 35 ). Primary cell cultures of mutant tumors showed
higher PRL secretion than wild-type tumors, and further-
more, adenoviral expression of SF3B1 R625H into GH3
and MMQ cell lines resulted in an increased cellular pro-
liferation and reduced apoptosis ( 35 ). SF3B1 R625H re-
sults in aberrant splicing of the estrogen-related receptor
gamma ( ESRRG ) gene, resulting in the generation of an
abnormal, cryptic mRNA ( 35 ). Patients carrying this so-
matic missense mutation appear to have higher PRL levels
and a decreased progression-free survival ( 35 ). POU class
6 homeobox 2 ( POU6F2 ) acts as a tumor suppressor and is
involved in the predisposition to Wilms tumor ( 36 ). Miao
Y, et al. reported the case of a male patient with an ag-
gressive macroprolactinoma resistant to dopamine agonist
therapy, who underwent debulking surgery and the excised
tumor was subjected to whole genome sequencing and was
found to have two biallelic POU6F2 mutations (p.P280L
and p.N292S) ( 37 ). In vitro expression studies revealed
that this biallelic mutation resulted in an increased PRL
secretion and cell viability ( 37 ). Thus far, the presence of
this molecular abnormality has not been reported by other
investigators ( Table 1 ).
Transcriptomics of Pituitary Tumors
In 2020, two independent groups of investigators reported
that transcriptomically, PT segregate into three distinct
clusters according to the expression of the transcription
factor that drives the terminal differentiation of the pitu-
itary cell type from which they originate ( 11 , 38 ). Accord-
ing to this paradigm, clinically nonfunctioning adenomas
of gonadotrope differentiation which are driven by NR5A1
constitute the first cluster; ACTH-secreting adenomas caus-
ing CD, which are driven by TBX19 , represent the second
cluster; and PRL-, GH- and TSH-secreting adenomas con-
form the third cluster and are driven by POU1F1 ( 11 , 38 ).
Silent corticotroph adenomas, which express TBX19 seem
Molecular Biology of Pituitary Tumors 5
Tab l e 1. Most common genomic mutation in PT
Gene Pituitary Tum o r
BRAF
USP48
CDKN1A
AURKA ACTH- secreting tumors
EGFR
PIK3CA
USP8
ATRX ACTH-secreting tumor and ACT H carcinoma
HSD3B1 CD-causing ACTH-adenomas, ACTH-secreting macroadenoma in a patient with Nelson’s syndrome, Crooke cell
macroadenoma, ACTH carcinoma and silent ACTH
TP53 CD-causing ACTH-adenomas, ACTH-secreting macroadenoma in a patient with Nelson’s syndrome, Crooke cell
macroadenoma and ACTH carcinoma
PDGFD
NDRG4
MAP3K20 FSH/LH-secreting tumor
IARS1
TRIP12
GNAS
PRKAA2
CACNA1H GH-secreting tumors
SUPV3L1
AIP
SMOX TSH-secreting tumors
SYTL3
SF3B1
POU6F2 PRL-secreting tumors
to share some transcriptomic features with the more fre-
quent clinically nonfunctioning adenoma of gonadotrope
differentiation ( 38 ). Interestingly, these two groups ar-
rived at the same conclusion using different methodolo-
gies which underscores the biological significance of their
findings. While Taniguchi-Ponciano K, et al. carried out a
whole coding transcriptome analysis at the gene and exon
level using a microarray that identified over 540 thousand
transcripts, including also long non-coding RNA (LIN-
CRNA), microRNA (miRNA) and circular RNA (circRNA)
( 38 ), while Neou M, et al. used direct RNA sequencing
( 11 ).
While there is an overexpression of the mRNA of
the anterior pituitary hormone specific for each tumor
type (i.e., PRL in prolactinomas, POMC in corticotrophi-
nomas, GH in somatotrophinomas, TSH in thyrotrophi-
nomas, LH/FSH in gonadotrophinomas), the mRNAs of
their corresponding cognate receptors are expressed in a
non-specific manner. Interestingly, the mRNA expression
of the receptors for the different hypothalamic releas-
ing hormones was variable ( 38 ). The thyrotropin releas-
ing hormone receptor ( TRH-R ) mRNA was clearly over-
expressed in thyrotrophinomas, gonadotrophinomas and to
a lesser extent somatotrophinomas (this explains the para-
doxical rise of GH upon thyrotropin releasing hormone
(TRH) stimulation in some patients with acromegaly),
while it was underexpressed in ACTH-secreting tumors.
The corticotropin releasing hormone receptor 1 ( CRH-R1 )
mRNA was distinctly overexpressed in corticotroph tu-
mors compared to other tumor types. The GHRH-R mRNA
was expressed in all tumor types with perhaps a slight
predominance in GH- and TSH-secreting tumors. The
gonadotropin-releasing hormone receptor ( GnRH-R ) was
also distinctly overexpressed in gonadotrophinomas com-
pared to other tumor types. TSH- and GH-secreting tumors
had the highest somatostatin receptor 2 ( SSTR2 ) mRNA ex-
pression, which are the tumor types that respond to first-
generation somatostatin analogs ( 38 ). In contrast, the so-
matostatin receptor 5 ( SSTR5 ) mRNA was overexpressed
in PRL-, TSH- and to a lesser extent in GH- and ACTH-
secreting tumors, which explains the favorable response
of corticotrophinomas to pasireotide, a second-generation
somatostatin analog ( 38 ). The expression of dopamine re-
ceptor mRNA was highest in prolactinomas, followed by
thyrotrophinomas, gonadotrophinomas, and somatotrophi-
nomas. Some genes, such as solute carrier family 25 mem-
ber 2 ( SLC25A2 ), miR590 , and LINC00412 were clearly
overexpressed in all PT types, compared to non-tumoral
gland ( 38 ).
Corticotrope Tumo rs
ACTH-secreting tumors causing CD and silent corti-
cotrophinomas, both express POMC and the transcription
factor TBX19 ; however, their transcriptomic profiles are
otherwise different. CD-causing ACTH-secreting adeno-
6 Marrero-Rodríguez et al. / Archives of Medical Research 54 (2023) 102915
mas are characterized by expressing arginine vasopressin
receptor 1B ( AVPR1B ), androgen receptor ( AR ), CRH-
R1, nuclear receptor subfamily 1, group F, member 2
( NR1F2 ), PLAG1 zinc finger ( PLAG1 ), and EPH receptor
A4 ( EPHA4 ) mRNA ( 38 ). Silent corticotrope adenomas
specifically overexpress calcium voltage-gated channel
auxiliary subunit alpha2delta 4 ( CACNA2D4 ), paired like
homeodomain 2 ( PITX2 ), retinoid X receptor gamma
( RxRG ), and EPH receptor B6 ( EPHB6 ) ( 38 ). Pathways
involved in ACTH-secreting tumors include enzymes in-
volved in drug metabolism, metabolism of xenobiotics by
cytochrome P450, and the renin-angiotensin-aldosterone
system, as well as tryptophan and pyrimidine metabolism
( 38 ). ACTH-secreting tumors bearing USP8 somatic
mutations overexpress SSTR5 mRNA, which seems to
correlate with the therapeutic response to pasireotide, as
mentioned above ( 11 , 38 ).
Clinically Non-functioning Pituitary Tumor s
Clinically nonfunctioning adenomas of gonadotrope differ-
entiation and null cell tumors share an important num-
ber of expressed genes, including neuronal differentiation
1 ( NEUROD1 ), zinc finger protein 804A ( ZNF804A ), zinc
finger BED-type containing 2 ( ZBED2 ), retina and anterior
neural fold homeobox 2 ( RAX2 ), ISL LIM homeobox 1
( ISL1 ) and high mobility group AT-hook 2 ( HMGA2 ) ( 11 ).
Genes such as CACNA2D4 and EPHB6 are upregulated
in gonadotrophinomas, silent ACTH tumors and null cell
adenomas ( 38 ). Pathways altered in gonadotrophinomas in-
clude WNT , estrogen and calcium signaling pathways, as
well as immune- related events such as Th17 cell differen-
tiation ( 38 ). Other investigators have found upregulation of
genes involved in the regulation of epithelial-mesenchymal
transition, particularly in fast-growing gonadotrope tumors
( 39 ).
GH-, TSH- and PRL-tumors
The cluster comprising GH-, PRL- and TSH-PT is char-
acterized by upregulation of genes encoding proteins such
as slit guidance ligand 1 ( SLIT1 ), PRL receptor ( PRLR )
and solute carrier family 6 member 6 ( SLC16A6 ) ( 38 ).
The GH-, TSH-tumors and POU1F1 -plurihormonal tumors
share the expression of SRY-box transcription factor 5
( SOX5 ), regulatory factor X4 ( RFX4 ), zinc finger protein
536 ( ZNF536 ), among others ( 11 ). Prolactinomas express
neuronal differentiation 4 ( NEUROD4 ) , estrogen receptor 1
( ESR1 ) and LIM homeobox 3 ( LHX3 ) ( 11 ). GH-secreting
tumors harboring GNAS mutations express higher amounts
of dopamine receptor D2 ( D2R ), which could be related to
a more favorable response to dopamine agonists ( 11 ).
Cell Populations Define Aggressiveness, Recurrence, and
Functionality
Recently, Zhang Q, et al. found that pituitary tumors
also may cluster according to their degree of differenti-
ation ( 40 ). The poorly differentiated cluster is character-
ized by an abnormal expression of genes involved in the
metabolism of lipids, as well as in cholesterol and long
chain fatty acid synthesis ( 40 ).
Single-cell RNA sequencing (scRNAseq) studies have
revealed that silent ACTH-tumors exhibit increased expres-
sion of epithelial-mesenchymal transition related genes,
along with the loss of transcripts related to hormonal bio-
genesis and secretion ( 41 ). In contrast, CD-causing cor-
ticotrophinomas show marked overexpression of secre-
togranin V ( SCG5 ) , g alanin and GMAP prepropeptide
( GAL ) , TIMP metallopeptidase inhibitor 1 ( TIMP1 ), and
adipocyte plasma membrane-associated protein ( APMAP )
which are related to secretory vesicles, ( 41 ) ( Table 2 ).
The Epigenetic Landscape of PT
Epigenetically, PT also cluster according to the tran-
scription factor that determines their terminal differenti-
ation ( 38 ). Approximately 649 genes have been found to
be potentially regulated by DNA methylation in ACTH-
secreting tumors causing CD. In these tumors, genes such
as AVPR1B, EPHA4 , and glutamate ionotropic receptor
NMDA type subunit 2B ( GRIN2B ) are upregulated due
to promoter demethylation ( 38 ). Silent corticotrope tumors
have a different methylation profile and are epigenetically
closer to nonfunctioning gonadotrophinomas than to clini-
cally eloquent ACTH-secreting adenomas ( 11 ). USP8 mu-
tation status does not appear to correlate with methyla-
tion profiles ( 11 ). CD-causing and silent ACTH tumors
also differ in their miRNA expression profile ( 42 ). Elo-
quent ACTH tumors overexpress miR-129-2-3p, miR-129-
5p, miR-124-3p, miR-132-5p, miR-129-1-3p, miR-135b-5p,
miR-27a-3p, miR-10b-5p, miR-9-3p, miR-6506-3p, miR-
6864-5p, let-7b-5p, miR-670-3p, miR-22-5p, miR-346 and
miR-9-5p, whereas miR-1909-3p, miR-4319, miR-181b-3p,
and miR4501 ( 38 , 42 ). These miRNAs are involved in the
regulation of steroid hormone receptor, nuclear receptor,
and ligand-activated transcription factor activity, as well as
steroid hormone-mediated signaling pathway. The differen-
tially expressed miRNAs in tumors causing CD to play a
role in the regulation of glucocorticoid receptor levels and
probably in reducing the effect of the negative feedback
mediated by corticosteroids ( 42 ).
Approximately 1066 genes have been found to be po-
tentially controlled by methylation in nonfunctioning ade-
nomas. Genes such as CACNA2D4, glutamate ionotropic
receptor AMPA type subunit 2 ( GRIA2 ), miR4774,
miR1179 and LINC01351 appear to be upregulated due
to lower methylation rates ( 38 ). Clinically nonfunction-
Molecular Biology of Pituitary Tumors 7
Tab l e 2. Differentially expressed genes in PT
Gene Regulation Pituitary Tum o r
SLC25A2 Upregulated All tumors
miR590
LINC00412
GHRH-R Upregulated All tumor with predominance in GH- and TSH- secreting tumors.
GnRH-R Upregulated FSH/LH secreting tumors
NR5A1
NEUROD1
ZNF804A
ZBED2
RAX2
ISL1
HMGA2
CACNA2DA Upregulated FSH/LH-, silent ACTH secreting tumor and null cell adenomas
EPHB6
SSTR2 Upregulated GH- and TSH-secreting tumors
POU1F1 Upregulated GH-, PRL- and TSH-secreting tumors
SLIT1
PRLR
SLC16A6
SOX5 Upregulated GH-, TSH- and POU1F1-plurihormonal tumor
RFX4
ZNF536
NEUROD4 Upregulated PRL-secreting tumors
ESR1
LHX3
D2R Downregulated GNAS mutation GH-secreting tumors
SSTR5 Upregulated PRL-, TSH- and to a lesser extent in GH- and ACTH-secreting tumors
USP8 somatic mutation ACTH-tumor
TRH-R Upregulated USP8 somatic mutation ACTH-tumor
SCG5 Upregulated CD-causing ACTH secreting tumors
GAL
TIMP1
APMAP
POMC Upregulated ACTH-secreting tumors and silent ACTH tumors
TBX19
AVPR1B Upregulated ACTH-secreting tumors
CRHR1
AR
NR1F2
PLAG1
EPHA4
PITX2 Upregulated Silent ACTH tumor
RxRG
ing adenomas of gonadotroph differentiation, null cell ade-
nomas and silent ACTH tumors share the overexpression
of miR-582, miR-1468-5p, miR-28, miR-532 and miR-137
( 11 ). Interestingly, the miRNA signature could potentially
discriminate patients with invasive from those with non-
invasive gonadotrope tumors. A gene set of six miRNAs
( miR184, miR181a-2-3p, miR93-3p, miR574-5p, miR185-
5p, and miR3200-5p ) may represent potential biomarkers
of invasiveness of clinically nonfunctioning gonadotrope
tumors, although this is still controversial and requires fur-
ther research ( 43 ).
GH-, PRL- and TSH-secreting adenomas have 184, 204,
and 68 distinct genes, respectively that could be poten-
tially regulated by methylation ( 38 ). In thyrotrophinomas,
these genes include SSTR2, GRIA2 and LINC01173 ; in so-
matotrophinomas, transmembrane protein 233 ( TMEM233 )
and glutamate ionotropic receptor AMPA type subunit
4 ( GRIA4 ); and in prolactinomas, glutamate ionotropic
receptor NMDA type subunit 3A ( GRIN3A ). All three
POU1F1 -derived tumors overexpress miR377 and miR136
( 38 ). GH-secreting tumors overexpress miR21-5p, miR34a,
miR145, miR23b and miR107 which in turn regulate
genes such as insulin-like growth factor 1 ( IGF1 ), SRY-
box transcription factor 7 ( SOX7 ), and MYCN proto-
oncogene, bHLH transcription factor ( MYCN ) involved
in cell proliferation and apoptosis ( 43 ). PRL-secreting
tumors characteristically overexpress miR72a, miR93,
miR1299 and miR130a-3p, which in turn regulate pro-
lactin production and resistance to dopamine agonists
(DA) ( 43 ).
8 Marrero-Rodríguez et al. / Archives of Medical Research 54 (2023) 102915
Chromatin Remodeling Proteins in Pituitary Tum ors
Chromatin remodeling refers to the post-translational mod-
ification of histones, including methylation, lysine addition,
acetylation and citrullination, which are crucial for DNA
packaging in the formation of nucleosomes ( 44 ). Chro-
matin remodeling complexes (CRCs) can be described as
specialized multiprotein machinery that allow access to
DNA by temporarily modifying the structure or compo-
sition of nucleosomes ( 44 ). Ikaros ( IK ) is a protein ex-
pressed in normal pituitaries that plays a critical role in
the development and expansion of corticotrophs and so-
matotrophs, putatively acting through the fibroblast growth
factor receptor 4 ( FGFR4 ). The IK1 and IK2/3 alterna-
tively spliced isoforms, as well as the dominant-negative
IK6 isoform are expressed in nearly 50% of human pitu-
itary adenomas ( 45 ). IK selectively deacetylates histone 3
residues on the proximal transfected or endogenous GH
promoter and limits access to the POU1F1 activator ( 45 ).
In contrast, IK acetylates histone 3 on the proximal PRL
promoter and facilitates POU1F1 binding to this region
in the same cells. These data provide evidence for IK-
mediated histone acetylation and chromatin remodeling in
the selective regulation of pituitary GH and PRL hormone
gene expression ( 45 ). MAGE family member A3 ( MAGE-
A3 ) transcripts are abundant in PT and have been impli-
cated in the transcriptional silencing of p53 through histone
deacetylation. Furthermore, MAGE-A3 downregulation re-
sults in p53 and p21 accumulation ( 45 ).
Concluding Remarks
The advent of genomics, transcriptomics, epigenetics, im-
munomics, metabolomics, proteomics, and their correlation
with clinical data, has provided researchers with an integral
perspective of the underlying complexity of PT, highlight-
ing their architecture, their molecular changes, and cellular
responses. Additionally, it has furthered our understanding
of the interactions between the tumor and its microenvi-
ronment and has broaden the therapeutic horizon with po-
tential, personalized molecular strategies that may be par-
ticularly useful in aggressive and recurrent neoplasms, as
well as in the rare cases of pituitary carcinoma. It is worth
mentioning that the main limitation of this review is the
significant heterogeneity of the data, which stems from the
different experimental approaches used in each of the re-
viewed studies. In our opinion, this data heterogeneity pre-
cludes us from drawing an integrative scenario encompass-
ing all the potential molecular alterations that contribute to
pituitary tumorigenesis.
Conflicts of Interest
There is no conflict of interest that could be perceived to
affect the impartiality of the reported research.
Acknowledgments
SVP and SAE are recipients of a grant from the Na-
tional Council for Humanities, Science and Technol-
ogy (CONAHCyT). All images were created in BioRen-
der.com.
References
1. Ashley E. Towar d s precision medicine. Nat Rev Genet
2016;17:507–522 .
2. Molitch M. Diagnosis and Treatment of Pituitary Adenomas: A Re-
view. JA MA 2017;317:516–524 .
3. Ramirez C, Cheng S, Va r g a s G, et al. Expression of Ki-67, PTTG1,
FGFR4, and SSTR 2, 3, and 5 in Nonfunctioning Pituitary Adeno-
mas: A High Throughput TMA, Immunohistochemical Study. J Clin
Endocrinol Metab 2012;97:1745–1751 .
4. Garcia-Guzman B, Portocarrero-Ortiz L, Dorantes-Argandar AA,
et al. Hereditary pituitary tumor syndomes: Genetic and clinical as-
pects. Rev invest Clin 2020;72:8–18 .
5. Denes J, Korbonits M. The clinical aspects of pituitary tumours ge-
netics. Endocrine 2021;71:663–674 .
6. Coopmans EC, Korbonits M. Molecular genetic testing in the man-
agement of pituitary disease. Clin Endocrinol 2022;97:424 .
7. Ho K, Fleseriu M, Kaiser U, et al. Pituitary Neoplasm nomencla-
ture Workshop:Does adenoma stand the test of time? J Endocr Soc
2021;5:bvaa205 .
8. Shao X, Lv N, Liao J, et al. Copynumber variation is highly cor-
related with differential gene expression: a pan-cancer study. BMC
Med Genet 2019;20:175 .
9. Pos O, Radvanszky J, Styk J, et al. Copy number variation: methods
and clinical applications. Appl Sci 2021;11:819 .
10. Ploski R. Next Generation Sequencing-General Information about the
Technology, Possibilities, and Limitations. Chapter 1. In: Clinical Ap-
plications for Next Generation sequencing. Academic Press; 2016.
p. 1–18 .
11. Neou M, Villa C, Armignacco R, et al. Pangenomic Classification of
Pituitary Neuroendocrine Tumors. Cancer Cell 2020;37 123–134.e5 .
12. Reincke M, Sbiera S, Hayakawa A, et al. Mutations in the deubiquiti-
nase gene USP8 cause Cushing’s disease. Nat Genet 2015;47:31–38 .
13. Andonegui-Elguera S, Silva-Román G, Peña-Martínez E, et al. The
Genomic Landscape of Corticotroph Tumors: From Silent Adenomas
to ACTH-Secreting Carcinomas. Int J Mol Sci 2022;23:4861 .
14. Albani A, Perez-Rivas LG, Dimopoulou Zoop S, et al. The USP8
mutational status may predict long-term remission in patients with
Cushing´s disease. Clin Endocrinol (Oxf) 2018;89:454–458 .
15. Wanichi I, Mariani B, Frassetto F, et al. Cushing’s disease due to
somatic USP8 mutations: a systematic review and meta-analysis. Pi-
tuitary 2019;22:435–442 .
16. Theodoropoulou M, Reincke M. Genetics of Cushing´s disease: from
the lab to clinical practice. Pituitary 2022;25:689–692 .
17. Chen J, Jian X, Deng S, et al. Identification of recurrent USP48 and
BRAF mutations in Cushing’s disease. Nat Commun 2018;9:3171 .
18. Casar-Borota O, Boldt H, Engström B, et al. Corticotroph Aggressive
Pituitary Tumo rs and Carcinomas Frequently Harbor ATRX Muta-
tions. J Clin Endocrinol Metab 2021;106:1183–1194 .
19. Lin Y, Jiang X, Shen Y, et al. Frequent mutations and amplifica-
tions of the PIK3CA gene in pituitary tumors. Endocr Relat Cancer
2009;16:301–310 .
20. Sbiera S, Perez-Rivas L, Taranets L, et al. Driver mutations in USP8
wild-type Cushing’s disease. Neuro Oncol 2019;21:1273–1283 .
21. Newey P, Nesbit M, Rimmer A, et al. Whole-exome sequencing stud-
ies of nonfunctioning pituitary adenomas. J Clin Endocrinol Metab
2013;98:E796–E800 .
Molecular Biology of Pituitary Tumors 9
22. Song Z, Reitman Z, Ma Z, et al. The genome-wide mutational land-
scape of pituitary adenomas. Cell Res 2016;26:1255–1259 .
23. Castellnou S, Vasiljevic A, Lapras V, et al. SST5 expression and
USP8 mutation in functioning and silent corticotroph pituitary tu-
mors. Endocr Connect 2020;9:243–253 .
24. Bujko M, Kober P, Boresowicz J, et al. USP8 mutations in
corticotroph adenomas determine a distinct gene expression pro-
file irrespective of functional tumour status. Eur J Endocrinol
2019;181:615–627 .
25. Perez-Rivas L, Theodoropoulou M, Ferraù F, et al. The Gene
of the Ubiquitin-Specific Protease 8 Is Frequently Mutated in
Adenomas Causing Cushing’s Disease. J Clin Endocrinol Metab
2015;100:E997–E1004 .
26. Drummond J, Roncaroli F, Grossman A, et al. Clinical and Patholog-
ical Aspects of Silent Pituitary Adenomas. J Clin Endocrinol Metab
2019;104:2473–2489 .
27. Spada A, Arosio M, Bochicchio D, et al. J Clin Endocrinol Metab
1990;71:1421–1426 .
28. Yasufuku-Takano J, Takano K, Morita K, et al. Does the prevalence
of gsp mutations in GH-secreting pituitary adenomas differ geograph-
ically or racially? Prevalence of gsp mutations in Japanecese patients
revisited. Clin Endocrinol 2006;64:91–96 .
29. Mendoza V, Sosa E, Espinosa-de-los-Monteros, et al. GSP αmuta-
tions in Mexican patients with acromegaly: potential impact on long
term prognosis. GH &IGF research 2005;15:28–32 .
30. Kim HJ, Kim MS, Park YJ, et al. Prevalence of Gs alpha mu-
tations in Korean patients with pituitary adenomas. J Endocrinol
2001;168:221–226 .
31. Landis CA, Masters SB, Spada A, et al. GTPase inhibiting mutations
activate the αchain of Gs and stimulate adenylyl cyclase in human
pituitary tumours. Nature 1989;340:692–696 .
32. Ronchi CL, Peverelli E, Herterich S, et al. Landscape of somatic mu-
tations in sporadic GH-secreting pituitary adenomas. Eur J Endocrinol
2016;174:363–372 .
33. Valimaki N, Demir H, Pitkanen E, et al. Whole-Genome Sequenc-
ing of Growth Hormone (GH-secreting) pituitary adenomas. J Clin
Endocrinol Metab 2015;100:3918–3927 .
34. Sapkota S, Horiguchi K, Tos aka M, et al. Whole-exome sequenc-
ing study of thyrotropin-secreting pituitary adenomas. Endo Soc
2017;102:565–575 .
35. Li C, Xie W, Rosenblum JS, et al. Somatic SF3B1 hotspot mutation
in prolactinomas. Nat Commun 2020;11:2506 .
36. Perotti D, De Vecchi G, Tes ti MA, et al. Germline mutations of
the POU6F2 gene in Wilms tumors with loss of heterozygosity on
chromosome 7p14. Hum mut 2004;24:400–407 .
37. Miao Y, Li C, Guo J, et al. Identification of a novel somatic mutation
of POU6F2 by whole-genome sequencing in prolactinoma. Mol Genet
Genomic Med 2019;7:e1022 .
38. Taniguchi-Ponciano K, Andonegui-Elguera S, Peña-Martínez E, et al.
Transcriptome and methylome analysis reveals three cellular origins
of pituitary tumors. Sci Rep 2020;10:19373 .
39. Oh J, Osorio R, Jung J, et al. Transcriptomic Profiles of Normal
Pituitary Cells and Pituitary Neuroendocrine Tumor Cells. Cancers
(Basel) 2022;15:110 .
40. Zhang Q, Yao B, Long X, et al. Single-cell sequencing identifies
differentiation-related markers for molecular classification and recur-
rence prediction of PitNET. Cell Rep Med 2023;4:100934 .
41. Zhang D, Hugo W, Bergsneider M, et al. Single-cell RNA sequenc-
ing in silent corticotroph tumors confirms impaired POMC process-
ing and provides new insights into their invasive behavior. Eur J
Endocrinol 2022;187:49–64 .
42. Mossakowska BJ, Kober P, Rusetska N, et al. Difference in miRNA
expression in functioning and silent corticotroph pituitary adenomas
indicates the role of miRNA in the regulation of corticosteroid. Int J
Mole Sci 2022;23:2867 .
43. Donati S, Aurilia C, Palmini G, et al. MicroRNAs as Potential
Biomarkers in Pituitary Adenomas. Noncoding RNA 2021;7:55 .
44. Magaña-Acosta M, Valadez-Graham V. Chromatin Remodelers in the
3D Nuclear Compartment. Front Genet 2020;11:600615 .
45. Tateno T, Zhu X, Asa SL, et al. Chromatin remodeling and histone
modifications in pituitary tumors. Mol cel endo 2010;326:66–70 .
... Unfortunately, due to technical restrictions in regard to tissue samples, we were unable to perform an experiment that may reveal the molecular identity of recurrent CNFPAs from these patients and explain the mechanistic modifications of the machinery of calcium signaling and gene expression of these adenomas. Our research group has identified some elements of the molecular alterations in other studies of pituitary adenomas, including CNFPAs [14,21,37,38]. However, further investigation is needed that traces, longitudinally, the evolution of adenoma recurrence, from the primary tumor to subsequent events of recurrence in patients, and compare them with non-recurrent CNFPAs and healthy tissues. ...
Article
Full-text available
Clinically non-functioning pituitary adenomas (CNFPAs) are the second most frequent sellar tumor among studies on community-dwelling adults. They are characterized by the absence of hormonal hypersecretion syndrome, and patients present with compressive symptoms, such as a headache and visual field defects. Immunohistochemically, most CNFPAs are of gonadotrope differentiation, with only a few of them being truly null cell adenomas. Although these tumors express receptors for one or more hypothalamic releasing hormones, to what extent this has an impact on the biological and clinical behavior of these neoplasms remains to be defined. In this research, we evaluated the basal and hypothalamic secretagogue-stimulated intracellular calcium mobilization in 13 CNFPAs, trying to correlate this response to the phenotypic features of the patients. Our results indicate that the recurrence of a CNFPA correlates positively with cellular responsiveness, as measured by spontaneous intracellular calcium activity and the ability to respond to multiple hypothalamic secretagogues. We conclude that this finding may be a useful tool for predicting the clinicopathologic behavior of CNFPAs, by testing the variation of cellular responsiveness to hypothalamic secretagogues.
Article
Full-text available
Pituitary neuroendocrine tumor (PitNET) is one of the most common intracranial tumors with variable recurrence rate. Currently, the recurrence prediction is unsatisfying and can be improved by understanding the cellular origins and differentiation status. Here, to comprehensively reveal the origin of PitNET, we perform comparative analysis of single-cell RNA sequencing data from 3 anterior pituitary glands and 21 PitNETs. We identify distinct genes representing major subtypes of well and poorly differentiated PitNETs in each lineage. To further verify the predictive value of differentiation biomarkers, we include an independent cohort of 800 patients with an average follow-up of 7.2 years. In both PIT1 and TPIT lineages, poorly differentiated groups show significantly higher recurrence rates while well-differentiated groups show higher recurrence rates in SF1 lineage. Our findings reveal the possible origin and differentiation status of PitNET based on which new differentiation classification is proposed and verified to predict tumor recurrence.
Article
Full-text available
Simple Summary The molecular pathogenesis of pituitary neuroendocrine tumors (PitNETs) involves the interplay of many genes and transcription factors in the setting of a diverse altered tumor microenvironment. Recent advancements in molecular technologies, such as single-cell RNA sequencing, became essential in delineating specific cell types and identifying altered genes in both normal physiological states and diseases in the pituitary. This review discusses the recent progress made in characterizing the transcriptomic profiles of the normal pituitary gland and sporadic PitNETs and their significance. Abstract The pituitary gland is one of the most cellularly diverse regions of the brain. Recent advancements in transcriptomic biology, such as single-cell RNA sequencing, bring an unprecedented glimpse into the molecular composition of the pituitary, both in its normal physiological state and in disease. Deciphering the normal pituitary transcriptomic signatures provides a better insight into the ontological origin and development of five types of endocrine cells, a process involving complex cascades of transcription factors that are still being established. In parallel with these observations about normal pituitary development, recent transcriptomic findings on pituitary neuroendocrine tumors (PitNETs) demonstrate both preservations and changes in transcription factor expression patterns compared to those seen during gland development. Furthermore, recent studies also identify differentially expressed genes that drive various tumor behaviors, including hormone hypersecretion and tumor aggression. Understanding the comprehensive multiomic profiles of PitNETs is essential in developing molecular profile-based therapies for PitNETs not curable with current treatment modalities and could eventually help align PitNETs with the breakthroughs being made in applying precision medicine to other tumors.
Article
Full-text available
Cushing’s disease is a rare, but devastating condition, caused by corticotroph tumors. It rarely manifests as syndrome and very few isolated cases present with germline mutations. Instead, the vast majority of corticotroph tumors are sporadic monoclonal neoplasms. At present, the major recurrent somatic driver mutations are found in the USP8 gene, which encodes for a deubiquitinase that rescues proteins regulating ACTH synthesis. Almost half of functional corticotroph tumors carry somatic USP8 mutations that associate with a distinct transcriptomic and clinical profile. Other genes mutated in a small fraction of corticotroph tumors include the deubiquitinase encoding gene USP48 and the glucocorticoid receptor expressing NR3C1 . Recent reports on somatic TP53 and ATRX mutations in corticotroph macroadenomas and carcinomas indicate that within specific patient subpopulations they are not as rare as assumed.
Article
Full-text available
Corticotroph cells give rise to aggressive and rare pituitary neoplasms comprising ACTH-producing adenomas resulting in Cushing disease (CD), clinically silent ACTH adenomas (SCA), Crooke cell adenomas (CCA) and ACTH-producing carcinomas (CA). The molecular pathogenesis of these tumors is still poorly understood. To better understand the genomic landscape of all the lesions of the corticotroph lineage, we sequenced the whole exome of three SCA, one CCA, four ACTH-secreting PA causing CD, one corticotrophinoma occurring in a CD patient who developed Nelson syndrome after adrenalectomy and one patient with an ACTH-producing CA. The ACTH-producing CA was the lesion with the highest number of single nucleotide variants (SNV) in genes such as USP8, TP53, AURKA, EGFR, HSD3B1 and CDKN1A. The USP8 variant was found only in the ACTH-CA and in the corticotrophinoma occurring in a patient with Nelson syndrome. In CCA, SNV in TP53, EGFR, HSD3B1 and CDKN1A SNV were present. HSD3B1 and CDKN1A SNVs were present in all three SCA, whereas in two of these tumors SNV in TP53, AURKA and EGFR were found. None of the analyzed tumors showed SNV in USP48, BRAF, BRG1 or CABLES1. The amplification of 17q12 was found in all tumors, except for the ACTH-producing carcinoma. The four clinically functioning ACTH adenomas and the ACTH-CA shared the amplification of 10q11.22 and showed more copy-number variation (CNV) gains and single-nucleotide variations than the nonfunctioning tumors.
Article
Full-text available
Corticotroph pituitary adenomas commonly cause Cushing’s disease (CD), but some of them are clinically silent. The reason why they do not cause endocrinological symptoms remains unclear. We used data from small RNA sequencing in adenomas causing CD (n = 28) and silent ones (n = 20) to explore the role of miRNA in hormone secretion and clinical status of the tumors. By comparing miRNA profiles, we identified 19 miRNAs differentially expressed in clinically functioning and silent corticotroph adenomas. The analysis of their putative target genes indicates a role of miRNAs in regulation of the corticosteroid receptors expression. Adenomas causing CD have higher expression of hsa-miR-124-3p and hsa-miR-135-5p and lower expression of their target genes NR3C1 and NR3C2. The role of hsa-miR-124-3p in the regulation of NR3C1 was further validated in vitro using AtT-20/D16v-F2 cells. The cells transfected with miR-124-3p mimics showed lower levels of glucocorticoid receptor expression than control cells while the interaction between miR-124-3p and NR3C1 3′ UTR was confirmed using luciferase reporter assay. The results indicate a relatively small difference in miRNA expression between clinically functioning and silent corticotroph pituitary adenomas. High expression of hsa-miR-124-3p in adenomas causing CD plays a role in the regulation of glucocorticoid receptor level and probably in reducing the effect of negative feedback mediated by corticosteroids.
Article
Full-text available
Pituitary adenomas (PAs) are one of the most common lesions of intracranial neoplasms, occurring in approximately 15% of the general population. They are typically benign, although some adenomas show aggressive behavior, exhibiting rapid growth, drug resistance, and invasion of surrounding tissues. Despite ongoing improvements in diagnostic and therapeutic strategies, late first diagnosis is common, and patients with PAs are prone to relapse. Therefore, earlier diagnosis and prevention of recurrence are of importance to improve patient care. MicroRNAs (miRNAs) are short non-coding single stranded RNAs that regulate gene expression at the post-transcriptional level. An increasing number of studies indicate that a deregulation of their expression patterns is related with pituitary tumorigenesis, suggesting that these small molecules could play a critical role in contributing to tumorigenesis and the onset of these tumors by acting either as oncosuppressors or as oncogenes, depending on the biological context. This paper provides an overview of miRNAs involved in PA tumorigenesis, which might serve as novel potential diagnostic and prognostic non-invasive biomarkers, and for the future development of miRNA-based therapeutic strategies for PAs.
Article
Full-text available
The WHO Classification of Endocrine Tumours designates pituitary neoplasms as adenomas. A proposed nomenclature change to neuroendocrine tumors (PitNETs) has been met with concern by some stakeholder groups. The Pituitary Society coordinated the Pituitary Neoplasm Nomenclature (PANOMEN) Workshop to address the topic. Experts in pituitary developmental biology, pathology, neurosurgery, endocrinology, and oncology, including representatives nominated by the Endocrine Society, European Society of Endocrinology, European Neuroendocrine Association, Growth Hormone Research Society, and International Society of Pituitary Surgeons.Clinical epidemiology, disease phenotype, management, and prognosis of pituitary adenomas differ from that of most NETs. The vast majority of pituitary adenomas are benign and do not adversely impact life expectancy. A nomenclature change to PitNET does not address the main challenge of prognostic prediction, assigns an uncertain malignancy designation to benign pituitary adenomas, and may adversely affect patient. Due to pandemic restrictions, the workshop was conducted virtually, with audio-visual lectures and written précis on each topic provided to all participants. Feedback was collated and summarized by Content Chairs and discussed during a virtual writing meeting moderated by Session Chairs, which yielded an evidence-based draft document sent to all participants for review and approval. There is not yet a case for adopting the PitNET nomenclature. The PANOMEN Workshop recommends that the term adenoma be retained and that the topic be revisited as new evidence on pituitary neoplasm biology emerges
Article
Full-text available
Background Pituitary tumours are usually benign and relatively common intracranial tumours, with under- and overexpression of pituitary hormones and local mass effects causing considerable morbidity and increased mortality. While most pituitary tumours are sporadic, around 5% of the cases arise in a familial setting, either isolated [familial isolated pituitary adenoma, related to AIP or X-linked acrogigantism], or in a syndromic disorder, such as multiple endocrine neoplasia type 1 or 4, Carney complex, McCune–Albright syndrome, phaeochromocytoma/paraganglioma with pituitary adenoma, DICER1 syndrome, Lynch syndrome, and USP8-related syndrome. Genetically determined pituitary tumours usually present at younger age and show aggressive behaviour, and are often resistant to different treatment modalities. Subject In this practical summary, we take a practical approach: which genetic syndromes should be considered in case of different presentation, such as tumour type, family history, age of onset and additional clinical features of the patient. Conclusion The identification of the causative mutation allows genetic and clinical screening of relatives at risk, resulting in earlier diagnosis, a better therapeutic response and ultimately to better long-term outcomes.
Article
Objective: Provide insights into the defective POMC processing and invasive behavior in silent pituitary corticotroph tumors. Design and methods: Single cell RNAseq was used to compare the cellular makeup and transcriptome of silent and active corticotroph tumors. Results: A series of transcripts related to hormone processing peptidases and genes involved in the structural organization of secretory vesicles were reduced in silent compared to active corticotroph tumors. Most relevant to their invasive behavior, silent corticotroph tumors exhibited several features of epithelial-to-mesenchymal transition (EMT), with increased expression of mesenchymal genes along with the loss of transcripts which regulate hormonal biogenesis and secretion. Silent corticotroph tumor vascular smooth muscle cell and pericyte stromal cell populations also exhibited plasticity in their mesenchymal features. Conclusions: Our findings provide novel insights into the mechanisms of impaired POMC processing and invasion in silent corticotroph tumors and suggest that a common transcriptional reprogramming mechanism simultaneously impairs POMC processing and activates tumor invasion.
Article
Objective: Most pituitary tumours occur sporadically without a genetically identifiable germline abnormality, a small but increasing proportion present with a genetic defect that predisposes to pituitary tumour development, either isolated (e.g., aryl hydrocarbon receptor-interacting protein, AIP) or as part of a tumour-predisposing syndrome (e.g., multiple endocrine neoplasia (MEN) type 1, Carney complex, McCune-Albright syndrome or pituitary tumour and paraganglioma association). Genetic alterations in sporadic pituitary adenomas may include somatic mutations (e.g., GNAS, USP8). In this review, we take a practical approach: which genetic syndromes should be considered in case of different presentation, such as tumour type, family history, age of onset and additional clinical features of the patient. Design: Review of the recent literature in the field of genetics of pituitary tumours. Results: Genetic testing in the management of pituitary disease is recommended in a significant minority of the cases. Understanding the genetic basis of the disease helps to identify patients and at-risk family members, facilitates early diagnosis and therefore better long-term outcome and opens up new pathways leading to tumorigenesis. Conclusion: We provide a concise overview of the genetics of pituitary tumours and discuss the current challenges and implications of these genetic findings in clinical practice.