Functional characterization of the MTC-associated germline RET-K666E mutation: evidence of oncogenic potential enhanced by the G691S polymorphism ...

Page created by Gary Ford
 
CONTINUE READING
Functional characterization of the MTC-associated germline RET-K666E mutation: evidence of oncogenic potential enhanced by the G691S polymorphism ...
Endocrine-Related Cancer (2011) 18 519–527

Functional characterization of the MTC-
associated germline RET-K666E mutation:
evidence of oncogenic potential enhanced
by the G691S polymorphism
Maria Grazia Borrello1†, Antonella Aiello2†, Bernard Peissel 3,
Maria Grazia Rizzetti1, Piera Mondellini1, Debora Degl’Innocenti1,
Veronica Catalano1, Morena Gobbo2, Paola Collini 2, Italia Bongarzone1,
Marco A Pierotti 4, Angela Greco1‡ and Ettore Seregni 5‡
1
 Operative Unit Molecular Mechanisms, Department of Experimental Oncology, 2Pathology and Laboratory Medicine, 3Unit of
Medical Genetics, Department of Preventive and Predictive Medicine, 4Scientific Directorate and 5Nuclear Medicine, Therapy and
Endocrinology, IRCCS Istituto Nazionale dei Tumori Foundation, Milan, Italy
(Correspondence should be addressed to M G Borrello; Email: mariagrazia.borrello@istitutotumori.mi.it)
†
(M G Borrello and A Aiello contributed equally to this work)
‡
(A Greco and E Seregni share senior authorship)

Abstract
Activating mutations of RET, a gene encoding two isoforms of a tyrosine kinase receptor
physiologically expressed in several neural crest-derived cell lineages, are associated with
the inherited forms of medullary thyroid carcinoma (MTC). The identification and character-
ization of novel RET mutations involved in MTC is valuable, as RET gene testing plays a crucial
role in the management of these patients. In an MTC patient, we have identified a germline
c.1996AOG transition in heterozygosis leading to K666E substitution. In addition, the
conservative S904S (c.2712COG) and the non-conservative functional G691S (c.2071GOA)
polymorphisms have been identified. Through functional studies, we demonstrate for the first
time that K666E is a gain-of-function mutation with oncogenic potential, based on its ability to
transform NIH3T3 cells. It was not possible to define whether K666E is a de novo or inherited
RET variant in the patient, as the family history was negative for MTC, and the carrier status of
family members could not be tested. Our results, together with a recent report of co-segregation
of the mutation in three MTC families, suggest that K666E is a causative MTC mutation. As we
have shown that the same patient allele carries both K666E and G691S variants, the latter
known to increase downstream RET signaling, a possible role for the G691S polymorphism has
also been investigated. We have demonstrated that, although RET-G691S is not oncogenic
per se, it enhances the transforming activity of the RET-K666E mutant, thus suggesting a
modifier role for this functional polymorphism.
Endocrine-Related Cancer (2011) 18 519–527

Introduction                                                              (RET51) and a shorter protein of 1072 residues
The RET (rearranged during transfection) gene                             displaying nine unrelated C-terminal-specific amino
encodes a tyrosine kinase receptor with a crucial role                    acids (RET9), as shown in Fig. 1A. The RET receptor
in development. RET comprises 21 exons and                                system also includes the GFRa1–4 alternative
generates a transcript subjected to alternative splicing                  co-receptors and GDNF family ligands, including
leading to two main isoforms: a protein of 1114                           GDNF, neurturin, persephin, and artemin (reviewed
residues displaying 51 C-terminal-specific amino acids                    in Arighi et al. (2005)).
Endocrine-Related Cancer (2011) 18 519–527                                                         Downloaded from Bioscientifica.com at 09/25/2020 03:50:51PM
                                                                                                                      DOI: 10.1530/ERC-10-0306
                                                                                                                                                 via free access
1351–0088/11/018–519 q 2011 Society for Endocrinology Printed in Great Britain           Online version via http://www.endocrinology-journals.org
M G Borrello et al.: Germline RET K666E and G691S mutations

A                       B                    Lys666Glu                      C                                 RET9
                            150                 160                  170

                                                                                                               1S -
                                                                                                            69 E
                                                                                                           G 666
                                                                                                               8T
                                                                                                       R

                                                                                                               6E

                                                                                                               1S
                                                                                          k

                                                                                                     34

                                                                                                              K
                                                                                                             91
                                                                                        oc

                                                                                                            66

                                                                                                            69
                                                                                                   C6
                                                                                       t
                                                                                      M

                                                                                                           M
              CDL

                                                                                      w

                                                                                                           K

                                                                                                           G
                                                                           RET

                                                                           pERK1/2
    C634      CR
                                                                           ERK1/2
              TM
    K666
    G691      JM                             Gly691Ser
                                      230                  240

              TK
 M918

 RET9
            RET51

Figure 1 (A) Schematic representation of RET protein. The codons mutated in the patient (K666 and G691) and the codons
corresponding to MEN2A RET-C634R and MEN2B RET-M918T mutations are indicated. CL, cadherin-like domain; CR, Cys-rich
domain; TM, transmembrane domain; JM, juxtamembrane domain; TK, tyrosine kinase domain. (B) Sequence electropherogram for
missense mutations found in exon 11. The mutations are marked by arrows. (C) Western blot analysis of RET protein and associated
ERK signaling of transiently transfected RET gene variants RET-K666E, RET-G691S, and RET-K666E–G691S. Lysates from
HEK293T cells transiently transfected with recombinant plasmids of the indicated RET mutants were analyzed by western blot with
anti-RET, anti p-ERK1/2, and anti-ERK1/2 antibodies.

   RET is involved in thyroid neoplasias through                  largely exploited to optimize the diagnostic and
different gain-of-function alterations. Somatic                   clinical management of hereditary MTC (Chiefari
rearrangements of RET are present in about one-third              et al. 1998, Marx 2005, Gagel & Marx 2008). The
of papillary thyroid carcinomas (reviewed in Greco                presence of a known MEN2-associated RET mutation
et al. (2009)), whereas specific germline activating              in the germline of an MTC patient identifies hereditary
mutations of RET are causally involved in hereditary              MTC disease, thus allowing preclinical identification
medullary thyroid carcinoma (MTC), including                      of family members at risk of developing MTC, as well
multiple endocrine neoplasia type 2A (MEN2A,                      as providing information about the risk of the proband
OMIM 171400), type 2B (MEN2B, OMIM 162300),                       developing other tumors associated with MEN2
and familial MTC (FMTC; OMIM 155240) syndromes                    syndromes. Moreover, the RET mutation type, assessed
(Lodish & Stratakis 2008). Activating RET mutations               by RET genetic testing, guides clinical decisions, as
involved in hereditary MTC forms are also present in a            different RET mutants have been associated with
fraction of sporadic MTCs.                                        different risk profiles. Consequently, prophylactic
   MTC, which accounts for around 5% of thyroid                   thyroidectomy for asymptomatic mutation carriers is
tumors, is a neuroendocrine neoplasia arising from the            recommended at early ages (from the first months of
thyroid parafollicular C cells. Physiologically, RET,             life to 5 years) for higher risk mutations (codons
GFRa4, and persephin appear to be necessary for                   883/918 and 634 respectively), whereas for milder
migration of neural crest calcitonin producing cells              mutations surgery in carriers may be delayed
into the developing thyroid gland: markedly dimin-                (Frank-Raue et al. 2010). Moreover, the identification
ished parafollicular C cells are found in RETK/K mice             and functional characterization of novel activating
(Lindahl et al. 2000, Lindfors et al. 2006).                      RET mutations responsible for the disease contributes
   MTC is relatively unresponsive to radiotherapy and             to the knowledge of genetic–phenotypic relationships
conventional chemotherapy: thyroidectomy is the                   in hereditary MTC.
standard treatment and the only curative therapy                     In this study, we report the identification, in a
(Wells & Santoro 2009). RET mutation analysis is                  48-year-old woman carrying unicentric MTC with
                                                                                          Downloaded from Bioscientifica.com at 09/25/2020 03:50:51PM
                                                                                                                                        via free access
520                                                                                                   www.endocrinology-journals.org
Endocrine-Related Cancer (2011) 18 519–527

C-cell hyperplasia, of the RET-K666E germline variant       tumors/disorders, or pheochromocytomas in her
as well as two polymorphisms: the G691S non-                family. Her father died at the age of 66 years from
synonymous variant and the conservative S904S               liver cirrhosis. Her mother was healthy at the age of 82
(TCC/TCG) variant. K666E and G691S RET                      years and her brother at the age of 52 years. Her sister
mutations occur in the juxtamembrane domain, a              developed breast cancer at the age of 50 years. The
region not frequently affected by mutations. In this        patient had no offspring. The patient and her sister
study, the co-segregation of the K666E mutation with        provided a written informed consent for RET mutation
MTC in our patient’s family was not verifiable.             analysis and for research purposes approved by the
However, K666E has previously been reported in              local ethical committee. To date, no other family
three unrelated families and it is thought to be a          members have consented to testing.
pathogenic mutation (Ahmed et al. 2005). In this study,
we have demonstrated the in vitro oncogenic potential       Mutation analysis
associated with the RET-K666E mutant, based on its
                                                            DNA was extracted from peripheral venous blood using
ability to transform NIH3T3 cells, thus suggesting that
                                                            a commercial kit (Qiagen Blood Extraction Kit) as
this RET variant is indeed responsible for hereditary
                                                            recommended by the manufacturer. The screening for
MTC. Moreover, our results, showing that the
                                                            germline RET gene mutations was carried out in exons
oncogenic activity of RET-K666E is enhanced by the
                                                            8, 10, 11, 13, 14, 15, and 16, where most known
presence of the polymorphism G691S, suggest a
                                                            mutations take place. RET exons 10, 11, 13, and 14 were
possible modifier role for this known non-synonymous
                                                            analyzed by double-gradient denaturing gradient gel
RET polymorphism.
                                                            electrophoresis (DG-DGGE) with primers and con-
                                                            ditions previously described (Hofstra et al. 1996). RET
                                                            exons 8, 15, and 16 (together with exon 11 that showed a
Materials and methods                                       DG-DGGE migration pattern suggestive of base
Patient presentation and analysis                           substitution) were amplified with exon flanking primers
                                                            and analyzed by automated sequencing (ABIprism 377;
The patient, a 48-year-old woman, underwent neck
                                                            Applied Biosystems, Foster City, CA, USA). The
ultrasound after clinical demonstration of cervical
                                                            amplified exon 11 PCR product was cloned in pCRII-
lymphadenomegaly. At the inferior region of the left
                                                            TOPO vector (Invitrogen) according to the manufac-
thyroid lobe, a 6!5!5 mm nodule with irregular
                                                            turer’s instructions, and single clones were sequenced.
margins was detected. A fine needle aspiration biopsy
gave a cytological diagnosis of follicular proliferation
                                                            Construction of the RET mutants
with oxyphilic metaplasia, and for this reason, a left
hemithyroidectomy was performed. The histological           Recombinant plasmids carrying RET9-WT (the short
examination, however, showed that the patient dis-          isoform of the proto-RET gene), RET51-WT (the long
played a unicentric MTC of 5 mm, associated with            isoform of the proto-RET gene), RET9-C634R
C-cell hyperplasia, and vascular invasion. Before           (containing an MEN2A causing mutation), and
completing the thyroidectomy, abdomen computed              RET9-M918T (containing the main MEN2B causing
tomography (CT) and 24 h urinary catecholamine              mutation) are described elsewhere (Arighi et al. 2004).
determination were performed in order to exclude the        Site-directed mutagenesis was performed, using an
presence of pheochromocytoma. In addition, C-cell           in vitro oligonucleotide mutagenesis system (Quik-
hyperplasia and lymphocytic chronic thyroiditis were        Change XL site-directed mutagenesis; Stratagene, La
histologically detected in the right lobe. The patient is   Jolla, CA, USA), on RET9-WT and RET51-WT
currently in active follow-up, which includes laboratory    cDNAs cloned in the eukaryotic expression vector
tests (e.g. s-calcitonin; carcinoembryonic antigen          pCDNA3 (Invitrogen), to obtain RET9-K666E,
(s-CEA), s-chromogranin A; s-calcium; s-parathyroid         RET51-K666E, RET9-G691S and RET51-G691S
hormone (s-PTH), s-25(OH)D, u-catecholamines) and           constructs. The K666E single mutants were subjected
instrumental examination (e.g. neck and abdomen             to a second cycle of site-directed mutagenesis to obtain
ultrasound; chest and abdomen CT). At present, 3            the double mutants RET9-K666E–G691S and RET51-
years after surgery, these diagnostic procedures provide    K666E–G691S. The mutant clones were identified by
no evidence of clinical recurrence or metastases.           direct sequencing and then entirely sequenced to
   The patient attended genetic counseling and family       exclude possible additional mutations. Plasmid DNA
history data were collected. The clinical investiga-        was extracted using the MAXI PREP Kit (Qiagen) as
tion did not reveal any history of MTC, thyroid             suggested by the supplier.
                                                                               Downloaded from Bioscientifica.com at 09/25/2020 03:50:51PM
                                                                                                                             via free access
www.endocrinology-journals.org                                                                                             521
M G Borrello et al.: Germline RET K666E and G691S mutations

Cell culture, transfections, focus formation                  Results
assay, and anchorage-independent assay
                                                              Patient’s and family’s RET mutation analysis
Human HEK293T cells were maintained in DMEM
                                                              DG-DGGE analysis and/or direct DNA sequencing
with 10% FCS. The recombinant plasmids were
                                                              after PCR amplification of RET exons 8, 10, 11, 13, 14,
transfected using Lipofectamine 2000 (Invitrogen)
                                                              15, and 16, from the patient’s peripheral blood, has
according to the manufacturer’s instructions.
                                                              shown three variations in the RET gene sequence: a
   NIH3T3 cells were cultured in DMEM supplemented
                                                              c.1996AOG transition in heterozygosis, leading to the
with 10% FCS. Stable transfection of NIH3T3 cells was
                                                              missense K666E mutation, a c.2071GOA transversion
performed using the CaPO4 method (Bongarzone et al.
                                                              in heterozygosis, leading to a missense mutation
1989) and using 250 ng plasmid DNA together with
                                                              corresponding to the known non-conservative functional
30 mg NIH3T3-derived DNA carrier. Transfected cells
                                                              G691S polymorphism (SNIP rs1799939), both in exon
were grown in DMEM with 10% FCS and selected in
                                                              11 (Fig. 1B), and a c.2712COG transversion, a silent
the presence of G418 antibiotic (650 mg/ml) to
                                                              mutation leading to the conservative S904S polymorphic
determine the transfection efficiency; transforming
                                                              variant (SNIP rs1800863) in exon 15 (not shown). The
activity was determined in medium containing 5%
                                                              patient’s sister, the only family member who gave
serum in the presence or absence of 10 ng/ml GDNF
                                                              informed consent, carried the G691S polymorphism
(AlomoneLabs, Jerusalem, Israel). Both G418-resistant
                                                              but not the K666E genetic variant (not shown).
colonies and transformed foci were fixed and counted.
                                                                 Most mutations associated with hereditary MTC map
   For anchorage-independent assay, cell lines NIH3T3
                                                              in the Cys-rich region, such as the classical MEN2A-
expressing RET mutants were suspended in DMEMC
                                                              associated C634R mutation, or in the kinase domain,
10% FCS and plated at 1000–10 000 cells per 60 mm
                                                              such as the prevalent MEN2B-associated mutation
dish in the same medium 0.33% final agar. After
                                                              M918T (Fig. 1A); at variance, the RET-K666E variant
2 weeks, the colonies were stained with p-iodonitrote-
                                                              maps in the juxtamembrane region of the receptor.
trazolium violet, photographed, and counted.
                                                                 To assess whether the putative MTC-associated
                                                              K666E mutation and the polymorphic G691S mutation
Western blot analysis                                         carried by the proband were on the same allele, the
Cells were serum-starved for 24 h, then lysed in              amplified exon 11 PCR product was cloned in pCR2.1-
ice-cold RIPA buffer (20 mM Tris, pH 7.4, 137 mM              TOPO vector. A total of 11 clones were sequenced: six
NaCl, 10% glycerol, 0.1% SDS, 0.5% sodium deox-               presented both wt codons and five presented both
ycholate, 1% Triton X-100, 2 mM EDTA, pH 8.0)                 mutated codons, thus indicating that the two mutations
supplemented with protease/phosphatase inhibitors.            are on the same allele.
Protein samples were boiled in NuPAGE LDS sample
buffer (Invitrogen), separated by 7% NuPAGE Novex             Transforming activity of RET-K666E, RET-G691S,
Bis–Tris Gels with the appropriate running buffer             and RET-K666E/G691S gene variants
(Invitrogen), then transferred onto nitrocellulose filters,
and immunoblotted with the indicated antibodies. For          Site-directed mutagenesis was performed on pCDNA3
densitometric analyses, blots were analyzed using the         eukaryotic expression vector carrying the two RET
Image Quant 5.2 Software (Molecular Dynamics, GE              isoforms (RET9 or RET51) to obtain the following
Healthcare, CA, USA). The following antibodies were           mutants: RET9-K666E, RET51-K666E, RET9-G691S,
used in blotting experiments: anti-RET (C-19) from            RET51-G691S, RET9-K666E–G691S, and RET51-
Santa Cruz Biotechnology (Santa Cruz, CA, USA),               K666E–G691S.
anti-phospho-RET (Tyr905) from Cell Signaling Tech-              To assess the quality and efficiency of recombinant
nologies (Beverly, MA, USA), anti-phospho-Tyr 4G10            plasmids, all the constructs were transiently transfected
from Upstate Biotechnology (Lake Placid, NY, USA)             into HEK293T cells to analyze RET protein
anti-MAP kinase (ERK1/2), and anti-MAP kinase                 expression, as well as their associated downstream
activated (pERK1/2) from Sigma–Aldrich.                       signaling on ERK1/2, as read-out of RET activation, in
                                                              comparison with RET-wt, and the MEN2A- and
                                                              MEN2B-associated RET-C634R and RET-M918T
Statistical analysis
                                                              mutants (Fig. 1C). All the constructs express proteins
Excel software was used to analyze all the data.              of the expected molecular weight. RET protein
Significance of differences was determined by two-            displaying K666E amino acid variant shows a stronger
tailed Student’s t-test with unequal variance.                ERK phosphorylation than RET-wt. The RET protein
                                                                                    Downloaded from Bioscientifica.com at 09/25/2020 03:50:51PM
                                                                                                                                  via free access
522                                                                                             www.endocrinology-journals.org
Endocrine-Related Cancer (2011) 18 519–527

encoded by the double mutants, RET K666E–G691S,                                          significantly lower than RET carrying the classical
shows an even stronger ERK phosphorylation, similar                                      C634R mutation (P!0.01). At variance, no transform-
to mutant proteins encoded by RET-C634R and RET-                                         ing activity was associated with the non-synonymous
M918T controls. The respective long isoforms behave                                      polymorphic variant G691S carried by either RET
similarly (data not shown). This preliminary analysis                                    isoforms. Interestingly, RET gene isoforms carrying
prompted us to analyze the biological activity of either                                 both variants RET9- and RET51-K666E–G691S,
single- or double-mutated RET forms.                                                     mimicking the mutated allele of the patient, proved
   To define the oncogenic potential of the K666E                                        to be significantly more efficient (P!0.01) in focus
mutation, the RET-K666E constructs, together with                                        formation compared with the respective single mutants
vectors expressing the short and the long isoforms of                                    (Fig. 2A). Moreover, RET9-K666E–G691S generates
RET-wt and RET-C634R, were stably transfected in                                         foci larger than single mutant RET9-K666E and more
NIH3T3 cells for a focus formation assay in the                                          similar to those obtained with C634R mutation, as
presence or absence of chronic stimulation with the                                      shown in representative plates for each RET mutant
RET ligand GDNF. G418 selection was also performed                                       (Fig. 2B). Similar data, but with lower foci numbers
to normalize the results. Both isoforms of RET were                                      (about 1/4), were obtained without GDNF ligand
tested, as we have demonstrated that long isoforms of                                    stimulation (data not shown).
oncogenic RETs display a stronger transforming                                              Several NIH3T3-transformed cell lines carrying
activity compared with the short ones (Borrello et al.                                   RET-K666E or RET-K666E–G691S mutants were
2002), thus enhancing the possibility of identifying                                     isolated and analyzed. At variance with RET-wt
mild activating mutations. The results of three                                          expressing cells, all the NIH3T3 cells expressing
transfections, performed in triplicate and normalized                                    RET mutants are able to grow in soft agar. The
on the number of NIH3T3 G418-resistant colonies, are                                     RET-K666E–G691S agar clones were generally
shown in Fig. 2. Both RET isoforms carrying the                                          bigger than the RET-K666E ones and comparable to
K666E mutation show transforming activity, although                                      RET-C634R clones (Fig. 2C).

                                                                                                                   *
                      A 4.5                                                                             *
                                                     *P
M G Borrello et al.: Germline RET K666E and G691S mutations

                            K666E           K666E–G691S          Discussion
                   Mock 1   2   3   4   5     6    7      8      A complete classification and functional character-
RET                                                              ization of all RET mutations involved in inherited
                                                                 MTC is essential for an effective genetic analysis in
pTyr-905
                                                                 clinical practice. Timely thyroidectomy in family
pERK1/2                                                          members carrying an inherited mutated RET allele
                                                                 prevents MTC, the most common cause of death in
ERK1/2                                                           MEN2 syndromes (Modigliani et al. 1998).
                                    *P=0.02                         In this study, we report the identification of three
             1.2                                                 RET variants in an MTC patient: the K666E mutation,
               1                                                 the non-synonymous polymorphism G691S, and
  pERK/ERK

             0.8                                                 the conservative polymorphism S904S. Through
             0.6                                                 functional analysis of the corresponding RET mutants,
             0.4                                                 we have demonstrated the in vitro oncogenic potential
             0.2                                                 associated with RET-K666E, based on its ability to
               0
                                                                 transform NIH3T3 cells, thus suggesting that this RET
                                                                 variant is indeed responsible for hereditary MTC.
Figure 3 Western blot analysis of RET protein and associated
signaling in NIH3T3 cells stably expressing RET gene variants    Moreover, we have shown that the oncogenic activity
RET9-K666E and RET9-K666E–G691S. Lysates from                    of RET-K666E is enhanced by the presence of G691S,
NIH3T3 cell lines were analyzed by western blot with anti-RET,   suggesting a possible modifier role of this non-
anti-p-RET (Tyr905), anti-ERK1/2, and anti-p-ERK1/2
antibodies. *Statistically significant result.                   synonymous RET polymorphism.
                                                                    Sequence variations in the intracellular juxtamem-
                                                                 brane domain of RET have been infrequently reported
   The same NIH3T3-derived cell lines were analyzed              and affect codons 665 and 666 (Ahmed et al. 2005,
for RET protein expression and Tyr905 phospho-                   Cordella et al. 2006, Muzza et al. 2010). Both K666N
rylation, as well as for ERK1/2 phosphorylation, as              and the complex K666NinsS mutations have been
read-out of RET receptor activation (Fig. 3). All the            shown to display oncogenic potential (Cordella et al.
analyzed transformed cell lines, either expressing               2006, Muzza et al. 2010). In particular, K666N and
RET-K666E–G691S or RET-K666E, show different                     K666E mutants show a comparable transforming
levels of RET protein and are able to elicit a stronger          activity; in fact, in both cases, their transforming activity
ERK1/2 phosphorylation than the mock-transfected                 is one half compared to that of the C634R mutant
NIH3T3 cells. The cell lines expressing RET-K666E–               (Muzza et al. 2010). Moreover, biochemical analysis of
G691S receptor are significantly more efficient in               K666E shows higher amounts of fully glycosylated
ERK1/2 phosphorylation (PZ0.02) than those expres-               mutant receptor compared to RET-wt and other mutants.
sing RET-K666E receptor, in keeping with the                     This feature was also previously displayed by the
described biological effect of the respective RET                K666NinsS mutant (Cordella et al. 2006).
mutants. RET-K666E clones show a certain correlation                The K666E variant has already been described in
between RET expression and phosphorylation (eval-                three unrelated families (Ahmed et al. 2005) and it is
uated either as total Tyr- (data not shown) or as Tyr905-        reported as a pathogenic mutation in the web-based
phosphorylation) and ERK1/2 activation levels.                   ARUP online Scientific Resource RET database http://
At variance, ERK1/2 activation is high in all the RET-           www.arup.utah.edu/database/MEN2/MEN2_search.
K666E–G691S expressing clones, despite the different             php (Margraf et al. 2009). Pathogenicity was deduced
levels of RET protein and autophosphorylation, thus              from clinical and segregation analysis, although
highlighting the capability of double mutant to strongly         penetrance was incomplete and one of the MEN2A/
activate ERK1/2, even at low expression level, as                FMTC families had one discordant member displaying
shown by clone 5.                                                no mutation and a positive pentagastrin stimulation. A
   Overall, our in vitro analyses suggest that K666E             functional analysis for the RET-K666E variant has not
is a gain-of-function alteration of RET receptor kinase          been performed earlier. We have now demonstrated for
with a weaker oncogenic potential than the MEN2A-                the first time that K666E is a gain-of-function mutation
associated C634R mutation; however, its oncogenic                with oncogenic potential, as it displays transforming
ability is enhanced by the presence of the G691S                 activity by NIH3T3 focus formation assay. It is not
polymorphism.                                                    known whether K666E is a de novo or inherited RET
                                                                                        Downloaded from Bioscientifica.com at 09/25/2020 03:50:51PM
                                                                                                                                      via free access
524                                                                                                 www.endocrinology-journals.org
Endocrine-Related Cancer (2011) 18 519–527

variation in the presented patient, as the family history      The RET-G691S polymorphism has been suggested
was negative for MTC, and the carrier status of family      as a genetic modifier in MEN2A (Gil et al. 2002),
members could be tested only for the patient’s sister,      but this issue is controversial (Lesueur et al. 2006).
who proved negative for the mutation.                       Conflicting results are also reported with regard to a
   However, the RET-K666E variant has a clear-cut           possible role for this polymorphism in susceptibility to
transforming activity, displayed by short and long          sporadic MTC ((Fugazzola et al. 2008) and references
isoforms of RET and in the presence/absence of the          herein). Interestingly, the G691S polymorphism has
RET ligand GDNF. This is consistent with the fact that      been suggested to play a role in a specific mutation
the presence of lysine residue at codon 666 is highly       affecting K666 residue. In fact, a complex in frame
conserved among species, and with a recent report           insertion–deletion mutation (insTTGTdelG) at codon
showing the presence of the wt codon for Lys in a series    666 (resulting in replacement of the lysine by
of 400 normal control alleles (Muzza et al. 2010).          asparagine and serine insertion) was detected in the
   Our in vitro studies show a milder transforming          germline of a 12-year-old boy displaying MTC, and in
activity of the K666E mutation compared with the            his unaffected mother and maternal grandfather.
classical MEN2A-associated C634R mutation. The              Interestingly, the MTC-affected boy, but not the
clinical data obtained from three families (Ahmed et al.    unaffected family members, displayed the G691S
2005), along with the patient described in this study,      polymorphic variant allele inherited from his father
showed that among 15 carriers of the K666E variant,         (Vandenbosch et al. 2005). Thus, the G691S poly-
five developed MTC (35 to 64 years old), one                morphism seems to enhance RET oncogenicity of
presented pheochromocytoma and elevated calcitonin          distinct mutations affecting RET codon 666, by
at 35 years old, two developed C cell hyperplasia at 35     increasing its penetrance in the clinical onset
and 71 years old, and seven remained asymptomatic           (Vandenbosch et al. 2005), and its transforming
individuals (2–66 years old). Altogether, this varia-       activity in in vitro assay (our study). Overall, these
bility suggests that K666E could be a mild mutation         data strengthen the potential role of the RET-G691S
with variable expressivity and penetrance. The              polymorphism in MTC. This is the first demonstration
presence of modifier genes or of an undetected              of an in vitro effect of the G691S variant on the
segregating mutation could explain the segregation          transforming activity of a RET mutant. This effect may
pattern observed in these families. RET polymorphic         be related to the capacity of the polymorphic variant
variants might be possible modifiers contributing to the    G691S to enhance ERK1/2 activation triggered by
variable penetrance and expressivity associated with        RET-K666E mutant receptor, demonstrated by our
K666E mutation (see below).                                 biochemical analysis of NIH3T3 cell lines expressing
   The patient in this study presents the non-              RET-K666E or RET-K666E–G691S, and previously
synonymous polymorphism RET-G691S in the same               shown for RET-wt (Sawai et al. 2005, Narita et al. 2009).
allele carrying the K666E mutation. RET-G691S is               It remains to be investigated whether the effect of
a common polymorphic variant with a reported                G691S on in vitro transforming activity is relevant for
allele frequency of 11–33% (ARUP database). It was          all the gain-of-function RET mutations causing
shown that RET-G691S is able to increase downstream         hereditary MTC. Different mutations might be differ-
signaling compared with RET-wt (Sawai et al. 2005,          entially affected by the polymorphism, depending on
Narita et al. 2009). However, this variant is not           their ‘strength’. Alternatively, as the variation at 691
oncogenic in focus formation assays, as previously          residue has been hypothesized to modify the protein
shown by Fugazzola et al. (2008) for the RET9               structure (Lesueur et al. 2006), it might differentially
isoform and now confirmed in this paper for both            synergize with specific gain-of-function mutations.
RET isoforms. Even in the presence of GDNF ligand,          Future research on this issue could contribute toward
no valuable transforming activity different from            clarifying the controversial role of G691S as a genetic
RET-wt background was appreciated. Regarding                modifier. It is worth noting that strong modifying
S904S polymorphism, also detected in the presented          effects of different RET variations on the oncogenic
patient, strong co-segregation with G691S poly-             RET-V804M mutant have been reported. In particular,
morphism has already been demonstrated (Gil et al.          the double mutants V804M–E805K, V804M–Y806C,
2002, Robledo et al. 2003). Both polymorphisms have         and V804M–S940C, at variance with V804M
been shown to be under-represented in Hirschsprung’s        causing FMTC, have been found in MEN2B patients
disease (HSCR, OMIM 142623) patients compared               (Miyauchi et al. 1999, Menko et al. 2002, Cranston
with controls, thus suggesting that they might protect      et al. 2006). Thus, the risk level associated with the
against the development of HSCR (Borrego et al. 1999).      double mutants is significantly higher (Kloos et al.
                                                                                Downloaded from Bioscientifica.com at 09/25/2020 03:50:51PM
                                                                                                                              via free access
www.endocrinology-journals.org                                                                                              525
M G Borrello et al.: Germline RET K666E and G691S mutations

2009). The V804M–V778I double mutant has also                 Arighi E, Borrello MG & Sariola H 2005 RET tyrosine
been reported, and it has been classified at higher risk         kinase signaling in development and cancer. Cytokine &
level than V804M (Kasprzak et al. 2001). However, at             Growth Factor Reviews 16 441–467. (doi:10.1016/j.
variance with our report, in these cases, the mutations          cytogfr.2005.05.010)
in tandem with V804M are classified as ‘uncertain’            Bongarzone I, Pierotti MA, Monzini N, Mondellini P,
variants and not as polymorphic variants, and in some            Manenti G, Donghi R, Pilotti S, Grieco M, Santoro M,
                                                                 Fusco A et al. 1989 High frequency of activation of
cases, they display in vitro transforming activity
                                                                 tyrosine kinase oncogenes in human papillary thyroid
(ARUP database).
                                                                 carcinoma. Oncogene 4 1457–1462.
   In conclusion, we have demonstrated the in vitro
                                                              Borrego S, Saez ME, Ruiz A, Gimm O, Lopez-Alonso M,
oncogenic potential of a mutation located in the                 Antinolo G & Eng C 1999 Specific polymorphisms in the
juxtamembrane domain of RET detected in the germ-                RET proto-oncogene are over-represented in patients with
line of three families and in our patient. Moreover, we          Hirschsprung disease and may represent loci modifying
have demonstrated for the first time the possibility that        phenotypic expression. Journal of Medical Genetics 36
the in vitro oncogenic potential of a given gain of              771–774. (doi:10.1136/jmg.36.10.771)
function RET mutation might be enhanced by the                Borrello MG, Mercalli E, Perego C, Degl’Innocenti D,
presence of the functional non-oncogenic poly-                   Ghizzoni S, Arighi E, Eroini B, Rizzetti MG & Pierotti
morphism G691S, thus suggesting a modifier role for              MA 2002 Differential interaction of enigma protein with
this polymorphic RET variant. Our results also suggest           the two RET isoforms. Biochemical and Biophysical
that integration of patients/families genotyping with            Research Communications 296 515–522. (doi:10.1016/
functional characterization of RET mutants can provide           S0006-291X(02)00886-0)
useful information to optimize the diagnostic and             Chiefari E, Russo D, Giuffrida D, Zampa GA, Meringolo D,
clinical management of hereditary MTC.                           Arturi F, Chiodini I, Bianchi D, Attard M, Trischitta V
                                                                 et al. 1998 Analysis of RET proto-oncogene abnormal-
                                                                 ities in patients with MEN2A, MEN2B, familial or
Declaration of interest                                          sporadic medullary thyroid carcinoma. Journal of
The authors declare that there is no conflict of interest        Endocrinological Investigation 21 358–364.
that could be perceived as prejudicing the impartiality of    Cordella D, Muzza M, Alberti L, Colombo P, Travaglini P,
the research reported.                                           Beck-Peccoz P, Fugazzola L & Persani L 2006 An
                                                                 in-frame complex germline mutation in the juxtamem-
                                                                 brane intracellular domain causing RET activation in
Funding                                                          familial medullary thyroid carcinoma. Endocrine-Related
This work was partially supported by grants from Associa-        Cancer 13 945–953. (doi:10.1677/erc.1.01144)
zione Italiana per la Ricerca sul Cancro (AIRC), Fondazione   Cranston A, Carniti C, Oakhill K, Radzio-Andzelm E, Stone
Guido Berlucchi, Institutional Strategic Projects ‘contri-       EA, McCallion AS, Hodgson S, Clarke S, Mondellini P,
bution 5 per mille’ Fondazione IRCCS Istituto Nazionale          Leyland J et al. 2006 RET is constitutively activated by
Tumori.                                                          novel tandem mutations that alter active site resulting in
                                                                 MEN2B. Cancer Research 66 10179–10187. (doi:10.
                                                                 1158/0008-5472.CAN-06-0884)
Acknowledgements                                              Frank-Raue K, Rondot S & Raue F 2010 Molecular genetics
We thank Silvia Grassi for secretarial assistance and            and phenomics of RET mutations: impact on prognosis of
Dr Laura Fugazzola for helpful discussion.                       MTC. Molecular and Cellular Endocrinology 322 2–7.
                                                                 (doi:10.1016/j.mce.2010.01.012)
                                                              Fugazzola L, Muzza M, Mian C, Cordella D, Barollo S,
References                                                       Alberti L, Cirello V, Dazzi D, Girelli ME, Opocher G
Ahmed SA, Snow-Bailey K, Highsmith WE, Sun W,                    et al. 2008 RET genotypes in sporadic medullary thyroid
   Fenwick RG & Mao R 2005 Nine novel germline gene              cancer: studies in a large Italian series. Clinical
   variants in the RET proto-oncogene identified in twelve       Endocrinology 69 418–425. (doi:10.1111/j.1365-2265.
   unrelated cases. Journal of Molecular Diagnostics 7           2008.03218.x)
   283–288. (doi:10.1016/S1525-1578(10)60556-9)               Gagel RF & Marx SJ 2008 Multiple endocrine neoplasia. In
Arighi E, Popsueva A, Degl’Innocenti D, Grazia BM, Carniti       Williams Textbook of Endocrinology, pp 1705–1746. Eds
   C, Perala NM, Pierotti MA & Sariola H 2004 Biological         HM Kronenberg, J Melamed, K Polonsky & P Larsen.
   effects of the dual phenotypic janus mutation of RET          Philadelphia, PA, USA: Saunders, Elsevier Science.
   co-segregating with both multiple endocrine neoplasia      Gil L, Azanedo M, Pollan M, Cristobal E, Arribas B, Garcia-
   type 2 and Hirschsprung’s disease. Molecular Endo-            Albert L, Garcia-Saiz A, Maestro ML, Torres A,
   crinology 18 1004–1017. (doi:10.1210/me.2003-0173)            Menarguez J et al. 2002 Genetic analysis of RET, GFR
                                                                                     Downloaded from Bioscientifica.com at 09/25/2020 03:50:51PM
                                                                                                                                   via free access
526                                                                                              www.endocrinology-journals.org
Endocrine-Related Cancer (2011) 18 519–527

   alpha 1 and GDNF genes in Spanish families with            Menko FH, Van der Luijt RB, de Valk IA, Toorians AW,
   multiple endocrine neoplasia type 2A. International          Sepers JM, van Diest PJ & Lips CJ 2002 Atypical MEN
   Journal of Cancer 99 299–304. (doi:10.1002/ijc.10298)        type 2B associated with two germline RET mutations on
Greco A, Borrello MG, Miranda C, Degl’Innocenti D &             the same allele not involving codon 918. Journal of
   Pierotti MA 2009 Molecular pathology of differentiated       Clinical Endocrinology and Metabolism 87 393–397.
   thyroid cancer. Quarterly Journal of Nuclear Medicine        (doi:10.1210/jc.87.1.393)
   and Molecular Imaging 53 440–453.                          Miyauchi A, Futami H, Hai N, Yokozawa T, Kuma K, Aoki
Hofstra RM, Sijmons RH, Stelwagen T, Stulp RP, Kousseff         N, Kosugi S, Sugano K & Yamaguchi K 1999 Two
   BG, Lips CJ, Steijlen PM, Van Voorst Vader PC &              germline missense mutations at codons 804 and 806 of the
   Buys CH 1996 RET mutation screening in familial              RET proto-oncogene in the same allele in a patient with
   cutaneous lichen amyloidosis and in skin amyloidosis         multiple endocrine neoplasia type 2B without codon 918
   associated with multiple endocrine neoplasia. Journal of     mutation. Japanese Journal of Cancer Research 90 1–5.
   Investigative Dermatology 107 215–218. (doi:10.1111/         (doi:10.1111/j.1349-7006.1999.tb00658.x)
   1523-1747.ep12329651)                                      Modigliani E, Cohen R, Campos JM, Conte-Devolx B, Maes
Kasprzak L, Nolet S, Gaboury L, Pavia C, Villabona C,           B, Boneu A, Schlumberger M, Bigorgne JC, Dumontier P,
   Rivera-Fillat F, Oriola J & Foulkes WD 2001 Familial         Leclerc L et al. 1998 Prognostic factors for survival and
   medullary thyroid carcinoma and prominent corneal            for biochemical cure in medullary thyroid carcinoma:
   nerves associated with the germline V804M and V778I          results in 899 patients. The GETC Study Group. Groupe
   mutations on the same allele of RET. Journal of Medical      d’etude des tumeurs a calcitonine. Clinical Endocrinology
   Genetics 38 784–787. (doi:10.1136/jmg.38.11.784)             48 265–273. (doi:10.1046/j.1365-2265.1998.00392.x)
Kloos RT, Eng C, Evans DB, Francis GL, Gagel RF, Gharib       Muzza M, Cordella D, Bombled J, Bressac-de PB, Guizzardi
   H, Moley JF, Pacini F, Ringel MD, Schlumberger M et al.      F, Francis Z, Beck-Peccoz P, Schlumberger M, Persani L
   2009 Medullary thyroid cancer: management guidelines         & Fugazzola L 2010 Four novel RET germline variants in
   of the American Thyroid Association. Thyroid 19              exons 8 and 11 display an oncogenic potential in vitro.
   565–612. (doi:10.1089/thy.2008.0403)                         European Journal of Endocrinology 162 771–777.
Lesueur F, Cebrian A, Robledo M, Niccoli-Sire P, Svensson       (doi:10.1530/EJE-09-0929)
   KA, Pinson S, Leyland J, Whittaker J, Pharoah PD &         Narita N, Tanemura A, Murali R, Scolyer RA, Huang S,
   Ponder BA 2006 Polymorphisms in RET and its                  Arigami T, Yanagita S, Chong KK, Thompson JF, Morton
   coreceptors and ligands as genetic modifiers of multiple     DL et al. 2009 Functional RET G691S polymorphism in
   endocrine neoplasia type 2A. Cancer Research 66              cutaneous malignant melanoma. Oncogene 28
   1177–1180. (doi:10.1158/0008-5472.CAN-05-2995)               3058–3068. (doi:10.1038/onc.2009.164)
Lindahl M, Timmusk T, Rossi J, Saarma M & Airaksinen MS       Robledo M, Gil L, Pollan M, Cebrian A, Ruiz S, Azanedo M,
   2000 Expression and alternative splicing of mouse Gfra4      Benitez J, Menarguez J & Rojas JM 2003 Polymorphisms
   suggest roles in endocrine cell development. Molecular       G691S/S904S of RET as genetic modifiers of MEN2A.
   and Cellular Neurosciences 15 522–533. (doi:10.1006/         Cancer Research 63 1814–1817.
   mcne.2000.0845)                                            Sawai H, Okada Y, Kazanjian K, Kim J, Hasan S, Hines OJ,
Lindfors PH, Lindahl M, Rossi J, Saarma M & Airaksinen          Reber HA, Hoon DS & Eibl G 2005 The G691S RET
   MS 2006 Ablation of persephin receptor glial cell line-      polymorphism increases glial cell line-derived neuro-
   derived neurotrophic factor family receptor alpha4           trophic factor-induced pancreatic cancer cell invasion by
   impairs thyroid calcitonin production in young mice.         amplifying mitogen-activated protein kinase signaling.
   Endocrinology 147 2237–2244. (doi:10.1210/en.2005-           Cancer Research 65 11536–11544. (doi:10.1158/0008-
   1620)                                                        5472.CAN-05-2843)
Lodish MB & Stratakis CA 2008 RET oncogene in MEN2,           Vandenbosch K, Renard M, Uyttebroeck A, Sciot R, Matthijs
   MEN2B, MTC and other forms of thyroid cancer. Expert         G & Legius E 2005 Medullary thyroid carcinoma in a
   Review of Anticancer Therapy 8 625–632. (doi:10.1586/        child with a new RET mutation and a RET polymorphism.
   14737140.8.4.625)                                            Genetic Counseling 16 95–100.
Margraf RL, Crockett DK, Krautscheid PM, Seamons R,           Wells SA Jr & Santoro M 2009 Targeting the RET pathway
   Calderon FR, Wittwer CT & Mao R 2009 Multiple                in thyroid cancer. Clinical Cancer Research 15
   endocrine neoplasia type 2 RET protooncogene database:       7119–7123. (doi:10.1158/1078-0432.CCR-08-2742)
   repository of MEN2-associated RET sequence variation
   and reference for genotype/phenotype correlations.
   Human Mutation 30 548–556. (doi:10.1002/humu.20928)        Received in final form 14 June 2011
Marx SJ 2005 Molecular genetics of multiple endocrine         Accepted 20 June 2011
   neoplasia types 1 and 2. Nature Reviews. Cancer 5          Made available online as an Accepted Preprint
   367–375. (doi:10.1038/nrc1610)                             20 June 2011

                                                                                  Downloaded from Bioscientifica.com at 09/25/2020 03:50:51PM
                                                                                                                                via free access
www.endocrinology-journals.org                                                                                                527
You can also read