ArticlePDF AvailableLiterature Review

Noonan Syndrome and Related Disorders: A Review of Clinical Features and Mutations in Genes of the RAS/MAPK Pathway

Authors:

Abstract and Figures

Noonan syndrome (NS) is one of the most common syndromes transmitted by a mendelian mode. In recent years, germline mutations that affect components of the RAS-MAPK (mitogen-activated protein kinase) pathway were shown to be involved in the pathogenesis of NS and four rare syndromes with clinical features overlapping with NS: Leopard syndrome, cardio-facio-cutaneous syndrome, Costello syndrome and neurofibromatosis type 1. Several hormones act through receptors that stimulate the RAS-MAPK pathway, and therefore, NS and related disorders represent a remarkable opportunity to study the implication of the RAS-MAPK pathway in different endocrine systems. Additionally, children with NS frequently are referred to the endocrinologist because of short stature, delayed puberty and/or undescended testes in males. In this paper, we review the diagnostic, clinical and molecular aspects of NS and NS-related disorders.
Content may be subject to copyright.
Fax +41 61 306 12 34
E-Mail karger@karger.ch
www.karger.com
Mini Review
Horm Res 2009;71:185–193
DOI: 10.1159/000201106
Noonan Syndrome and Related Disorders:
A Review of Clinical Features and Mutations
in Genes of the RAS/MAPK Pathway
Alexander A.L. Jorge Alexsandra C. Malaquias Ivo J.P. Arnhold
Berenice B. Mendonca
Unidade de Endocrinologia do Desenvolvimento, Laboratorio de Hormonios e Genetica Molecular LIM/42,
Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de São Paulo, São Paulo , Brazil
Introduction
Noonan syndrome (NS; OMIM 163950) is one of the
most common syndromes transmitted by a mendelian
mode. The incidence of affected individuals is estimated
to be between 1:
1,000 and 1: 2,500 [1, 2] . NS is a clinically
heterogeneous disorder predominantly characterized by
dysmorphic facial features ( fig. 1 ), congenital heart dis-
ease (most commonly pulmonary valve stenosis, hyper-
trophic cardiomyopathy and atrial septal defects), pro-
portionate post-natal short stature, neck abnormalities
and chest deformities
[1, 3 , 4] ( table 1 ). Mild mental retar-
dation, bleeding diathesis, lymphedema, hearing difficul-
ty and cryptorchidism are also occasionally observed in
affected individuals. NS has an equal ma le to female ratio
[5, 6] . Familial cases correspond to approximately 20% of
the cases
[1] and presented mostly an autosomal domi-
nant inheritance with a near complete penetrance
[7] .
In recent years, germline mutations that affect compo-
nents of the RAS-MAPK (mitogen-activated protein ki-
nase) pathway were shown to be involved in the patho-
genesis of NS and of four rare syndromes with NS over-
lapping features: Leopard syndrome (OMIM 151100),
cardio-facio-cutaneous syndrome (CFC; OMIM 115150),
Costello syndrome (OMIM 218040) and neurofibroma-
tosis type 1 (NF1; OMIM 162200)
[8] .
Key Words
Noonan syndrome Genetics MAPK PTPN11 SOS1
RAF1 Short stature Genetics
Abstract
Noonan syndrome (NS) is one of the most common syn-
dromes transmitted by a mendelian mode. In recent years,
germline mutations that affect components of the RAS-
MAPK (mitogen-activated protein kinase) pathway were
sh ow n t o b e in vo lv ed i n t he pa tho ge ne si s o f NS an d f ou r r are
syndromes with clinical features overlapping with NS: Leop-
ard syndrome, cardio-facio-cutaneous syndrome, Costello
syndrome and neurofibromatosis type 1. Several hormones
act through receptors that stimulate the RAS-MAPK path-
way, and therefore, NS and related disorders represent a re-
markable opportunity to study the implication of the RAS-
MAPK pathway in different endocrine systems. Additionally,
children with NS frequently are referred to the endocrinolo-
gist because of short stature, delayed puberty and/or unde-
scended testes in males. In this paper, we review the diag-
nostic, clinical and molecular aspects of NS and NS-related
disorders. Copyr ight © 2009 S. Karger AG, B asel
Received: August 13, 2008
Accepted : October 30, 2008
Published online: March 4, 2009
H
O
RM
O
NE
RESEARCH
Alexander A .L. Jorge
Hospital das Clínic as, Laboratorio de Hormonios e G enetica Molecular
Av Dr Eneas d e Carvalho Aguiar 155 PAMB, 2 andar Bloc o 6
São Pau lo 05403-00 0 (Brazil)
Tel. +55 11 3069 7512, Fax +55 11 3069 7519, E-Mail a lexj@usp.br or beremen@usp.br
© 200 9 S. Karger AG, Basel
0301– 0163/09/0 714–0185 $26.0 0/0
Accessible online at:
www.karger.com/hre
Jorge/Malaquias/Arnhold/Mendonca
Horm Res 2009;71:185–193
186
Several hormones, including growth hormone (GH)
and insulin-like growth factor 1 (IGF-1), act through re-
ceptors that stimulate the RAS-MAPK pathway, and
therefore NS and related disorders represent a remarkable
opportunity to study the implication of the RAS-MAPK
pathway in different endocrine systems. Additionally,
children with NS are frequently referred to endocrinolo-
gists because of short stature, delayed puberty and/or un-
descended testes in males. Therefore, NS is important in
the differential diagnosis of short stature and has impor-
tant implications in GH therapy
[9–11] .
Differential Diagnosis of Noonan Syndrome
A lthough NS gi rls ca n be misdiagnosed as Turner syn-
drome (TS) because of clinical feature similarities be-
tween these two syndromes, the presence of hypergonad-
otrophic hypogonadism and abnormal karyotype in TS
girls facilitates the distinction between these two syn-
dromes. Several other disorders, with normal gonadal
function and normal karyotype, resemble NS phenotypi-
cally and constitute the true differential diagnosis of NS.
Leopard syndrome is an autosomal dominant disor-
der that shares some clinical characteristics with NS. The
acronym refers to the major features: Lentigines, ECG
conduction abnormalities, Ocular hypertelorism, Pul-
monic stenosis, Abnormal genitalia, Retardation of
growth and sensorineural Deafness
[12, 13] . The pres-
ence of café-au-lait spots in early infancy and generalized
multiple lentigines after 5–6 years of age are the main
characteristics of Leopard syndrome ( fig. 2 ).
In addition to Leopard syndrome, two other rare syn-
dromes, Costello syndrome and CFC syndrome, present
facial features and cardiac malformations that resemble
Fig. 1. NS features: ( a ) eye abnormalities: ptosis, hypertelorism,
epicanthal folds; (
b ) webbed neck, and ( c ) ear abnormalities: low-
set posteriorly rotated ears and thick helix.
Fig. 2. Generalized multiple lentigines in a patient with Leopard
syndrome.
Noonan Syndrome and Related Disorders Horm Res 2009;71:185–193
187
NS. In comparison with NS, Costello and CFC syndromes
present a coarser face and both syndromes are associated
with more frequent and severe developmental delay. Cos
-
tello syndrome patients usually have macrocephaly, cutis
laxa, nasal and perioral papillomata, deep palmar and
plantar creases, diffuse skin hyperpigmentation and nail
dy smorpholog y, as wel l as an inc rea sed risk of ma lig nanc y,
especia lly rhabdomysarcoma . CFC syndrome patients ty p-
ically have ectodermal abnormalities such as sparse hair
and eyebrows, follicular hyperkeratosis, palmoplantar hy-
perkeratosis and an ichthyosis-like condition
[14, 15] .
Clinical Diagnosis of Noonan Syndrome
Diagnosis of NS is primarily based on clinical find-
ings. Generally, classical facial features or a typical car-
diac malformation trigger suspicions of NS. In the new-
born, facial features can be less apparent, but generalized
edema, exce ss nu ch al fo ld an d c ong en it al he ar t d ef ec t c an
suggest the diagnosis.
Widely variable facial appearance is observed among
NS patients, even among patients from the same family
[16] or with the same molecular defect [17] . Furthermore,
a marked change of phenotype with age from newborn
period, infancy, childhood and adolescence to adulthood
was classically documented, resulting in a mild pheno-
type in adult patients
[3] . All these facts can contribute to
misdiagnosis, especially in patients without congenital
heart disease, with mild forms and/or at an older age.
In 1981, Duncan et a l. [4] proposed a scoring system for
NS diagnosis with 26 items devised on the basis of fre-
quency and severity of NS features in 23 typical patients.
This complex first scoring system was difficult for routine
use by non-geneticist specialists. In 1993, Sharland et al.
[18] proposed that the diagnosis should be based on the
pres ence of typical fa cia l feat ures in a pat ient wit h norma l
chromosomes plus short stature (height ! 10th centile for
sex and age) and/or cardiac defect and/or undescended
testicles in males. These new criteria did not allow the di-
agnosis in patients with mild facial features. Finally, in
1994, van der Burgt et al.
[16] proposed a simple and ac-
curate scoring system for the diagnosis of NS based on the
variable clinical NS presentation in one family ( table 2 ).
The van der Burgt scoring system is the most used model
to select patients for molecular studies in recent studies
[19 –21] . In this scoring system, patients are first classified
ac cording to f aci al feature s as hav ing ty pical or sugg est ive
NS characteristics. Typical face and any other major sign
or two minor signs establishes the diagnosis of NS, where-
as patients with suggestive NS face need two major or
three minor criteria to confirm the NS diagnosis. Analy-
ses of clinical features in NS patients, who had their diag-
nosis confirmed by molecular study, demonstrated that
Tab le 1. Phenotypic abnormalities associated to NS [1, 3, 4]
Charac-
teristics
Sign
Inheritance Autosomal dominant
Growth Short stature (postnatal onset) (50–80%)
Failure to thrive in infancy (40%)
Head and
neck
Triangular face
Ear abnormalities (44–90%): low-set posteriorly ro-
tated ears and thick helix
Eye abnormalities (95%): ptosis, hypertelorism,
epicanthal folds, down-slanting palpebral fissures,
strabismus, proptosis, myopia and nystagmus
Deeply grooved philtrum with high peaks of upper lip
vermilion border (95%)
Neck abnormalities (95%): short or webbed neck
High-arched palate (45–34%)
Dental malocclusion (35%)
Low posterior hairline (32%)
Micrognathia (22%)
Cardio-
vascular
Congenital heart defect (50–75%): pulmonary valve
stenosis (50%), hypertrophic cardiomyopathy (10%),
atrial septal defects (10%) and other (aortic stenosis,
ventricular septal defects and mitral insufficiency)
Electrocardiogram with left axis deviation and a neg-
ative pattern in the left precordial leads
Chest Thoraxic abnormalities (53–70%): flat, funnel, shield
or deformed chest, pectus carinatum superiorly and/
or pectus excavatum inferiorly
Genito-
urinary
Cryptorchidism (60–69%)
Puberty delay
Skeletal Cubitus valgus (47%)
Hand abnormalities: clinodactyly and brachydactyly
and blunt fingertips (30%)
Vertebral abnormalities (25%)
Neurologic Motor developmental delay (26%), language delay
(20%) and learning disability (15%)
Mental retardation, generally mild (25–35%)
Hema-
tology
Bleeding anomalies (20%), including factor XI or XII
deficiencies, von Willebrand’s disease, platelet dys-
function and leukemia (in especial juvenile myelo-
monocytic leukemia – JMML)
Other Peripheral lymphedema, splenomegaly, deafness
Values in parentheses show percent frequency.
Jorge/Malaquias/Arnhold/Mendonca
Horm Res 2009;71:185–193
188
no is ol ate d cl in ic al ch ar ac te ri st ic ca n e ns ur e t he d ia gn osis
of NS; however, t he van der Burg t et al. criteria, which take
in account the facial features, growth pattern, chest defor-
mity and cardiac defects, have been shown to be an accu-
rate tool for NS diagnosis
[22] . For these reasons, we rec-
ommend the use of this scoring system by endocrinolo-
gists during the evaluation of short stature children to
facilitate the recognition of patients with NS.
Genetic Heterogeneity of Noonan Syndrome
In 1994, Jamieson et al. [23] performed a genome-wide
scan in a large Dutch NS family and demonstrated a link-
age with several markers located at chromosome 12q22-
qter. In parallel, they demonstrated the existence of
genetic heterogeneity in NS by haplotype analysis of
another NS family. Subsequent studies reduced the chro-
mosome 12 interval which contains the NS candidate
gene region
[24, 25] and several positional candidate ap-
proaches were taken to identify the NS disease gene.
Finally, in 2001, Tartaglia et al.
[26] demonstrated the
presence of heterozygous missense mutations in PTPN11
(protein tyrosine phosphatase, non-receptor type 11;
OMIM 176876), a gene mapped to chromosome 12q24.1,
in patients with NS. Several further studies confirmed
PTPN11 as the most affected gene in NS patients and
demonstrated a mutation frequency of 38–100 and 37–
52% in individuals with familial and sporadic NS, respec-
tively
[7, 19, 27, 28] .
G e r m l i n e PTPN11 mutations have also been detected
in Leopard syndrome (OMIM 151100)
[12, 13] , Noonan-
like/multiple giant cell lesion syndrome (OMIM 163955)
[29] and in p atients wit h is olate d congenit al h ear t dise ase
[30] showing that mutations in this gene exhibit a broad
phenotype spectrum. Furthermore, it is noteworthy that,
during the screening of mutations in parents and rela-
tives of NS patients with PTPN11 mutations, some indi-
viduals carried the same PTPN11 mutations found in the
index case, but exhibited mild NS phenotypes that did
not fulfill the NS diagnostic criteria
[17, 26, 31, 32] . For
these reasons, we investigated 50 children with idiopath-
ic short stature that presented some NS-associated signs,
without fulfilling the van der Burgt et al. criteria for NS
diagnosis. No mutations were found in this cohort, sug-
gesting that PTPN11 mutations are not involved in the
pathogenesis of idiopathic short stature
[22] .
Specific somatic PTPN11 mutations were also found in
leukemia patients, especially in juvenile myelomonocytic
leukemia
[33] . Additionally, somatic mutations are iden-
tified at low frequency in several human cancers
[34] .
T h e PTPN11 protein product, SHP-2 (Src homology re-
gion 2-domain phosphatase 2), is a ubiquitously expressed
cytoplasmic protein member of a subfamily of protein ty-
rosine phosphatases that contains two Src homology 2 (C-
SH2 and N-SH2) domains. Through SH2 domains, SHP-2
binds to activated receptors and adapter proteins that pres-
ent specific phosphorylated tyrosines. In addition to tyro-
sine phosphatase actions, SHP-2 may also act as an adapt-
er molecule through phosphorylation of a tyrosine residue
at the amino terminus region, thus working as a docking
site for other SH-2-containing molecules
[35] . Both func-
tions, tyrosine phosphatases and adapter molecules, are
obviously relevant to several intracellular signal pathways,
including signal transduction of growth factors and cyto-
kines
[36 , 37] . From the endocrinological point of view,
Tab le 2. Noonan syndrome diagnostic criteria (adapted from van der Burgt et al. [16])
Clinical characteristics Major Minor
1 Facial typical face suggestive face
2 Cardiac pulmonary valve stenosis and/or typical ECG other defects
3 Height <3rd centile <10th centile
4 Chest wall pectus carinatum/excavatum broad thorax
5 Family history first-degree relative with definite diagnosis first-degree relative with suggestive diagnosis
6 Other:
Mental retardation
Cryptorchidism
Lymphatic dysplasia
all 3 any of the 3
Definite NS: typical face + one major or two minor clinical characteristics or suggestive face + two major or three minor clinical
characteristics.
Noonan Syndrome and Related Disorders Horm Res 2009;71:185–193
189
SHP-2 is implicated in GH [38, 39] , IGF-1 [4 0] , insulin [41]
and leptin
[42] signaling. The effects of NS PTPN11 muta-
tion on these hormone actions are still to be evaluated.
The PTPN11 mutations identified in patients with NS
or leukemia are predicted to be gain-of-function changes
that augment the capacity of tyrosine dephosphorylation
[26] . The SHP-2 mutants positively regulate the signal f lux
through the RAS/MAPK pathway induced by EGF (epi-
derma l grow th f actor), FGF (fibroblas t grow th fac tor), IL-
1 (interleukin-1) and TNF-1 (tumor necrosis factor-1), but
negatively regulate JAK/STAT signaling
[7] . Interestingly,
somatic PTPN11 mutations associated with leukemia pre-
sented a higher phosphatase activity than germline muta-
tions identified in NS patients
[20, 36] . Additionally, so-
matic mutations commonly found in leukemias rarely oc-
cur as germline mutation in NS patients and vice versa
[20, 36] , suggesting that somatic PTPN11 mutations with
higher tyrosine phosphatase activity and consequently
with more leukemogenic properties could cause embry-
onic lethality if they occur as germline mu tations.
Several other molecules in the RAS/MAPK pathway
were involved in NS patients without PTPN11 mutations:
germline KRAS [43] , SOS1 [44, 45] , RAF1 [46, 47] and
MEK
[15] mutations were found in 2.3, 21, 10 and 4.3% of
NS patients who did not harbor mutations in PTPN11
gene, respectively. Furthermore, mutations in other RAS/
MAPK molecules were also found in patients with NS-
like syndromes ( table 3 ). All these mutations, except
those found in patients with Leopard syndrome
[12, 13] ,
are characterized by an increase of constitutive function
of each mutated protein and consequently increased sig-
nal transduction via RAS/MAPK pathway. Due to the
clinical and molecular disease mechanisms and similari-
ties between NS and NS-like disorders, it is possible to
categorize these sy ndromes as a group of disorders caused
by RAS/MAPK pathway dysregulation.
R A S - M A P K P a t h w a y
RAS proteins (HRAS, NRAS and KRAS) are small
guanosine-binding proteins which act as signal switch
molecules that integrate extracellular inputs and activate
downstream effectors ( fig. 3 ) [reviewed in
8, 48 ]. Cell
stimulation promotes cycling between inactive GDP-
bound to active GTP-bound conformations (RAS-GDP
and RAS-GTP). The counterbalancing activities of gua-
nosine nucleotide exchange factors (GNEFs), which fa-
cilitate RAS-GTP conformation, and GTPase-activat-
ing proteins (GAPs), which increase the RAS intrinsic
GTPase activity and act in favor of the RAS-GDP confor-
mation, control RAS activity in vivo.
SOS1 (son of sevenless, drosophila , homolog 1 – OMIM
182530), the major GNEF, is recruited to protein com-
plexes that assemble on activated growth receptors. SOS1
binds to either RAS-GTP or RAS-GDP and displaces
guanine nucleotide. Because GTP is much more abun-
dant than GDP in the cytosol, this nucleotide exchange
Tab le 3. Genetic causes and summary of major features of NS and related disorders
Disorder Causative gene Phenotype
Noonan syndrome PTPN11, SOS1, RAF1, MEK1, KRAS See table 1
Neurofibromatosis type 1 NF1 Familial cancer syndrome; hallmark features include hyperpig-
mented skin lesions and benign neurofibromas; learning disabili-
ties are common
Leopard syndrome PTPN11, RAF1 Multiple lentigines, electrocardiographic conduction abnormali-
ties, ocular hypertelorism, pulmonic stenosis, abnormal genitalia,
growth retardation and deafness
Costello syndrome HRAS, KRAS, BRAF, MEK1 Mental retardation, high birth weight, neonatal feeding problems,
curly hair, coarse face, thick lips, nasal papillomata, diffuse skin
hyperpigmentation, and nail dystrophy
Cardio-facio-cutaneous
syndrome
KRAS, BRAF, MEK1, MEK2 Coarse face, congenital heart defects, ectodermal anomalies (fol-
licular and palmar hyperkeratosis), short stature, variable degrees
of mental retardation (moderate to severe) and facial features rem-
iniscent of NS and Costello syndrome
Jorge/Malaquias/Arnhold/Mendonca
Horm Res 2009;71:185–193
190
increases intracel lular RAS- GTP levels. In its GTP-bound
form, RAS can activate several intracellular pathways, in-
cluding the MAPK pathway. The RAF-MEK-ERK cas-
cade is the best characterized RAS effector pathway.
There are three RAF serine/threonine kinases (ARAF,
BRAF and RAF1) that activate the MEK-ERK kinase cas-
cade. ERK kinase can phosphorylate both cytosolic and
nuclear substrates, which include transcription factors
that control the cell cycle
[8, 48] .
Mutations in several molecules involved in this cas-
cade were identified in NS and other related syndromes
( table 3 ). It has been proposed that these disorders should
be classified together as neuro-cardio-facial-cutaneous
syndromes based on a constellation of similar pheno-
typic features and the central role of hyperactive RAS/
MAPK in their pathogenesis
[49] . Based on genotype-
phenotype correlation, three clusters of genes were pro-
posed
[15] :
The first group comprises genes outside the RAS-
RAF-MEK chain, which encompasses those upstream of
RAS. Most patients with PTPN11 or SOS1 mutation have
NS or Leopard syndrome. NF1 is caused by mutations in
neurofibromin, a GAP protein involved in RAS inactiva-
tion. Mutations in this group of genes usually lead to a NS
phenotype, with a low rate of mental impairment and a
low rate of keratinization disorders, but a tendency to
patchy skin hyperpigmentation, and, at least for NF1 and
PTPN11 , a slightly increased risk of leukemia
[15] .
The second group comprises KRAS and the cascading
genes downstream. Mutations in these genes usually af-
fect the cognitive functions, have more influence on so-
matic growth, skin redundancy, keratinization, and hair
development, and usually result in a CFC syndrome phe-
notype. Malignancy risk appears to be low, but could in-
clude leukemias
[15] .
The third group is restricted to HRAS. Diffuse hyper-
pigmentation, papillomata, chaotic atrial fibrillation and
a tendency for soft-tissue tumors are the most distin-
guishing phenotypic features in this group and usually
result in a Costello syndrome phenotype
[15] .
Germline Mutations in Genes of RAS/MAPK Pathway
and Risk of Neoplasia
Somatic activating RAS mutations occur in 30% of
huma n cancers: mutations in KRAS are commo n in pan-
creatic, colorectal, endometrial, biliary tract, lung and
cervical cancer, KRAS and NRAS mutations are preva-
lent in myeloid malignancies, whereas NRAS and HRAS
mutations are found in melanoma and bladder cancer,
respectively
[8] . BR AF mutations are also frequently
Fig. 3. RAS/MAPK pathway. Tyrosine-
phosphorylated domains of activated ty-
rosine kinase receptors act as docki ng sites
for several intracellular proteins, which
contain SH2 domains such as SHC, SHP-2
and GRB2. These molecules recruit SOS1
which promotes cycling between inactive
RAS-GDP to active RAS-GTP. RAS-GTP
directly activates the MAPK pathway
(RAF-MEK-ERK cascade). ERK kinase
can phosphorylate both cytosolic and nu-
clear substrates, which include transcrip-
tion factors that control the cell cycle.
SHC = Signaling and transforming pro-
tein contai ning Src homology 2 and 3 (SH2
and SH3) domains; SHP-2 = Src homology
region 2-domain phosphatase 2; GRB2 =
growth factor receptor-bound protein 2;
SOS 1 = son of sevenless 1; NF1 = neurofi-
bromin; RAS = rat sarcoma v iral oncogene
homolog; RA F = muri ne sarcoma viral on-
cogene homolog; MEK = mitogen-activat-
ed kinase kinase; ERK = mitogen-activat-
ed kinase.
Noonan Syndrome and Related Disorders Horm Res 2009;71:185–193
191
found in thyroid, colorectal and ovarian cancers [8] and
somatic PTPN11 mutation is responsible for one third of
juvenile myelomonocytic leukemia (JMML) and is less
frequent in other leukemias or solid tumors
[34] .
In contrast with higher malignancy potential of so-
matic mutations in RAS/MAPK components, germline
mutations presented more variable effects and depend on
which specific genes are affected. Patients with Costello
syndrome are predisposed to rhabdomyosarcoma, gan-
glioneuroblastoma and bladder cancer due to germline
HRAS mutations, whereas patients with NF1 mutations
frequently presented neurofibromas and show an in-
creased risk of neurofibrosarcoma, astrocytoma, pheo-
chromocytoma and JMML
[8] .
NS patients principally present an increase in the
prevalence of JMML and multiple giant cell lesions
(MGCL), although both conditions affect only a small per-
centage of patients. It is postulated that PTPN11 mutations
associated with NS-JMML present a higher increase in ty-
rosine phosphatases activity than mutations only associ-
ated with NS and not found in leukemias
[20, 36, 50] .
MGCL are benign tumor-like lesions most frequently af-
fecting the jaws and are associated with PTPN11 , SOS1 ,
BRAF and MEK1 mutations [51] . It is postulated that dys-
regulation of the RAS/MAPK pathway represents the
common and basic molecular event which predisposes to
MGCL.
The finding that the same molecular defects can be
found in NS patients with or without concomitant JMML
or MGCL indicates that NS-related heterozygous muta-
tions in RAS/MAPK pathway components are not suf-
ficient to produce both conditions and an additional
unknown factor (modifier gene, epigenetic factors or so-
matic second hit) is necessary for JMML or MGCL devel-
opment
[51] .
RAS/MAPK Pathway Dysregulation and Its
Consequences for the Endocrine System
The consequences of NS and NS-related disorders
with molecular RAS/MAPK pathway defects for the en-
docrine system remain mostly unexplored. One of the
cardinal signs of NS is short stature
[3, 4] , although the
physiopathological cause of growth impairment remains
u nclea r. G H i s e ss ent ia l f or nor ma l pos t-n at al gr ow th a nd
exerts its action after binding to a specific receptor that
phosphorylates several tyrosine residues located in the
intracellular domain. Tyrosine dephosphorylation leads
to the physiological interruption of the GH pathway.
It has been consistently documented that SHP-2, the
PTPN11 gene-encoded protein, negatively regulates
GHR-JAK2-STAT5 signa ling
[39, 52] . Thus, t he increased
tyrosine phosphatase action of the SHP-2 protein, ob-
served in PTPN11 -mutated NS children, is expected to
cause decreased GH action and consequently negatively
influence individual stature. We demonstrated that NS
children with PTPN11 gain-of-function mutations pre-
sented lower growth velocity and lesser height SDS gain
during GH therapy than patients with NS without PTPN11
mutations
[10] . These clinical f indings were also observed
in two other studies
[9–11] , suggesting that PTPN11 mu-
tations can cause partial GH insensitivity at post-recep-
tor level and appears to be a pharmacogenetic predictor
of GH responsiveness
[53] .
Add itiona lly, NS pat ient s with SOS1 mutations, a mol-
ecule that was not directly involved in the GHR-JAK2-
STAT5 pat hway, more f req uently have normal h eig ht
[44,
45] . These NS patients also presented a high frequency of
fetal macrosomia and macrocephaly, possibly due to the
increase in the RAS-MAPK pathway caused by NS-asso-
ciated SOS1 mutations and its positive inf luences on fetal
growth
[44, 45] .
Patients with NS often present delayed puberty and NS
boys frequently have cryptorchidism. Several cytokines,
tyrosine and G-protein-coupled receptors are involved in
the hypothalamic-pituitary-gonadal axis. Insulin acts
through a tyrosine kinase receptor, and thus, the repro-
ductive and metabolic effects of different NS-related mo-
lecular defects are still to be elucidated.
In summary, NS is a common condition and an im-
portant differential diagnosis in children with short stat-
ure, cryptorchidism and delayed puberty, conditions fre-
quently seen by endocrinologists. Germline mutations
that affect several components of RAS-MAPK pathway
are involved in the pathogenesis of NS and can be diag-
nosed by molecular studies. Genotype-phenotype corre-
lation studies are still necessary to better characterize the
effect of RAS/MAPK pathway dysregulation on endo-
crine and metabolic systems.
A c k n o w l e d g m e n t s
T h is wo rk w a s s up po rt ed by g ra nt s f ro m F un d ac ao de Am pa ro
a Pesquisa do Estado de São Paulo (FAPESP) (05/04726-0 and
07/59555-0 to A.C.M.) and from Conselho Nacional de Desen-
volvimento Cientifico e Tecnologico (CNPq) (301246/95-5 to
B.B.M., 300938/06-3 to I.J.P.A. and 307951/06-5 to A.A.L.J.). The
authors thank Prof. Martin O. Savage for his useful suggestions.
Jorge/Malaquias/Arnhold/Mendonca
Horm Res 2009;71:185–193
192
References
1 Mendez HM, Opitz JM: Noonan syndrome:
a review. Am J Med Genet 1985;
21: 493–506.
2 Nora JJ, Nora AH, Sinha AK, Spangler RD,
Lubs HA: The Ullrich-Noonan syndrome
(Turner phenotype). Am J Dis Child 1974;
127: 48–55.
3 Allanson JE: Noonan syndrome. J Med Ge-
net 1987;
24: 9–13.
4 Duncan W J, Fowler RS, Farkas LG, Ross RB,
Wright AW, Bloom KR, Huot DJ, Sond-
heimer HM, Rowe R D: A comprehensive
scoring system for evaluating Noonan syn-
drome. Am J Med Genet 1981;
10: 37–50.
5 Jongmans M, Sistermans EA, Rikken A,
Nillesen WM, Tamminga R, Patton M, Mai-
er EM, Tartaglia M, Noordam K, van der
Burgt I: Genotypic and phenoty pic charac-
terization of Noonan syndrome: new data
and rev iew of the literatu re. Am J Med Ge ne t
A 2005;
134: 165–170.
6 Shaw AC, Kalidas K, Crosby AH, Jeffery S,
Patton MA: The natural history of Noonan
syndrome : a long-term follow-up study. Arch
Dis Child 2007;
92: 128–132.
7 Tartaglia M, Gelb BD: Noonan syndrome
and related disorders: genetics and patho-
genesis. Annu Rev Genomics Hum Genet
2005;
6: 45–68.
8 Schubber t S, Shannon K, Boll ag G: Hyperac-
tive Ras i n developmental di sorders and can-
cer. Nat Rev Cancer 2007;
7: 295–308.
9 Binder G, Neuer K, Ranke MB, Wittekindt
NE: PTPN11 mutations are associated with
mild growth hormone resistance in individ-
uals w ith Noonan sy ndrome. J Clin Endoc ri-
nol Metab 2005;
90: 5377–5381.
10 Ferreira LV, Souza SA, Arnhold IJ, Mendon-
ca BB, Jorge AA: PTPN11 (protein tyrosine
phosphatase, non-receptor ty pe 11) muta-
tions and response to growth hormone ther-
apy in children wit h Noonan syndrome. J
Clin Endocrinol Metab 2005;
90: 5156–5160.
11 Limal JM, Parfait B, Cabrol S, Bonnet D, Le-
heup B, Lyonnet S, Vidaud M, Le Bouc Y:
Noonan syndrome: relationships between
genoty pe, growth, a nd growth fac tors. J Clin
Endocrinol Metab 2006;
91: 300–306.
12 Legius E, Schrander-Stumpel C, Schollen E,
Pulles-Heintzberger C, Gewillig M, Fryns
JP: PTPN11 mutations i n Leopard sy ndrome.
J Med Genet 2002;
39: 571–574.
13 Digilio MC, Conti E, Sarkozy A, Mingarelli
R, Dottorini T, Marino B, Pizzuti A, Dal-
lapiccola B: Groupi ng of multiple-lentigi nes/
Leopard and Noonan syndromes on the
PTPN11 gene. Am J Hum Genet 2002;
71:
389–394.
14 Schubbert S, Bollag G, Shannon K: Deregu-
lated Ras signaling in developmental disor-
ders: new tricks for an old dog. Curr Opin
Genet Dev 2007;
17: 15–22.
15 Nava C, Hanna N, Michot C, Perei ra S, Pou-
vreau N, Nii hori T, Aoki Y, Matsubara Y, Ar-
veiler B, Lacombe D, Pasmant E, Parfait B,
Baumann C, Heron D, Sigaudy S, Toutain A,
Rio M, Goldenberg A, Leheup B, Verloes A,
Cave H: Cardio-facio-cutaneous and Noon-
an syndromes due to mutations in the R AS/
MAPK signalling pathway: genotype-phe-
notype relationships and overlap w ith
Costello syndrome. J Med Genet 2007;
44:
763–771.
16 Van der Burgt I, Berends E, Lommen E, va n
Beersum S, Hamel B, Mariman E: Clinical
and molecular studies in a large Dutch fam-
ily wit h Noonan syndrome . Am J Med Genet
1994;
53: 187–191.
17 Zenker M, Buheitel G, Rauch R, Koenig R,
Bosse K, Kress W, Tietze HU, Doerr HG,
Hofbeck M, Singer H, Reis A, Rauch A: Ge-
notype-phenotype correlations in Noonan
syndrome. J Pediatr 2004;
144 : 368–374.
18 Sharland M, Morgan M, Smith G, Burch M,
Patton MA: Genetic counselling in Noonan
syndrome. Am J Med Genet 1993;
45: 437
440.
19 Bertola DR Pereira AC, Albano LM, De
Oliveira PS, Kim CA, Krieger JE: PTPN11
gene analysis in 74 Brazilian patients with
Noonan sy ndrome or Noonan-like pheno-
type. Genet Test 2006;
10: 18 6–191.
20 Tartaglia M, Martinelli S, Stella L, Bocchin-
fuso G, Flex E, Cordeddu V, Zampino G,
Burgt I, Pa lleschi A, Petrucci TC, Sorcini M,
Schoch C, Foa R, Emanuel PD, Gelb BD: Di-
versity and functional consequences of
germline and somatic PTPN11 mutations in
human disease. Am J Hum Genet 2006;
78:
279–290.
21 Yoshida R, Hasegawa T, Hasegawa Y, Nagai
T, Kinoshita E, Tanaka Y, Kanegane H,
Ohy ama K , Oni shi T, Ha new K , Oku yama T,
Horikawa R, Tanaka T, Ogata T: Protein-ty-
rosine phosphatase, non-receptor type 11
mutation analysis and clinical assessment in
45 patients w ith Noona n syndrome. J Clin
Endocrinol Metab 2004;
89: 3359–3364.
22 Ferreira LV, Souza SC, Montenegro LR,
Malaquias AC, Arnhold IJ, Mendonca BB,
Jorge AA: Analysis of PTPN11 gene in idio-
pathic shor t stature children and Noonan
syndrome patients. Clin Endocrinol (Oxf)
2008;69:
426–431.
23 Jamieson CR, van der Burgt I, Brady AF, van
Reen M, Elsawi MM, Hol F, Jeffery S, Patton
MA, Mariman E: Mapping a gene for Noon-
an synd rome to the long arm of chromo some
12. Nat Genet 1994;
8: 357–360.
24 Brady AF, Jamieson CR, van der Burgt I,
Cros by A, va n Reen M, Kre mer H, Ma riman
E, Patton MA, Jeffer y S: Further delineation
of the critical region for Noonan syndrome
on the long arm of chromosome 12. Eur J
Hum Genet 1997;
5: 336–337.
25 Legius E, Schollen E, Matthijs G, Fryns JP:
Fine mapping of Noonan/cardio-facio-cuta-
neous sy ndrome in a large fam ily. Eur J Hum
Genet 1998;
6: 32–37.
2 6 Tar ta gl ia M , Me hl er E L, Gol dbe rg R , Z amp i-
no G, Brun ner HG, Kremer H, van der Burgt
I, Crosby AH, Ion A , Jeffery S, K alidas K, Pat-
ton MA, Kucherlapati RS, Gelb BD: Muta-
tions in PTPN11 , encoding the protein tyro-
sine phosphatase SHP-2, cause Noonan
syndrome. Nat Genet 2001;
29: 465–468.
27 Ferrei ra LV, Souza SA, Montenegro L R, Arn-
hold IJ, Pasqualini T, Heinrich JJ, Keselman
AC, Mendonca BB, Jorge AA: Phenoty pe
variability in Noonan syndrome patients
with and without PTPN11 mutation (in Por-
tugue se). Arq Bras Endocri nol Metabol 2007;
51: 450–456.
28 Zenker M, Voss E, Reis A: Mild variable
Noonan syndrome in a family with a novel
PTPN11 mutation. Eu r J Med Genet 2007;
50:
43–47.
29 Ber tola DR, Kim CA, Perei ra AC, Mota GFA,
Krieger J E, Vieira IC, Valente M, Loreto M R,
Magalhães RP, Gonzáles CH: Are Noonan
syndrome and Noonan-like/multiple giant
cell lesion syndrome distinct entities? Am J
Med Genet 2001;
98: 230–234.
30 Weismann CG, Hager A, Kaemmerer H,
Maslen CL, Morris CD, Schranz D, Kreuder
J, Gelb BD: PTPN11 mutations play a minor
role in isolated congenital heart disease. Am
J Med Genet A 2005;
136: 14 6–151.
31 Bertola DR, Pereira AC, de Oliveira PS,
Kim CA, Krieger JE: Clinical variability
in a Noonan syndrome family with a new
PTPN11 gene mutation. Am J Med Genet
2004;
130A:378–383.
32 Kitsiou-Tzeli S, Papadopoulou A, Kanaka-
Gantenbein C, Fretzayas A, Daskalopoulos
D, Kanavakis E, Nicolaidou P: Does the rare
A172G mutation of PTPN11 gene convey a
mild Noonan syndrome phenotype? Horm
Res 2006;
66: 124–131.
33 Tartaglia M, Niemeyer CM, Fragale A, Song
X, Buechner J, Jung A, Hahlen K, Hasle H,
Licht JD, Gelb BD: Somatic mutations in
PTPN11 in juvenile myelomonocytic leuke-
mia, myelodysplastic syndromes and acute
myeloid leukemia. Nat Genet 2003;
34: 148
150.
34 Bentires-Alj M, Paez JG, David FS, Keilhack
H, Halmos B, Naoki K, Maris JM, Richard-
son A, Bardelli A, Sugarbaker DJ, Richards
WG, Du J, Girard L , Minna JD, Loh ML ,
Fisher DE, Velculescu VE, Vogelstein B,
Meyerson M, Sellers WR, Neel BG: Activat-
ing mutations of the Noonan syndrome-as-
sociated SHP2/PTPN11 gene in human solid
tumors and adult acute myelogenous leuke-
mia. Cancer Res 20 04;
64: 8816–8820.
Noonan Syndrome and Related Disorders Horm Res 2009;71:185–193
193
35 Van Vactor D, O’Reilly AM, Neel BG: Ge-
netic ana lysis of protein tyrosine phospha-
tases. Curr Opin Genet Dev 1998;
8: 112–
126.
36 Niihori T, Aoki Y, Ohashi H, Kurosawa K,
Kondoh T, Ishikiriyama S, Kawame H, Ka-
masaki H, Yama naka T, Takada F, Nishio K,
Sakurai M, Tamai H, Nagashima T, Suzuki
Y, Kure S, Fujii K, Imaizumi M, Matsubara
Y: Functional analysis of PTPN11/SHP-2 mu-
tants identified in Noonan syndrome and
childhood leukemia. J Hum Genet 2005;
50:
192–202.
37 Schubbert S, Lieuw K, Rowe SL, Lee CM, Li
X, Loh ML, Clapp DW, Shannon KM: Func-
tional analysis of leukemia-associated
PTPN11 mutations in primary hematopoi-
etic cells. Blood 2005;
106: 311–317.
38 Kim SO, Jiang J, Yi W, Feng GS, Frank SJ:
Involvement of the Src homolog y 2-contain-
ing tyrosine phosphatase SHP-2 in grow th
hormone signaling. J Biol Chem 1998;
273:
2344–2354.
39 Stofega MR, Herrington J, Billestrup N,
Carter-Su C: Mutation of the SHP-2 binding
site in grow th hormone (GH) receptor pro-
longs GH-promoted tyrosyl phosphoryla-
tion of GH receptor, JAK 2, and STAT5B. Mol
Endocrinol 2000;
14: 1338–1350.
40 Maile LA, Clemmons DR: Regulation of in-
sulin-like growth factor I receptor dephos-
phorylation by SHPS-1 and the tyrosine
phosphatase SHP-2. J Biol Chem 2002;
277:
8955–8960.
41 Lima MH, Ueno M, Thirone AC, Rocha EM,
Carvalho CR, Saad MJ: Regulation of IRS-
1/SHP2 interaction and AKT phosphoryla-
tion in animal models of insulin resistance.
Endocrine 2002;
18: 1–12 .
42 Carpenter LR, Farruggella TJ, Symes A,
Karow ML, Yancopoulos GD, Stahl N: En-
hancing leptin response by preventing SH2-
containing phosphatase 2 interaction with
Ob receptor. Proc Natl Acad Sci USA 1998;
95: 6061–6066.
43 Schubbert S, Zenker M, Rowe SL, Boll S,
Klein C, Bollag G, van der Burgt I, Musante
L, Kalscheuer V, Wehner LE, Nguyen H,
West B, Zhang KY, Sistermans E, Rauch A,
Niemeyer CM, Sha nnon K, Kratz CP: Germ-
line KRAS mutations cause Noonan syn-
drome. Nat Genet 2006;
38: 331–336.
44 Roberts AE, Araki T, Swanson KD, Mont-
gomery KT, Schi ripo TA, Joshi VA, Li L, Yas-
sin Y, Tamburino AM , Neel BG, Kucherlapa-
ti RS: Germline gain-of-function mutations
in SOS1 cause No onan syndrome. Nat Gene t
2007;
39: 70–74.
45 Tartaglia M, Pennacchio LA, Zhao C, Yadav
KK, Fodale V, Sarkozy A, Pandit B, Oishi K,
Martinelli S, Schackwitz W, Ustaszewska A,
Martin J, Bristow J, Carta C, Lepri F, Neri C,
Vasta I, Gibson K, Curry CJ, Siguero JP, Di-
gilio MC, Zampino G, Dallapiccola B, Bar-
Sagi D, Gelb BD: Gain-of-function SOS1
mutations cause a distinctive form of Noon-
an syndrome. Nat Genet 2007;
39: 75–79.
46 Pandit B, Sarkozy A, Pennacchio LA, Carta
C, Oishi K, Martinelli S, Pogna EA, Schack-
witz W, Ustaszewska A, Landstrom A, Bos
JM, Ommen SR, Esposito G, Lepri F, Faul C,
Mundel P, Lopez Siguero JP, Tenconi R, Seli-
corni A, Ros si C, Mazzant i L, Torrente I, Ma-
rino B, Digilio MC, Zampino G, Ackerman
MJ, Dallapiccola B, Tartaglia M, Gelb BD:
Gain-of-function raf1 mutations cause
Noonan and Leopard syndromes with hy-
pert rophic cardiomyopat hy. Nat Genet 20 07;
39: 1007–1012.
47 Razzaque MA, Nishizawa T, Komoike Y,
Yagi H, Furutani M, Amo R, Kamisago M,
Mom ma K, K atay ama H , Nak agawa M, Fuji-
w a r a Y , M a t s u s h i m a M , M i z u n o K , T o k u y a m a
M, Hirota H, Muneuchi J, Higashinakagawa
T, Matsuoka R: Germline gain-of-function
mutations i n RAF1 cause No onan synd rome.
Nat Genet 2007;
39: 1013–1017.
48 Shannon K, Bollag G: Sending out an SOS.
Nat Genet 2007;
39: 8–9.
49 Bentires-Alj M, Kontaridis MI, Neel BG:
Stops along the RAS pathway in human ge-
netic disease. Nat Med 2006;
12: 283–285.
50 Keil hack H, David FS, McGregor M, C antley
LC, Neel BG : Diverse biochemica l properties
of SHP2 mutants. Implications for disease
phenotypes. J Biol Chem 2005;
280: 30984–
30993.
51 Neumann TE, Allanson J, Kavamura I, Kerr
B, Neri G , Noonan J, C ordeddu V, Gibson K,
Tzschach A, Kruger G, Hoeltzenbein M,
Goecke TO, Kehl HG , Albrecht B, Luczak K,
S as ia d ek MM , M u sa nt e L, L au ri e R, Pe te r s H ,
Tartaglia M, Zenker M, Kalscheuer V: Mul-
tiple giant cell lesions in patients with Noon-
an syndrome and cardio-facio-cutaneous
syndrome. Eur J Hum Genet 2008; [Epub
ahead of print].
52 Chen Y, Wen R, Yang S, Schuman J, Zhang
EE, Yi T, Feng GS, Wang D: Identification of
SHP-2 as a STAT5A phosphatase. J Biol
Chem 2003;
278: 1652 0–16527.
53 Padidela R, Ca macho-Hubner C, Attie KM,
Savage MO: Abnormal g rowt h in Noonan
syndrome: genetic and endocrine features
and optimal treatment. Horm Res 2008;
70:
129–136.
... 7,8 In addition, CGCGs have been shown to be associated with several genetic syndromes. Noonan syndrome involving multiple genes such as PTPN11 and SOS1 that regulate the Ras/mitogen-activated protein kinase (RAS/MAPK) pathway, [9][10][11][12] neurofibromatosis 1 caused by pathogenic variants in NF1, and cherubism due to variants in SH3BP2 have been described to present with CGCG, [13][14][15][16] implying a potential genetic etiology. ...
... 11,25,26 These genes are involved in the RAS/MAPK pathway. [9][10][11][12] Dysregulation and activation of RAS/MAPK pathway have been postulated to be a common mechanism contributing to the development of CGCG in these syndromes. 11 It has also been proposed that pathogenic variants in NF1 may cause a decrease in type 1 collagen expression, which alters bone formation leading to intraosseous defects. ...
Article
Full-text available
Central giant cell granuloma (CGCG) is a rare disease characterized by sporadic, benign, intraosseous mandibular lesions of unknown etiology. Histologically, these lesions are indistinguishable from brown tumors of hyperparathyroidism and cherubism, and occasionally have been associated with different syndromes raising a question for genetic etiology. The CGCG has varied presentation ranging from nonaggressive and indolent to aggressive, destructive, and recurrent, often posing diagnostic and therapeutic challenges. Herein, we present the first case of a 10-year-old boy with CGCG and 16p13.11 microdeletion syndrome, highlight the diagnostic challenges inherent to this heterogeneous disorder, and discuss the genetics and treatment approaches of these complex lesions.
... The MAPKs are a collection of essential threonine kinases produced under high temperatures by producing cytokines, ischemia, and ROS. MAPKs are stimulated under stress conditions and affect many cellular procedures like cell-mediated cell death, survival, adjustment, and cell multiplication [86]. ERK, c-Jun N-terminal kinases (JNK), and p38 MAPK are the three most important members of the family of MAPK. ...
... Claudin-2 reduces the rigidity of the epithelial barrier. Due to inflammation of the intestine, claudin-2 protein expression is increased [86]. Tight junction permeability is connected with the claudin expression. ...
Article
Full-text available
The potential threat of global warming in the 21st century is on the ecosystem through many aspects, including the negative impact of rising global temperature on the health of humans and animals, especially domestic animals. The damage caused by heat stress to animals has been more and more significant as the worldwide climate continues to rise, along with the breeding industry’s expanding scale and stocking density, and it has become the most important stress-causing factor in southern China. In this review, we described the effects of heat stress on animal immune organs and immune system. The much-debated topic is how hyperthermia affects the tight junction barrier. Heat stress also induces inflammation in the body of animals causing low body weight and loss of appetite. This review also discussed that heat stress leads to hepatic disorder, and it also damages the intestine. The small intestine experiences ischemia, and the permeability of the intestine increases. Furthermore, the oxidative stress and mitogen-activated protein kinase (MAPK) pathways have a significant role in stress-induced cellular and organ injury. The study has shown that MAPK activity in the small intestine was increased by heat stress. Heat stress caused extreme small intestine damage, enhanced oxidative stress, and activated MAPK signaling pathways.
... Noonan syndrome has no chromosome abnormality, resulting in a normal karyotype assessment. The genetics behind the generation of Noonan syndrome is due to mutations in the RAS/MAPK signal pathway (frequently affecting PTPN11, SOS1, RAF1 or RIT1 genes) 16,18 . ...
Preprint
Full-text available
It is important to be familiar with the presentation of Turner syndrome and to understand the common causes of death associated with the disease. Using the autopsy case as an example, this case report will outline the classic presentation of Turner syndrome, go over its physiology, epidemiology, pathogenesis and explain the cause of death in this particular case while also highlighting other common causes of death/risk factors within Turner syndrome. Differential diagnosis and mimicking disease processes will also be discussed. This report will also highlight the molecular tests used for the diagnosis of Turner syndrome, discuss developments using in situ hybridization, and discuss why this method is best for the determination of Turner syndrome over conventional karyotyping.
... The majority of the CSCF patients showed clinical features in part similar to NS, where the RAS-MAPK pathway is hyperactivated (van der Burgt, 2007;Jorge et al., 2009). We therefore assessed whether the CSCF-related MAP3K7 variants upregulated the RAS-MAPK pathway. ...
Preprint
Mitogen-Activated Protein 3 Kinase 7 (MAP3K7, MIM 602614) encodes the ubiquitously expressed transforming growth factor β (TGF-β)–activated kinase 1 (TAK1), which plays a crucial role in many cellular processes. Variants in the MAP3K7 gene have been linked to 2 distinct disorders: frontometaphyseal dysplasia type 2 (FMD2, MIM #617137) and cardiospondylocarpofacial syndrome (CSCF, MIM #157800). The fact that different variants can induce 2 distinct phenotypes suggests a phenotype/genotype correlation, but no side-by-side comparison has been done thus far to confirm this. Here we significantly expand the cohort and the description of clinical phenotypes for individuals with CSCF and FMD2 who carry variants in MAP3K7. We show that in contrast to FMD2-causing variants, CSCF-causing variants in MAP3K7 have a loss-of-function effect. Additionally, patients with pathogenic variants in MAP3K7 are at risk for cardiac disease, have symptoms associated with connective tissue disease and we show overlap in clinical phenotypes of CSCF with Noonan syndrome. Together, we provide evidence for a molecular fingerprint of FMD2- versus CSCF-causing MAP3K7 variants and conclude that variants in MAP3K7 should be considered in the differential diagnosis of patients with syndromic congenital cardiac defects and/or cardiomyopathy, syndromic connective tissue disorders and in the differential diagnosis of Noonan syndrome.
... Incomplete penetrance is observed in OI (OMIM 166200, 166210, 259420, 166220), Ehlers-Danlos syndrome (EDS) (OMIM 130060, 225320, 617821) and Muenke syndrome (OMIM 602849) (Pereira et al., 1994;Deodhar and Woolf, 2000;Solomon and Muenke, 2011;Cortini et al., 2017). In contrast, a near complete penetrance is common for achondroplasia (OMIM 100800), Noonan (OMIM 163950), CHARGE (OMIM 214800) and Apert (OMIM 101200) syndromes (Jorge et al., 2009;Shirley and Ain, 2013;Van Ravenswaaij-Arts et al., 2015;Conrady et al., 2020). As clinical and genetic diagnostics develop, the understanding of penetrance in different disorders will change. ...
Article
BACKGROUND With the help of ART, an advanced parental age is not considered to be a serious obstacle for reproduction anymore. However, significant health risks for future offspring hide behind the success of reproductive medicine for the treatment of reduced fertility associated with late parenthood. Although an advanced maternal age is a well-known risk factor for poor reproductive outcomes, understanding the impact of an advanced paternal age on offspring is yet to be elucidated. De novo monogenic disorders (MDs) are highly associated with late fatherhood. MDs are one of the major sources of paediatric morbidity and mortality, causing significant socioeconomic and psychological burdens to society. Although individually rare, the combined prevalence of these disorders is as high as that of chromosomal aneuploidies, indicating the increasing need for prenatal screening. With the help of advanced reproductive technologies, families with late paternity have the option of non-invasive prenatal testing (NIPT) for multiple MDs (MD-NIPT), which has a sensitivity and specificity of almost 100%. OBJECTIVE AND RATIONALE The main aims of the current review were to examine the effect of late paternity on the origin and nature of MDs, to highlight the role of NIPT for the detection of a variety of paternal age-associated MDs, to describe clinical experiences and to reflect on the ethical concerns surrounding the topic of late paternity and MD-NIPT. SEARCH METHODS An extensive search of peer-reviewed publications (1980–2021) in English from the PubMed and Google Scholar databases was based on key words in different combinations: late paternity, paternal age, spermatogenesis, selfish spermatogonial selection, paternal age effect, de novo mutations (DNMs), MDs, NIPT, ethics of late fatherhood, prenatal testing and paternal rights. OUTCOMES An advanced paternal age provokes the accumulation of DNMs, which arise in continuously dividing germline cells. A subset of DNMs, owing to their effect on the rat sarcoma virus protein–mitogen-activated protein kinase signalling pathway, becomes beneficial for spermatogonia, causing selfish spermatogonial selection and outgrowth, and in some rare cases may lead to spermatocytic seminoma later in life. In the offspring, these selfish DNMs cause paternal age effect (PAE) disorders with a severe and even life-threatening phenotype. The increasing tendency for late paternity and the subsequent high risk of PAE disorders indicate an increased need for a safe and reliable detection procedure, such as MD-NIPT. The MD-NIPT approach has the capacity to provide safe screening for pregnancies at risk of PAE disorders and MDs, which constitute up to 20% of all pregnancies. The primary risks include pregnancies with a paternal age over 40 years, a previous history of an affected pregnancy/child, and/or congenital anomalies detected by routine ultrasonography. The implementation of NIPT-based screening would support the early diagnosis and management needed in cases of affected pregnancy. However, the benefits of MD-NIPT need to be balanced with the ethical challenges associated with the introduction of such an approach into routine clinical practice, namely concerns regarding reproductive autonomy, informed consent, potential disability discrimination, paternal rights and PAE-associated issues, equity and justice in accessing services, and counselling. WIDER IMPLICATIONS Considering the increasing parental age and risks of MDs, combined NIPT for chromosomal aneuploidies and microdeletion syndromes as well as tests for MDs might become a part of routine pregnancy management in the near future. Moreover, the ethical challenges associated with the introduction of MD-NIPT into routine clinical practice need to be carefully evaluated. Furthermore, more focus and attention should be directed towards the ethics of late paternity, paternal rights and paternal genetic guilt associated with pregnancies affected with PAE MDs.
Article
Full-text available
Mitogen‐Activated Protein 3 Kinase 7 (MAP3K7) encodes the ubiquitously expressed transforming growth factor β (TGF‐β)–activated kinase 1 (TAK1), which plays a crucial role in many cellular processes. Mutationsin the MAP3K7 gene have been linked to 2 distinct disorders: frontometaphyseal dysplasia type 2 (FMD2) and cardiospondylocarpofacial syndrome (CSCF). The fact that different mutations can induce 2 distinct phenotypes suggests a phenotype/genotype correlation, but no side‐by‐side comparison has been done thus far to confirm this. Here we significantly expand the cohort and the description of clinical phenotypes for patients with CSCF and FMD2 who carry mutations in MAP3K7. Our findings support that in contrast to FMD2‐causing mutations, CSCF‐causing mutations in MAP3K7 have a loss‐of‐function effect. Additionally, patients with pathogenic mutations in MAP3K7 are at risk for (severe) cardiac disease, have symptoms associated with connective tissue disease and we show overlap in clinical phenotypes of CSCF with Noonan syndrome. Together, we confirm a molecular fingerprint of FMD2‐ versus CSCF‐causing MAP3K7 mutations and conclude that mutations in MAP3K7 should be considered in the differential diagnosis of patients with syndromic congenital cardiac defects and/or cardiomyopathy, syndromic connective tissue disorders and in the differential diagnosis of Noonan syndrome. This article is protected by copyright. All rights reserved.
Article
Introduction: Pubertal delay is described as one of the clinical features in Noonan Syndrome (NS) and it may be one of the factors causing short adult height in those patients. The present study aims at characterizing pubertal development in NS and identifying pubertal delay predictors. Methods: We analyzed 133 individuals with a molecular diagnosis of NS and clinical puberty evaluation. We characterized delayed puberty as pubertal onset after 12 years in girls and 13.5 years in boys, according to parameters of the Brazilian population. To investigate its predictors, we correlated the age at onset of puberty with several characteristics and genotype in a multilevel regression model. For comprehending pubertal development in NS, we assessed age and anthropometric measures at each Tanner stage and adult age. Results: The mean age at puberty onset for girls was 11.9±1.9 years and for boys, 12.5±1.7 years, significantly later than the Brazilian population (p=0.025; p<0.001). Girls (49.1%) presented delayed puberty more frequently than boys (27.9%, p=0.031). BMI SDS and IGF1 SDS at puberty onset significantly predicted later puberty entry. Height gain from the onset of puberty to adult height was lower in children with pubertal delay. Conclusion: Pubertal delay is characteristically found in children with NS, more frequently in females. The low weight of patients with NS could modulate the age of puberty, just as the increase in overweight/obesity in the general population has shown an effect on reducing the age of onset of puberty.
Chapter
Heart disease is one of the commonest abnormalities of the fetus and infant. Malformation of the heart is the commonest congenital malformation and accounts for significant morbidity and mortality in utero and postnatally. This chapter gives a brief overview of cardiac development and extensive discussion of the methods of pathological examination of the heart, including histological sampling. Congenital heart disease has a reputation as a particularly difficult area of pathology. With care and following a few simple rules, all but the most complex cases can be confidently tackled. All the commoner forms are described and illustrated together with an account of heart disease in the fetus. The cardiomyopathies are covered in detail, especially metabolic and mitochondrial cardiomyopathy. Myocarditis, ischemia and infraction, tumors and abnormalities of the cardiac rhythm are all discussed in detail. Already many gene mutations have been identified for cardiomyopathy and channelopathies in particular and the commoner defects are listed.
Article
Full-text available
Background Pectus excavatum (PE) could be part of a genetic disorder, which then has implications regarding comorbidity, the surgical correction of PE, and reproductive choices. However, referral of a patient presenting with PE for genetic analysis is often delayed because additional crucial clinical signs may be subtle or even missed in syndromic patients. We reviewed the literature to inventory known genetic disorders associated with PE and create a standardized protocol for clinical evaluation. Methods A systematic literature search was performed in electronic databases. Genetic disorders were considered associated with PE if studies reported at least five cases with PE. Characteristics of each genetic disorder were extracted from the literature and the OMIM database in order to create a practical guide for the clinician. Results After removal of duplicates from the initial search, 1632 citations remained. Eventually, we included 119 full text articles, representing 20 different genetic disorders. Relevant characteristics and important clinical signs of each genetic disorder were summarized providing a standardized protocol in the form of a scoring list. The most important clinical sign was a positive family history for PE and/or congenital heart defect. Conclusions Twenty unique genetic disorders have been found associated with PE. We have created a scoring list for the clinician that systematically evaluates crucial clinical signs, thereby facilitating decision making for referral to a clinical geneticist.
Article
Full-text available
Binding of GH to GH receptor (GHR) rapidly and transiently activates multiple signal transduction pathways that contribute to the growth-promoting and metabolic effects of GH. While the events that initiate GH signal transduction, such as activation of the Janus tyrosine kinase JAK2, are beginning to be understood, the signaling events that terminate GH signaling, such as dephosphorylation of tyrosyl-phosphorylated signaling molecules, are poorly understood. In this report, we examine the role of the SH2 (Src homology-2) domain-containing protein tyrosine phosphatase SHP-2 in GH signaling. We demonstrate that the SH2 domains of SHP-2 bind directly to tyrosyl phosphorylated GHR from GH-treated cells. Tyrosine-to-phenylalanine mutation of tyrosine 595 of rat GHR greatly diminishes association of the SH2 domains of SHP-2 with GHR, and tyrosine-to-phenylalanine mutation of tyrosine 487 partially reduces association of the SH2 domains of SHP-2 with GHR. Mutation of tyrosine 595 dramatically prolongs the duration of tyrosyl phosphorylation of the signal transducer and activator of transcription STAT5B in response to GH, while mutation of tyrosine 487 moderately prolongs the duration of STAT5B tyrosyl phosphorylation. Consistent with the effects on STAT5B phosphorylation, tyrosine-to-phenylalanine mutation of tyrosine 595 prolongs the duration of tyrosyl phosphorylation of GHR and JAK2. These data suggest that tyrosine 595 is a major site of interaction of GHR with SHP-2, and that GHR-bound SHP-2 negatively regulates GHR/JAK2 and STAT5B signaling.
Article
Full-text available
Noonan syndrome is a developmental disorder characterized by short stature, facial dysmorphia, congenital heart defects and skeletal anomalies1. Increased RAS-mitogen-activated protein kinase (MAPK) signaling due to PTPN11 and KRAS mutations causes 50% of cases of Noonan syndrome2, 3, 4, 5, 6. Here, we report that 22 of 129 individuals with Noonan syndrome without PTPN11 or KRAS mutation have missense mutations in SOS1, which encodes a RAS-specific guanine nucleotide exchange factor. SOS1 mutations cluster at codons encoding residues implicated in the maintenance of SOS1 in its autoinhibited form. In addition, ectopic expression of two Noonan syndrome–associated mutants induces enhanced RAS and ERK activation. The phenotype associated with SOS1 defects lies within the Noonan syndrome spectrum but is distinctive, with a high prevalence of ectodermal abnormalities but generally normal development and linear growth. Our findings implicate gain-of-function mutations in a RAS guanine nucleotide exchange factor in disease for the first time and define a new mechanism by which upregulation of the RAS pathway can profoundly change human development.
Article
Full-text available
Noonan syndrome (NS) and cardio-facio-cutaneous syndrome (CFCS) are related developmental disorders caused by mutations in genes encoding various components of the RAS-MAPK signaling cascade. NS is associated with mutations in the genes PTPN11, SOS1, RAF1, or KRAS, whereas CFCS can be caused by mutations in BRAF, MEK1, MEK2, or KRAS. The NS phenotype is rarely accompanied by multiple giant cell lesions (MGCL) of the jaw (Noonan-like/MGCL syndrome (NL/MGCLS)). PTPN11 mutations are the only genetic abnormalities reported so far in some patients with NL/MGCLS and in one individual with LEOPARD syndrome and MGCL. In a cohort of 75 NS patients previously tested negative for mutations in PTPN11 and KRAS, we detected SOS1 mutations in 11 individuals, four of whom had MGCL. To explore further the relevance of aberrant RAS-MAPK signaling in syndromic MGCL, we analyzed the established genes causing CFCS in three subjects with MGCL associated with a phenotype fitting CFCS. Mutations in BRAF or MEK1 were identified in these patients. All mutations detected in these seven patients with syndromic MGCL had previously been described in NS or CFCS without apparent MGCL. This study demonstrates that MGCL may occur in NS and CFCS with various underlying genetic alterations and no obvious genotype-phenotype correlation. This suggests that dysregulation of the RAS-MAPK pathway represents the common and basic molecular event predisposing to giant cell lesion formation in patients with NS and CFCS rather than specific mutation effects.
Article
Ras genes are the most common targets for somatic gain-of-function mutations in human cancer. Recently, germline mutations that affect components of the Ras-Raf-mitogen-activated and extracellular-signal regulated kinase kinase (MEK)-extracellular signal-regulated kinase (ERK) pathway were shown to cause several developmental disorders, including Noonan, Costello and cardio-facio-cutaneous syndromes. Many of these mutant alleles encode proteins with aberrant biochemical and functional properties. Here we will discuss the implications of germline mutations in the Ras-Raf-MEK-ERK pathway for understanding normal developmental processes and cancer pathogenesis.
Article
Nat. Genet. 34, 148–150 (2003). The subpanels in Figure 1 were labeled incorrectly. A corrected version appears below.
Article
We report on a family with typical clinical findings of Noonan syndrome associated with giant cell lesions in maxilla and mandible.We discuss the obvious clinical overlap between Noonan syndrome and Noonan-like/multiple giant cell lesion syndrome, and we give further clinical and molecular support that these two entities could be allelic conditions. © 2001 Wiley-Liss, Inc.
Article
Genetic analysis has enhanced our understanding of the biological roles of many protein tyrosine kinases (PTKs). More recently, studies utilizing both spontaneous mutants and mutants induced by homologous recombination techniques have begun to yield key insights into the role of specific protein tyrosine phosphatases (PTPs) and to suggest how PTKs and PTPs interact. Specific PTPs in Saccharomyces cerevesiae and Schizomyces pombe regulate MAP kinase pathways. Several Drosophila receptor PTPs control axonal targeting pathways, whereas the non-receptor PTP Corkscrew (Csw), plays an essential positive signaling role in multiple developmental pathways directed by receptor PTKs. The vertebrate homolog of Csw, SHP-2, also is required for growth factor signaling and normal development. Finally, very recent studies of other mammalian PTPs suggest that they have critical roles in processes as diverse as hematopoiesis and liver and pituitary development.
Article
Noonan syndrome (NS) is a phenotypically heterogeneous syndrome which is frequently associated with short stature. Recent genetic investigations have identified mutations in five genes, namely PTPN11, KRAS, SOS1, NF1 and RAF1 in patients with the NS phenotype. PTPN11 is the commonest, being present in approximately 50% of cases. The degree of short stature in children does not associate closely with the presence of mutations, however some PTPN11-positive patients have decreased GH-dependent growth factors consistent with mild GH insensitivity. GH therapy, using doses similar to those approved for Turner syndrome (TS), induced short-term increases in height velocity over 1-3 years, and may improve final adult height with longer-term treatment.