29
MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short stature Jan M Wit 1 , Wilma Oostdijk 1 , Monique Losekoot 2 , Hermine A van Duyvenvoorde 2 , Claudia A L Ruivenkamp 2 and Sarina G Kant 2 Departments of 1 Paediatrics and 2 Clinical Genetics, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands Correspondence should be addressed to J M Wit Email [email protected] Abstract The fast technological development, particularly single nucleotide polymorphism array, array-comparative genomic hybridization, and whole exome sequencing, has led to the discovery of many novel genetic causes of growth failure. In this review we discuss a selection of these, according to a diagnostic classification centred on the epiphyseal growth plate. We successively discuss disorders in hormone signalling, paracrine factors, matrix molecules, intracellular pathways, and fundamental cellular processes, followed by chromosomal aberrations including copy number variants (CNVs) and imprinting disorders associated with short stature. Many novel causes of GH deficiency (GHD) as part of combined pituitary hormone deficiency have been uncovered. The most frequent genetic causes of isolated GHD are GH1 and GHRHR defects, but several novel causes have recently been found, such as GHSR, RNPC3, and IFT172 mutations. Besides well-defined causes of GH insensitivity (GHR, STAT5B, IGFALS, IGF1 defects), disorders of NFkB signalling, STAT3 and IGF2 have recently been discovered. Heterozygous IGF1R defects are a relatively frequent cause of prenatal and postnatal growth retardation. TRHA mutations cause a syndromic form of short stature with elevated T 3 /T 4 ratio. Disorders of signalling of various paracrine factors (FGFs, BMPs, WNTs, PTHrP/IHH, and CNP/NPR2) or genetic defects affecting cartilage extracellular matrix usually cause disproportionate short stature. Heterozygous NPR2 or SHOX defects may be found in w3% of short children, and also rasopathies (e.g., Noonan syndrome) can be found in children without clear syndromic appearance. Numerous other syndromes associated with short stature are caused by genetic defects in fundamental cellular processes, chromosomal abnormalities, CNVs, and imprinting disorders. European Journal of Endocrinology (2016) 174, R145–R173 Introduction The fast technological development has caused a flood of novel discoveries in genetic causes of congenital disorders, including syndromic and non-syndromic forms of short stature. In the first decade of the 21st century, the genetic toolbox was expanded by whole genome single nucleotide polymorphism (SNP) array (1) and array-comparative Invited Author’s profile Professor Jan Maarten Wit is currently Professor Emeritus and honorary staff member of the Department of Paediatrics at Leiden University Medical Centre, The Netherlands. Trained as a paediatric endocrinologist, he served as an Associate Professor of Paediatric Endocrinology in Utrecht and Full Professor/Chairman of Paediatrics in Leiden. Most of his research has been focused on the diagnosis and management of growth disorders. Shortly after his PhD thesis (Responses to growth hormone therapy), he founded the Dutch Growth Hormone Advisory Group and the Dutch Growth Hormone Research Foundation’s bureau, instrumental in conducting numerous multicentre clinical trials on the efficacy and safety of growth hormone treatment. In Leiden, he led research projects on regulation of the epiphyseal growth plate and on referral criteria and diagnostic guidelines for short children, but the main subject focus over the last 10 years has been the elucidation of novel genetic causes of short and tall stature. European Journal of Endocrinology Review J M Wit and others Genetics of short stature 174 :4 R145–R173 www.eje-online.org Ñ 2016 European Society of Endocrinology DOI: 10.1530/EJE-15-0937 Printed in Great Britain Published by Bioscientifica Ltd. Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PM via free access

MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

  • Upload
    others

  • View
    7

  • Download
    0

Embed Size (px)

Citation preview

Page 1: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

ReviewJ M Wit and others Genetics of short stature 174 :4 R145–R173

MECHANISMS IN ENDOCRINOLOGY

Novel genetic causes of short stature

Jan M Wit1, Wilma Oostdijk1, Monique Losekoot2, Hermine A van Duyvenvoorde2,

Claudia A L Ruivenkamp2 and Sarina G Kant2

Departments of 1Paediatrics and 2Clinical Genetics, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden,

The Netherlands

Invited Author’s profile

Professor Jan Maarten Wit is currently Professor Emeritus and honorary staff member of the DepPaediatrics at Leiden University Medical Centre, The Netherlands. Trained as a paediatric endocrinserved as an Associate Professor of Paediatric Endocrinology in Utrecht and Full Professor/Chairman ofin Leiden. Most of his research has been focused on the diagnosis and management of growth disordeafter his PhD thesis (Responses to growth hormone therapy), he founded the Dutch Growth HormonGroup and the Dutch Growth Hormone Research Foundation’s bureau, instrumental in conductingmulticentre clinical trials on the efficacy and safety of growth hormone treatment. In Leiden, he leprojects on regulation of the epiphyseal growth plate and on referral criteria and diagnostic guidelinechildren, but the main subject focus over the last 10 years has been the elucidation of novel genetic cauand tall stature.

www.eje-online.org � 2016 European Society of EndocrinologyDOI: 10.1530/EJE-15-0937 Printed in Great Britain

Published by Bioscientifica Ltd.

Download

Correspondence

should be addressed

to J M Wit

Email

[email protected]

Abstract

The fast technological development, particularly single nucleotide polymorphism array, array-comparative genomic

hybridization, and whole exome sequencing, has led to the discovery of many novel genetic causes of growth failure.

In this review we discuss a selection of these, according to a diagnostic classification centred on the epiphyseal growth plate.

We successively discuss disorders in hormone signalling, paracrine factors, matrix molecules, intracellular pathways, and

fundamental cellular processes, followed by chromosomal aberrations including copy number variants (CNVs) and imprinting

disorders associated with short stature. Many novel causes of GH deficiency (GHD) as part of combined pituitary hormone

deficiency have been uncovered. The most frequent genetic causes of isolated GHD are GH1 and GHRHR defects, but several

novel causes have recently been found, such as GHSR, RNPC3, and IFT172 mutations. Besides well-defined causes of GH

insensitivity (GHR, STAT5B, IGFALS, IGF1 defects), disorders of NFkB signalling, STAT3 and IGF2 have recently been discovered.

Heterozygous IGF1R defects are a relatively frequent cause of prenatal and postnatal growth retardation. TRHA mutations

cause a syndromic form of short stature with elevated T3/T4 ratio. Disorders of signalling of various paracrine factors

(FGFs, BMPs, WNTs, PTHrP/IHH, and CNP/NPR2) or genetic defects affecting cartilage extracellular matrix usually cause

disproportionate short stature. Heterozygous NPR2 or SHOX defects may be found in w3% of short children, and also

rasopathies (e.g., Noonan syndrome) can be found in children without clear syndromic appearance. Numerous other

syndromes associated with short stature are caused by genetic defects in fundamental cellular processes, chromosomal

abnormalities, CNVs, and imprinting disorders.

artoloParse Anuds f

ses

ed

European Journal of

Endocrinology

(2016) 174, R145–R173

Introduction

The fast technological development has caused a flood of

novel discoveries in genetic causes of congenital disorders,

including syndromic and non-syndromic forms of short

stature. In the first decade of the 21st century, the genetic

toolbox was expanded by whole genome single nucleotide

polymorphism (SNP) array (1) and array-comparative

ment ofgist, he

ediatrics. Shortlydvisorymerous

researchor shortof short

from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 2: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R146

genomic hybridization (array-CGH) (2) for the detection

of microdeletions or microduplications (copy number

variants (CNVs)), the former of which is also able to detect

uniparental disomy. In the second decade an even more

successful tool became available – whole exome sequen-

cing (WES) – for the detection of gene variants as possible

causes of congenital disorders (3, 4, 5, 6), with a good

diagnostic yield in well-selected patients (6). In general,

WES is performed in an index patient and his/her parents

(a ‘trio’), and (if available) affected and non-affected

siblings, to limit the number of informative variants in

the bioinformatic analysis.

At the same time, information about genes associated

with linear growth was collected through non-clinical

research, in particular through genome-wide association

studies (GWAS) and animal and in vitro experiments on

epiphyseal growth plate (GP) function. GWAS have shown

that common SNPs at over 400 loci contribute to variation

in normal adult stature, albeit with a small effect size per

locus (7). Many of these genes, but also others, have

appeared in gene expression studies in the various zones

of the GP (8, 9).

For this review we chose to focus on discoveries made

in the last 10 years (up to August 2015), against the

background of earlier findings, as summarized in previous

reviews by our group (10, 11, 12, 13) and others (5, 14, 15,

16, 17) (for search strategy see section at the end of the

article). The tables offer the formal names of the disorders

and codes according to online Mendelian inheritance in

man (OMIM) (http://www.ncbi.nlm.nih.gov/omim), and

we aimed at providing the most recent relevant references.

In line with a recent review paper (17), we structured

this review according to a diagnostic classification centred

on the GP. In the GP, chondrocytes proliferate, hyper-

trophy, and secrete cartilage extracellular matrix, under

the influence of endocrine and paracrine factors. Thus, in

this review successively hormones, paracrine factors,

matrix molecules, intracellular pathways, and fundamen-

tal cellular processes will be discussed, followed by CNVs

and imprinting disorders. Because the GP is the structure

where linear growth takes place, we prefer this patho-

physiologic classification above the multiple reported

alternative classifications, for example proportionate vs

disproportionate short stature; with or without micro-

cephaly (18); prenatal vs postnatal onset of growth

retardation (19); or growth hormone (GH) deficiency or

insensitivity (20).

A complicating factor in the classification of mono-

genic disorders is that a variety of mutations in one gene

can result in a broad phenotypic spectrum, sometimes

www.eje-online.org

including different clinical entities, previously defined as

separate conditions (‘allelic heterogeneity’). On the other

hand, one clinical disorder can be caused by mutations in

different genes (‘locus heterogeneity’) (14). Furthermore,

mutations in some genes not only impair GP development

and/or function but also non-skeletal structures, causing

associated congenital anomalies (syndromic short

stature) (17).

The last decades have taught us that with time the

clinical phenotype of genetic defects tends to become

more variable than initially assumed. The rapid increase

of the use of SNP arrays and WES in the coming years, and

the expected appearance of whole genome sequencing

(WGS), RNA sequencing, and methylation assays, will

certainly lead to the discovery of many more novel causes

of short stature, as well as a further expansion of the

clinical phenotypes associated with genetic and epigenetic

variants.

Genetic defects of the GH–insulin-likegrowth factor 1 axis

The GH––insulin-like growth factor 1 (IGF1) axis is an

important pathway in the regulation of linear growth, and

defects have been found in virtually all components of this

cascade. Tables 1 and 2 show conditions associated with

GH or IGF1 signalling, divided into three categories: i) GH

deficiency (GHD); ii) GH insensitivity (GHI) and decreased

expression or biologic activity of IGF1 or IGF2; and iii)

IGF1 insensitivity. For various genes, a publicly available

database has been established (www.growthgenetics.com)

(21), and clinicians and geneticists are encouraged to

upload clinical and genetic data of additional cases.

GH deficiency

Table 1 shows the gene defects that have been associated

with GHD. Many of the proteins encoded by these

genes are associated with GHD as part of combined

pituitary hormone deficiency (CPHD), and function as

pituitary transcription factors (for detailed information on

associated clinical features and MRI appearances see (5, 22,

23, 24)). A novel endocrine syndrome discovered by our

group, immunoglobulin superfamily member 1 (IGSF1)

deficiency syndrome, is primarily characterized by central

hypothyroidism and macroorchidism, but can also

present with hypoprolactinaemia and transient partial

GHD (25, 26). The association of Netherton syndrome

with GH and prolactin deficiency suggests that a defect of

LEKT1 (encoded by SPINK5) may increase the degradation

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 3: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Table 1 Causes of GHD.

Disordera Gene(s) Clinical features Inheritance References

GHD and potential for CPHDCPHD-1 (613038) POU1F1 GH, PRL, var. TSH def. AR, AD (5, 22, 23)CPHD-2 (262600) PROP1 GH, PRL, TSH, LH, FSH, var. ACTH def.

Pituitary can be enlarged.AR (5, 22, 23)

CPHD-3 (221750) LHX3 GH, TSH, LH, FSH, PRL def. Sensorineural hearingloss, cervical abnormalities, short stiff neck

AR (5, 22, 23)

CPHD-4 (262700) LHX4 GH, TSH, ACTH def. AD, AR (5, 22, 23)Septo-optic dysplasia (CPHD-5)(182230)

HESX1 Optic nerve hypoplasia, pituitary hypoplasia,midline abnormalities of brain, absent corpuscallosum and septum pellucidum

AR, AD (5, 22, 24)

CPHD-6 (613986) OTX2 TSH, GH, LH, FSH, var. ACTH and PRL def. AD (5, 22, 24)Axenfeld–Rieger syndrome type 1(180500)

PITX Coloboma, glaucoma, dental hypoplasia,protuberant umbilicus, brain abnormalities,var. pituitary def.

AD (22)

Optic nerve hypoplasia andabnormalities of the centralnervous system (206900)

SOX2 Var. GHD, hypogonadism, anophthalmia,developmental delay

AD (22, 24)

X-linked panhypopituitarism(312000, 300123)

SOX3dupb GHD or CHPD, mental retardation XLR (5, 22, 24)

Dopa-responsive dystonia due tosepiapterin reductase deficiency(612716)

SPR Diurnally fluctuating movement disorder,cognitive delay, neurologic dysfunction,GH and TSH def.

AR (237)

Holoprosencephaly 9 (610829) GLI2 Holoprosencephaly, craniofacial abnormalities,polydactyly, single central incisor, partial agen-esis corpus callosum (or hypopituitarism only)

AD (5, 22)

IGSF1 deficiency syndrome(300888)

IGSF1 TSH, var. GH and PRL def.; macroorchidism XLR (26)

Netherton syndrome (256500) SPINK5 Var. GH and PRL def. AR (27)Pallister–Hall syndrome (146510) GLI3 Hypothalamic hamartoma, central polydactyly,

visceral malformationsAD (5)

FGF8 Holoprosencephaly, septo-optic dysplasia,Moebius syndrome

AR (5, 24)

FGFR1 Hypoplasia pituitary, corpus callosum, oculardefects

AD (5, 238)

PROKR2 Var. hypopituitarism AD (238)HMGA2 Severe GHD, ectopic posterior pituitary AD (239, 240)GRP161 Pituitary stalk interruption syndrome, intellectual

disability, sparse hair in frontal area, hypo-telorism, broad nasal root, thick alae nasi, nailhypoplasia, short fifth finger, 2–3 toe syndactyly,hypopituitarism

AR (241)

Isolated GHD or bioinactivityIsolated GHD, type IB (612781) GHRHR Low serum GH AR (240, 242)Isolated GHD, type 1A (262400) GH1 No serum GH, often anti-GH ab AR (240, 242)Isolated GHD, type IB (612781) GH1 Low serum GH AR (240, 242)Isolated GHD, type II (173100) GH1 Var. height deficit and pituitary size; other pituitary

deficits can developAD (240, 242)

Isolated GHD, type III (307200) BTK, SOX3 GHD with agammaglobulinemia XLR (240, 242)Isolated partial GHD (615925) GHSR Var. serum GH and IGF1 AR, AD (39, 41)Kowarski syndrome (bioinactiveGH syndrome) (262650)

GH1 high GH; def. of IGF1, IGFBP-3, and ALS AD (242)

Almstrom syndrome (203800) ALMS1 50% of cases are GHD AR (35)RNPC3 Severe GHD, hypoplasia anterior pituitary AR (33)IFT172 Functional GHD, retinopathy, metaphyseal

dysplasia, hypertensionAR (34)

AD, autosomal dominant; AR, autosomal recessive; def., deficiency; GHBP, growth hormone binding protein; GHD, growth hormone deficiency;IGF1, insulin-like growth factor 1; IGFBP-3, IGF binding protein-3; PRL, prolactin; var., variable; XLR, X-linked recessive.aName (number) according to OMIM. For clinical and radiological features of the various conditions, see (5, 22, 23, 24).bThis condition can also be caused by SOX3 polyalanine deletions and expansions.

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R147

www.eje-online.org

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 4: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Table 2 Causes of GH insensitivity or IGF insensitivity.

Disordera Gene(s) Clinical features Inheritance References

GH insensitivityLaron syndrome (262500) GHR Variable height deficit and GHBP,

midfacial hypoplasia;[GH, YIGF1,IGFBP-3 and ALS

AR (AD) (46, 242)

GH insensitivity with immuno-deficiency (245590)

STAT5B Midfacial hypoplasia, immuno-deficiency; [GH and PRL; YIGF1,IGFBP-3 and ALS

AR (55)

Multisystem, infantile-onsetautoimmune disease (615952)

STAT3 (act) Associated with early-onset multi-organautoimmune disease

AD (68, 69)

X-linked severe combinedimmunodeficiency (300400)

IL2RG GH normal, low IGF1, non-respondingto GH injections

XLR (243, 244)

IGF1 deficiency (608747) IGF1 SGA, microcephaly, deafness; [GH andIGFBP-3; variable IGF1

AR (13)

Severe growth restriction withdistinctive facies (616489)

IGF2 Y[/nl GH, IGFBP3; nl IGF1 Pat inheritance (82)

ALS deficiency (615961) IGFALS Mild height deficit; GH?,YIGF1, IGFBP-3and ALS

AR (59)

PAPP-A2 Microcephaly, skeletal abnormalities,[GH, IGF1, IGFBP-3, and ALS

AR (84)

Immunodeficiency 15 (615592) IKBKB Immunodeficiency; YIGF1 and IGFBP-3 AR, AD (65)IGF insensitivityResistance to insulin-like growthfactor 1

IGF1R SGA, microcephaly; [/nl GH, IGF1, andIGFBP-3

AD, AR (85)

act, activating; AD, autosomal dominant; ALS, acid-labile subunit; AR, autosomal recessive; GH, growth hormone; GHBP, growth hormone binding protein;IGF1, insulinlike growth factor 1; IGFBP-3, insulin-like growth factor binding protein 3; SGA, small for gestational age; XLR, X-linked recessive.aName (number) according to OMIM.

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R148

of these hormones in pituitary cells by human tissue

kallikreins before they enter the circulation (27). Other

causes of CPHD include mutations in GLI3, FGF8, FGFR1,

PROKR2, HMGA2, and GRP161 (Table 1).

Isolated GHD mutations in the genes encoding GH

(GH1) or GH releasing hormone receptor (GHRHR) can be

found in up to 34% in familial cases (28). GH1 mutations

can either lead to classical GHD (types IA, IB, and II) or

bioinactive GH syndrome. While in the past the latter

diagnosis was used without good experimental evidence,

recent reports have shown that this is a real condition,

characterized by normal or even elevated circulating GH

levels, and in some cases also associated with partial GHI

(28, 29, 30).

The most common cause of type IA GHD is a

homozygous GH1 deletion; in most of such patients

anti-GH antibodies develop with GH treatment. However,

several other aberrations of GH1 have been described. The

less severe type IB GHD is caused by mutations of GH1 or

GHRHR, and a dominant form of GHD (type II) is usually

caused by skipping of exon 3 resulting in production of a

17.5-kDa isoform of GH with a dominant negative effect

(28). The X-linked type III GHD is associated with

agammaglobulinaemia, and has been associated with

mutations in BTK (31) and SOX3 (32).

www.eje-online.org

Isolated GHD can also be caused by biallelic mutations

in RNPC3, which encodes a minor spliceosome protein

required for U11/U12 small nuclear ribonucleoprotein

(snRNP) formation and splicing of U12-type introns (33).

Compound heterozygosity for a gene encoding a protein

important for ciliary function (IFT172) can cause

functional GHD, pituitary hypoplasia, and ectopic pos-

terior pituitary (34), and also Alstrom syndrome, caused

by a mutation of ALMS1 encoding a protein localized to

the centrosomes and basal bodies of ciliated cells (35) is

associated with GHD. GHD has also been documented

in a congenital malformation syndrome associated with a

paternal deletion of 6q24.2–q25.2 (36), complete general-

ized glucocorticoid resistance (37), and mitochondrial

diseases (38).

A still insufficiently defined cause of GHD is a

mutation of the gene encoding the Ghrelin receptor

(GHSR) (reviewed in (39)). The variability of clinical

phenotypes (GHD, idiopathic short stature (ISS) and

constitutional delay of growth and puberty (CDGP)) and

incomplete segregation of the mutations with the pheno-

type still cast doubt on the role of GHSR mutations in

causing short stature, although functional studies do

suggest that GHSR mutations may decrease GH secretion

(40, 41, 42), implying that GHSR mutations may

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 5: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R149

contribute to the genetic aetiology of children originally

considered ISS (41).

GHI and decreased expression or biologic activity of

IGF1or IGF2

Table 2 shows the various syndromes presenting with

insensitivity to GH or IGF1. The first discovered cause

of GHI was Laron syndrome, usually caused by a

homozygous mutation of the gene encoding the GH

receptor (GHR) (43, 44, 45). Since then more than 70

mutations in O300 cases have been found with mutations

in extracellular, transmembrane, and intracellular parts of

the GHR (46, 47). In most cases serum GH binding protein

(GHBP) is absent, except in cases with a mutation in the

intracellular or transmembrane part of the protein. While

the classical form causes severe growth failure, there are

milder forms as well, for example caused by an intronic

base change leading to the activation of a pseudoexon

sequence and insertion of 36 new amino acids within the

receptor extracellular domain (48, 49, 50) or by hetero-

zygous GHR mutations causing a dominant negative effect

(51, 52, 53).

In 2003 the first patient with a homozygous loss-

of-function mutation of the gene encoding the main

component of the intracellular GH signalling pathway

(STAT5B) was found (54), and since then ten patients have

been reported in seven families (55). Most have an

additional immunodeficiency and pulmonary fibrosis

(56). Heterozygosity for a STAT5B mutation leads to a

slightly lower height (57).

Another well-defined cause of GHI is a defect in

IGFALS, encoding acid-labile subunit (ALS) which forms

with IGF binding protein 3 (or 5) and IGF1 (or IGF2) a

ternary complex in the circulation (58, 59). Children with

ALS deficiency show a mild growth failure, delayed

puberty, undetectable serum ALS, low serum IGF1, and

even lower IGF binding protein 3 (IGFBP-3) (59), and

variable osteopenia and hyperinsulinism (60, 61, 62).

Heterozygosity for IGFALS variants causes a one S.D. lower

height (60, 62, 63) and may be responsible for a subset of

children previously considered having ISS (64).

GHI may also be caused by a mutation in the gene

encoding IkBa (IKBKB), presenting with short stature,

GHI, severe immune deficiency and other features (65) or a

PRKCA duplication, in a patient with a mosaic de novo

duplication of 17q21–25 (66) (reviewed in (67)). Further-

more, activating STAT3 mutations may be not only

associated with early-onset multi-organ autoimmune

disease, but also with growth failure (68, 69).

Homozygous deletions or missense mutations of

IGF1 (encoding IGF1) resulting in a complete loss-of-

function (70, 71) cause a severe prenatal and postnatal

growth failure, developmental delay, microcephaly, and

sensorineural deafness. Patients with a homozygous

hypomorphic mutation (72) or specific heterozygous

mutations (73, 74) presented with less severe growth failure

and normal hearing (reviewed in (75)). Heterozygous

carriers of IGF1 mutations or deletions are w1 S.D. shorter

than non-carriers (71, 73, 74, 76).

With regard to IGF2, it is assumed that in most

children with Silver–Russell syndrome the pre- and post-

natal growth restriction is caused by deficient expression

of the paternally expressed gene encoding IGF2 (IGF2) (77,

78), usually through H19 hypomethylation. Such children

can have relatively high serum IGF1 and IGFBP-3,

suggesting partial IGF1 resistance (79, 80). In contrast,

Silver–Russell syndrome patients carrying a maternal

uniparental disomy of chromosome 7 (UPD7) usually

present with low levels of IGF1 (79, 81). Very recently, the

first family with a paternally inherited IGF2 mutation

and growth restriction was reported, indicating that

IGF2 not only is a mediator of intrauterine development

but also contributes to postnatal growth (82). This

confirmed an earlier observation of a patient with a

paternally transmitted severe intrauterine growth retar-

dation (IUGR) with a translocation breakpoint disrupting

regulation of IGF2 (83).

Another novel finding is that a homozygous mutation

of the gene encoding the protease PAPPA-2 (PAPPA2) is

associated with mild short stature, presumably by insuffi-

cient availability of free IGF1 (84).

IGF1 insensitivity

Numerous cases have been reported of heterozygous

mutations or deletions of the gene encoding the receptor

for IGF1 (and IGF2) (IGF1R) (reviewed in (75, 85)). Clinical

features include prenatal growth failure persisting after

birth, microcephaly, and serum IGF1 in the upper half of,

or above, the normal range. On GH treatment serum IGF1

can become very high, which may probably be accepted

because of the decreased sensitivity. We estimate that

IGF1R defects can be found in w3% of short children born

small for gestational age (SGA) (86). A homozygous or

compound heterozygous IGF1R mutation leads to a more

severe phenotype (87, 88, 89, 90). In theory, IGF1

insensitivity may also be caused by mutations downstream

of the IGF receptor, or by defective microRNA regulation

of IGF1 signalling (91).

www.eje-online.org

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 6: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R150

Genetic defects affecting signalling of otherhormones regulating GP function

Congenital disorders of thyroid hormone signalling

include primary hypothyroidism (thyroid dysgenesis or

dyshormonogenesis) and thyroid hormone resistance.

If undiagnosed, congenital hypothyroidism leads to very

severe growth failure (92), but in most middle- and high-

income countries early detection by neonatal screening

will prevent this, as well as the severe consequences for

mental development. Presently known genetic causes of

thyroid dysgenesis and dyshormonogenesis have recently

been reviewed (17, 93).

Children with thyroid hormone resistance caused by

mutations of THRB (encoding the beta form of the thyroid

hormone receptor (TRb)) usually show normal growth, but

in severe cases short stature has been observed (94).

In contrast, all reported children with mutations in

THRA (encoding TRa) are short. Further clinical features

include delayed mental and bone development, consti-

pation, and relatively low serum T4 and high serum T3

levels (elevated T3/T4 ratio) (95, 96). An opposite serum

thyroid hormone profile (elevated T4 and low-normal or

slightly decreased T3) is seen in a homozygous or

compound heterozygous mutation of SECISBP2 (SBP2)

(encoding an iodothyronine deiodinase), associated with

short stature and responding to GH and T3 treatment (97).

It is well known that growth failure can be caused by

excessive exposure to glucocorticoids, due to Cushing

syndrome or pharmacological doses of corticosteroids.

A discussion of newly discovered genetic causes of ACTH-

dependent and independent Cushing syndrome is outside

the scope of this paper (for recent findings, see (98, 99)).

Homozygous or compound heterozygous mutations of the

gene encoding the insulin receptor (INSR) cause Donohue

syndrome (Leprechaunism) (100).

Genetic defects affecting paracrine factorsin the GP

Paracrine regulation plays a major role in the GP, and only

part of its complexity is presently understood. Most of the

genetic defects of paracrine pathways result in some form

of skeletal dysplasia, of which 436 conditions, caused by

defects in 364 genes, have been listed in the 2015 revision

of the nosology of genetic skeletal disorders (101).

Disproportionate short stature is one of the main features

of most of these conditions. Therefore, in the clinical

assessment of the short individual, not only accurate

measurements of height and head circumference have to

www.eje-online.org

be carried out, but also of sitting height and arm span, and

the same measurements should be performed in the

parents. The length of upper and lower arms and legs,

and hands and feet, should be at least visually assessed,

and possibly measured and compared with normative

charts (a relatively short upper arm and leg is called

rhizomelia, in contrast to mesomelia if forearm and lower

leg are relatively short). A series of skeletal radiographs

usually gives important clues for the diagnosis (15, 102,

103, 104). Most forms of skeletal dysplasia show short-

limb dwarfism, in contrast to type I and II collageno-

pathies which are characterized by short-trunk dwarfism

(14). Because a comprehensive review of these conditions

is beyond the scope of this article, only a few relatively

common conditions are discussed (Table 3).

Fibroblast growth factor signalling

Several fibroblast growth factors (FGFs) and their receptors

play a role in the GP (9, 105). Best known is the FGF

receptor-3 (encoded by FGFR3), which acts as a negative

regulator of GP chondrogenesis (106, 107). Consequently,

heterozygous activating mutations in FGFR3 impair bone

elongation and lead to a spectrum of disorders, reflecting

the degree of activation of the FGFR3 mutation. The best

known examples are thanatophoric dysplasia, achondro-

plasia, and hypochondroplasia, each associated with

different locations of the mutation. The clinical presen-

tation of hypochondroplasia is milder and more variable

than achondroplasia and includes rhizomelic limb

shortening, limitation of elbow extension, brachydactyly,

relative macrocephaly, generalized laxity, and specific

radiologic features (5, 108). We recently reported a novel

activating FGFR3 mutation in a family with proportionate

short stature (109).

Bone morphogenetic protein signalling

Bone morphogenetic proteins (BMPs), also known as

growth and differentiation factors (GDFs), belong to the

transforming growth factor-beta (TGFb) superfamily of

paracrine factors. The BMPs regulate a multitude of

processes in skeletal development, including spatial

regulation of proliferation and differentiation in the GP,

and a BMP signalling gradient across the GP may

contribute to the progressive differentiation of resting to

proliferative to hypertrophic chondrocytes (9). Inacti-

vating mutations in the genes for several BMPs, their

receptors, and antagonists cause various forms of skeletal

dysplasias, particularly brachydactylies.

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 7: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Table 3 Examples of genetic defects affecting paracrine factors in the growth plate.

Disordera Gene(s) Clinical features Inheritance References

FGF signalingPfeiffer syndrome, acrocephalo-syndactyly, type V (101600)

FGFR1, FGFR2 Craniosynostosis with characteristicanomalies of the hands and feet(three types)

AD (245)

Thanatophoric dysplasia type I(187600)

FGFR3 (act) Severe short-limb dwarfism syndrome usuallylethal in the perinatal period

AD (9)

Achondroplasia (100800) FGFR3 (act) Rhizomelic limb shortening, frontal bossing,midface hypoplasia, exaggeratedlumbar lordosis, limited elbow extension,genu varum, trident hand

AD (9)

Hypochondroplasia (146000) FGFR3 (act) Short-limbed dwarfism, lumbar lordosis,short and broad bones, caudal narrowingof interpediculate distance of lumbar spine

AD (9, 108, 246)

Short stature FGFR3 (act) Relative macrocephaly for height AD (109)BMP signalingBrachydactyly A1 (112500) IHH, GDF5,

BMPR1BMiddle phalanges rudimentary or fused with

terminal phalanges, short proximalphalanges thumbs and big toes

AD (247)

Brachydactyly A2 (112600) BMPR1B, BMP2,GDF5

Malformations of middle phalanx of indexfinger, anomalies of second toe

AD (248)

Brachydactyly C (113100) GDF5, CDMP1 Deformity of middle and proximal phalanges(II, III), hypersegmentation of proximalphalanx

AD (249)

WNT signalingRobinow syndrome (268310) ROR2, WNT5A Frontal bossing, hypertelorism, broad nose,

short-limbed dwarfism, vertebralsegmentation, genital hypoplasia

AR, AD (112)

Brachydactyly, Type B1 (113000) ROR2 Short middle phalanges, terminal phalangesrudimentary or absent; deformed thumbs,big toes

AD (113)

PTHrP-IHH pathwayBrachydactyly, type E2 (613382) PTHLH Short stature and learning difficulties AD (116)Blomstrand chondro-dysplasia(215045)

PTHR1 Short limbs, polyhydramnios, hydrops fetalis,facial anoma-lies, increased bone density,advanced skeletal maturation

AR (117)

Jansen type of meta-physealchondrodys-plasia (156400)

PTHR1 (act) Severe short stature, short bowed limbs,clinodactyly, prominent upper face,small mandible; hypercalcemia andhypophosphatemia

AD (118)

Brachydactyly type A1 (112500) IHH, GDF5,BMPR1B

Middle phalanges rudimentary or fused withterminal phalanges. Short proximalphalanges of thumbs, big toes

AD (119)

Acrocapitofemoral dysplasia(607778)

IHH Variable short stature, short limbs withbrachydactyly, relatively large headcircumference

AR (119)

Albright hereditary osteodystrophy(103580)

GNAS Pseudohypoparathyroidism, type Ia/c.Caused by loss of function of Gs-alphaisoform of GNAS on maternal allele.For further details see Table 8

Imprinted (228)

Acrodysostosis 1 (101800) PRKAR1A Severe brachydactyly, facial dysostosis, nasalhypoplasia, advanced bone age, obesity,resistance to multiple hormones

AD (121)

CNP-NPR2 pathwayAcromesomelic dysplasia,Maroteaux type (602875)

NPR2 Disproportionate shortening of middlesegments (forearms and forelegs) anddistal segments (hands and feet)

AR (124)

(Dis)proportionate short stature NPR2 Moderate short stature, short forearms andforelegs

AD (130)

AD, autosomal dominant; AR, autosomal recessive; act, activating.aName (number) according to OMIM.

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R151

www.eje-online.org

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 8: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R152

WNT signalling

The receptor tyrosine kinase-like orphan receptor 2 (RoR2) is

part of a conserved family of tyrosine kinase-like receptors

that serve as receptors for noncanonical WNT ligands,

participating in developmental processes like cell move-

mentand cellpolarity (110, 111). Homozygous mutations of

ROR2 or heterozygous mutations of WNT5A cause Robinow

syndrome (112) and dominant ROR2 mutations cause

brachydactyly, Type B1 (113).

PTHrP–IHH pathway

Parathyroid hormone related peptide (PTHrP) and Indian

Hedgehog (IHH) form a negative feedback loop within the

GP that regulates chondrocyte hypertrophy and prolifer-

ation (114, 115). Heterozygous loss-of-function mutations

in PTHLH, encoding PTHrP, and inactivating and activat-

ing mutations in PTHR1 (encoding the parathyroid

hormone receptor-1) cause various short stature syn-

dromes (116, 117, 118), as well as inactivating and

activating mutations of IHH (119, 120). Heterozygous

missense mutations in PRKAR1A and PDE4D cause

acrodysostosis 1 and 2 respectively, with or without

hormone resistance (121, 122).

CNP–NPR2 pathway

One of the most interesting breakthroughs in the field

of growth genetics is the unravelling of the role of

C-natriuretic peptide (CNP, encoded by NPPC) and its

receptors in GP function. CNP is a local, positive regulator

of GP function, and SNPs in NPPC and in the gene encoding

one of its receptors (NPR3) show a significant association

with adult height in GWAS (123). Homozygous inactivat-

ing mutations of NPR2 (encoding the main CNP receptor)

cause a severe skeletal dysplasia, acromesomelic dysplasia,

Maroteaux type (124). Initial observations that relatives

heterozygous for NPR2 mutations of patients with acrome-

somelic dysplasia are shorter than non-carriers (125), were

confirmed by recent studies (126, 127, 128, 129). The

phenotype of heterozygous NPR2 mutations is similar to

that of patients with SHOX haploinsufficiency (Leri–Weill

syndrome), with short forearms and lower legs

(mesomelia), except for the absence of Madelung deformity

(130). Heterozygous NPR2 mutations may explain 2–3% of

cases with assumed ISS (129) and probably more if one of

the parents has a similar phenotype.

Unravelling of the role of this pathway in linear

growth has led to potential therapeutic consequences for

www.eje-online.org

children with achondroplasia. Binding of CNP to NPR2

stimulates the receptor guanylyl cyclase activity thereby

increasing synthesis of cGMP, activating the type II cGMP-

dependent protein kinase (131), which in turn leads to

inhibition of the MAPK pathway, thus antagonizing FGFR

signalling (132). In a mouse model of achondroplasia,

CNP had beneficial effects (133), and clinical trials with a

long-acting CNP analogue are in progress in children with

achondroplasia.

Genetic defects affecting cartilageextracellular matrix

A unique characteristic of chondrocytes is that they

secrete an extracellular matrix containing specific

collagens, non-collagenous proteins and proteoglycans,

which are vital to normal GP function. This extracellular

matrix not only provides the compressible, resilient

structural properties of cartilage, but also interacts with

signalling molecules to regulate GP chondrogenesis (17).

Mutations in several genes encoding matrix proteins

and proteoglycans have been shown to lead to growth

disorders (Table 4). Mutations in ACAN, encoding aggre-

can, show a gene-dose effect: homozygous mutations

cause a severe skeletal dysplasia, spondyloepimetaphyseal

dysplasia aggrecan type (134), while heterozygous

mutations can present as a milder skeletal dysplasia,

spondyloepiphyseal dysplasia type Kimberley, or as short

stature without evident radiographic skeletal dysplasia

(135). This latter form is associated with an advanced bone

age and early cessation of growth (17, 135).

Some disorders, such as the genetically heterogeneous

brachyolmia, tend to affect the spine more than the long

bones, for example mutations in PAPSS2 encoding a

sulphotransferase, required for sulphation of a variety of

molecules, including cartilage glycosaminoglycans and

DHEA (136, 137).

Genetic defects of intracellular pathways

Various intracellular pathways play a role in chondrocyte

differentiation in the GP, and examples of disorders in

such pathways are listed in Table 5.

For the clinician, the relatively frequent aberrations of

the gene encoding short stature homeobox (SHOX)

(located at the tip of the X and Y chromosome, and

transmitted in a pseudoautosomal fashion) are most

relevant. SHOX acts as a transcriptional activator and,

like in NPR2 mutations, a gene-dose effect is apparent:

homozygous or compound heterozygous inactivating

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 9: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Table 4 Examples of genetic defects affecting cartilage extracellular matrix.

Disordera Gene(s) Clinical features Inheritance References

Acromicric dysplasia (102370) FBN1 Severe short stature, short hands and feet,joint limitations, skin thickening

AD (250, 251)

Geleophysic dysplasia-2 (614185) FBN1 Severe short stature, short hands and feet,joint limitations, skin thickening, heartinvolvement

AD (250, 251)

Brachyolmia type 4 with mildepiphyseal and metaphysealchanges (spondyloepimeta-physeal dysplasia, Pakistanitype) (612847)

PAPSS2 Short trunk, normal intelligence and facies;rectangular vertebral bodies with irregularendplates and narrow intervertebral discs,precocious calcification of rib cartilages,short femoral neck, mildly shortenedmetacarpals, and mild epiphyseal andmetaphyseal changes of the tubular bones

AR (137, 252)

Hurler syndrome (607014) IDUA Skeletal deformities, corneal clouding,hepatosplenomegaly, psychomotor delay

AR (253)

Metaphyseal chondro-dysplasia,Schmid type (156500)

COL10A1 Short stature, widened growth plates,bowing of long bones

AD (254)

Multiple epiphyseal dysplasia 1–6 COMP, COL9A2,COL9A3, SLC26A2,MATN3, COL9A1

Short-limbed dwarfism, joint pain andstiffness and early onset osteoarthritis

AD (255)

Pseudoachondro-plasia (177170) COMP Disproportionate short stature, deformity oflower limbs, brachydactyly, loose joints,ligamentous laxity, vertebral anomalies,osteoarthritis

AD (256)

Spondyloepiphyseal dysplasiacongenita (183900)

COL2A1 Multiple presentations AD (257)

Spondyloepimetaphy-sealdysplasia aggrecan type(612813)

ACAN Relative macrocephaly, severe midfacehypoplasia, almost absent nasal cartilage,relative prognathism, slightly low-set,posteriorly rotated ears; short neck, barrelchest, mild lumbar lordosis; rhizomelia andmesomelia

AR (134)

Spondyloepiphyseal dysplasiatype Kimberley (608361)

ACAN Proportionate short stature, stocky habitus,progressive osteoarthropathy

AD (258)

Short stature with advancedbone age

ACAN Advanced bone age, premature growthcessation

AD (135)

Weill–Marchesani syndrome(613195, 608328)

ADAMTS10, FBN1 Spherophakia, lenticular myopia, ectopialentis, joint stiffness, brachydactyly

AR (259)

AD, autosomal dominant; AR, autosomal recessive.aName (number) according to OMIM.

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R153

SHOX mutations cause Langer mesomelic dysplasia, while

heterozygous mutations or deletions of SHOX cause a

milder skeletal dysplasia, Leri–Weill dyschondrosteosis

(with the classic Madelung deformity of the wrist) or

present clinically as ISS. It is assumed that most of the

growth failure characteristic for Turner syndrome is

caused by heterozygous SHOX deletion. Body proportions

are usually mildly affected (mesomelia) but can be within

the normal range (138). Various clinical prediction rules

have been proposed to select patients for testing (139, 140,

141, 142), but the high variability of the clinical

presentation limits their predictive value (5). SHOX

mutations account for 2–15% of individuals presenting

with ISS (143). Since usually SHOX defects are transmitted

from one of the parents, physical examination of the

parents is essential, including height, sitting height,

arm span, forearm length, and presence of Madelung

deformity.

Heterozygous deletions of the downstream and

upstream enhancer of SHOX cause a similar phenotype as

defects of SHOX itself (144, 145, 146, 147, 148, 149), and the

growth response to GH treatment is even better in children

carrying a deletion of the SHOX enhancer than in carriers

of a SHOX defect (150). The consequences of increased

copies of SHOX are less clear (146, 150, 151, 152, 153).

A second intracellular pathway that plays a role in

cellular proliferation and differentiation of GP chondro-

cytes is the Ras/MAPK signalling pathway, which inte-

grates signals from several growth factors including GH,

FGFs, CNP, and EGF (154, 155). Activation of this pathway

results in a number of overlapping syndromes, called

‘rasopathies’, including Noonan, LEOPARD, Costello,

www.eje-online.org

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 10: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Table 5 Examples of genetic defects affecting intracellular pathways.

Disordera Gene(s) Clinical features Inheritance References

SHOX aberrationsLanger mesomelic dysplasia (249700) SHOX Severe limb aplasia or hypoplasia of

the ulna and fibula, and a thickenedand curved radius and tibia

AR (138)

Leri–Weill dyschon-drosteosis(127300)

SHOX Mesomelia, Madelung wrist deform-ity, or mild body disproportion

AD (138, 149)

RasopathiesNoonan syndrome 1–8 PTPN11, KRAS,

SOS1, RAF1,NRAS, BRAF,RIT1

Facial dysmorphism, wide spectrum ofcongenital heart defects

AD (157, 260, 261)

LEOPARD syndrome1 (151100)2 (611554)3 (613707)

PTPN11,RAF1,BRAF

Multiple lentigines, electrocardio-graphic conduction abnormalities,ocular hypertelorism, pulmonicstenosis, abnormal genitalia,sensorineural deafness

AD (260)

Costello syndrome (218040) HRAS Coarse facies, distinctive hand postureand appearance, feeding difficulty,failure to thrive, cardiac anomalies,developmental delay

AD (260)

Cardio-facio-cutaneous syndrome(115150)

BRAF, KRAS Distinctive facial appearance, heartdefects, mental retardation

AD (260)

Neurofibromatosis-Noonansyndrome (601321)

NF1 Features of both conditions AD (260)

Neurofibromatosis type I (162200) NF1 Cafe-au-lait spots, Lisch nodules ineye, fibroma-tous skin tumours;short in 13%; large headcircumference in 24%

AD (262)

Coffin–Lowry syndrome (303600) RPS6KA3 Mental retardation, skeletalmalformations, hearing deficit,paroxysmal movement disorders

XLR (261)

Other syndromesAarskog–Scott syndrome(faciogenital dysplasia) (305400)

FGD1 Hypertelorism, shawl scrotum,brachydactyly

XLR (263)

Alstrom syndrome (203800) ALMS1 Retinal photoreceptor degeneration,sensorineural hearing imparment,obesity, insulin resistance

AR (35)

Campomelic dysplasia (114290) SOX9 Congenital bowing and angulation oflong bones, other skeletal andextraskeletal defects

AD (264)

Congenital disorders of glycosylation Multiple genes(O76)

Multisystem disorders caused bydefects in biosynthesis of glyco-conjugates

AR (168)

Kabuki syndrome 1 (147920) and 2(300867)

KMT2D, KDM6A Long palpebral fissures, eversion oflateral third of the lower eyelids,broad and depressed nasal tip, largeprominent earlobes, cleft or high-arched palate, scoliosis, short fifthfinger, persistence of fingerpads,radiographic abnormalities ofvertebrae, hands, and hip joints,recurrent otitis media in infancy

AD (265)

Kenny–Caffey syndrome type 1(244460) and 2 (127000)

TBCE, FAM111A Craniofacial anomalies, small handsand feet, hypocalcemia, hypopara-thyroidism, cortical thickening oflong bones with medullary stenosis,delayed closure of anterior fonta-nel, eye abnormalities, transienthypocalcemia. Gene encodestubulin-specific chaperone E.

ARAD

(6, 266)

AD, autosomal dominant; AR, autosomal recessive; XLR, X-linked recessive.aName (number) according to OMIM.

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R154

www.eje-online.org

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 11: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R155

cardio-facio-cutaneous, and neurofibromatosis–Noonan

syndrome, all characterized by postnatal growth failure

of varying degree (156, 157). Mutations in these genes can

also cause short stature without obvious clinical features

(158). Inhibition of IGF1 release via GH-induced ERK

hyperactivation or EGF-induced PI3K/AKT/GSK-3b stimu-

lation may contribute to short stature in patients with

PTPN11 mutations (159, 160).

Genetic aberrations in several other intracellular

pathways play a role in short stature syndromes. For

example, mutations in FGD1, encoding a guanine nucleo-

tide exchange factor of the Rho/Rac family of small

GTP-binding proteins, cause the X-linked form of

Aarskog–Scott syndrome (faciogenital dysplasia) (161),

although in only 18% of clinically suspected cases a

mutation was found (162). FGD1 activates MAP3K mixed-

lineage kinase 3 (MLK3), which regulates ERK and p38

MAPK, which in turn phosphorylate and activate the

master regulator of osteoblast differentiation, RUNX2

(163). FGD1 is involved in the regulation of the formation

and function of invadopodia and podosomes, which are

cellular structures devoted to degradation of the extra-

cellular matrix in tumour and endothelial cells (164).

Inactivating mutations in SOX9 cause a severe skeletal

dysplasia, campomelic dysplasia. The encoded protein

and its distant relatives SOX5 and SOX6 also activate the

genes for cartilage-specific extracellular matrix com-

ponents (165).

Congenital disorders of glycosylation (CDG) are a

rapidly expanding family of genetic diseases due to defects

in the synthesis of the glycan moiety of glycoproteins and

glycolipids and in their attachment to proteins and lipids.

Most CDG are multisystem disorders, and many are

associated with skeletal abnormalities, including short

stature and microcephaly (166, 167, 168).

Genetic defects in fundamental cellularprocesses

Mutations in genes encoding proteins involved in

fundamental cellular processes can produce severe global

growth deficiencies, termed primordial dwarfisms, which

affect not just the GP but multiple other tissues and

typically impair both pre- and post-natal growth (17).

Several of these syndromes are associated with a normal

head circumference, but many are microcephalic. In some

syndromes, DNA repair defects are prominent. Some

examples are presented in Tables 6 and 7, classified

according to head size and DNA repair.

Syndromes with (usually) normal head circumference

CHARGE syndrome is caused by heterozygous mutations

in CHD7 (169) or SEMA3E (170). CHD7 is a transcriptional

regulator that binds to enhancer elements in the nucleo-

plasm, and also functions as a positive regulator of rRNA

biogenesis in the nucleolus (171).

Patients diagnosed with Coffin–Siris syndrome have a

broad clinical variability, and at present mutations in six

genes have been reported, all encoding components of the

SWI/SNF complex (172, 173). The gene associated with

Floating–Harbor syndrome (SRCAP) encodes a component

of SWI/SNF chromatin remodelling complexes (174, 175).

The KBG syndrome is caused by a heterozygous

mutation in ANKRD11 (176), encoding a member of a

family of ankyrin repeat-containing cofactors that

interacts with p160 nuclear receptor coactivators and

inhibits ligand-dependent transcriptional activation (177).

Mulibrey nanism (referring to muscle, liver, brain and

eye) is caused by homozygous mutations in TRIM37,

which encodes a peroxisomal protein that mono-ubiqui-

tinates histone H2A, a chromatin modification associated

with transcriptional repression (178). In contrast to a

promising short-term effect of GH treatment, the effect on

adult height is modest (5 cm) (179).

SHORT syndrome is caused by mutations in PIK3R1

(p85-alpha). In addition to regulating PI3K function,

p85-alpha and p85-beta regulate the function of XBP-1,

a transcription factor that orchestrates the unfolded protein

response following endoplasmic reticulum stress (180).

SOFT syndrome, caused by homozygous POC1A

mutations, is associated with severe pre- and post-natal

short stature, symmetric shortening of long bones,

triangular facies, sparse hair, and short, thickened distal

phalanges (181, 182).

Three-M syndrome is caused by defects in one of three

genes: CUL7 (encoding a ubiquitin ligase) (183), OBSL1

(encoding a cytoskeletal adaptor) (184) or CCDC8 (encoding

a protein possibly linked to CUL7 through the adaptor

protein OBSL1) (185, 186). The products of these genes play

a critical role in maintaining microtubule integrity with

defects leading to aberrant cell division (17, 187).

Microcephalic primordial dwarfism

Microcephalic primordial dwarfism is characterized

by severe pre- and post-natal growth retardation accom-

panied by microcephaly (18).

For Cornelia de Lange syndrome, five types have been

distinguished, and the same applies to Meier–Gorlin

www.eje-online.org

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 12: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Table 6 Examples of genetic defects in fundamental cellular processes.

Disordera Gene(s) Clinical features Inheritance References

Syndromes with (usually) normal head circumferenceCHARGE syndrome(214800)

CHD7, SEMA3E Choanal atresia, malformations of heart,inner ear and retina

AD (267)

Coffin–Siris syndrome(135900)

SMARCB1, SMARCA4,SMARCA2, ARID1A,ARID1B

Developmental delay, speech impairment,coarse facial features, hypertrichosis,hypoplastic fifth fingernails or toenails,agenesis of the corpus callosum

AD (172)

Floating–Harbor syndrome(136140)

SRCAP Delayed bone age and speech, triangular face,deep-set eyes, long eyelashes, bulbous nose,wide columella, short philtrum, thin lips

AD (174, 175)

KBG syndrome (148050) ANKRD11 Macrodontia of upper central incisors, distinc-tive craniofacial findings, skeletal anomalies,global developmental delay, seizures,intellectual disability

AD (176)

Mulibrey nanism (253250) TRIM37 Progressive cardiomyopathy, characteristic facialfeatures, failure of sexual maturation, insulinresistance with DM2, increased risk forWilms tumor

AR (178)

SHORT syndrome (269880) PIK3R1 hyperextensibility of joints, inguinal hernia,ocular depression, teething delay

AD (180)

Short stature, onycho-dysplasia, facial dys-morphism, hypotri-chosis(SOFT, 614813)

POC1A Severely short long bones, peculiar faciesassociated with paucity of hair, triangularfacies, nail anomalies, short, thickened distalphalanges. Relative macrocephaly inchildhood, microcephaly in adulthood

AR (181, 182)

Three-M syndrome 1(273750), 2 (612921),3 (614205)

CUL7, OBSL1, CCDC8 Facial features, normal mental development,long, slender tubular bones, reducedanteroposterior diameter of vertebralbodies, delayed bone age

AR (183, 184,185, 186)

Microcephalic primordial dwarfismCornelia de Langesyndrome 1–5

NIPBL, SMC1A, SMC3,RAD21, HDAC8

Low anterior hairline, arched eyebrows,synophrys, ante-verted nares, maxillaryprognathism, long philtrum, thin lips, ‘carp’mouth, upper limb anomalies.

AD (190)

Meier–Gorlin syndrome 1–5 ORC1, ORC4, ORC6,CDT1, CDC6

Bilateral microtia, and aplasia or hypoplasia ofthe patellae, normal intelligence

AR (192, 268)

MOPD I (210710) U4atac Neurologic abnormalities, including mentalretardation, brain malformations, ocular/auditory sensory deficits

AR (5, 193)

MOPD II (210720) PCNT Radiologic abnormalities, absent or mild mentalretardation in comparison to Seckelsyndrome, truncal obesity, diabetes,moyamoya, small loose teeth

AR (5, 194, 269)

Microcephaly and chorio-retinopathy, 1 (251270),2 (616171)

TUBGCP6, PLK4 Retinopathy. The gene encodes PLK4 kinase,a master regulator of centriole duplication.

AR (270)

Rett syndrome (312750) MECP2 Almost exclusively in females. Arresteddevelopment (6–18 months), loss of speech,stereotypic movements, microcephaly,seizures, mental retardation.

XLD (271)

Rubinstein–Taybi syndrome1 (180849), 2 (613684)

CREBBP, EP300 Mental retardation, broad thumbs and halluces,dysmorphic facial features

AD (272)

Seckel syndrome 1–8 ATR, RBBP8, CENPJ,CEP152, CEP63, NIN,DNA2, ATRIP

Mental retardation, characteristic ‘bird-headed’facial appearance

AR (5, 18, 195)

Short stature with micro-cephaly and distinctivefacies (615789)

CRIPT Frontal bossing, high forehead, sparse hair andeyebrows, telecanthus, proptosis, antevertednares, flat nasal bridge

AR (273)

AD, autosomal dominant; AR, autosomal recessive; DM2, diabetes mellitus type 2; MOPD, microcephalic osteodysplastic primordial dwarfism; IUGR,intrauterine growth retardation; XLR, X-linked recessive.aName (number) according to OMIM.

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R156

www.eje-online.org

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 13: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Table 7 Examples of genetic defects in fundamental cellular processes: DNA repair defects.

Disordera Gene(s) Clinical features Inheritance References

Bloom syndrome (210900) RECQL3 Sun-sensitive, telangiectatic, hypo- andhyperpigmented skin, predisposition tomalignancy,chromosomal instability

AR (274)

Cockayne syndrome A, B, XPG/CS(five types)

ERCC8, ERCC6, ERCC5,ERCC3, ERCC4

Cutaneous photosensitivity, thin, dry hair,progeroid appearance, pigmentary retino-pathy, sensorineural hearing loss, dentalcaries

AR (272)

Fanconi anemia (multiple types) FANCA and multiplegenes

Heterogeneous disorder causing genomicinstability, abnormalities in major organsystems, bone marrow failure, highpredisposition to cancer

AR (275, 276)

Hutchinson–Gilford progeriasyndrome (176670)

LMNA Low body weight, early loss of hair, lipo-dystrophy, scleroderma, decreased jointmobility, osteolysis, facial features thatresemble aged persons

AD (277)

Hypomorphic PCNA mutation PCNA Hearing loss, premature aging, telangiecta-sia, neurodegeneration, photosensitivityby nucleotide excision repair defect

AR (278)

Immunoosseous dysplasia,Schimke type (242900)

SMARCAL1 Spondyloepiphyseal dysplasia, numerouslentigines, slowly progressive immunedefect, immune-complex nephritis

AR (279)

Natural killer cell and gluco-corticoid deficiency with DNArepair defect (609981)

MCM4 Variant of familial glucocorticoid deficiency:hypocortisolemia, increased chromosomalbreakage, NK cell deficiency

AR (280, 281)

Nijmegen breakage syndrome(251260)

NBS1 Microcephaly, growth retardation, immuno-deficiency, predisposition to cancer

AR (282)

Ovarian dysgenesis 4 MCM9 Hypergonadotropic hypogonadism, genomicinstability

AR (283)

Rothmund–Thomson syndrome RECQL4 Skin atrophy, telangiectasia, hyper- andhypopigmentation, congenital skeletalabnormalities, premature aging

AR (284)

X-linked mental retardation-hypotonic facies syndrome(309580)

ATRX Mental retardation, dysmorphic facies,hypogonadism, deafness, renal anomalies,mild skeletal defects

XLR (285)

Defective nonhomologous end-joining (NHEJ) DNA damagerepair

LIG4, NHEJ1, ARTEMIS,DNA-PKCs, XRCC4,PRKDC

Radiosensitive, severe combined immuno-deficiency

AR (197, 198,199, 273,286, 287)

AD, autosomal dominant; AR, autosomal recessive; DM2, diabetes mellitus type 2; IUGR, intrauterine growth retardation; XLR, X-linked recessive; MOPD,Microcephalic osteodysplastic primordial dwarfism.aName (number) according to OMIM.

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R157

syndrome (188, 189, 190, 191, 192). Microcephalic osteo-

dysplastic primordial dwarfism (MOPD) type I is caused by

mutations in RNU4ARAC, encoding a small nuclear RNA

that is part of the minor spliceosome and necessary for

proper splicing of U12-dependent introns (193). Mutations

in the gene encoding pericentrin (PCNT) cause MOPD type

II (5, 194). Seckel syndrome is caused by mutations in many

different genes encoding proteins involved in DNA damage

response or centrosomal function (reviewed in (5, 18, 195)).

DNA repair defects

Many syndromes associated with abnormal DNA repair

present with short stature (Table 7). The best known

example is Bloom syndrome, caused by a mutation in the

gene encoding DNA helicase RecQ protein-like-3 (RECQL3).

Cells of these patients show an increased frequency of

chromosomal breaks, and the elevation in the rate of sister

chromatid exchanges is used as a diagnostic test. Other

syndromes include Cockayne syndrome, Fanconi anaemia,

and Rothmund–Thomson syndrome. Fanconi anaemia is a

clinically and genetically heterogeneous disorder that

causes genomic instability. Characteristic clinical features

include developmental abnormalities in major organ

systems, early-onset bone marrow failure, and a high

predisposition to cancer. The cellular hallmark is hypersen-

sitivity to DNA crosslinking agents and high frequency of

chromosomal aberrations.

An important pathway for the repair of DNA double-

stranded breaks is non-homologous end-joining (196),

www.eje-online.org

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 14: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R158

and mutations in several genes encoding proteins

involved in this process have been discovered in short

individuals, including LIG4 and XRCC4 mutations

(197, 198, 199). XRCC4 mutations are also associated

with hypergonadotrophic hypogonadism (199).

Chromosomal abnormalities, CNVs, andimprinting disorders associated with shortstature

Chromosomal abnormalities

Most guidelines on clinical workup of children with short

stature advise to perform routine karyotyping in females

with unexplained short stature, to detect Turner syndrome.

Indeed, it is very important to diagnose Turner syndrome,

given the comorbidities (partly potentially life-threaten-

ing) and efficacy of GH treatment. However, the diagnostic

yield in females with isolated short stature is low (estimated

at 4% (200)), so that several clinicians have doubted if this

would be cost-effective (201, 202, 203, 204). In fact, even in

the presence of clear guidelines for diagnostic studies in

short children, karyotyping was only performed in w50%

of cases in a Dutch study (205). Potentially useful criteria for

a cost-effective selection of short girls for this expensive test

may include a large distance between height SDS and target

height SDS (e.g., O2 S.D.) (206), delayed puberty and any

indication of physical stigmata. Deletions of the long arm

of the Y chromosome, or X/XY mosaicisms in phenotypic

females or males, are associated with short stature

(207, 208, 209, 210). However, in short males the

diagnostic yield of karyotyping is low (3%) (200).

Besides numerical aberrations of sex chromosomes,

several other chromosome abnormalities associated

with short stature are detectable with routine karyotyping,

e.g., Down syndrome (trisomy 21), Edwards syndrome

(trisomy 18), Patau syndrome (trisomy 13), and trisomy 17

mosaicism (211).

Copy number variants

As alluded to in the introduction, CNVs can be detected by

array-CGH (2) or SNP arrays (1). With these methods,

many new microdeletion and microduplication syn-

dromes have been identified, and several novel genes

associated with short stature as part of contiguous

gene syndromes have been discovered. Examples include

the observation that EPHA4 haploinsufficiency is

responsible for short stature observed in children with

Waardenburg syndrome caused by a chromosome

www.eje-online.org

2q35q36.2 deletion (212), and a possible role of dupli-

cations of EPAS and RHOQ on chromosome 2p21 in severe

short stature and delayed bone age (213). For Dubowitz

syndrome, a presumed autosomal recessive disorder

characterized by microcephaly, developmental delay,

growth failure, and a predisposition to allergies and

eczema, no unifying genetic alteration has been identified,

but in a subset of individuals diagnosed with this

syndrome deletions at 19q13 were found (214). Relatively

frequent contiguous gene deletion (and occasionally

duplication) syndromes are listed in Table 8.

Apart from these relatively well documented syn-

dromes, there may be many more. Four recent studies

(153, 215, 216, 217) showed that w10% of patients with ISS

carry a disease-causing CNV, and in short children

microdeletions (in contrast to microduplications) are

significantly more frequent than in controls (218). However,

for individual cases one often remains uncertain whether

their growth failure is due to the encountered CNV, and

which of the genes is responsible for it. Comparison with

previously reported patients and databases like DatabasE of

Chromosomal Imbalance and Phenotype in Humans using

Ensembl Resources (DECIPHER; http://decipher.sanger.uk)

and European Cytogeneticists Association Register of

Unbalanced Chromosome Aberrations (ECARUCA; http://

www.ecaruca.net) may give hints for candidate genes.

Imprinting disorders and uniparental disomy

Examples of imprinting disorders are shown in Table 9.

The best known example of a growth disorder associated

with an imprinting disorder is the Silver–Russell syn-

drome, which is most commonly caused by hypomethyla-

tion of an imprinting control region on the paternal allele

of chromosome 11p15.5, controlling the methylation of

the IGF2 and H19 genes (219). However, also multilocus

loss-of-methylation can occur (220, 221). Other genetic

causes include uniparental (maternal) disomy of chromo-

some 7 (UPD7) (79) and a mutation in the paternally

imprinted gene CDKN1C (222). CDKN1C mutations are

also associated with the IMAGe syndrome, characterized

by intrauterine growth restriction, metaphyseal dysplasia,

congenital adrenal hypoplasia, and genital anomalies

(223), and a syndrome of pre and postnatal growth failure

and early-onset diabetes mellitus (224). The clinical

spectrum of Silver–Russell syndrome is considerably

broader than thought before, and lack of intrauterine

growth restriction should not automatically result in

exclusion from molecular testing (225).

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 15: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Table 8 Examples of contiguous gene deletion or duplication syndromes associated with short stature.

Disordera Location Clinical features References

Recurrent rearrangements of 1q21.1 1q21.1del Intellectual disability, autism spectrum disorder,microcephaly, cardiac abnormalities, cataracts

(288)

2p16p22 microduplication syndrome 2p16p22dup Delayed bone age, facial dysmorphism. Role ofEPAS and RHOQ?

(213)

Wolf–Hirschhorn syndrome (194190) 4p16.3del ‘Greek warrior helmet’, epicanthal folds, shortphiltrum, downturned corners of mouth,micrognathia, seizures. Mitochondrial defect byLETM1 haploinsufficiency

(289, 290)

Chromosome 4q21 deletion syndrome(613509)

4q21del Neonatal muscular hypotonia, severe psychomotorretardation, severely delayed speech, broadforehead, frontal bossing, hypertelorism, shortphiltrum, downturned corners of mouth

(291)

Cri-du-chat syndrome (123450) 5p15.2ter del High-pitched catlike cry, microcephaly, round face,ocular hypertelorism, micrognathia, epicanthalfolds, low-set ears, hypotonia, severe psychomotorretardation. CTNND2?

(292)

Short stature, microce-phaly, speechdelay

5q35.2q35.3dup Microcephaly, speech delay. Reciprocal to commonSotos syndrome deletion (increased NSD1function?)

(293)

Williams–Beuren syndrome (194050) 7q11.23del Supravalvular aortic stenosis, intellectual disability,distinctive facial features

(294)

Trichorhinophalangeal syndrome, type II(Langer–Giedion syndrome) (150230)

8q21.11q24.13del Large, laterally protruding ears, bulbous nose,elongated upper lip, sparse scalp hair, wingedscapulae, multiple cartilaginous exostoses,redundant skin, intellectual disability. TRPS1, EXT1?

(295)

WAGR syndrome (194072) 11p13del Aniridia, hemihypertrophy, Wilms tumor,cryptorchidism. PAX6, WT1?

(296)

12q14 microdeletion syndrome 12q14del Developmental delay, osteopoikilosis. HMGA2? (297, 298)Chromosome 13q14 deletion syndrome

(613884)13q14del Retinoblastoma, mental impairment, high forehead,

prominent philtrum, anteverted earlobes(299)

Frias syndrome (609640) 14q22.1q22.3del Exophthalmia, palpebral ptosis, hypertelorism, shortsquare hands, small broad great toes. BMP4?

(300)

Distal 14q duplication syndrome 14q32.2-qter Mild developmental delay, high forehead, hyper-telorism, dysplastic ear helices, short philtrum, cupidbow upper lip, broad mouth, micrognathia

(230)

Smith–Magenis syndrome (182290) 17p11.2del Brachycephaly, midface hypoplasia, prognathism,hoarse voice, speech delay, hearing loss, psycho-motor retardation, behavioral problems. RAI1?Can be associated with GHD

(301)

Miller–Dieker lissencephaly syndrome(247200)

17p13.3del Lissencephaly, microcephaly, wrinkled skin overglabella and frontal suture, prominent occiput,narrow forehead, downward slanting palpebralfissures, small nose and chin, cardiac malformations,hypoplastic male external genitalia, seizures. CRK?

(302, 303)

17q21q25 duplication syndrome 17q2125dup Developmental delay, distal arthrogryposis.GH insensitivity, disturbed STAT5B, PI3K, andNF-kappaB signaling. Role of PRKCA mRNAoverexpression?

(66, 304)

Chromosome 18p deletion syndrome(146390)

18p11del Intellectual disability, round face, dysplastic ears, widemouth, abnormalities of teeth, limbs, genitalia,brain, eyes, heart

(305)

Chromosome 18q deletion syndrome(601808)

18q22.3q23del Congenital aural atresia, GHD, intellectual disability,reduced white-matter myelination, foot deformities

(306, 307)

Velocardiofacial syndrome (192430) 22q11.2del Highly variable phenotype. Central deletions: cardiacdisorders, learning delays, dysmorphic facialfeatures, hypernasal speech, velopalatalinsufficiency, hypocalcemia, hypoparathyroidism,psychiatric disorders; roleof TBX1? Distal: role of MAPK1?

(308, 309)

GHD, growth hormone deficiency; WAGR syndrome, Wilms tumor, Aniridia, genitourinary anomalies, and mental retardation syndrome.aName (number) according to OMIM.

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R159

www.eje-online.org

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 16: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Table 9 Examples of imprinting disorders.

Disordera Genetics Clinical features References

Silver–Russell syndrome (180860) Hypomethylation of imprintingcontrol region on paternalallele of 11p15.5 controlingmethylation of IGF2 and H19

Severe IUGR, triangular shapedface, broad forehead, bodyasymmetry, variety of minormalformations

(79, 219, 220, 229, 310)

Maternal UPD7 (SRS, 7p11.2)Silver–Russell syndrome or IMAGe

syndrome (614732) or IUGR Cearly-onset diabetes mellitus

Mutation in paternally imprintedgene CDKN1C

IUGR, metaphyseal dysplasia,adrenal hypoplasia congenita,genital anomalies; or onlySilver–Russell syndrome; orIUGR and early-adulthood-onset diabetes with normaladrenal function

(222, 224)

Prader–Willi syndrome (176270) Loss of expression of paternalcopies of imprinted genes(SNRPN, NDN), and others(15q11–q13) by deletion,maternal UPD, imprintingcenter defect, or Robertsoniantranslocation

Intellectual disability, seizures,poor gross and fine motorcoordination, behavioralproblems, sleep disturbances,high pain threshold

(226)

Pseudohypoparathyroidismtype 1a/c (103580)

Heterozygous GNAS1 (20q13.32)mutation inherited frommother

Resistance to parathyroidhormone and other hormones

(228)

Pseudohypoparathyroidismtype 1b (603233)

Both alleles have a paternal-specific imprinting pattern onboth parental alleles

Resistance to PTH is present with-out signs of Albright hereditaryosteodystrophy

Pseudopseudohypopara –thyroidism (612463)

Heterozygous GNAS1 mutationinherited from father

Albright hereditary osteodystro-phy without multiple hormoneresistance, brachydactyly

Temple syndrome (616222) Maternal UPD14 (14q32) Low birth weight, hypotonia,motor delay, feeding problemsearly in life, early puberty,reduced adult height, broadforehead, short nose with widenasal tip, small hands and feet

(153, 229)

IUGR, intrauterine growth retartdation.aName (number) according to OMIM.

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R160

Another well-known example is Prader–Willi syn-

drome, a contiguous gene syndrome. There are three

main genetic subtypes: a paternal chromosome 15q11q13

deletion (65–75% of cases), a maternal UPD of chromo-

some 15 (20–30% of cases), and an imprinting defect

(1–3%). It is now thought that deletion of the paternal

copies of the imprinted genes SNRPN, NDN, and possibly

others within the chromosome region 15q11q13, are

responsible for the phenotype (226). GH secretion can be

low and GH treatment has positive effects on linear

growth and body composition (227).

Loss-of-function mutations of GNAS, coding for the

a-subunit of the Gs protein, is associated with a spectrum

of growth disorders (228). The term pseudohypopara-

thyroidism indicates a group of heterogeneous disorders

whose common feature is represented by impaired

signalling of various hormones (primarily PTH) that

www.eje-online.org

activate cAMP-dependent pathways via Gsa protein. The

two main subtypes of PHP (types Ia and Ib) are caused by

molecular alterations within or upstream of the imprinted

GNAS gene, which encodes Gsa and other translated and

untranslated products. Patients who inherited a GNAS

mutation from their father develop Albright hereditary

osteodystrophy (AHO) without multiple hormone resist-

ance (pseudopseudohypoparathyroidism), characterized

by brachydactyly and short stature. In contrast, patients

who inherited the mutation from their mother, addition-

ally develop resistance to PTH and other hormones

(pseudohypoparathyroidism type 1a or 1c). This difference

is caused by the tissue-specific imprinting of GNAS.

In pseudohypoparathyroidism type 1b only resistance to

PTH is present without signs of AHO, due to an imprinting

defect of GNAS with silencing of the maternal allele,

affecting mainly the renal tubules.

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 17: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R161

Besides UPD7, there is another UPD syndrome that is

associated with short stature and various additional clinical

features: maternal UPD14 (Temple syndrome) (229). This

syndrome shares similarities with the distal 14q dupli-

cation phenotype (Table 8) (230). We showed that such

diagnoses can be found using SNP array technology in

children who had been considered ISS (153).

There may be many more epigenetic disorders

associated with short stature. In a study on 79 patients

with suspected Silver–Russell syndrome or unexplained

short stature/intrauterine growth restriction, 37% showed

a methylation abnormality in eleven imprinted loci. The

commonest finding was a loss of methylation at H19, and

a gain of methylation at IGF2R was significantly more

observed than in controls (231). Another example is the

epigenetic control of several parts of the IGF1 signalling

pathway. The IGF1 P2 promotor is an epigenetic quan-

titative trait locus (QTL), and methylation of a cluster of

six CGs located within the proximal part of this promoter

shows a strong negative association with serum IGF1 and

growth (232). In children with ISS CG-137 methylation in

this promoter contributed 30% to the variance of the IGF1

response to GH in an IGF1 generation test (233).

Diagnostic approach

In agreement with Dauber et al. (5), we believe that genetic

testing to identify rare monogenic causes of short stature is

important for various reasons: i) it can end the diagnostic

workup and the family’s uncertainty about the cause of

the condition; ii) it may alert the clinician to other

medical comorbidities; iii) it is invaluable for genetic

counselling; and iv) it may have implications for therapy

(e.g., some conditions, such as Bloom syndrome, are

contraindications for GH treatment (234)). With respect

to the question of who should undergo genetic testing, the

clinician should take several factors into consideration

that increase the likelihood of a monogenic cause of short

stature (5). The severity of the growth failure, presence of

additional abnormalities, presence of sibling or parent

with similar features, and consanguinity may be the most

important indicators.

The genetic evaluation of short stature is well

described in a recent review, which also presents a useful

diagnostic algorithm (5) in a step-wise fashion. If a

particular genetic aetiology or syndrome is suspected,

based on clinical features such as birth size, head

circumference, body proportions, and inheritance pattern,

a single gene-based test or gene panel is usually indicated.

We estimate that this applies to a limited number of

patients, since in the majority short stature is probably

of polygenic origin. If there is no strong suspicion on a

certain genetic diagnosis, or if initial testing showed no

abnormality – while a monogenic disorder appears very

likely – the clinician can either accept the diagnosis

‘apparent ISS’ or proceed on a hypothesis-free approach.

To arrive at this decision, various considerations apply,

including the availability of DNA from other family

members, informed consent, local infrastructure, and

financial aspects. It is noteworthy that presently limited

information is available about the sensitivity, specificity

and cost-effectiveness of this approach, while it is

important that the ethical aspects are properly dealt

with, for example appropriate informed consent forms

including information about handling incidental find-

ings. For details we refer to recently published guidelines

for diagnostic next-generation sequencing (235).

The hypothesis-free approach consists of two steps.

First, an array-cGH or SNP array is carried out, to search for

CNVs and uniparental disomies (with SNP-arrays) (1). Even

if no CNV is found, the results are useful for the analysis

of the second step, WES. For example, SNP arrays provide

information about homozygous regions, which can be

used in the bioinformatic analyses of the WES data,

particularly if a recessive condition is suspected. If a

potentially causative gene variant is found, it should be

confirmed by Sanger sequencing. After confirmation,

cosegregation studies in affected and non-affected relatives

should be performed, and if confirmatory, functional

studies are usually indicated to provide final proof.

However, we expect that in the coming years further

reduction of costs of next generation sequencing

technologies will render this step-wise approach super-

fluous, so that WES will be used as a tool to identify

small mutations as well as CNVs and homozygous

regions. The next step that the field will probably take

is WGS which, in combination with RNA sequencing of

the whole transcriptome and sequencing-based DNA

methylation analysis of the whole genome, will provide

additional information. It will probably lead to further

novel insights in the causes of short stature, if the ability

to interpret sequence variants outside the exome can be

improved.

Conclusion

In the past decade, many novel gene defects have been

found in association with multiple clinical disorders

associated with short stature, which has enormously

expanded the ability of clinicians to obtain a diagnosis

www.eje-online.org

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 18: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R162

in their patients. A more widespread use of currently

available genetic tools will certainly lead to a further

increase of clinical syndromes associated with genetic

aberrations. We agree with Lu et al. (236) and Dauber et al.

(5) that clinical use of sequencing data may reduce the cost

of care, result in more specific treatment guidelines and

avoidance of costly diagnostic and therapeutic procedures,

and reduce variance in diagnosis and treatment outcomes

between academic medical centres and community

hospitals and clinics.

Declaration of interest

J M Wit has served as consultant for Pfizer, Biopartners, OPKO, Versartis,

Teva, Merck-Serono, and Ammonett and has received speaker’s honoraria

from Pfizer, Versartis, Merck-Serono, Lilly and Sandoz. W Oostdijk received

unrestricted grant support from Novo Nordisk, Ipsen and Ferring. The other

authors have nothing to disclose.

Funding

This review did not receive any specific grant from any funding agency in

the public, commercial or not-for-profit sector.

Search Strategy

The search strategy started with updating information on genetic causes of

short stature described in previous reviews (10, 11, 13) and others (5, 14, 15,

16, 17), through OMIM and PubMed. Novel genetic causes were found with

the following search strategy (courtesy J. Schoones, Leiden):

(("body size"[Majr] OR Body Size[ti] OR "body height"[Majr] OR body

height[Ti] OR "body height"[ti] OR (growth[ti] NOT ("growth factor"[ti]

OR "growth factors"[ti]))) AND ("child"[MeSH Terms] OR child[Text Word]

OR children[Text Word] OR "infant"[MeSH Terms] OR infant[Text Word] OR

infants[Text Word] OR pediatric[tiab] OR paediatric[tiab]) NOT (obese

OR obesity OR obes* OR mice[tiab] OR animal[tiab] OR animals[tiab] OR

cattle[tiab] OR bovine[tiab] OR cows[tiab] OR pigs[tiab] OR birds[tiab]

OR fish[tiab] OR snakes[tiab] OR squirrels[tiab] OR cow[tiab] OR pig[tiab]

OR bird[tiab] OR fishes[tiab] OR snake[tiab] OR squirrel[tiab]) AND

english[la] AND ("genetics"[Subheading] OR "genetics"[tw] OR "genetics"

[mesh] OR "Genetic Techniques"[mesh])) AND ("2005/01/01"[PDAT]:

"3000/12/31"[PDAT]) NOT ("cell growth"[tw] OR "Cell Transformation,

Neoplastic"[mesh] OR "Cell Proliferation"[mesh] OR "Gene Expression

Regulation, Neoplastic"[mesh] OR "Cell Movement"[mesh]).

References

1 Gijsbers AC & Ruivenkamp CA. Molecular karyotyping: from

microscope to SNP arrays. Hormone Research in Pædiatrics 2011 76

208–213. (doi:10.1159/000330406)

2 Kharbanda M, Tolmie J & Joss S. How to use. microarray comparative

genomic hybridisation to investigate developmental disorders.

Archives of Disease in Childhood. Education and Practice Edition 2015 100

24–29. (doi:10.1136/archdischild-2014-306022)

3 Yang Y, Muzny DM, Reid JG, Bainbridge MN, Willis A, Ward PA,

Braxton A, Beuten J, Xia F, Niu Z et al. Clinical whole-exome

sequencing for the diagnosis of mendelian disorders.

www.eje-online.org

New England Journal of Medicine 2013 369 1502–1511. (doi:10.1056/

NEJMoa1306555)

4 Biesecker LG & Green RC. Diagnostic clinical genome and exome

sequencing. New England Journal of Medicine 2014 370 2418–2425.

(doi:10.1056/NEJMra1312543)

5 Dauber A, Rosenfeld RG & Hirschhorn JN. Genetic evaluation of short

stature. Journal of Clinical Endocrinology and Metabolism 2014 99 3080–

3092. (doi:10.1210/jc.2014-1506)

6 Guo MH, Shen Y, Walvoord EC, Miller TC, Moon JE, Hirschhorn JN &

Dauber A. Whole exome sequencing to identify genetic causes of short

stature. Hormone Research in Pædiatrics 2014 82 44–52. (doi:10.1159/

000360857)

7 Wood AR, Esko T, Yang J, Vedantam S, Pers TH, Gustafsson S, Chu AY,

Estrada K, Luan J, Kutalik Z et al. Defining the role of common

variation in the genomic and biological architecture of adult

human height. Nature Genetics 2014 46 1173–1186. (doi:10.1038/

ng.3097)

8 Lui JC, Nilsson O, Chan Y, Palmer CD, Andrade AC, Hirschhorn JN &

Baron J. Synthesizing genome-wide association studies and expression

microarray reveals novel genes that act in the human growth plate to

modulate height. Human Molecular Genetics 2012 21 5193–5201.

(doi:10.1093/hmg/dds347)

9 Lui JC, Nilsson O & Baron J. Recent research on the growth plate:

Recent insights into the regulation of the growth plate. Journal of

Molecular Endocrinology 2014 53 T1–T9. (doi:10.1530/JME-14-0022)

10 Kant SG, Wit JM & Breuning MH. Genetic analysis of short stature.

Hormone Research 2003 60 157–165. (doi:10.1159/000073226)

11 Walenkamp MJ & Wit JM. Genetic disorders in the growth hormone–

IGFI axis. Hormone Research 2006 66 221–230. (doi:10.1159/

000095161)

12 Wit JM, Ranke MB & Kelnar CJH. ESPE classification of paediatric

endocrine diagnoses. Hormone Research 2007 68 (Suppl 2) 1–120.

13 Wit JM, Kiess W & Mullis P. Genetic evaluation of short stature.

Best Practice & Research. Clinical Endocrinology & Metabolism 2011 25

1–17. (doi:10.1016/j.beem.2010.06.007)

14 Mortier GR, Graham JM Jr & Rimoin DL. Short stature syndromes.

In Growth Disorders, ch. 17, 2nd edn, pp 259–280. Eds CJH Kelnar,

MO Savage, P Saenger & CT Cowell. London, UK: Hodder Arnold, 2007.

15 Seaver LH & Irons M. ACMG practice guideline: genetic evaluation of

short stature. Genetics in Medicine 2009 11 465–470. (doi:10.1097/GIM.

0b013e3181a7e8f8)

16 Durand C & Rappold GA. Height matters-from monogenic disorders

to normal variation. Nature Reviews. Endocrinology 2013 9 171–177.

(doi:10.1038/nrendo.2012.251)

17 Baron J, Savendahl L, De Luca F, Dauber A, Phillip M, Wit JM &

Nilsson O. Short and tall stature: a new paradigm emerges.

Nature Reviews. Endocrinology 2015 11 735–746. (doi:10.1038/nrendo.

2015.165)

18 Klingseisen A & Jackson AP. Mechanisms and pathways of growth

failure in primordial dwarfism. Genes & development 2011 25

2011–2024. (doi:10.1101/gad.169037)

19 Clayton PE, Cianfarani S, Czernichow P, Johannsson G, Rapaport R &

Rogol A. Management of the child born small for gestational age

through to adulthood: a consensus statement of the International

Societies of Pediatric Endocrinology and the Growth Hormone

Research Society. Journal of Clinical Endocrinology and Metabolism 2007

92 804–810. (doi:10.1210/jc.2006-2017)

20 Rosenfeld RG. The molecular basis of idiopathic short stature.

Growth Hormone & IGF Research 2005 15 S3–S5. (doi:10.1016/j.ghir.

2005.06.014)

21 Rosenfeld RG & von Stein T. A database and website for molecular

defects of the GH–IGF axis: www.growthgenetics.com. Hormone

Research in Pædiatrics 2013 80 443–448. (doi:10.1159/000355543)

22 Alatzoglou KS & Dattani MT. Genetic forms of hypopituitarism and

their manifestation in the neonatal period. Early Human Development

2009 85 705–712. (doi:10.1016/j.earlhumdev.2009.08.057)

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 19: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R163

23 Pfaffle R & Klammt J. Pituitary transcription factors in the aetiology of

combined pituitary hormone deficiency. Best Practice & Research.

Clinical Endocrinology & Metabolism 2011 25 43–60. (doi:10.1016/j.

beem.2010.10.014)

24 McCabe MJ, Alatzoglou KS & Dattani MT. Septo-optic dysplasia and

other midline defects: the role of transcription factors: HESX1 and

beyond. Best Practice & Research. Clinical Endocrinology & Metabolism

2011 25 115–124. (doi:10.1016/j.beem.2010.06.008)

25 Sun Y, Bak B, Schoenmakers N, van Trotsenburg AS, Oostdijk W,

Voshol P, Cambridge E, White JK, Le Tissier P, Gharavy SN et al.

Loss-of-function mutations in IGSF1 cause an X-linked syndrome of

central hypothyroidism and testicular enlargement. Nature Genetics

2012 44 1375–1381. (doi:10.1038/ng.2453)

26 Joustra SD, Schoenmakers N, Persani L, Campi I, Bonomi M, Radetti G,

Beck-Peccoz P, Zhu H, Davis TM, Sun Y et al. The IGSF1 deficiency

syndrome: characteristics of male and female patients. Journal of

Clinical Endocrinology and Metabolism 2013 98 4942–4952.

(doi:10.1210/jc.2013-2743)

27 Aydin BK, Bas F, Tamay Z, Kilic G, Suleyman A, Bundak R, Saka N,

Ozkaya E, Guler N & Darendeliler F. Netherton syndrome associated

with growth hormone deficiency. Pediatric Dermatology 2014 31

90–94. (doi:10.1111/pde.12220)

28 Alatzoglou KS, Turton JP, Kelberman D, Clayton PE, Mehta A,

Buchanan C, Aylwin S, Crowne EC, Christesen HT, Hertel NT et al.

Expanding the spectrum of mutations in GH1 and GHRHR: genetic

screening in a large cohort of patients with congenital isolated growth

hormone deficiency. Journal of Clinical Endocrinology and Metabolism

2009 94 3191–3199. (doi:10.1210/jc.2008-2783)

29 Petkovic V, Godi M, Pandey AV, Lochmatter D, Buchanan CR,

Dattani MT, Eble A, Fluck CE & Mullis PE. Growth hormone (GH)

deficiency type II: a novel GH-1 gene mutation (GH-R178H) affecting

secretion and action. Journal of Clinical Endocrinology and Metabolism

2010 95 731–739. (doi:10.1210/jc.2009-1247)

30 Fritez N, Sobrier ML, Iraqi H, Vie-Luton MP, Netchine I, El Annas A,

Pantel J, Collot N, Rose S, Piterboth W et al. Molecular screening of a

large cohort of Moroccan patients with congenital hypopituitarism.

Clinical Endocrinology 2015 82 876–884. (doi:10.1111/cen.12706)

31 Duriez B, Duquesnoy P, Dastot F, Bougneres P, Amselem S &

Goossens M. An exon-skipping mutation in the btk gene of a patient

with X-linked agammaglobulinemia and isolated growth hormone

deficiency. FEBS Letters 1994 346 165–170. (doi:10.1016/0014-5793

(94)00457-9)

32 Burkitt Wright EM, Perveen R, Clayton PE, Hall CM, Costa T,

Procter AM, Giblin CA, Donnai D & Black GC. X-linked isolated

growth hormone deficiency: expanding the phenotypic spectrum of

SOX3 polyalanine tract expansions. Clinical Dysmorphology 2009 18

218–221. (doi:10.1097/MCD.0b013e32832d06f0)

33 Argente J, Flores R, Gutierrez-Arumi A, Verma B, Martos-Moreno GA,

Cusco I, Oghabian A, Chowen JA, Frilander MJ & Perez-Jurado LA.

Defective minor spliceosome mRNA processing results in isolated

familial growth hormone deficiency. EMBO Molecular Medicine 2014 6

299–306. (doi:10.1002/emmm.201303573)

34 Lucas-Herald AK, Kinning E, Iida A, Wang Z, Miyake N, Ikegawa S,

McNeilly J & Ahmed SF. A case of functional growth hormone

deficiency and early growth retardation in a child with IFT172

mutations. Journal of Clinical Endocrinology and Metabolism 2015 100

1221–1224. (doi:10.1210/jc.2014-3852)

35 Romano S, Maffei P, Bettini V, Milan G, Favaretto F, Gardiman M,

Marshall JD, Greggio NA, Pozzan GB, Collin GB et al. Alstrom

syndrome is associated with short stature and reduced GH reserve.

Clinical Endocrinology 2013 79 529–536. (doi:10.1111/cen.12180)

36 Stagi S, Lapi E, Pantaleo M, Carella M, Petracca A, De Crescenzo A,

Zelante L, Riccio A & de Martino M. A new case of de novo 6q24.2-q25.2

deletion on paternal chromosome 6 with growth hormone deficiency:

a twelve-year follow-up and literature review. BMC Medical Genetics

2015 16 69. (doi:10.1186/s12881-015-0212-z)

37 McMahon SK, Pretorius CJ, Ungerer JP, Salmon NJ, Conwell LS,

Pearen MA & Batch JA. Neonatal complete generalized glucocorticoid

resistance and growth hormone deficiency caused by a novel

homozygous mutation in Helix 12 of the ligand binding domain

of the glucocorticoid receptor gene (NR3C1). Journal of Clinical

Endocrinology and Metabolism 2010 95 297–302. (doi:10.1210/

jc.2009-1003)

38 Rocha V, Rocha D, Santos H & Marques JS. Growth hormone

deficiency in a patient with mitochondrial disease. Journal of Pediatric

Endocrinology & Metabolism 2015 28 1003–1004. (doi:10.1515/jpem-

2014-0315)

39 Wit JM, Oostdijk W & Losekoot M. Spectrum of insulin-like growth

factor deficiency. Endocrine Development 2012 23 30–41. (doi:10.1159/

000341739)

40 Pantel J, Legendre M, Nivot S, Morisset S, Vie-Luton MP, Le Bouc Y,

Epelbaum J & Amselem S. Recessive isolated growth hormone

deficiency and mutations in the ghrelin receptor. Journal of Clinical

Endocrinology and Metabolism 2009 94 4334–4341. (doi:10.1210/jc.

2009-1327)

41 Inoue H, Kangawa N, Kinouchi A, Sakamoto Y, Kimura C, Horikawa R,

Shigematsu Y, Itakura M, Ogata T & Fujieda K. Identification and

functional analysis of novel human growth hormone secretagogue

receptor (GHSR) gene mutations in Japanese subjects with short

stature. Journal of Clinical Endocrinology and Metabolism 2011 96

E373–E378. (doi:10.1210/jc.2010-1570)

42 Pugliese-Pires PN, Fortin JP, Arthur T, Latronico AC, Mendonca BB,

Villares SM, Arnhold IJ, Kopin AS & Jorge AA. Novel inactivating

mutations in the GH secretagogue receptor gene in patients with

constitutional delay of growth and puberty. European Journal of

Endocrinology/European Federation of Endocrine Societies 2011 165

233–241. (doi:10.1530/EJE-11-0168)

43 Laron Z, Pertzelan A & Mannheimer S. Genetic pituitary dwarfism

with high serum concentation of growth hormone – a new inborn

error of metabolism? Israel Journal of Medical Sciences 1966 2 152–155.

44 Amselem S, Duquesnoy P, Attree O, Novelli G, Bousnina S, Postel-

Vinay MC & Goossens M. Laron dwarfism and mutations of the

growth hormone-receptor gene. New England Journal of Medicine 1989

321 989–995. (doi:10.1056/NEJM198910123211501)

45 Godowski PJ, Leung DW, Meacham LR, Galgani JP, Hellmiss R, Keret R,

Rotwein PS, Parks JS, Laron Z & Wood WI. Characterization of the

human growth hormone receptor gene and demonstration of a partial

gene deletion in two patients with Laron-type dwarfism. PNAS 1989

86 8083–8087. (doi:10.1073/pnas.86.20.8083)

46 David A, Hwa V, Metherell LA, Netchine I, Camacho-Hubner C,

Clark AJ, Rosenfeld RG & Savage MO. Evidence for a continuum of

genetic, phenotypic, and biochemical abnormalities in children with

growth hormone insensitivity. Endocrine Reviews 2011 32 472–497.

(doi:10.1210/er.2010-0023)

47 Kurtoglu S & Hatipoglu N. Growth hormone insensitivity: diagnostic

and therapeutic approaches. Journal of Endocrinological Investigation

2015. In press.

48 Maamra M, Finidori J, Von Laue S, Simon S, Justice S, Webster J,

Dower S & Ross R. Studies with a growth hormone antagonist and

dual-fluorescent confocal microscopy demonstrate that the full-

length human growth hormone receptor, but not the truncated

isoform, is very rapidly internalized independent of Jak2-Stat5

signaling. Journal of Biological Chemistry 1999 274 14791–14798.

(doi:10.1074/jbc.274.21.14791)

49 Metherell LA, Akker SA, Munroe PB, Rose SJ, Caulfield M, Savage MO,

Chew SL & Clark AJ. Pseudoexon activation as a novel mechanism for

disease resulting in atypical growth-hormone insensitivity. American

Journal of Human Genetics 2001 69 641–646. (doi:10.1086/323266)

50 David A, Camacho-Hubner C, Bhangoo A, Rose SJ, Miraki-Moud F,

Akker SA, Butler GE, Ten S, Clayton PE, Clark AJ et al. An intronic

growth hormone receptor mutation causing activation of a

pseudoexon is associated with a broad spectrum of growth hormone

www.eje-online.org

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 20: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R164

insensitivity phenotypes. Journal of Clinical Endocrinology and

Metabolism 2007 92 655–659. (doi:10.1210/jc.2006-1527)

51 Ayling RM, Ross R, Towner P, Von Laue S, Finidori J, Moutoussamy S,

Buchanan CR, Clayton PE & Norman MR. A dominant-negative

mutation of the growth hormone receptor causes familial short stature

(letter). Nature Genetics 1997 16 13–14. (doi:10.1038/ng0597-13)

52 Iida K, Takahashi Y, Kaji H, Nose O, Okimura Y, Abe H & Chihara K.

Growth hormone (GH) insensitivity syndrome with high serum

GH-binding protein levels caused by a heterozygous splice site

mutation of the GH receptor gene producing a lack of intracellular

domain. Journal of Clinical Endocrinology and Metabolism 1998 83

531–537.

53 Aalbers AM, Chin D, Pratt KL, Little BM, Frank SJ, Hwa V &

Rosenfeld RG. Extreme elevation of serum growth hormone-binding

protein concentrations resulting from a novel heterozygous splice site

mutation of the growth hormone receptor gene. Hormone Research

2009 71 276–284. (doi:10.1159/000208801)

54 Kofoed EM, Hwa V, Little B, Woods KA, Buckway CK, Tsubaki J,

Pratt KL, Bezrodnik L, Jasper H, Tepper A et al. Growth hormone

insensitivity associated with a STAT5b mutation. New England

Journal of Medicine 2003 349 1139–1147. (doi:10.1056/

NEJMoa022926)

55 Hwa V, Nadeau K, Wit JM & Rosenfeld RG. STAT5b deficiency:

Lessons from STAT5b gene mutations. Best Practice & Research.

Clinical Endocrinology & Metabolism 2011 25 61–75. (doi:10.1016/j.

beem.2010.09.003)

56 Nadeau K, Hwa V & Rosenfeld RG. STAT5b deficiency: an unsuspected

cause of growth failure, immunodeficiency, and severe pulmonary

disease. Journal of Pediatrics 2011 158 701–708. (doi:10.1016/j.jpeds.

2010.12.042)

57 Scalco RC, Hwa V, Domene H, Jasper HG, Belgorosky A, Marino R,

Pereira AM, Tonelli C, Wit JM, Rosenfeld RG et al. STAT5B mutations

in heterozygous state have negative impact on height: another clue in

human stature heritability. European Journal of Endocrinology 2015 173

291–296. (doi:10.1530/EJE-15-0398)

58 Domene HM, Bengolea SV, Martinez AS, Ropelato MS, Pennisi P,

Scaglia P, Heinrich JJ & Jasper HG. Deficiency of the circulating

insulin-like growth factor system associated with inactivation of the

acid-labile subunit gene. New England Journal of Medicine 2004 350

570–577. (doi:10.1056/NEJMoa013100)

59 Domene HM, Hwa V, Jasper HG & Rosenfeld RG. Acid-labile subunit

(ALS) deficiency. Best Practice & Research. Clinical Endocrinology &

Metabolism 2011 25 101–113. (doi:10.1016/j.beem.2010.08.010)

60 van Duyvenvoorde HA, Kempers MJ, Twickler TB, van Doorn J,

Gerver WJ, Noordam C, Losekoot M, Karperien M, Wit JM &

Hermus AR. Homozygous and heterozygous expression of a novel

mutation of the acid-labile subunit. European Journal of

Endocrinology/European Federation of Endocrine Societies 2008 159

113–120. (doi:10.1530/EJE-08-0081)

61 Hess O, Khayat M, Hwa V, Heath KE, Teitler A, Hritan Y, Allon-Shalev S

& Tenenbaum-Rakover Y. A novel mutation in IGFALS, c.380TOC

(p.L127P), associated with short stature, delayed puberty, osteopenia

and hyperinsulinaemia in two siblings: insights into the roles of

insulin growth factor-1 (IGF1). Clinical Endocrinology 2013 79

838–844. (doi:10.1111/cen.12200)

62 Hogler W, Martin DD, Crabtree N, Nightingale P, Tomlinson J,

Metherell L, Rosenfeld R, Hwa V, Rose S, Walker J et al. IGFALS gene

dosage effects on serum IGF-I and glucose metabolism, body

composition, bone growth in length and width, and the pharmaco-

kinetics of recombinant human IGF-I administration. Journal of

Clinical Endocrinology and Metabolism 2014 99 E703–E712.

(doi:10.1210/jc.2013-3718)

63 Fofanova-Gambetti OV, Hwa V, Wit JM, Domene HM, Argente J,

Bang P, Hogler W, Kirsch S, Pihoker C, Chiu HK et al. Impact of

heterozygosity for acid-labile subunit (IGFALS) gene mutations on

stature: results from the international acid-labile subunit consortium.

www.eje-online.org

Journal of Clinical Endocrinology and Metabolism 2010 95 4184–4191.

(doi:10.1210/jc.2010-0489)

64 Domene HM, Scaglia PA, Martinez AS, Keselman AC, Karabatas LM,

Pipman VR, Bengolea SV, Guida MC, Ropelato MG, Ballerini MG

et al. Heterozygous IGFALS gene variants in idiopathic short stature

and normal children: impact on height and the IGF system.

Hormone Research in Pædiatrics 2013 80 413–423. (doi:10.1159/

000355412)

65 Wu S, Walenkamp MJ, Lankester A, Bidlingmaier M, Wit JM &

De Luca F. Growth hormone and insulin-like growth factor I

insensitivity of fibroblasts isolated from a patient with an IkBa

mutation. Journal of Clinical Endocrinology and Metabolism 2010 95

1220–1228. (doi:10.1210/jc.2009-1662)

66 Mul D, Wu S, de Paus RA, Oostdijk W, Lankester AC,

Duyvenvoorde HA, Ruivenkamp CA, Losekoot M, Tol MJ, De Luca LF

et al. A mosaic de novo duplication of 17q21-25 is associated with GH

insensitivity, disturbed in vitro CD28-mediated signaling, and

decreased STAT5B, PI3K, and NFkB activation. European Journal of

Endocrinology/European Federation of Endocrine Societies 2012 166

743–752. (doi:10.1530/EJE-11-0774)

67 Wit JM & De Luca F. Atypical defects resulting in growth hormone

insensitivity. Growth Hormone & IGF Research 2015. In press.

68 Flanagan SE, Haapaniemi E, Russell MA, Caswell R, Lango AH,

De Franco E, McDonald TJ, Rajala H, Ramelius A, Barton J et al.

Activating germline mutations in STAT3 cause early-onset multi-

organ autoimmune disease. Nature Genetics 2014 46 812–814.

(doi:10.1038/ng.3040)

69 Milner JD, Vogel TP, Forbes L, Ma CA, Stray-Pedersen A, Niemela JE,

Lyons JJ, Engelhardt KR, Zhang Y, Topcagic N et al. Early-onset

lymphoproliferation and autoimmunity caused by germline STAT3

gain-of-function mutations. Blood 2015 125 591–599. (doi:10.1182/

blood-2014-09-602763)

70 Woods KA, Camacho-Hubner C, Savage MO & Clark AJ. Intrauterine

growth retardation and postnatal growth failure associated with

deletion of the insulin-like growth factor I gene. New England

Journal of Medicine 1996 335 1363–1367. (doi:10.1056/

NEJM199610313351805)

71 Walenkamp MJ, Karperien M, Pereira AM, Hilhorst-Hofstee Y,

van Doorn J, Chen JW, Mohan S, Denley A, Forbes B,

van Duyvenvoorde HA et al. Homozygous and heterozygous

expression of a novel insulin-like growth factor-I mutation. Journal of

Clinical Endocrinology and Metabolism 2005 90 2855–2864.

(doi:10.1210/jc.2004-1254)

72 Netchine I, Azzi S, Houang M, Seurin D, Perin L, Ricort JM, Daubas C,

Legay C, Mester J, Herich R et al. Partial primary deficiency of insulin-

like growth factor (IGF)-I activity associated with IGF1 mutation

demonstrates its critical role in growth and brain development.

Journal of Clinical Endocrinology and Metabolism 2009 94 3913–3921.

(doi:10.1210/jc.2009-0452)

73 van Duyvenvoorde HA, van Setten PA, Walenkamp MJ, van Doorn J,

Koenig J, Gauguin L, Oostdijk W, Ruivenkamp CA, Losekoot M,

Wade JD et al. Short stature associated with a novel heterozygous

mutation in the insulin-like growth factor 1 gene. Journal of Clinical

Endocrinology and Metabolism 2010 95 E363–E367. (doi:10.1210/jc.

2010-0511)

74 Fuqua JS, Derr M, Rosenfeld RG & Hwa V. Identification of a novel

heterozygous IGF1 splicing mutation in a large kindred with familial

short stature. Hormone Research in Pædiatrics 2012 78 59–66.

(doi:10.1159/000337249)

75 Walenkamp MJ, Losekoot M & Wit JM. Molecular IGF-1 and IGF-1

receptor defects: from genetics to clinical management. Endocrine

Development 2013 24 128–137. (doi:10.1159/000342841)

76 Batey L, Moon JE, Yu Y, Wu B, Hirschhorn JN, Shen Y & Dauber A.

A novel deletion of IGF1 in a patient with idiopathic short stature

provides insight Into IGF1 haploinsufficiency. Journal of Clinical

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 21: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R165

Endocrinology and Metabolism 2014 99 E153–E159. (doi:10.1210/jc.

2013-3106)

77 Gicquel C, Rossignol S, Cabrol S, Houang M, Steunou V, Barbu V,

Danton F, Thibaud N, Le MM, Burglen L et al. Epimutation of the

telomeric imprinting center region on chromosome 11p15 in

Silver–Russell syndrome. Nature Genetics 2005 37 1003–1007.

(doi:10.1038/ng1629)

78 Binder G, Seidel AK, Weber K, Haase M, Wollmann HA, Ranke MB &

Eggermann T. IGF-II serum levels are normal in children with

Silver–Russell syndrome who frequently carry epimutations at the

IGF2 locus. Journal of Clinical Endocrinology and Metabolism 2006 91

4709–4712. (doi:10.1210/jc.2006-1127)

79 Binder G, Seidel AK, Martin DD, Schweizer R, Schwarze CP,

Wollmann HA, Eggermann T & Ranke MB. The endocrine phenotype

in Silver–Russell syndrome is defined by the underlying epigenetic

alteration. Journal of Clinical Endocrinology and Metabolism 2008 93

1402–1407. (doi:10.1210/jc.2007-1897)

80 Montenegro LR, Leal AC, Coutinho DC, Valassi HP, Nishi MY,

Arnhold IJ, Mendonca BB & Jorge AA. Post-receptor IGF1 insensi-

tivity restricted to the MAPK pathway in a Silver–Russell syndrome

patient with hypomethylation at the imprinting control region on

chromosome 11. European Journal of Endocrinology/European

Federation of Endocrine Societies 2012 166 543–550. (doi:10.1530/EJE-

11-0964)

81 Iliev DI, Kannenberg K, Weber K & Binder G. IGF-I sensitivity in

Silver–Russell syndrome with IGF2/H19 hypomethylation. Growth

Hormone & IGF Research 2014 24 187–191. (doi:10.1016/j.ghir.2014.

06.005)

82 Begemann M, Zirn B, Santen G, Wirthgen E, Soellner L, Buttel HM,

Schweizer R, van Workum W, Binder G & Eggermann T. Paternally

inherited IGF2 mutation and growth restriction. New England

Journal of Medicine 2015 373 349–356. (doi:10.1056/NEJMoa1415227)

83 Murphy R, Baptista J, Holly J, Umpleby AM, Ellard S, Harries LW,

Crolla J, Cundy T & Hattersley AT. Severe intrauterine growth

retardation and atypical diabetes associated with a translocation

breakpoint disrupting regulation of the insulin-like growth factor 2

gene. Journal of Clinical Endocrinology and Metabolism 2008 93

4373–4380. (doi:10.1210/jc.2008-0819)

84 Munoz-Calvo MT, Barrios V, Pozo J, Martos-Moreno GA, Hawkings

FG, Domene H, Jasper H, Yakar S, Conover CA, Kopchick.JE et al.

A new syndrome of short stature, mild microcephaly, skeletal

abnormalities and high circulating IGF1, IGFBP3 and ALS associated

with a homozygous mutation in the gene for pregnancy-associated

plasma protein A2 (PAPP-A2, pappalysin2). Endocrine Society Meeting

2015. Abstract.

85 Klammt J, Kiess W & Pfaffle R. IGF1R mutations as cause of SGA.

Best Practice & Research. Clinical Endocrinology & Metabolism 2011 25

191–206. (doi:10.1016/j.beem.2010.09.012)

86 Ester WA, van Duyvenvoorde HA, de Wit CC, Broekman AJ,

Ruivenkamp CA, Govaerts LC, Wit JM, Hokken-Koelega AC &

Losekoot M. Two short children born small for gestational age with

insulin-like growth factor 1 receptor haploinsufficiency illustrate the

heterogeneity of its phenotype. Journal of Clinical Endocrinology and

Metabolism 2009 94 4717–4727. (doi:10.1210/jc.2008-1502)

87 Abuzzahab MJ, Schneider A, Goddard A, Grigorescu F, Lautier C,

Keller E, Kiess W, Klammt J, Kratzsch J, Osgood D et al. IGF-I receptor

mutations resulting in intrauterine and postnatal growth retardation.

New England Journal of Medicine 2003 349 2211–2222. (doi:10.1056/

NEJMoa010107)

88 Fang P, Cho YH, Derr MA, Rosenfeld RG, Hwa V & Cowell CT. Severe

short stature caused by novel compound heterozygous mutations of

the insulin-like growth factor 1 receptor (IGF1R). Journal of Clinical

Endocrinology and Metabolism 2012 97 E243–E247. (doi:10.1210/jc.

2011-2142)

89 Gannage-Yared MH, Klammt J, Chouery E, Corbani S, Megarbane H,

Abou GJ, Choucair N, Pfaffle R & Megarbane A. Homozygous mutation

of the IGF1 receptor gene in a patient with severe pre- and postnatal

growth failure and congenital malformations. European Journal of

Endocrinology/European Federation of Endocrine Societies 2013 168

K1–K7. (doi:10.1530/EJE-12-0701)

90 Prontera P, Micale L, Verrotti A, Napolioni V, Stangoni G & Merla G.

A new homozygous IGF1R variant defines a clinically recognizable

incomplete dominant form of SHORT syndrome. Human Mutation

2015 36 1043–1047. (doi:10.1002/humu.22853)

91 Jung HJ & Suh Y. Regulation of IGF1 signaling by microRNAs.

Frontiers in Genetics 2014 5 472. (doi:10.3389/fgene.2014.00472)

92 Boersma B, Otten BJ, Stoelinga GB & Wit JM. Catch-up growth after

prolonged hypothyroidism. European Journal of Pediatrics 1996 155

362–367. (doi:10.1007/BF01955262)

93 Kuhnen P, Turan S, Frohler S, Guran T, Abali S, Biebermann H,

Bereket A, Gruters A, Chen W & Krude H. Identification of

PENDRIN (SLC26A4) mutations in patients with congenital

hypothyroidism and "apparent" thyroid dysgenesis. Journal of

Clinical Endocrinology and Metabolism 2014 99 E169–E176. (doi:10.

1210/jc.2013-2619)

94 Phillips SA, Rotman-Pikielny P, Lazar J, Ando S, Hauser P, Skarulis MC,

Brucker-Davis F & Yen PM. Extreme thyroid hormone resistance in a

patient with a novel truncated TR mutant. Journal of Clinical

Endocrinology and Metabolism 2001 86 5142–5147. (doi:10.1210/jcem.

86.11.8051)

95 Schoenmakers N, Moran C, Peeters RP, Visser T, Gurnell M &

Chatterjee K. Resistance to thyroid hormone mediated by defective

thyroid hormone receptor a. Biochimica et Biophysica Acta 2013 1830

4004–4008. (doi:10.1016/j.bbagen.2013.03.018)

96 van Mullem AA, Visser TJ & Peeters RP. Clinical consequences of

mutations in thyroid hormone receptor-a1. European Thyroid Journal

2014 3 17–24. (doi:10.1159/000360637)

97 Hamajima T, Mushimoto Y, Kobayashi H, Saito Y & Onigata K. Novel

compound heterozygous mutations in the SBP2 gene: characteristic

clinical manifestations and the implications of GH and triiodo-

thyronine in longitudinal bone growth and maturation. European

Journal of Endocrinology/European Federation of Endocrine Societies 2012

166 757–764. (doi:10.1530/EJE-11-0812)

98 Reincke M, Sbiera S, Hayakawa A, Theodoropoulou M, Osswald A,

Beuschlein F, Meitinger T, Mizuno-Yamasaki E, Kawaguchi K, Saeki Y

et al. Mutations in the deubiquitinase gene USP8 cause Cushing’s

disease. Nature Genetics 2015 47 31–38. (doi:10.1038/ng.3166)

99 Beuschlein F, Fassnacht M, Assie G, Calebiro D, Stratakis CA,

Osswald A, Ronchi CL, Wieland T, Sbiera S, Faucz FR et al. Constitutive

activation of PKA catalytic subunit in adrenal Cushing’s syndrome.

New England Journal of Medicine 2014 370 1019–1028. (doi:10.1056/

NEJMoa1310359)

100 Semple RK, Savage DB, Cochran EK, Gorden P & O’Rahilly S.

Genetic syndromes of severe insulin resistance. Endocrine Reviews 2011

32 498–514. (doi:10.1210/er.2010-0020)

101 Bonafe L, Cormier-Daire V, Hall C, Lachman R, Mortier G, Mundlos S,

Nishimura G, Sangiorgi L, Savarirayan R, Sillence D et al. Nosology and

classification of genetic skeletal disorders: 2015 revision. American

Journal of Medical Genetics. Part A 2015 167 2869–2892. (doi:10.1002/

ajmg.a.37365)

102 Kant SG, Grote F, de Ru MH, Oostdijk W, Zonderland HM,

Breuning MH & Wit JM. Radiographic evaluation of children with

growth disorders. Hormone Research 2007 68 310–315. (doi:10.1159/

000108399)

103 Veeramani AK, Higgins P, Butler S, Donaldson M, Dougan E,

Duncan R, Murday V & Ahmed SF. Diagnostic use of skeletal survey in

suspected skeletal dysplasia. Journal of Clinical Research in Pediatric

Endocrinology 2009 1 270–274. (doi:10.4274/jcrpe.v1i6.270)

104 Alanay Y & Lachman RS. A review of the principles of radiological

assessment of skeletal dysplasias. Journal of Clinical Research in Pediatric

Endocrinology 2011 3 163–178. (doi:10.4274/jcrpe.463)

www.eje-online.org

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 22: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R166

105 Grunwald T & De Luca F. Role of Fibroblast Growth Factor 21 (FGF21)

in the regulation of statural growth. Current Pediatric Reviews 2015 11

98–105. (doi:10.2174/1573396311666150702105152)

106 Foldynova-Trantirkova S, Wilcox WR & Krejci P. Sixteen years and

counting: the current understanding of fibroblast growth factor

receptor 3 (FGFR3) signaling in skeletal dysplasias. Human Mutation

2012 33 29–41. (doi:10.1002/humu.21636)

107 Krejci P. The paradox of FGFR3 signaling in skeletal dysplasia:

why chondrocytes growth arrest while other cells over proliferate.

Mutation Research. Reviews in Mutation Research 2014 759 40–48.

(doi:10.1016/j.mrrev.2013.11.001)

108 Song SH, Balce GC, Agashe MV, Lee H, Hong SJ, Park YE, Kim SG &

Song HR. New proposed clinico-radiologic and molecular criteria in

hypochondroplasia: FGFR 3 gene mutations are not the only cause of

hypochondroplasia. American Journal of Medical Genetics. Part A 2012

158A 2456–2462. (doi:10.1002/ajmg.a.35564)

109 Kant SG, Cervenkova I, Balek L, Trantirek L, Santen GW, de Vries MC,

van Duyvenvoorde HA, van der Wielen MJ, Verkerk AJ,

Uitterlinden AG et al. A novel variant of FGFR3 causes proportionate

short stature. European Journal of Endocrinology/European Federation of

Endocrine Societies 2015 172 763–770. (doi:10.1530/EJE-14-0945)

110 Oishi I, Suzuki H, Onishi N, Takada R, Kani S, Ohkawara B, Koshida I,

Suzuki K, Yamada G, Schwabe GC et al. The receptor tyrosine kinase

Ror2 is involved in non-canonical Wnt5a/JNK signalling pathway.

Genes to Cells: Devoted to Molecular & Cellular Mechanisms 2003 8

645–654. (doi:10.1046/j.1365-2443.2003.00662.x)

111 Cerpa W, Latorre-Esteves E & Barria A. RoR2 functions as a

noncanonical Wnt receptor that regulates NMDAR-mediated synaptic

transmission. PNAS 2015 112 4797–4802. (doi:10.1073/pnas.

1417053112)

112 Roifman M, Marcelis CL, Paton T, Marshall C, Silver R, Lohr JL,

Yntema HG, Venselaar H, Kayserili H, van Bon B et al. De novo WNT5A-

associated autosomal dominant Robinow syndrome suggests speci-

ficity of genotype and phenotype. Clinical Genetics 2015 87 34–41.

(doi:10.1111/cge.12401)

113 Habib R, Amin-ud-din M & Ahmad W. A nonsense mutation in the

gene ROR2 underlying autosomal dominant brachydactyly type B.

Clinical Dysmorphology 2013 22 47–50. (doi:10.1097/MCD.

0b013e32835c6c8c)

114 van der Eerden BC, Karperien M, Gevers EF, Lowik CW & Wit JM.

Expression of Indian hedgehog, parathyroid hormone-related protein,

and their receptors in the postnatal growth plate of the rat: evidence

for a locally acting growth restraining feedback loop after birth.

Journal of Bone and Mineral Research 2000 15 1045–1055. (doi:10.1359/

jbmr.2000.15.6.1045)

115 Kronenberg HM. Developmental regulation of the growth plate.

Nature 2003 423 332–336. (doi:10.1038/nature01657)

116 Klopocki E, Hennig BP, Dathe K, Koll R, de Ravel T, Baten E, Blom E,

Gillerot Y, Weigel JF, Kruger G et al. Deletion and point mutations of

PTHLH cause brachydactyly type E. American Journal of Human Genetics

2010 86 434–439. (doi:10.1016/j.ajhg.2010.01.023)

117 Hoogendam J, Farih-Sips H, Wynaendts LC, Lowik CW, Wit JM &

Karperien M. Novel mutations in the parathyroid hormone

(PTH)/PTH-related peptide receptor type 1 causing Blomstrand

osteochondrodysplasia types I and II. Journal of Clinical

Endocrinology and Metabolism 2007 92 1088–1095. (doi:10.1210/jc.

2006-0300)

118 Schipani E, Kruse K & Juppner H. A constitutively active mutant

PTH-PTHrP receptor in Jansen-type metaphyseal chondrodysplasia.

Science 1995 268 98–100. (doi:10.1126/science.7701349)

119 Byrnes AM, Racacho L, Grimsey A, Hudgins L, Kwan AC, Sangalli M,

Kidd A, Yaron Y, Lau YL, Nikkel SM et al. Brachydactyly A-1 mutations

restricted to the central region of the N-terminal active fragment of

Indian Hedgehog. European Journal of Human Genetics 2009 17

1112–1120. (doi:10.1038/ejhg.2009.18)

www.eje-online.org

120 Klopocki E, Lohan S, Brancati F, Koll R, Brehm A, Seemann P, Dathe K,

Stricker S, Hecht J, Bosse K et al. Copy-number variations involving the

IHH locus are associated with syndactyly and craniosynostosis.

American Journal of Human Genetics 2011 88 70–75. (doi:10.1016/j.

ajhg.2010.11.006)

121 Linglart A, Menguy C, Couvineau A, Auzan C, Gunes Y, Cancel M,

Motte E, Pinto G, Chanson P, Bougneres P et al. Recurrent PRKAR1A

mutation in acrodysostosis with hormone resistance. New England

Journal of Medicine 2011 364 2218–2226. (doi:10.1056/

NEJMoa1012717)

122 Lindstrand A, Grigelioniene G, Nilsson D, Pettersson M,

Hofmeister W, Anderlid BM, Kant SG, Ruivenkamp CA, Gustavsson P,

Valta H et al. Different mutations in PDE4D associated with

developmental disorders with mirror phenotypes. Journal of Medical

Genetics 2014 51 45–54. (doi:10.1136/jmedgenet-2013-101937)

123 Estrada K, Krawczak M, Schreiber S, van Duijn K, Stolk L, van Meurs JB,

Liu F, Penninx BW, Smit JH, Vogelzangs N et al. A genome-wide

association study of northwestern Europeans involves the C-type

natriuretic peptide signaling pathway in the etiology of human

height variation. Human Molecular Genetics 2009 18 3516–3524.

(doi:10.1093/hmg/ddp296)

124 Bartels CF, Bukulmez H, Padayatti P, Rhee DK, Ravenswaaij-Arts C,

Pauli RM, Mundlos S, Chitayat D, Shih LY, Al Gazali LI et al. Mutations in

the transmembrane natriuretic peptide receptor NPR-B impair skeletal

growth and cause acromesomelic dysplasia, type Maroteaux. American

Journal of Human Genetics 2004 75 27–34. (doi:10.1086/422013)

125 Olney RC, Bukulmez H, Bartels CF, Prickett TC, Espiner EA, Potter LR

& Warman ML. Heterozygous mutations in natriuretic peptide

receptor-B (NPR2) are associated with short stature. Journal of Clinical

Endocrinology and Metabolism 2006 91 1229–1232. (doi:10.1210/jc.

2005-1949)

126 Vasques GA, Amano N, Docko AJ, Funari MF, Quedas EP, Nishi MY,

Arnhold IJ, Hasegawa T & Jorge AA. Heterozygous mutations in

natriuretic peptide receptor-B (NPR2) gene as a cause of short stature

in patients initially classified as idiopathic short stature. Journal of

Clinical Endocrinology and Metabolism 2013 98 E1636–E1644.

(doi:10.1210/jc.2013-2142)

127 Amano N, Mukai T, Ito Y, Narumi S, Tanaka T, Yokoya S, Ogata T &

Hasegawa T. Identification and functional characterization of two

novel NPR2 mutations in Japanese patients with short stature.

Journal of Clinical Endocrinology and Metabolism 2014 99 E713–E718.

(doi:10.1210/jc.2013-3525)

128 Vasques GA, Arnhold IJ & Jorge AA. Role of the natriuretic peptide

system in normal growth and growth disorders. Hormone Research in

Pædiatrics 2014 82 222–229. (doi:10.1159/000365049)

129 Wang SR, Jacobsen CM, Carmichael H, Edmund AB, Robinson JW,

Olney RC, Miller TC, Moon JE, Mericq V, Potter LR et al. Heterozygous

mutations in natriuretic peptide receptor-B (NPR2) gene as a cause of

short stature. Human Mutation 2015 36 474–481. (doi:10.1002/humu.

22773)

130 Hisado-Oliva A, Garre-Vazquez AI, Santaolalla-Caballero F,

Belinchon A, Barreda-Bonis AC, Vasques GA, Ramirez J, Luzuriaga C,

Carlone G, Gonzalez-Casado I et al. Heterozygous NPR2 mutations

cause disproportionate short stature, similar to Leri-Weill dyschon-

drosteosis. Journal of Clinical Endocrinology and Metabolism 2015 100

E1133–E1142. (doi:10.1210/jc.2015-1612)

131 Teixeira CC, Agoston H & Beier F. Nitric oxide, C-type natriuretic

peptide and cGMP as regulators of endochondral ossification.

Developmental Biology 2008 319 171–178. (doi:10.1016/j.ydbio.2008.

04.031)

132 Krejci P, Masri B, Fontaine V, Mekikian PB, Weis M, Prats H &

Wilcox WR. Interaction of fibroblast growth factor and C-natriuretic

peptide signaling in regulation of chondrocyte proliferation and

extracellular matrix homeostasis. Journal of Cell Science 2005 118

5089–5100. (doi:10.1242/jcs.02618)

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 23: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R167

133 Lorget F, Kaci N, Peng J, Benoist-Lasselin C, Mugniery E, Oppeneer T,

Wendt DJ, Bell SM, Bullens S, Bunting S et al. Evaluation of the

therapeutic potential of a CNP analog in a Fgfr3 mouse model

recapitulating achondroplasia. American Journal of Human Genetics

2012 91 1108–1114. (doi:10.1016/j.ajhg.2012.10.014)

134 Tompson SW, Merriman B, Funari VA, Fresquet M, Lachman RS,

Rimoin DL, Nelson SF, Briggs MD, Cohn DH & Krakow D. A recessive

skeletal dysplasia, SEMD aggrecan type, results from a missense

mutation affecting the C-type lectin domain of aggrecan. American

Journal of Human Genetics 2009 84 72–79. (doi:10.1016/j.ajhg.2008.

12.001)

135 Nilsson O, Guo MH, Dunbar N, Popovic J, Flynn D, Jacobsen C, Lui JC,

Hirschhorn JN, Baron J & Dauber A. Short stature, accelerated bone

maturation, and early growth cessation due to heterozygous aggrecan

mutations. Journal of Clinical Endocrinology and Metabolism 2014 99

E1510–E1518. (doi:10.1210/jc.2014-1332)

136 Ahmad M, Faiyaz Ul Haque M, Ahmad W, Abbas H, Haque S,

Krakow D, Rimoin DL, Lachman RS & Cohn DH. Distinct, autosomal

recessive form of spondyloepimetaphyseal dysplasia segregating in an

inbred Pakistani kindred. American Journal of Medical Genetics 1998 78

468–473. (doi:10.1002/(SICI)1096-8628(19980806)78:5!468::AID-

AJMG13O3.0.CO;2-D)

137 Oostdijk W, Idkowiak J, Mueller JW, House PJ, Taylor AE,

O’Reilly MW, Hughes BA, de Vries MC, Kant SG, Santen GW et al.

PAPSS2 deficiency causes androgen excess via impaired DHEA

sulfation – in vitro and in vivo studies in a family harboring two novel

PAPSS2 mutations. Journal of Clinical Endocrinology and Metabolism

2015 100 E672–E680. (doi:10.1210/jc.2014-3556)

138 Malaquias AC, Scalco RC, Fontenele EG, Costalonga EF, Baldin AD,

Braz AF, Funari MF, Nishi MY, Guerra-Junior G, Mendonca BB et al.

The sitting height/height ratio for age in healthy and short individuals

and its potential role in selecting short children for SHOX analysis.

Hormone Research in Pædiatrics 2013 80 449–456. (doi:10.1159/

000355411)

139 Rappold GA, Fukami M, Niesler B, Schiller S, Zumkeller W,

Bettendorf M, Heinrich U, Vlachopapadoupoulou E, Reinehr T,

Onigata K et al. Deletions of the homeobox gene SHOX (short stature

homeobox) are an important cause of growth failure in children with

short stature. Journal of Clinical Endocrinology and Metabolism 2002 87

1402–1406. (doi:10.1210/jcem.87.3.8328)

140 Binder G, Ranke MB & Martin DD. Auxology is a valuable instrument

for the clinical diagnosis of SHOX haploinsufficiency in school-age

children with unexplained short stature. Journal of Clinical

Endocrinology and Metabolism 2003 88 4891–4896. (doi:10.1210/jc.

2003-030136)

141 Jorge AA & Arnhold IJ. Anthropometric evaluation of children with

SHOX mutations can be used as indication for genetic studies in

children of short stature. Journal of Medical Genetics 2007 44 e90.

142 Jorge AA, Souza SC, Nishi MY, Billerbeck AE, Liborio DC, Kim CA,

Arnhold IJ & Mendonca BB. SHOX mutations in idiopathic short

stature and Leri-Weill dyschondrosteosis: frequency and phenotypic

variability. Clinical Endocrinology 2007 66 130–135.

143 Binder G. Short stature due to SHOX deficiency: genotype,

phenotype, and therapy. Hormone Research in Pædiatrics 2011 75

81–89. (doi:10.1159/000324105)

144 Chen J, Wildhardt G, Zhong Z, Roth R, Weiss B, Steinberger D,

Decker J, Blum WF & Rappold G. Enhancer deletions of the SHOX

gene as a frequent cause of short stature: the essential role of a 250 kb

downstream regulatory domain. Journal of Medical Genetics 2009 46

834–839. (doi:10.1136/jmg.2009.067785)

145 Huber C, Rosilio M, Munnich A & Cormier-Daire V. High incidence of

SHOX anomalies in individuals with short stature. Journal of Medical

Genetics 2006 43 735–739. (doi:10.1136/jmg.2006.040998)

146 Benito-Sanz S, Barroso E, Heine-Suner D, Hisado-Oliva A, Romanelli V,

Rosell J, Aragones A, Caimari M, Argente J, Ross JL et al. Clinical and

molecular evaluation of SHOX/PAR1 duplications in Leri-Weill

dyschondrosteosis (LWD) and idiopathic short stature (ISS). Journal of

Clinical Endocrinology and Metabolism 2011 96 E404–E412.

(doi:10.1210/jc.2010-1689)

147 Benito-Sanz S, Aza-Carmona M, Rodriguez-Estevez A, Rica-

Etxebarria I, Gracia R, Campos-Barros A & Heath KE. Identification of

the first PAR1 deletion encompassing upstream SHOX enhancers in a

family with idiopathic short stature. European Journal of Human

Genetics 2012 20 125–127. (doi:10.1038/ejhg.2011.210)

148 Rosilio M, Huber-Lequesne C, Sapin H, Carel JC, Blum WF &

Cormier-Daire V. Genotypes and phenotypes of children with SHOX

deficiency in France. Journal of Clinical Endocrinology and Metabolism

2012 97 E1257–E1265. (doi:10.1210/jc.2011-3460)

149 Kant SG, Broekman SJ, de Wit CC, Bos M, Scheltinga SA, Bakker E,

Oostdijk W, van der Kamp HJ, van Zwet EW, van der Hout AH et al.

Phenotypic characterization of patients with deletions in the

3 0-flanking SHOX region. PeerJ 2013 1 e35. (doi:10.7717/peerj.35)

150 Donze SH, Meijer CR, Kant SG, Zandwijken GR, van der Hout AH,

van Spaendonk RM, van den Ouweland AM, Wit JM, Losekoot M &

Oostdijk W. The growth response to growth hormone treatment is

greater in patients with SHOX enhancer deletions compared to SHOX

defects. European Journal of Endocrinology 2015 173 611–621.

(doi:10.1530/EJE-15-0451)

151 Iughetti L, Capone L, Elsedfy H, Bertorelli R, Predieri B, Bruzzi P,

Forabosco A & El Kholy M. Unexpected phenotype in a boy with

trisomy of the SHOX gene. Journal of Pediatric Endocrinology &

Metabolism 2010 23 159–169. (doi:10.1515/JPEM.2010.23.1-2.159)

152 Caliebe J, Broekman S, Boogaard M, Bosch CA, Ruivenkamp CA,

Oostdijk W, Kant SG, Binder G, Ranke MB, Wit JM et al. IGF1, IGF1R

and SHOX mutation analysis in short children born small for

gestational age and short children with normal birth size (idiopathic

short stature). Hormone Research in Pædiatrics 2012 77 250–260.

(doi:10.1159/000338341)

153 Wit JM, van Duyvenvoorde HA, van Klinken JB, Caliebe J, Bosch CA,

Lui JC, Gijsbers AC, Bakker E, Breuning MH, Oostdijk W et al.

Copy number variants in short children born small for gestational age.

Hormone Research in Pædiatrics 2014 82 310–318. (doi:10.1159/

000367712)

154 Yasoda A, Komatsu Y, Chusho H, Miyazawa T, Ozasa A, Miura M,

Kurihara T, Rogi T, Tanaka S, Suda M et al. Overexpression of CNP in

chondrocytes rescues achondroplasia through a MAPK-dependent

pathway. Nature Medicine 2004 10 80–86. (doi:10.1038/nm971)

155 Cseh B, Doma E & Baccarini M. "RAF" neighborhood: protein-protein

interaction in the Raf/Mek/Erk pathway. FEBS Letters 2014 588

2398–2406. (doi:10.1016/j.febslet.2014.06.025)

156 Lee BH, Kim JM, Jin HY, Kim GH, Choi JH & Yoo HW. Spectrum of

mutations in Noonan syndrome and their correlation with pheno-

types. Journal of Pediatrics 2011 159 1029–1035. (doi:10.1016/j.jpeds.

2011.05.024)

157 Roberts AE, Allanson JE, Tartaglia M & Gelb BD. Noonan syndrome.

Lancet 2013 381 333–342. (doi:10.1016/S0140-6736(12)61023-X)

158 Wang SR, Carmichael H, Andrew SF, Miller TC, Moon JE, Derr MA,

Hwa V, Hirschhorn JN & Dauber A. Large-scale pooled next-

generation sequencing of 1077 genes to identify genetic causes of

short stature. Journal of Clinical Endocrinology and Metabolism 2013 98

E1428–E1437. (doi:10.1210/jc.2013-1534)

159 De Rocca Serra-Nedelec A, Edouard T, Treguer K, Tajan M, Araki T,

Dance M, Mus M, Montagner A, Tauber M, Salles JP et al. Noonan

syndrome-causing SHP2 mutants inhibit insulin-like growth factor 1

release via growth hormone-induced ERK hyperactivation, which

contributes to short stature. PNAS 2012 109 4257–4262. (doi:10.1073/

pnas.1119803109)

160 Edouard T, Combier JP, Nedelec A, Bel-Vialar S, Metrich M, Conte-

Auriol F, Lyonnet S, Parfait B, Tauber M, Salles JP et al. Functional

effects of PTPN11 (SHP2) mutations causing LEOPARD syndrome on

epidermal growth factor-induced phosphoinositide

www.eje-online.org

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 24: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R168

3-kinase/AKT/glycogen synthase kinase 3b signaling. Molecular and

Cellular Biology 2010 30 2498–2507. (doi:10.1128/MCB.00646-09)

161 Pasteris NG, Cadle A, Logie LJ, Porteous ME, Schwartz CE,

Stevenson RE, Glover TW, Wilroy RS & Gorski JL. Isolation and

characterization of the faciogenital dysplasia (Aarskog–Scott

syndrome) gene: a putative Rho/Rac guanine nucleotide exchange

factor. Cell 1994 79 669–678. (doi:10.1016/0092-8674(94)90552-5)

162 Orrico A, Galli L, Faivre L, Clayton-Smith J, Azzarello-Burri SM,

Hertz JM, Jacquemont S, Taurisano R, Arroyo Carrera I, Tarantino E

et al. Aarskog–Scott syndrome: clinical update and report of nine

novel mutations of the FGD1 gene. American Journal of Medical

Genetics. Part A 2010 152A 313–318. (doi:10.1002/ajmg.a.33199)

163 Zou W, Greenblatt MB, Shim JH, Kant S, Zhai B, Lotinun S, Brady N,

Hu DZ, Gygi SP, Baron R et al. MLK3 regulates bone development

downstream of the faciogenital dysplasia protein FGD1 in mice.

Journal of Clinical Investigation 2011 121 4383–4392. (doi:10.1172/

JCI59041)

164 Genot E, Daubon T, Sorrentino V & Buccione R. FGD1 as a central

regulator of extracellular matrix remodelling – lessons from

faciogenital dysplasia. Journal of Cell Science 2012 125 3265–3270.

(doi:10.1242/jcs.093419)

165 Akiyama H & Lefebvre V. Unraveling the transcriptional regulatory

machinery in chondrogenesis. Journal of Bone and Mineral Metabolism

2011 29 390–395. (doi:10.1007/s00774-011-0273-9)

166 Coman D, Irving M, Kannu P, Jaeken J & Savarirayan R. The skeletal

manifestations of the congenital disorders of glycosylation. Clinical

Genetics 2008 73 507–515. (doi:10.1111/j.1399-0004.2008.01015.x)

167 Morava E, Zeevaert R, Korsch E, Huijben K, Wopereis S, Matthijs G,

Keymolen K, Lefeber DJ, De Meirleir L & Wevers RA. A common

mutation in the COG7 gene with a consistent phenotype including

microcephaly, adducted thumbs, growth retardation, VSD and

episodes of hyperthermia. European Journal of Human Genetics 2007 15

638–645. (doi:10.1038/sj.ejhg.5201813)

168 Scott K, Gadomski T, Kozicz T & Morava E. Congenital disorders of

glycosylation: new defects and still counting. Journal of Inherited

Metabolic Disease 2014 37 609–617. (doi:10.1007/s10545-014-9720-9)

169 Vissers LE, van Ravenswaaij CM, Admiraal R, Hurst JA, de Vries BB,

Janssen IM, van der Vliet WA, Huys EH, de Jong PJ, Hamel BC et al.

Mutations in a new member of the chromodomain gene family cause

CHARGE syndrome. Nature Genetics 2004 36 955–957. (doi:10.1038/

ng1407)

170 Martin DM, Sheldon S & Gorski JL. CHARGE association with choanal

atresia and inner ear hypoplasia in a child with a de novo chromosome

translocation t(2;7)(p14;q21.11). American Journal of Medical Genetics

2001 99 115–119. (doi:10.1002/1096-8628(2000)9999:999!00::AID-

AJMG1126O3.0.CO;2-8)

171 Zentner GE, Hurd EA, Schnetz MP, Handoko L, Wang C, Wang Z,

Wei C, Tesar PJ, Hatzoglou M, Martin DM et al. CHD7 functions in the

nucleolus as a positive regulator of ribosomal RNA biogenesis. Human

Molecular Genetics 2010 19 3491–3501. (doi:10.1093/hmg/ddq265)

172 Santen GW, Aten E, Sun Y, Almomani R, Gilissen C, Nielsen M,

Kant SG, Snoeck IN, Peeters EA, Hilhorst-Hofstee Y et al. Mutations

in SWI/SNF chromatin remodeling complex gene ARID1B cause

Coffin–Siris syndrome. Nature Genetics 2012 44 379–380.

(doi:10.1038/ng.2217)

173 Tsurusaki Y, Okamoto N, Ohashi H, Kosho T, Imai Y, Hibi-Ko Y,

Kaname T, Naritomi K, Kawame H, Wakui K et al. Mutations

affecting components of the SWI/SNF complex cause

Coffin–Siris syndrome. Nature Genetics 2012 44 376–378. (doi:10.

1038/ng.2219)

174 Hood RL, Lines MA, Nikkel SM, Schwartzentruber J, Beaulieu C,

Nowaczyk MJ, Allanson J, Kim CA, Wieczorek D, Moilanen JS et al.

Mutations in SRCAP, encoding SNF2-related CREBBP activator

protein, cause Floating–Harbor syndrome. American Journal of Human

Genetics 2012 90 308–313. (doi:10.1016/j.ajhg.2011.12.001)

www.eje-online.org

175 Nikkel SM, Dauber A, de Munnik S, Connolly M, Hood RL,

Caluseriu O, Hurst J, Kini U, Nowaczyk MJ, Afenjar A et al. The

phenotype of Floating–Harbor syndrome: clinical characterization of

52 individuals with mutations in exon 34 of SRCAP. Orphanet

Journal of Rare Diseases 2013 8 63. (doi:10.1186/1750-1172-8-63)

176 Sirmaci A, Spiliopoulos M, Brancati F, Powell E, Duman D, Abrams A,

Bademci G, Agolini E, Guo S, Konuk B et al. Mutations in ANKRD11

cause KBG syndrome, characterized by intellectual disability, skeletal

malformations, and macrodontia. American Journal of Human Genetics

2011 89 289–294. (doi:10.1016/j.ajhg.2011.06.007)

177 Zhang A, Yeung PL, Li CW, Tsai SC, Dinh GK, Wu X, Li H & Chen JD.

Identification of a novel family of ankyrin repeats containing

cofactors for p160 nuclear receptor coactivators. Journal of Biological

Chemistry 2004 279 33799–33805. (doi:10.1074/jbc.M403997200)

178 Bhatnagar S, Gazin C, Chamberlain L, Ou J, Zhu X, Tushir JS,

Virbasius CM, Lin L, Zhu LJ, Wajapeyee N et al. TRIM37 is a new

histone H2A ubiquitin ligase and breast cancer oncoprotein. Nature

2014 516 116–120. (doi:10.1038/nature13955)

179 Karlberg N, Jalanko H & Lipsanen-Nyman M. Growth and growth

hormone therapy in subjects with mulibrey nanism. Pediatrics 2007

120 e102–e111. (doi:10.1542/peds.2006-2686)

180 Park SW, Zhou Y, Lee J, Lu A, Sun C, Chung J, Ueki K & Ozcan U. The

regulatory subunits of PI3K, p85a and p85b, interact with XBP-1 and

increase its nuclear translocation. Nature Medicine 2010 16 429–437.

(doi:10.1038/nm.2099)

181 Sarig O, Nahum S, Rapaport D, Ishida-Yamamoto A, Fuchs-Telem D,

Qiaoli L, Cohen-Katsenelson K, Spiegel R, Nousbeck J, Israeli S et al.

Short stature, onychodysplasia, facial dysmorphism, and

hypotrichosis syndrome is caused by a POC1A mutation. American

Journal of Human Genetics 2012 91 337–342. (doi:10.1016/j.ajhg.2012.

06.003)

182 Barraza-Garcia J, Ivan Rivera-Pedroza C, Salamanca L, Belinchon A,

Lopez-Gonzalez V, Sentchordi-Montane L, Del PA, Santos-Simarro F,

Campos-Barros A, Lapunzina P et al. Two novel POC1A mutations in

the Primordial dwarfism, SOFT syndrome: clinical homogeneity but

also unreported malformations. American Journal of Medical Genetics

2016 170 210–216. (doi:10.1002/ajmg.a.37393)

183 Huber C, Dias-Santagata D, Glaser A, O’Sullivan J, Brauner R, Wu K,

Xu X, Pearce K, Wang R, Uzielli ML et al. Identification of mutations

in CUL7 in 3-M syndrome. Nature Genetics 2005 37 1119–1124.

(doi:10.1038/ng1628)

184 Hanson D, Murray PG, Sud A, Temtamy SA, Aglan M, Superti-Furga A,

Holder SE, Urquhart J, Hilton E, Manson FD et al. The primordial

growth disorder 3-M syndrome connects ubiquitination to the

cytoskeletal adaptor OBSL1. American Journal of Human Genetics 2009

84 801–806. (doi:10.1016/j.ajhg.2009.04.021)

185 Hanson D, Murray PG, O’Sullivan J, Urquhart J, Daly S, Bhaskar SS,

Biesecker LG, Skae M, Smith C, Cole T et al. Exome sequencing

identifies CCDC8 mutations in 3-M syndrome, suggesting that

CCDC8 contributes in a pathway with CUL7 and OBSL1 to control

human growth. American Journal of Human Genetics 2011 89 148–153.

(doi:10.1016/j.ajhg.2011.05.028)

186 Clayton PE, Hanson D, Magee L, Murray PG, Saunders E, Abu-

Amero SN, Moore GE & Black GC. Exploring the spectrum of 3-M

syndrome, a primordial short stature disorder of disrupted

ubiquitination. Clinical Endocrinology 2012 77 335–342. (doi:10.1111/

j.1365-2265.2012.04428.x)

187 Yan J, Yan F, Li Z, Sinnott B, Cappell KM, Yu Y, Mo J, Duncan JA,

Chen X, Cormier-Daire V et al. The 3M complex maintains

microtubule and genome integrity. Molecular Cell 2014 54 791–804.

(doi:10.1016/j.molcel.2014.03.047)

188 Bicknell LS, Bongers EM, Leitch A, Brown S, Schoots J, Harley ME,

Aftimos S, Al-Aama JY, Bober M, Brown PA et al. Mutations in the

pre-replication complex cause Meier–Gorlin syndrome. Nature

Genetics 2011 43 356–359. (doi:10.1038/ng.775)

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 25: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R169

189 Guernsey DL, Matsuoka M, Jiang H, Evans S, Macgillivray C,

Nightingale M, Perry S, Ferguson M, LeBlanc M, Paquette J et al.

Mutations in origin recognition complex gene ORC4 cause

Meier–Gorlin syndrome. Nature Genetics 2011 43 360–364.

(doi:10.1038/ng.777)

190 Boyle MI, Jespersgaard C, Brondum-Nielsen K, Bisgaard AM &

Tumer Z. Cornelia de Lange syndrome. Clinical Genetics 2015 88 1–12.

(doi:10.1111/cge.12499)

191 de Munnik SA, Otten BJ, Schoots J, Bicknell LS, Aftimos S, Al-Aama JY,

van Bever Y, Bober MB, Borm GF, Clayton-Smith J et al. Meier–Gorlin

syndrome: growth and secondary sexual development of a micro-

cephalic primordial dwarfism disorder. American Journal of Medical

Genetics. Part A 2012 158A 2733–2742. (doi:10.1002/ajmg.a.35681)

192 Barbelanne M & Tsang WY. Molecular and cellular basis of autosomal

recessive primary microcephaly. BioMed Research International 2014

2014 547986. (doi:10.1155/2014/547986)

193 He H, Liyanarachchi S, Akagi K, Nagy R, Li J, Dietrich RC, Li W,

Sebastian N, Wen B, Xin B et al. Mutations in U4atac snRNA,

a component of the minor spliceosome, in the developmental disorder

MOPD I. Science 2011 332 238–240. (doi:10.1126/science.1200587)

194 Rauch A, Thiel CT, Schindler D, Wick U, Crow YJ, Ekici AB,

van Essen AJ, Goecke TO, Al-Gazali L, Chrzanowska KH et al.

Mutations in the pericentrin (PCNT) gene cause primordial dwarfism.

Science 2008 319 816–819. (doi:10.1126/science.1151174)

195 Ogi T, Walker S, Stiff T, Hobson E, Limsirichaikul S, Carpenter G,

Prescott K, Suri M, Byrd PJ, Matsuse M et al. Identification of the first

ATRIP-deficient patient and novel mutations in ATR define a clinical

spectrum for ATR-ATRIP Seckel Syndrome. PLoS Genetics 2012 8

e1002945. (doi:10.1371/journal.pgen.1002945)

196 Davis AJ & Chen DJ. DNA double strand break repair via non-

homologous end-joining. Translational Cancer Research 2013 2

130–143.

197 Murray JE, Bicknell LS, Yigit G, Duker AL, van Kogelenberg M,

Haghayegh S, Wieczorek D, Kayserili H, Albert MH, Wise CA et al.

Extreme growth failure is a common presentation of ligase IV deficiency.

Human Mutation 2014 35 76–85. (doi:10.1002/humu.22461)

198 Murray JE, van der Burg M, IJspeert H, Carroll P, Wu Q, Ochi T,

Leitch A, Miller ES, Kysela B, Jawad A et al. Mutations in the NHEJ

component XRCC4 cause primordial dwarfism. American Journal of

Human Genetics 2015 96 412–424. (doi:10.1016/j.ajhg.2015.01.013)

199 de Bruin C, Mericq V, Andrew SF, van Duyvenvoorde HA, Verkaik NS,

Losekoot M, Porollo A, Garcia H, Kuang Y, Hanson D et al. An XRCC4

splice mutation associated with severe short stature, gonadal failure,

and early-onset metabolic syndrome. Journal of Clinical Endocrinology

and Metabolism 2015 100 E789–E798. (doi:10.1210/jc.2015-1098)

200 Moreno-Garcia M, Fernandez-Martinez FJ & Barreiro ME. Chromo-

somal anomalies in patients with short stature. Pediatrics International

2005 47 546–549. (doi:10.1111/j.1442-200x.2005.02120.x)

201 Eggert P, Pankau R & Oldigs HD. How necessary is a chromosomal

analysis in growth-retarded girls? Clinical Genetics 1990 37 351–354.

(doi:10.1111/j.1399-0004.1990.tb03518.x)

202 Savendahl L & Davenport ML. Delayed diagnoses of Turner’s

syndrome: proposed guidelines for change. Journal of Pediatrics 2000

137 455–459. (doi:10.1067/mpd.2000.107390)

203 Partsch CJ, Raffenberg U & Sippell WG. Screening for Turner’s syndrome

by chromosome analysisof all girls with short stature. Journal of Pediatrics

2002 140 140–141. (doi:10.1067/mpd.2002.119172)

204 Massa GG, Verlinde V & Heinrichs C. Screening for Turner’s syndrome

by chromosome analysis of all girls with short stature. Journal of

Pediatrics 2002 140 141–142. (doi:10.1067/mpd.2002.119173)

205 Grote FK, Oostdijk W, De Muinck Keizer-Schrama SM, Van

Dommelen P, Van Buuren S, Dekker FW, Ketel AG, Moll HA & Wit JM.

The diagnostic work up of growth failure in secondary health care;

an evaluation of consensus guidelines. BMC Pediatrics 2008 8 21.

(doi:10.1186/1471-2431-8-21)

206 Grote FK, Van Dommelen P, Oostdijk W, De Muinck Keizer-

Schrama SM, Verkerk PH, Wit JM & Van Buuren S. Developing

evidence-based guidelines for referral for short stature. Archives of

Disease in Childhood 2008 93 212–217. (doi:10.1136/adc.2007.120188)

207 Kirsch S, Weiss B, Schon K & Rappold GA. The definition of the Y

chromosome growth-control gene (GCY) critical region: relevance of

terminal and interstitial deletions. Journal of Pediatric Endocrinology &

Metabolism 2002 15 (Suppl 5) 1295–1300.

208 Richter-Unruh A, Knauer-Fischer S, Kaspers S, Albrecht B,

Gillessen-Kaesbach G & Hauffa BP. Short stature in children with an

apparently normal male phenotype can be caused by 45,X/46,XY

mosaicism and is susceptible to growth hormone treatment.

European Journal of Pediatrics 2004 163 251–256. (doi:10.1007/s00431-

004-1406-0)

209 Tosson H, Rose SR & Gartner LA. Children with 45,X/46,XY karyotype

from birth to adult height. Hormone Research in Pædiatrics 2010 74

190–200. (doi:10.1159/000281468)

210 Lindhardt JM, Hagen CP, Rajpert-De ME, Kjaergaard S, Petersen BL,

Skakkebaek NE, Main KM & Juul A. 45,X/46,XY mosaicism:

phenotypic characteristics, growth, and reproductive function –

a retrospective longitudinal study. Journal of Clinical Endocrinology

and Metabolism 2012 97 E1540–E1549. (doi:10.1210/jc.2012-1388)

211 Utermann B, Riegel M, Leistritz D, Karall T, Wisser J, Meisner L,

Fauth C, Baldinger R, Johnson J, Erdel M et al. Pre- and postnatal

findings in trisomy 17 mosaicism. American Journal of Medical Genetics.

Part A 2006 140 1628–1636. (doi:10.1002/ajmg.a.31319)

212 Li C, Chen R, Fan X, Luo J, Qian J, Wang J, Xie B, Shen Y & Chen S.

EPHA4 haploinsufficiency is responsible for the short stature of a

patient with 2q35-q36.2 deletion and Waardenburg syndrome.

BMC Medical Genetics 2015 16 23. (doi:10.1186/s12881-015-0165-2)

213 Blassnig-Ezeh A, Bandelier C, Fruhmesser A, Revencu N, Krabichler B,

Beauloye V, Ravoet M, Fauth C, Zschocke J, Simma B et al. Severe

growth retardation, delayed bone age, and facial dysmorphism in two

patients with microduplications in 2p16/p22. American Journal of

Medical Genetics. Part A 2013 161A 3176–3181. (doi:10.1002/ajmg.a.

36176)

214 Urquhart JE, Williams SG, Bhaskar SS, Bowers N, Clayton-Smith J &

Newman WG. Deletion of 19q13 reveals clinical overlap with

Dubowitz syndrome. Journal of Human Genetics 2015 60 781–785.

(doi:10.1038/jhg.2015.111)

215 Zahnleiter D, Uebe S, Ekici AB, Hoyer J, Wiesener A, Wieczorek D,

Kunstmann E, Reis A, Doerr HG, Rauch A et al. Rare copy number

variants are a common cause of short stature. PLoS Genetics 2013 9

e1003365. (doi:10.1371/journal.pgen.1003365)

216 van Duyvenvoorde HA, Lui JC, Kant SG, Oostdijk W, Gijsbers AC,

Hoffer MJ, Karperien M, Walenkamp MJ, Noordam C, Voorhoeve PG

et al. Copy number variants in patients with short stature. European

Journal of Human Genetics 2014 22 602–609. (doi:10.1038/ejhg.2013.203)

217 Canton AP, Costa SS, Rodrigues TC, Bertola DR, Malaquias AC,

Correa FA, Arnhold IJ, Rosenberg C & Jorge AA. Genome-wide

screening of copy number variants in children born small for

gestational age reveals several candidate genes involved in growth

pathways. European Journal of Endocrinology/European Federation of

Endocrine Societies 2014 171 253–262. (doi:10.1530/EJE-14-0232)

218 Dauber A, Yu Y, Turchin MC, Chiang CW, Meng YA, Demerath EW,

Patel SR, Rich SS, Rotter JI, Schreiner PJ et al. Genome-wide association of

copy-number variation reveals an association between short stature and

the presence of low-frequency genomic deletions. American Journal of

Human Genetics 2011 89 751–759. (doi:10.1016/j.ajhg.2011.10.014)

219 Netchine I, Rossignol S, Dufourg MN, Azzi S, Rousseau A, Perin L,

Houang M, Steunou V, Esteva B, Thibaud N et al. 11p15 imprinting

center region 1 loss of methylation is a common and specific cause of

typical Russell–Silver syndrome: clinical scoring system and epi-

genetic-phenotypic correlations. Journal of Clinical Endocrinology and

Metabolism 2007 92 3148–3154. (doi:10.1210/jc.2007-0354)

www.eje-online.org

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 26: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R170

220 Azzi S, Rossignol S, Steunou V, Sas T, Thibaud N, Danton F, Le Jule M,

Heinrichs C, Cabrol S, Gicquel C et al. Multilocus methylation analysis

in a large cohort of 11p15-related foetal growth disorders (Russell

Silver and Beckwith Wiedemann syndromes) reveals simultaneous

loss of methylation at paternal and maternal imprinted loci. Human

Molecular Genetics 2009 18 4724–4733. (doi:10.1093/hmg/ddp435)

221 Wakeling EL. Silver–Russell syndrome. Archives of Disease in Childhood

2011 96 1156–1161. (doi:10.1136/adc.2010.190165)

222 Brioude F, Oliver-Petit I, Blaise A, Praz F, Rossignol S, Le Jule M,

Thibaud N, Faussat AM, Tauber M, Le Bouc Y et al. CDKN1C mutation

affecting the PCNA-binding domain as a cause of familial Russell–

Silver syndrome. Journal of Medical Genetics 2013 50 823–830.

(doi:10.1136/jmedgenet-2013-101691)

223 Arboleda VA, Lee H, Parnaik R, Fleming A, Banerjee A, Ferraz-de-Souza B,

Delot EC, Rodriguez-Fernandez IA, Braslavsky D, Bergada I et al.

Mutations in the PCNA-binding domain of CDKN1C cause IMAGe

syndrome. Nature Genetics 2012 44 788–792. (doi:10.1038/ng.2275)

224 Kerns SL, Guevara-Aguirre J, Andrew S, Geng J, Guevara C, Guevara-

Aguirre M, Guo M, Oddoux C, Shen Y, Zurita A et al. A novel variant in

CDKN1C is associated with intrauterine growth restriction, short

stature, and early-adulthood-onset diabetes. Journal of Clinical

Endocrinology and Metabolism 2014 99 E2117–E2122. (doi:10.1210/jc.

2014-1949)

225 Eggermann T, Gonzalez D, Spengler S, Arslan-Kirchner M, Binder G &

Schonherr N. Broad clinical spectrum in Silver–Russell syndrome and

consequences for genetic testing in growth retardation. Pediatrics 2009

123 e929–e931. (doi:10.1542/peds.2008-3228)

226 Angulo MA, Butler MG & Cataletto ME. Prader–Willi syndrome:

a review of clinical, genetic, and endocrine findings. Journal of

Endocrinological Investigation 2015 38 1249–1263. (doi:10.1007/

s40618-015-0312-9)

227 Bakker NE, Kuppens RJ, Siemensma EP, Tummers-de Lind van

Wijngaarden RF, Festen DA, Bindels-de Heus GC, Bocca G, Haring DA,

Hoorweg-Nijman JJ, Houdijk EC et al. Eight years of growth hormone

treatment in children with Prader–Willi syndrome: maintaining the

positive effects. Journal of Clinical Endocrinology and Metabolism 2013

98 4013–4022. (doi:10.1210/jc.2013-2012)

228 Turan S & Bastepe M. GNAS spectrum of disorders. Current Osteoporosis

Reports 2015 13 146–158. (doi:10.1007/s11914-015-0268-x)

229 Hoffmann K & Heller R. Uniparental disomies 7 and 14. Best Practice &

Research. Clinical Endocrinology & Metabolism 2011 25 77–100.

(doi:10.1016/j.beem.2010.09.004)

230 Thiel CT, Dorr HG, Trautmann U, Hoyer J, Hofmann K, Kraus C,

Ekici AB, Reis A & Rauch A. A de novo 7.6Mb tandem duplication of

14q32.2-qter associated with primordial short stature with neuro-

secretory growth hormone dysfunction, distinct facial anomalies and

mild developmental delay. European Journal of Medical Genetics 2008

51 362–367. (doi:10.1016/j.ejmg.2008.03.001)

231 Turner CL, Mackay DM, Callaway JL, Docherty LE, Poole RL, Bullman H,

Lever M, Castle BM, Kivuva EC, Turnpenny PD et al. Methylation

analysis of 79 patients with growth restriction reveals novel patterns of

methylation change at imprinted loci. European Journal of Human

Genetics 2010 18 648–655. (doi:10.1038/ejhg.2009.246)

232 Ouni M, Gunes Y, Belot MP, Castell AL, Fradin D & Bougneres P.

The IGF1 P2 promoter is an epigenetic QTL for circulating IGF1 and

human growth. Clinical Epigenetics 2015 7 22. (doi:10.1186/s13148-

015-0062-8)

233 Ouni M, Castell AL, Linglart A & Bougneres P. Genetic and epigenetic

modulation of growth hormone sensitivity studied with the IGF1

generation test. Journal of Clinical Endocrinology and Metabolism 2015

100 E919–E925. (doi:10.1210/jc.2015-1413)

234 Renes JS, Willemsen RH, Wagner A, Finken MJ & Hokken-Koelega AC.

Bloom syndrome in short children born small for gestational age: a

challenging diagnosis. Journal of Clinical Endocrinology and Metabolism

2013 98 3932–3938. (doi:10.1210/jc.2013-2491)

www.eje-online.org

235 Matthijs G, Souche E, Alders M, Corveleyn A, Eck S, Feenstra I, Race V,

Sistermans E, Sturm M, Weiss M et al. Guidelines for diagnostic next-

generation sequencing. European Journal of Human Genetics 2016 24

2–5. (doi:10.1038/ejhg.2015.226)

236 Lu JT, Campeau PM & Lee BH. Genotype–phenotype correlation –

promiscuity in the era of next-generation sequencing. New

England Journal of Medicine 2014 371 593–596. (doi:10.1056/

NEJMp1400788)

237 Zielonka M, Makhseed N, Blau N, Bettendorf M, Hoffmann GF &

Opladen T. Dopamine-responsive growth-hormone deficiency and

central hypothyroidism in sepiapterin reductase deficiency. In: JIMD

Reports vol 24 pp 109–113. Eds: J Zschocke, M Baumgartner, EMorava,

M Patterson, S Rahman and V Peters Springer Verlag: Heidelberg

2015. (doi:10.1007/8904_2015_450)

238 Correa FA, Trarbach EB, Tusset C, Latronico AC, Montenegro LR,

Carvalho LR, Franca MM, Otto AP, Costalonga EF, Brito VN et al.

FGFR1 and PROKR2 rare variants found in patients with combined

pituitary hormone deficiencies. Endocrine Connections 2015 4 100–107.

(doi:10.1530/EC-15-0015)

239 Gorbenko del BD, de Graaff LC, Posthouwer D, Visser TJ & Hokken-

Koelega AC. Isolated GH deficiency: mutation screening and copy

number analysis of HMGA2 and CDK6 genes. European Journal of

Endocrinology/European Federation of Endocrine Societies 2011 165

537–544. (doi:10.1530/EJE-11-0478)

240 Alatzoglou KS, Webb EA, Le Tissier P & Dattani MT. Isolated growth

hormone deficiency (GHD) in childhood and adolescence:

recent advances. Endocrine Reviews 2014 35 376–432. (doi:10.1210/er.

2013-1067)

241 Karaca E, Buyukkaya R, Pehlivan D, Charng WL, Yaykasli KO,

Bayram Y, Gambin T, Withers M, Atik MM, Arslanoglu I et al.

Whole-exome sequencing identifies homozygous GPR161 mutation

in a family with pituitary stalk interruption syndrome. Journal of

Clinical Endocrinology and Metabolism 2015 100 E140–E147.

(doi:10.1210/jc.2014-1984)

242 Mullis PE. Genetics of GHRH, GHRH-receptor, GH and GH-receptor:

Its impact on pharmacogenetics. Best Practice & Research. Clinical

Endocrinology & Metabolism 2011 25 25–41. (doi:10.1016/j.beem.2010.

06.006)

243 Ursini MV, Gaetaniello L, Ambrosio R, Matrecano E, Apicella AJ,

Salerno MC & Pignata C. Atypical X-linked SCID phenotype

associated with growth hormone hyporesponsiveness. Clinical and

Experimental Immunology 2002 129 502–509. (doi:10.1046/j.1365-

2249.2002.01823.x)

244 Adriani M, Garbi C, Amodio G, Russo I, Giovannini M, Amorosi S,

Matrecano E, Cosentini E, Candotti F & Pignata C. Functional

interaction of common gamma-chain and growth hormone receptor

signaling apparatus. Journal of Immunology 2006 177 6889–6895.

(doi:10.4049/jimmunol.177.10.6889)

245 Flottmann R, Knaus A, Zemojtel T, Robinson PN, Mundlos S,

Horn D & Spielmann M. FGFR2 mutation in a patient without

typical features of Pfeiffer syndrome – the emerging role of

combined NGS and phenotype based strategies. European

Journal of Medical Genetics 2015 58 376–380. (doi:10.1016/j.ejmg.

2015.05.007)

246 Heuertz S, Le Merrer M, Zabel B, Wright M, Legeai-Mallet L, Cormier-

Daire V, Gibbs L & Bonaventure J. Novel FGFR3 mutations creating

cysteine residues in the extracellular domain of the receptor cause

achondroplasia or severe forms of hypochondroplasia. European

Journal of Human Genetics 2006 14 1240–1247. (doi:10.1038/sj.ejhg.

5201700)

247 Racacho L, Byrnes AM, MacDonald H, Dranse HJ, Nikkel SM,

Allanson J, Rosser E, Underhill TM & Bulman DE. Two novel

disease-causing variants in BMPR1B are associated with brachydactyly

type A1. European Journal of Human Genetics 2015 23 1640–1645.

(doi:10.1038/ejhg.2015.38)

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 27: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R171

248 Liu X, Gao L, Zhao A, Zhang R, Ji B, Wang L, Zheng Y, Zeng B,

Valenzuela RK, He L et al. Identification of duplication downstream

of BMP2 in a Chinese family with brachydactyly type A2 (BDA2).

PLoS ONE 2014 9 e94201. (doi:10.1371/journal.pone.0094201)

249 Al-Qattan MM, Al-Motairi MI & Al Balwi MA. Two novel homozygous

missense mutations in the GDF5 gene cause brachydactyly type C.

American Journal of Medical Genetics. Part A 2015 167 1621–1626.

(doi:10.1002/ajmg.a.37040)

250 Le GC, Mahaut C, Wang LW, Allali S, Abhyankar A, Jensen S,

Zylberberg L, Collod-Beroud G, Bonnet D, Alanay Y et al. Mutations in

the TGFb binding-protein-like domain 5 of FBN1 are responsible for

acromicric and geleophysic dysplasias. American Journal of Human

Genetics 2011 89 7–14. (doi:10.1016/j.ajhg.2011.05.012)

251 Cain SA, McGovern A, Baldwin AK, Baldock C & Kielty CM. Fibrillin-1

mutations causing Weill–Marchesani syndrome and acromicric and

geleophysic dysplasias disrupt heparan sulfate interactions. PLoS ONE

2012 7 e48634. (doi:10.1371/journal.pone.0048634)

252 Miyake N, Elcioglu NH, Iida A, Isguven P, Dai J, Murakami N,

Takamura K, Cho TJ, Kim OH, Hasegawa T et al. PAPSS2 mutations

cause autosomal recessive brachyolmia. Journal of Medical Genetics

2012 49 533–538. (doi:10.1136/jmedgenet-2012-101039)

253 Gardner CJ, Robinson N, Meadows T, Wynn R, Will A, Mercer J,

Church HJ, Tylee K, Wraith JE & Clayton PE. Growth, final height and

endocrine sequelae in a UK population of patients with Hurler

syndrome (MPS1H). Journal of Inherited Metabolic Disease 2011 34

489–497. (doi:10.1007/s10545-010-9262-8)

254 Makitie O, Susic M, Ward L, Barclay C, Glorieux FH & Cole WG.

Schmid type of metaphyseal chondrodysplasia and COL10A1

mutations – findings in 10 patients. American Journal of Medical

Genetics. Part A 2005 137A 241–248. (doi:10.1002/ajmg.a.30855)

255 Briggs MD, Brock J, Ramsden SC & Bell PA. Genotype to phenotype

correlations in cartilage oligomeric matrix protein associated chon-

drodysplasias. European Journal of Human Genetics 2014 22 1278–1282.

(doi:10.1038/ejhg.2014.30)

256 Posey KL, Alcorn JL & Hecht JT. Pseudoachondroplasia/COMP –

translating from the bench to the bedside. Matrix Biology 2014 37

167–173. (doi:10.1016/j.matbio.2014.05.006)

257 Terhal PA, Nievelstein RJ, Verver EJ, Topsakal V, van Dommelen P,

Hoornaert K, Le MM, Zankl A, Simon ME, Smithson SF et al. A study of

the clinical and radiological features in a cohort of 93 patients with a

COL2A1 mutation causing spondyloepiphyseal dysplasia congenita or

a related phenotype. American Journal of Medical Genetics. Part A 2015

167A 461–475. (doi:10.1002/ajmg.a.36922)

258 Gleghorn L, Ramesar R, Beighton P & Wallis G. A mutation in the

variable repeat region of the aggrecan gene (AGC1) causes a form of

spondyloepiphyseal dysplasia associated with severe, premature

osteoarthritis. American Journal of Human Genetics 2005 77 484–490.

(doi:10.1086/444401)

259 Morales J, Al-Sharif L, Khalil DS, Shinwari JM, Bavi P, Al-Mahrouqi RA,

Al-Rajhi A, Alkuraya FS, Meyer BF & Al Tassan N. Homozygous mutations

in ADAMTS10 and ADAMTS17 cause lenticular myopia, ectopia lentis,

glaucoma, spherophakia, and short stature. American Journal of Human

Genetics 2009 85 558–568. (doi:10.1016/j.ajhg.2009.09.011)

260 Bezniakow N, Gos M & Obersztyn E. The RASopathies as an example

of RAS/MAPK pathway disturbances – clinical presentation and

molecular pathogenesis of selected syndromes. Developmental Period

Medicine 2014 18 285–296.

261 Chen PC, Yin J, Yu HW, Yuan T, Fernandez M, Yung CK, Trinh QM,

Peltekova VD, Reid JG, Tworog-Dube E et al. Next-generation

sequencing identifies rare variants associated with Noonan syndrome.

PNAS 2014 111 11473–11478. (doi:10.1073/pnas.1324128111)

262 Szudek J, Birch P & Friedman JM. Growth in North American white

children with neurofibromatosis 1 (NF1). Journal of Medical Genetics

2000 37 933–938. (doi:10.1136/jmg.37.12.933)

263 Aten E, Sun Y, Almomani R, Santen GW, Messemaker T, Maas SM,

Breuning MH & den Dunnen JT. Exome sequencing identifies a

branch point variant in Aarskog–Scott syndrome. Human Mutation

2013 34 430–434. (doi:10.1002/humu.22252)

264 Mattos EP, Sanseverino MT, Magalhaes JA, Leite JC, Felix TM,

Todeschini LA, Cavalcanti DP & Schuler-Faccini L. Clinical and

molecular characterization of a Brazilian cohort of campomelic

dysplasia patients, and identification of seven new SOX9 mutations.

Genetics and Molecular Biology 2015 38 14–20. (doi:10.1590/S1415-

475738120140147)

265 Dentici ML, Di PA, Lepri FR, Gnazzo M, Lombardi MH, Auriti C,

Petrocchi S, Pisaneschi E, Bellacchio E, Capolino R et al. Kabuki

syndrome: clinical and molecular diagnosis in the first year of life.

Archives of Disease in Childhood 2015 100 158–164. (doi:10.1136/

archdischild-2013-305858)

266 Isojima T, Doi K, Mitsui J, Oda Y, Tokuhiro E, Yasoda A, Yorifuji T,

Horikawa R, Yoshimura J, Ishiura H et al. A recurrent de novo

FAM111A mutation causes Kenny–Caffey syndrome type 2.

Journal of Bone and Mineral Research 2014 29 992–998. (doi:10.1002/

jbmr.2091)

267 Dorr HG, Madeja J & Junghans C. Spontaneous postnatal growth is

reduced in children with CHARGE syndrome. Acta Paediatrica 2015

104 e314–e318. (doi:10.1111/apa.12980)

268 de Munnik SA, Hoefsloot EH, Roukema J, Schoots J, Knoers NV,

Brunner HG, Jackson AP & Bongers EM. Meier–Gorlin syndrome.

Orphanet Journal of Rare Diseases 2015 10 114. (doi:10.1186/s13023-

015-0322-x)

269 Griffith E, Walker S, Martin CA, Vagnarelli P, Stiff T, Vernay B,

Al Sanna N, Saggar A, Hamel B, Earnshaw WC et al. Mutations in

pericentrin cause Seckel syndrome with defective ATR-dependent

DNA damage signaling. Nature Genetics 2008 40 232–236.

(doi:10.1038/ng.2007.80)

270 Martin CA, Ahmad I, Klingseisen A, Hussain MS, Bicknell LS, Leitch A,

Nurnberg G, Toliat MR, Murray JE, Hunt D et al. Mutations in PLK4,

encoding a master regulator of centriole biogenesis, cause micro-

cephaly, growth failure and retinopathy. Nature Genetics 2014 46

1283–1292. (doi:10.1038/ng.3122)

271 Tarquinio DC, Motil KJ, Hou W, Lee HS, Glaze DG, Skinner SA,

Neul JL, Annese F, McNair L, Barrish JO et al. Growth failure and

outcome in Rett syndrome: specific growth references. Neurology 2012

79 1653–1661. (doi:10.1212/WNL.0b013e31826e9a70)

272 Seltzer LE & Paciorkowski AR. Genetic disorders associated with

postnatal microcephaly. American Journal of Medical Genetics. Part C,

Seminars in Medical Genetics 2014 166C 140–155. (doi:10.1002/ajmg.c.

31400)

273 Shaheen R, Faqeih E, Ansari S, Abdel-Salam G, Al-Hassnan ZN,

Al-Shidi T, Alomar R, Sogaty S & Alkuraya FS. Genomic analysis of

primordial dwarfism reveals novel disease genes. Genome Research

2014 24 291–299. (doi:10.1101/gr.160572.113)

274 Arora H, Chacon AH, Choudhary S, McLeod MP, Meshkov L, Nouri K

& Izakovic J. Bloom syndrome. International Journal of Dermatology

2014 53 798–802. (doi:10.1111/ijd.12408)

275 Petryk A, Kanakatti SR, Giri N, Hollenberg AN, Rutter MM, Nathan B,

Lodish M, Alter BP, Stratakis CA & Rose SR. Endocrine disorders in

Fanconi anemia: recommendations for screening and treatment.

Journal of Clinical Endocrinology and Metabolism 2015 100 803–811.

(doi:10.1210/jc.2014-4357)

276 Manandhar M, Boulware KS & Wood RD. The ERCC1 and ERCC4

(XPF) genes and gene products. Gene 2015 569 153–161. (doi:10.1016/

j.gene.2015.06.026)

277 Gonzalo S & Kreienkamp R. DNA repair defects and genome instability

in Hutchinson-Gilford Progeria Syndrome. Current Opinion in Cell

Biology 2015 34 75–83. (doi:10.1016/j.ceb.2015.05.007)

278 Baple EL, Chambers H, Cross HE, Fawcett H, Nakazawa Y, Chioza BA,

Harlalka GV, Mansour S, Sreekantan-Nair A, Patton MA et al.

Hypomorphic PCNA mutation underlies a human DNA repair

disorder. Journal of Clinical Investigation 2014 124 3137–3146.

(doi:10.1172/JCI74593)

www.eje-online.org

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 28: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R172

279 Morimoto M, Yu Z, Stenzel P, Clewing JM, Najafian B, Mayfield C,

Hendson G, Weinkauf JG, Gormley AK, Parham DM et al. Reduced

elastogenesis: a clue to the arteriosclerosis and emphysematous

changes in Schimke immuno-osseous dysplasia? Orphanet Journal of

Rare Diseases 2012 7 70. (doi:10.1186/1750-1172-7-70)

280 Hughes CR, Guasti L, Meimaridou E, Chuang CH, Schimenti JC,

King PJ, Costigan C, Clark AJ & Metherell LA. MCM4 mutation causes

adrenal failure, short stature, and natural killer cell deficiency in

humans. Journal of Clinical Investigation 2012 122 814–820.

(doi:10.1172/JCI60224)

281 Gineau L, Cognet C, Kara N, Lach FP, Dunne J, Veturi U, Picard C,

Trouillet C, Eidenschenk C, Aoufouchi S et al. Partial MCM4

deficiency in patients with growth retardation, adrenal insufficiency,

and natural killer cell deficiency. Journal of Clinical Investigation 2012

122 821–832. (doi:10.1172/JCI61014)

282 Chrzanowska KH, Gregorek H, Dembowska-Baginska B, Kalina MA &

Digweed M. Nijmegen breakage syndrome (NBS). Orphanet Journal of

Rare Diseases 2012 7 13. (doi:10.1186/1750-1172-7-13)

283 Wood-Trageser MA, Gurbuz F, Yatsenko SA, Jeffries EP, Kotan LD,

Surti U, Ketterer DM, Matic J, Chipkin J, Jiang H et al. MCM9

mutations are associated with ovarian failure, short stature, and

chromosomal instability. American Journal of Human Genetics 2014 95

754–762. (doi:10.1016/j.ajhg.2014.11.002)

284 Smeets MF, DeLuca E, Wall M, Quach JM, Chalk AM, Deans AJ,

Heierhorst J, Purton LE, Izon DJ & Walkley CR. The Rothmund–

Thomson syndrome helicase RECQL4 is essential for hematopoiesis.

Journal of Clinical Investigation 2014 124 3551–3565. (doi:10.1172/

JCI75334)

285 Gibbons RJ & Higgs DR. Molecular-clinical spectrum of the ATR-X

syndrome. American Journal of Medical Genetics 2000 97 204–212. (doi:10.

1002/1096-8628(200023)97:3!204::AID-AJMG1038O3.0.CO;2-X)

286 Woodbine L, Gennery AR & Jeggo PA. The clinical impact of

deficiency in DNA non-homologous end-joining. DNA Repair 2014 16

84–96. (doi:10.1016/j.dnarep.2014.02.011)

287 Mathieu AL, Verronese E, Rice GI, Fouyssac F, Bertrand Y, Picard C,

Chansel M, Walter JE, Notarangelo LD, Butte MJ et al. PRKDC

mutations associated with immunodeficiency, granuloma, and

autoimmune regulator-dependent autoimmunity. Journal of Allergy

and Clinical Immunology 2015 135 1578–1588. (doi:10.1016/j.jaci.

2015.01.040)

288 Mefford HC, Sharp AJ, Baker C, Itsara A, Jiang Z, Buysse K, Huang S,

Maloney VK, Crolla JA, Baralle D et al. Recurrent rearrangements of

chromosome 1q21.1 and variable pediatric phenotypes. New England

Journal of Medicine 2008 359 1685–1699. (doi:10.1056/

NEJMoa0805384)

289 Maas NM, Van Buggenhout G, Hannes F, Thienpont B, Sanlaville D,

Kok K, Midro A, Andrieux J, Anderlid BM, Schoumans J et al.

Genotype–phenotype correlation in 21 patients with Wolf–Hirsch-

horn syndrome using high resolution array comparative genome

hybridisation (CGH). Journal of Medical Genetics 2008 45 71–80.

(doi:10.1136/jmg.2007.052910)

290 Hart L, Rauch A, Carr AM, Vermeesch JR & O’Driscoll M. LETM1

haploinsufficiency causes mitochondrial defects in cells from humans

with Wolf–Hirschhorn syndrome: implications for dissecting the

underlying pathomechanisms in this condition. Disease Models &

Mechanisms 2014 7 535–545. (doi:10.1242/dmm.014464)

291 Bonnet C, Andrieux J, Beri-Dexheimer M, Leheup B, Boute O,

Manouvrier S, Delobel B, Copin H, Receveur A, Mathieu M et al.

Microdeletion at chromosome 4q21 defines a new emerging syn-

drome with marked growth restriction, mental retardation and absent

or severely delayed speech. Journal of Medical Genetics 2010 47

377–384. (doi:10.1136/jmg.2009.071902)

292 Sardina JM, Walters AR, Singh KE, Owen RX & Kimonis VE.

Amelioration of the typical cognitive phenotype in a patient with the

5pter deletion associated with Cri-du-chat syndrome in addition to a

www.eje-online.org

partial duplication of CTNND2. American Journal of Medical Genetics.

Part A 2014 164A 1761–1764. (doi:10.1002/ajmg.a.36494)

293 Franco LM, de Ravel T, Graham BH, Frenkel SM, Van Driessche J,

Stankiewicz P, Lupski JR, Vermeesch JR & Cheung SW. A syndrome of

short stature, microcephaly and speech delay is associated with

duplications reciprocal to the common Sotos syndrome deletion.

European Journal of Human Genetics 2010 18 258–261. (doi:10.1038/

ejhg.2009.164)

294 Honjo RS, Dutra RL, Furusawa EA, Zanardo EA, Costa LS,

Kulikowski LD, Bertola DR & Kim CA. Williams–Beuren Syndrome:

a clinical study of 55 Brazilian patients and the diagnostic use of

MLPA. BioMed Research International 2015 2015 903175. (doi:10.1155/

2015/903175)

295 Maas SM, Shaw AC, Bikker H, Ludecke HJ, van der Tuin K, Badura-

Stronka M, Belligni E, Biamino E, Bonati MT, Carvalho DR et al.

Phenotype and genotype in 103 patients with tricho-rhino-phalan-

geal syndrome. European Journal of Medical Genetics 2015 58 279–292.

(doi:10.1016/j.ejmg.2015.03.002)

296 Adams DJ & Clark DA. Common genetic and epigenetic syndromes.

Pediatric Clinics of North America 2015 62 411–426. (doi:10.1016/j.pcl.

2014.11.005)

297 Buysse K, Reardon W, Mehta L, Costa T, Fagerstrom C, Kingsbury DJ,

Anadiotis G, McGillivray BC, Hellemans J, de Leeuw N et al. The 12q14

microdeletion syndrome: additional patients and further evidence

that HMGA2 is an important genetic determinant for human height.

European Journal of Medical Genetics 2009 52 101–107. (doi:10.1016/j.

ejmg.2009.03.001)

298 Alyaqoub F, Pyatt RE, Bailes A, Brock A, Deeg C, McKinney A,

Astbury C, Reshmi S, Shane KP, Thrush DL et al. 12q14 microdeletion

associated with HMGA2 gene disruption and growth restriction.

American Journal of Medical Genetics. Part A 2012 158A 2925–2930.

(doi:10.1002/ajmg.a.35610)

299 Mitter D, Ullmann R, Muradyan A, Klein-Hitpass L, Kanber D,

Ounap K, Kaulisch M & Lohmann D. Genotype–phenotype corre-

lations in patients with retinoblastoma and interstitial 13q deletions.

European Journal of Human Genetics 2011 19 947–958. (doi:10.1038/

ejhg.2011.58)

300 Martinez-Fernandez ML, Bermejo-Sanchez E, Fernandez B,

MacDonald A, Fernandez-Toral J & Martinez-Frias ML. Haploinsuffi-

ciency of BMP4 gene may be the underlying cause of Frias syndrome.

American Journal of Medical Genetics. Part A 2014 164A 338–345.

(doi:10.1002/ajmg.a.36224)

301 Dubourg C, Bonnet-Brilhault F, Toutain A, Mignot C, Jacquette A,

Dieux A, Gerard M, Beaumont-Epinette MP, Julia S, Isidor B et al.

Identification of nine new RAI1-truncating mutations in Smith–

Magenis syndrome patients without 17p11.2 deletions. Molecular

Syndromology 2014 5 57–64. (doi:10.1159/000357359)

302 Yu YR, You LR, Yan YT, Chen CM et al. Role of OVCA1/DPH1

in craniofacial abnormalities of Miller–Dieker syndrome.

Human Molecular Genetics 2014 23 5579–5596. (doi:10.1093/hmg/

ddu273)

303 Ostergaard JR, Graakjaer J, Brandt C & Birkebaek NH. Further

delineation of 17p13.3 microdeletion involving CRK. The effect of

growth hormone treatment. European Journal of Medical Genetics 2012

55 22–26. (doi:10.1016/j.ejmg.2011.09.004)

304 Lukusa T & Fryns JP. Pure de novo 17q25.3 micro duplication

characterized by micro array CGH in a dysmorphic infant with growth

retardation, developmental delay and distal arthrogryposis.

Genetic Counseling 2010 21 25–34.

305 Wester U, Bondeson ML, Edeby C & Anneren G. Clinical and

molecular characterization of individuals with 18p deletion: a

genotype–phenotype correlation. American Journal of Medical Genetics.

Part A 2006 140 1164–1171. (doi:10.1002/ajmg.a.31260)

306 Feenstra I, Vissers LE, Pennings RJ, Nillessen W, Pfundt R, Kunst HP,

Admiraal RJ, Veltman JA, van Ravenswaaij-Arts CM, Brunner HG et al.

Disruption of teashirt zinc finger homeobox 1 is associated with

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access

Page 29: MECHANISMS IN ENDOCRINOLOGY Novel genetic causes of short

Eu

rop

ean

Jou

rnal

of

En

do

crin

olo

gy

Review J M Wit and others Genetics of short stature 174 :4 R173

congenital aural atresia in humans. American Journal of Human Genetics

2011 89 813–819. (doi:10.1016/j.ajhg.2011.11.008)

307 Cody JD, Hasi M, Soileau B, Heard P, Carter E, Sebold C,

O’Donnell L, Perry B, Stratton RF & Hale DE. Establishing a

reference group for distal 18q-: clinical description and molecular

basis. Human Genetics 2014 133 199–209. (doi:10.1007/s00439-

013-1364-6)

308 Rump P, de Leeuw N, van Essen AJ, Verschuuren-Bemelmans CC,

Veenstra-Knol HE, Swinkels ME, Oostdijk W, Ruivenkamp C,

Reardon W, de Munnik S et al. Central 22q11.2 deletions. American

Journal of Medical Genetics. Part A 2014 164A 2707–2723. (doi:10.1002/

ajmg.a.36711)

309 Hacihamdioglu B, Hacihamdioglu D & Delil K. 22q11 deletion

syndrome: current perspective. Application of Clinical Genetics 2015 8

123–132. (doi:10.2147/TACG.S82105)

310 Penaherrera MS, Weindler S, Van Allen MI, Yong SL, Metzger DL,

McGillivray B, Boerkoel C, Langlois S & Robinson WP. Methylation

profiling in individuals with Russell–Silver syndrome. American

Journal of Medical Genetics. Part A 2010 152A 347–355. (doi:10.1002/

ajmg.a.33204)

Received 18 September 2015

Revised version received 2 November 2015

Accepted 16 November 2015

www.eje-online.org

Downloaded from Bioscientifica.com at 05/07/2022 08:51:50PMvia free access