Download as pdf or txt
Download as pdf or txt
You are on page 1of 24

Received: 7 August 2015 | Revised: 30 December 2016 | Accepted: 6 January 2017

DOI 10.1002/ajmg.a.38159

RESEARCH REVIEW

Genetic advances in craniosynostosis

Wanda Lattanzi1,2 | Marta Barba1 | Lorena Di Pietro1 | Simeon A. Boyadjiev3


1 Institute
of Anatomy and Cell Biology,
Università Cattolica del Sacro Cuore, Rome, Italy Craniosynostosis, the premature ossification of one or more skull sutures, is a clinically and
2 Latium Musculoskeletal Tıssue Bank, Rome, genetically heterogeneous congenital anomaly affecting approximately one in 2,500 live births.
Italy In most cases, it occurs as an isolated congenital anomaly, that is, nonsyndromic
3 Division of Genomic Medicine, Department of craniosynostosis (NCS), the genetic, and environmental causes of which remain largely
Pediatrics, Davis Medical Center, University of
unknown. Recent data suggest that, at least some of the midline NCS cases may be explained by
California, Sacramento, California
two loci inheritance. In approximately 25–30% of patients, craniosynostosis presents as a
Correspondence
Simeon A. Boyadjiev, Division of Genomic feature of a genetic syndrome due to chromosomal defects or mutations in genes within
Medicine, Department of Pediatrics, University interconnected signaling pathways. The aim of this review is to provide a detailed and
of California, Sacramento, CA 95817. comprehensive update on the genetic and environmental factors associated with NCS,
Email: sboyd@ucdavis.edu
integrating the scientific findings achieved during the last decade. Focus on the neuro-
Funding information
developmental, imaging, and treatment aspects of NCS is also provided.
NIH-NIDCR, Grant number: R01DE16886;
“Federazione GENE” patients’ association
KEYWORDS
craniofacial, malformation, craniosynostosis, gene mutations, skull sutures

1 | INTRODUCTION According to an etiological classification, CS can be defined as


primary, due to intrinsic genetic causes acting alone or in combination
Major anomalies affect 1 in 33 newborns or about 125,000 live with environmental factors; or secondary, due to disorders affecting
births annually in the U.S. and are a leading cause of infant mortality the developing suture (Higashino & Hirabayashi, 2013; Shetty,
(Parker et al., 2010; Petrini et al., 2002). Craniofacial anomalies Thomas, Rao, & Vargas, 1998). Conditions associated with secondary
account for close to one-third of all anomalies. They contribute CS include hyperthyroidism, hypercalcemia, hypophosphatasia, sickle
significantly to infant morbidity and disability and the expenditure of cell disease and thalassemia, vitamin D deficiency, renal osteodys-
millions of dollars annually in health care costs (Dolk, Loane, & trophy, and Hurler syndrome, among others. Finally, secondary CS
Garne, 2010; Wehby & Cassell, 2010; Weiss et al., 2009). may also be due to conditions that restrict growth across the open
Craniosynostosis (CS) is one of the most common anomalies suture, such as microcephaly, encephalocele, and shunted hydroceph-
occurring in approximately 1 in 2,500 live births (Boulet, Rasmussen, alus (Khanna, Thapa, Iyer, & Prasad, 2011).
& Honein, 2008; Boyadjiev, 2014: Cohen, 2000; Gorlin, Cohen, & Primary craniosynostoses are usually classified, based on the
Hennekam, 2001), and represents a clinically and genetically possible association with additional clinical features, as syndromic or
heterogeneous congenital anomaly. nonsyndromic (NCS). Syndromic CS manifests with additional
The main phenotypic manifestation of CS is an abnormal head anomalies and/or developmental delays, and is thought to account
shape due to restriction of skull growth in a direction, that is, for 25–30% of all cases (Passos-Bueno, Serti Eacute, Jehee,
perpendicular to the closed suture (Persing, Jane, & Shaffrey, 1989; Fanganiello, & Yeh, 2008; Wilkie et al., 2007, 2010). Monogenic
Virchow, 1851) (Figure 1). This congenital anomaly may lead to skull mutations or chromosomal defects are typically found in 75–80% of
constraints, which may affect brain growth, resulting in a number of the syndromic patients (Johnson & Wilkie, 2011). Conversely, NCS
morphologic and functional abnormalities. True CS is to be differenti- occurs in the absence of associated anomalies or developmental
ated from positional plagiocephaly, which represents a deformation of delays (Boyadjiev & International Craniosynostosis Consortium,
the skull in the absence of sutural fusion (Komotar, Zacharia, Ellis, 2007).
Feldstein, & Anderson, 2006; Liu et al., 2008). This is an important, but NCS occurs in approximately 75% of all patients with CS
not always easy task as posterior skull flattening may affect close to (Cohen, 2000; Greenwood, Flodman, Osann, Boyadjiev, & Kimonis,
20% of healthy infants at 4 months of age (Rogers, 2011). 2014). It is usually classified based on the type of suture fusion

1406 | © 2017 Wiley Periodicals, Inc. wileyonlinelibrary.com/journal/ajmga Am J Med Genet. 2017;173A:1406–1429.


LATTANZI ET AL.
| 1407

F I G UR E 1 Craniofacial features and three-dimensional computed tomography images of individuals with craniosynostosis. (A–C)
Nonsyndromic sagittal craniosynostosis. (D–F) Nonsyndromic metopic craniosynostosis. (G–I) Nonsyndromic unilateral right coronal
craniosynostosis. (J–L) Nonsyndromic bilateral coronal craniosynostosis. (M–O) Nonsyndromic unilateral left lambdoid craniosynostosis. (P–R)
Nonsyndromic bilateral lambdoid craniosynostosis. (S–W) Images of individuals with Muenke syndrome demonstrate extreme clinical
variability and overlap of the milder cases with nonsyndromic coronal craniosynostosis. (X) Mild case of Saethre–Chotzen syndrome resembles
unicoronal craniosynostosis. The images on the bottom row demonstrate the need of molecular testing of the coronal cases for proper
diagnosis

(sagittal, metopic, bi- or unicoronal, and bi- or unilambdoid Bicoronal synostosis, leading to brachycephaly, is approximately
synostoses), that leads to an abnormal skull shape (dolichocephaly, half as frequent as the unicoronal form (Lajeunie, Le Merrer,
trigonocephaly, brachycephaly, anterior or posterior plagiocephaly, Bonaïti-Pellie, Marchac, & Renier, 1995). Lambdoid synostosis,
respectively). Sagittal synostosis is the most frequent form, resulting in posterior plagiocephaly, is the least common type,
accounting for 45–58% of all NCSs (1,9–2,3 per 10,000 live births) representing 1% of all CS and 3% of NCS (Heuzé, Holmes, Peter,
(Di Rocco, Arnaud, Meyer, Sainte-Rose, & Renier, 2009; Lajeunie, Le Richtsmeier, & Jabs, 2014; Lattanzi et al., 2012). Although NCS are
Merrer, Bonaïti-Pellie, Marchac, & Renier, 1996; Lattanzi et al., mainly sporadic, a positive family history has been reported in 2–
2012; Ursitti et al., 2011). Recent epidemiological studies have 10% of the patients depending on the involved suture (Greenwood
suggested that metopic synostosis has become the second most et al., 2014).
prevalent form (occurring in over 25% of all NCS patients), as a NCS appears to be a heterogeneous group of anomalies based on
result of a significantly increased incidence in the Western World the type of suture closure patterns as suggested by the various
over the last two decades. This may be due to a number of factors, population incidences and discrepant male/female ratios (2.5:1–3:1
including increased exposure to environmental risk factors or for sagittal and metopic NCS and around 1:2 for unicoronal NCS)
improved diagnosis (Selber et al., 2008; van der Meulen et al., 2009). (Selber et al., 2008). NCS is thought to be a multifactorial condition,
Unicoronal synostosis has become the third most prevalent NCS, with both genetic and environmental factors contributing to the
accounting for less than 15% of NCS in the large cohorts analyzed to phenotype. The evidence for genetic determinants of NCS is based on
date (Di Rocco, Arnaud, Meyer, et al., 2009, Di Rocco, Arnaud, & observations of multiplex families, the increased recurrence risk in the
Renier, 2009; Selber et al., 2008; van der Meulen et al., 2009). affected families, and twin studies documenting increased
1408 | LATTANZI ET AL.

concordance rates among monozygotic as compared to dizygotic & Boyadjiev, 2007; Lattanzi et al., 2012) and provides the reader with a
twins (60.9% vs. 5.3%) (Lakin, Sinkin, Chen, Koltz, & Girotto, 2012). broader insight into CS, its developmental aspects, the diagnostic
However, the lack of complete concordance among monozygotic imaging techniques and available surgical options.
twins suggests that environmental, epistatic, and/or epigenetic factors
play a role in the etiology of CS. We have found that the highest
incidence of recurrence among first-degree relatives, mostly in 2 | SKULL AND SUTURE ORIGIN AND
siblings, was for metopic NCS, followed by complex NCS, sagittal GROWTH
NCS, lambdoid NCS, and coronal NCS (Greenwood et al., 2014). In this
same cohort, patients with complex NCS showed the highest Human skull development begins around 23–26 days post-fertiliza-
frequency of associated symptoms (including ear infections, palate tion, when a multipotent cranial neural crest cell population migrates
abnormalities, and hearing problems) while sagittal NCS presented from the dorsal aspect of the neural tube into the head region of the
mostly as a truly isolated form (Greenwood et al., 2014). Previous embryo to give rise to craniofacial structures. Upon completion of
epidemiological studies (Alderman et al., 1988; Boyadjiev, 2014; Gill neural crest migration the head has mesenchyme originating from both
et al., 2012; Kallen, 1999; Reefhuis, Honein, Shaw, & Romitti, 2003; paraxial mesoderm and cranial neural crest (Noden, 1983, 1986).
Reefhuis & Honein, 2004; Sanchez-Lara et al., 2010; Zeiger et al., While the neural crest is widely recognized as the embryonic origin of
2002) have suggested that male sex, non-Hispanic white race/ the facial bones (Couly, Coltey, & Le Douarin, 1992; Couly, Coltey, &
ethnicity, advanced maternal age, and maternal age <20 years are Le Douarin, 1993; Opperman, 2000), the origin of the other
associated with NCS. Such studies showed mixed results for twinning, intramembranous calvarial bones is still debatable. Experiments with
intrauterine head constraint, plurality, parity, prematurity, and macro- a quail-chick chimera system have suggested that the intramembra-
somia. Prenatal exposures to maternal tobacco smoking and other nous bones of the cranial vault and the sutures originate from paraxial
hypoxemia-inducing agents (Carmichael et al., 2008), sodium val- mesoderm (Noden, 1986). However, these data were later disputed by
proate (Lajeunie et al., 2001), retinoic acid (James, Levi, Xu, Carre, & studies using the same experimental system that suggested the
Longaker, 2010), antidepressants (Alwan, Reefhuis, Rasmussen, cephalic neural crest cells were an initial source of the membranous
Olney, & Friedman, 2007), and infertility medications (Ardalan et al., bones of the skull (Couly et al., 1992, 1993). Even if there is no
2012) have been suggested, but have not been conclusively proven to contribution of neural crest cells to the membranous bones, the dura,
be related. The equivocal findings for environmental exposures may be which arises from neural crest cells, plays a major role in regulating the
due to small sample sizes, limited co-variable data, limited subtype growth of the membranous vault, and maintaining suture patency
analysis, and differently defined intervals for the critical period of (Bifari et al., 2015; Lee et al., 2006; Yu, Hess, Horning, Brown, &
exposure. Korsmeyer, 1995). The current view of the embryonic origin of the
The public health burden of CS is substantial. Infants with CS skull is that the neural crest gives rise to the frontal bone and the
typically require extensive surgical treatment in the first year of life. interparietal portion of the occipital bone, while the rest of the skull
The most frequent and serious perioperative complication is the vault is derived mostly from mesoderm, with some minor contribution
occurrence of intraoperative hemorrhages that can cause significant from the cranial neural crest cell population (Jiang, Iseki, Maxson,
blood loss and require transfusions in approximately 80% of patients Sucov, & Morriss-Kay, 2002; Ting et al., 2009).
(Mathijssen & Arnaud, 2007). Re-synostosis occurs in approximately The growth of the skull is driven by the growth of the underlying
6% of NCS patients and in 15% of those with syndromic CS (Foster, brain, which reaches 90% of adult size in the first year and 95% of adult
Frim, & McKinnon, 2008). Patients with complex and/or syndromic size by 6 years of age (Williams, 2008). The mature skull has two main
forms typically require multiple surgeries, with increased burden and components: the neurocranium and the viscerocranium. The neuro-
consequently worse prognosis (Wilkie et al., 2010). With current cranium includes the membranous skull roof that surrounds the brain
surgical treatment methods, the mortality is less than 1% (Nguyen, and is comprised of the frontal bone, the paired parietal and temporal
Hernandez-Boussard, Khosla, & Curtin, 2013) but even after bones, and the occipital bone, which has both cartilaginous and
successful surgery, children with sagittal NCS or metopic NCS can membranous parts. It also includes the cartilaginous skull base with the
experience ongoing medical problems, such as increased intracranial greater wings of the sphenoid, the ethmoid, and the petrous part of the
pressure (Shimoji, Shimabukuro, Sugama, & Ochiai, 2002; Thompson, temporal bones (Shapiro & Robinson, 1976). The viscerocranium
Harkness, et al., 1995; Thompson, Malcolm, et al., 1995), develop- consists of the cartilaginous facial bones that articulate with the skull
mental disabilities (Chieffo et al., 2010; Magge, Westerveld, Pruzinsky, base, and may be distorted to a various degree in CS.
& Persing, 2002; Shipster et al., 2003), vision problems (Gupta et al., The cranial sutures form around the 16–18th week post-
2003), Chiari I malformation (Tubbs, Elton, Blount, & Oakes, 2001), and fertilization as dense fibrous tissue membranes bridging the gaps
even sudden death (Rabl, Tributsch, & Ambach, 1990). between the cranial bones (Mathijssen et al., 1999; Wilkie, 1997).
In this paper, we provide a comprehensive review of the genetic The calvarial sutures allow molding of the head in the birth canal,
causes of CS, with a specific focus on the molecular mechanisms serve as growth sites of the skull, and absorb mechanical trauma
associated with NCS and newly categorized syndromes. This work is an in the first years of life. During that period, the cells within the
update of our previous reviews on this topic (Boyadjiev & International suture must proliferate without differentiating to maintain
Craniosynostosis Consortium, 2007; Kimonis, Gold, Hoffman, Panchal, patency while those at the bone margins differentiate to ensure
LATTANZI ET AL.
| 1409

growth (Lana-Elola, Rice, Grigoriadis, & Rice, 2007; Morriss-Kay & etiopathogenesis can also be gathered from chromosomal aberra-
Wilkie, 2005). This balance between cell proliferation and tions found in patients. These are observed in about 14% of patients
differentiation ensures suture patency and the coordinated with predominantly complex/syndromic CS, the majority of these
growth of the cranial vault. Indeed, slow cycling, self-renewing being of uncertain significance (Wilkie et al., 2010). The incidence of
stem cells are found within the suture midline which represent a chromosomal aberrations detected by routine karyotype and
calvarial-specific skeletogenic niche, ensuring bone modeling, various molecular techniques was as high as 42% in a group of
remodeling, regeneration, and healing over an extensive time 45 patients with syndromic CS, while submicroscopic defects were
period during skeletal development (Lattanzi, Parolisi, Barba, & present in 28% of those with normal karyotypes (Jehee et al., 2008).
Bonfanti, 2015; Maruyama, Jeong, Sheu, & Hsu, 2016; Zhao & The causative role of many of these, however, is uncertain
Chai, 2015). The metopic suture is the first to close at considering the large number of neutral copy number variants in
approximately 9 months of age (Blaser, 2008) while the fusion the genome. Hence, the true incidence of causative chromosomal
of the remaining calvarial sutures does not occur until the third defects is yet to be determined.
decade of life. In contrast, the sutures of the viscerocranium Several loci have been recurrently described, but their significance
remain open until the seventh or eighth decade of life. The also remains partially unclear (Lattanzi et al., 2012). In particular, 1p36.3
postnatal shape of the skull is characterized by the cephalic index dosage imbalance has been studied in detail in the attempt to identify
(CI = head width/head length ×100), with the normal range 76–81. candidate genes involved in suture closure. Deletion of chromosome
Anything less than 76 is labeled dolichocephaly and above 81 is 1p36, the most common terminal deletion observed in humans, is
brachycephaly (Hall, Allanson, Gripp, & Slavotinek, 2013). associated with delayed suture closure, along with additional craniofacial
It is important to point out that CS is not the sole result of an abnormal features, intellectual disability, hearing impairment, seizures, growth
growth of the calvarial bones. A recent report indicated that congenital impairment, and heart defects (Gajecka et al., 2005). Conversely,
absence of the sagittal suture does not necessarily manifest with duplication and triplication of this locus are extremely rare; they have
dolichocephaly (Padmalayam, Tubbs, Loukas, & Cohen-Gadol, 2013). This been associated with complex malformation syndromes, which include
suggests that CS may not be an entirely skeletal phenotype; deviations from either metopic (Heilstedt, Shapira, Gregg, & Shaffer, 1999) or coronal
the normal brain growth patterns with subsequent abnormal distention of craniosynostosis (Garcia-Heras et al., 1999). Increased copy number of
the dura may also be implicated in suture fusion (Aldridge, Marsh, Govier, & the matrix metalloproteinase-23 (MMP23) −A and −B genes which are
Richtsmeier, 2002; Yu, Lucas, Fryberg, & Borke, 2001). involved in extracellular matrix remodeling and map to 1p36, play a role in
cranial suture patency and closure (Gajecka et al., 2005).
The short arm of chromosome 7 has also been found to be involved
3 | GEN ETIC E TIOPAT HOGEN ESIS OF in interstitial deletions found in patients with complex phenotypes,
often resembling Saethre–Chotzen syndrome (De Marco et al., 2011;
CRA NI O S Y NOS TO S I S
Fryssira et al., 2011; Kosaki et al., 2005). Indeed the 7p21 locus houses
In the attempt to comprehensively dissect the molecular causes of CS, TWIST1 along with the HOXA gene cluster; hence, plausible deletions
we will first briefly introduce the genetic causes of the best defined CS cause haploinsufficiency leading to a complex impairment of systemic

syndromes in this section. We will then focus on the most recent development (Chotai et al., 1994; Tsuji, Narahara, Yokoyama, Grzeschik,
findings regarding the genetic etiology of previously undefined simple & Kunz, 1995; Zneimer, Cotter, & Stewart, 2000).
and complex CS cases, including truly nonsyndromic forms, with the The 9p22.3 region represents a specific hotspot for metopic CS
intent of depicting possible genotype/phenotype correlations be- (OMIM #158170) (Choucair et al., 2015; Jehee et al., 2005; Kawara
tween mutated genes and pattern of suture closure. et al., 2006; Swinkels et al., 2008; Vissers et al., 2011). Deletions at this
locus are found in over 15% of patients with syndromic trigonocephaly
who may also have additional suture involvement, developmental
3.1 | Syndromic CS: genes and chromosomal
delay and facial dysmorphisms, including midface hypoplasia (Jehee
aberrations
et al., 2005). A careful revision of the clinical and the neuroradiological
Significant progress in the last few decades has been made in findings of the 9p deletion syndrome has been recently proposed to
understanding the genetic causes of syndromic forms of CS (Johnson assist with the differential diagnosis of those patients wth trigono-
& Wilkie, 2011; Twigg & Wilkie, 2015; Wilkie et al., 2007). In recent cephaly who are more likely to have an underlying chromosomal
years, converging evidence suggests a stronger influence of genetic aberration (Spazzapan et al., 2016). The FRAS1-related extracellular
factors in the etiopathogenesis of NCS thanks to genome-wide analyses matrix 1 (FREM1) gene is the most likely candidate in this region.
aimed at identifying new mutations and disease-associated loci in large FREM1 is believed to bind fibroblast growth factors (FGFs) within the
patient cohorts. The incomplete list of genetic syndromes with a CS intrasutural mesenchyme. Therefore, with FREM1 haploinsufficiency
phenotype currently includes around 170 entries in the OMIM database the availability of FGF growth factors is expected to increase, which
(http://www.ncbi.nlm.nih.gov/omim/?term=craniosynostosis) (Heuzé ultimately leads to premature ossification (Vissers et al., 2011; Yu et al.,
et al., 2014; Lattanzi, 2016; Twigg & Wilkie, 2015). 2001). More recently, the receptor-type protein tyrosine phosphatase
Although syndromic craniosynostoses are mostly regarded as gene (PTPRD), located in the 9p24.1-p23 region, was deleted in a
monogenic disorders, reliable information on the molecular patient with trigonocephaly, dysmorphic facial features, and severe
1410 | LATTANZI ET AL.

intellectual disability (Choucair et al., 2015). This gene is expressed in anomaly (Dean et al., 1998; De Silva et al., 1995). The molecular
cortical neurons and plays an important role in synaptic organization. etiopathogenesis of the CS phenotype observed in this contiguous
Besides its role in cognitive functions, it is also postulated to represent gene syndrome has not been clarified to date. Finally, metopic
an additional plausible candidate in the 9p22.3 region for metopic CS synostosis has also been described in a single patient with mosaic
(Choucair et al., 2015; Mitsui et al., 2013). trisomy 13 (Aypar et al., 2011). Table 1 summarizes the recurrent
Interstitial duplications involving chromosome 11q have been chromosomal loci discussed in this section and provides a tentative
associated with syndromic CS phenotypes. In particular, 11q11-q13.3 association with the pattern of suture closure. Taken together,
duplications have been reported in patients with either coronal (Grillo these data seem to suggest that the prevalence of chromosomal
et al., 2014) or multisuture CS (Jehee et al., 2007), featuring a plethora aberrations is around 50% when the CS affects the metopic suture.
of additional signs including congenital heart defects and develop- Given the recurrence and the extreme heterogeneity of chromo-
mental delay. Fibroblast growth factor-3 (FGF3) and −4 (FGF4), somal and submicroscopic rearrangements, chromosomal microarray
mapping to 11q13.3, represent the most plausible candidates causing has been recommended as a first-tier genetic test for patients with
abnormal craniofacial development as a result of increased dosage congenital anomalies and/or developmental delays (Miller et al., 2010),
(Grillo et al., 2014; Jehee et al., 2007). and is justified for all patients with any unclassified syndromic CS.
11q23-q24 represents another recurrent hotspot in syndromic CS Whether chromosomal microarray is indicated for patients with
with characteristic facial dysmorphisms and psychomotor delay sporadic NCS is debatable, but may be reasonable for patients with
(Jacobsen syndrome, OMIM#147791), particularly involving the familial occurance of NCS.
metopic suture (Azimi et al., 2003; Foley, McAuliffe, Mullarkey, &
Reardon, 2007; Jehee et al., 2005; Passariello et al., 2013). Genes
3.2 | Molecular genetics of NCS
mapping in this chromosome region include the lysine methyltransfer-
ase 2A (KMT2A) gene, which interacts in multiprotein complexes that NCS is often considered a multifactorial disorder rather than a truly
regulate the expression of multiple HOX genes, among others (Milne genetic condition; however, the putative role of environmental risk
et al., 2002; Nakamura et al., 2002). Interestingly, Kmt2a heterozygous factors is still widely debated (Boyadjiev & International Craniosynostosis
deletion in mice causes homeotic transformation of the axial skeleton Consortium, 2007; Di Rocco, Arnaud, Meyer, et al., 2009; Lajeunie,
and malformations of the membranous bones (Yu et al., 1995). Crimmins, Arnaud, & Renier, 2005). In recent years, evidence from
CS is observed also as a recurrent feature in 22q11 deletion genome-wide association studies and exome sequencing analyses
syndrome (Dean, De Silva, & Reardon, 1998). Variable degrees of CS suggests a stronger influence of genetic factors in the etiopathogenesis
severity have been described in this condition, ranging from of NCS. In a limited number of patients, selected mutations in known
metopic (Yamamoto et al., 2006), or bicoronal CS (McDonald- genomic hotspots and in new loci have been described (Boyadjiev et al.,
McGinn et al., 2005; Rojnueangnit & Robin, 2013), to severe 2002; Twigg & Wilkie, 2015). In some patients, low penetrance mutations
multisutural CS (Al-Hertani, Hastings, McGowan-Jordan, Hurteau, & in genes associated with syndromic forms may contribute to the etiology
Graham, 2013), indicating the markedly variable expressivity of the of NCS (Heuzé et al., 2014). Recently Timberlake et al. (2016) documented

TABLE 1 Chromosomal hotspot loci for CS, with putative candidate genes
Affected suturea

Chromosomal Candidate
locus gene(s) Sagittal Metopic Coronalb Multiple References
1p36 MMP23A + + Garcia-Heras (1999);Heilstedtl, (1999)
MMP23B
9p22.3 FREM1 + Choucair (2015); Jehee (2005); Kawara (2006); Mitsui (2013);
Swinkels (2008); Vissers (2011)
PTPRD
11q11-q13.3 FGF3 + + Jehee (2007); Grillo (2014); Yu, Hess, Horning, Brown, and
Korsmeyer (1995)
FGF4
11q23-q24 KMTM2A? + Azimi(2003); Foley, McAuliffe, Mullarkey, and Reardon (2007);
Jehee (2005); Milne (2002); Nakamura (2002); Passariello (2013)
22q11 CGSc + + + Al-Hertani, Hastings, McGowan-Jordan, Hurteau, and Graham
(2013); McDonald-McGinn et al. (2005); Rojnueangnit and Robin
(2013); Yamamoto (2006)
a
Always in syndromic phenotypes.
b
Both uni- and bicoronal cases are considered.
c
Contiguous gene syndrome.
LATTANZI ET AL.
| 1411

SMAD6 mutations in 7% of patients with midline (sagittal and/or metopic) SMAD6 mutations with reduced penetrance is augmented by the
NCS by using an exome sequencing approach. Importantly, transmitted previously identified risk allele of the common SNP rs1884302 in the
SMAD6 mutations were present in 25% (4 of 17) of the patients with vicinity of BMP2 (Justice et al., 2012). These observations would place at
familial occurance of midline CS. These authors suggested two-loci least a portion of NCS in the relatively small list of human disorders with
inheritance for NCS due to epistatic interaction where the effect of digenic inheritance (Lupski, 2012).

TABLE 2 Genes associated with different types of craniosynostosis


Affected suture

Gene Sagittal Metopic Coronal Lambdoid Multiple Syndromic References


ALX4 + no Mavrogiannis et al. (2001)
BBS9 + no Justice et al. (2012)
BMP2 + no Justice et al. (2012)
EFNA4 + no Merrill et al. (2006)
EFNB1 + no Twigg and Wilkie (2015); Twigg et al. (2015);
+ yes
ERF + + + yes+ Twigg et al. (2013)
FGFR1 + no Kress et al. (2000); Ye (2016)
+ no+
FGFR2 + yes/no Johnson, Wall, Mann, and Wilkie (2000); Weber et al. (2001);
Wilkie et al. (2010)
+ yes/no
+ yes/no
FGFR3 + yes/no Wilkie et al. (2010)
FLNA + + Fennell et al. (2015)
FREM1 + no Vissers et al. (2011)
IGF1R + + no Cunningham et al. (2011)
MSX2 + + yes Janssen et al. (2013)
PTH2R + + yes Kim et al. (2015)
RAB23 + yes Jenkins et al. (2007)
RUNX2 + no+ Mefford et al. (2010)
SIX2 + yes Hufnagel et al. (2016)
SMAD6 + + + no Timberlake et al. (2016)
SMURF1 + no Timberlake et al. (2016)
SPRY1 + no Timberlake et al. (2016)
SPRY4 + no Timberlake et al. (2016)
TCF12 + yes/no Di Rocco et al. (2014); Le Tanno et al. (2014); Paumard-
Hernandez et al. (2015); Piard et al. (2015); Sharma et al.
(2013)
+ yes
+ yes
+ yes
+ yes
TWIST1 + + no Ko, Jeong, Yang, Park, and Yoon (2012); Sauerhammer et al.
(2014);Seto et al. (2007); Tahiri et al., (2015); Ye et al.
(2016)
+ yes +
+ no +
+ no
+ no
ZIC1 + yes Twigg et al. (2015)

no+, only minor syndromic features present; yes+, syndromic in most cases, rare NCS cases are described; yes/no, both syndromic and NSC cases are
described.
1412 | LATTANZI ET AL.

Given that the genetic component is believed to be suture-specific single patient (out of 70) with a truly nonsyndromic form of sagittal
(Zeiger et al., 2002), here, we will attempt to categorize all recent craniosynostosis.
molecular genetic findings in NCS, including simple/complex (multi- The mutations in the above genes account for approximately 10%
suture) NCS phenotypes with subtle associated features that are not of patients with sNCS, and appear to have incomplete penetrance
yet classified as syndromes. Table 2 provides a detailed list of genes confirming the perception that NCS is a multifactorial or oligogenic
involved in different suture closure patterns in such cases. disorder with considerable genetic heterogeneity.

3.2.1 | Sagittal NCS 3.2.2 | Metopic NCS


Sagittal NCS (sNCS) occurs in 1 in 5,000 live births, being the most The metopic NCS (mNCS) incidence is reported to range between 1/
prevalent NCS. The genetic causes in the majority of patients remain 7,000 and 1/15,000 births, according to recent updates (van der
unknown. Our group has completed the first genome wide association Meulen, 2012) and seems to have a stronger genetic component
study of 130 patient-parent trios with sagittal NCS and identified compared to sNCS. Familiar recurrence has been described in up to
strong and reproducible associations with BMP2 and BBS9 (Justice 10% of patients and the incidence rate of the phenotype observed in
et al., 2012). Preliminary data suggest that BMP2 is upregulated due to first-degree relatives is 6.4%, a rate higher than in any other NCS
a putative enhancer effect of the associated region (Justice, 2014). (Greenwood et al., 2014).
Rare SMAD6 mutations in combination with the risk C allele of Cytogenetic abnormalities are relatively frequent among the
rs1884302 near BMP2 were identified in 2.7% (3 of 113) of patients causes of metopic CS, and are believed to occur in at least 6% of the
with NCS (Timberlake et al., 2016). The same authors found SPRY1 and patients (Kini et al., 2010). Since these rearrangements can be subtle, a
SPRY4 mutations in one sporadic and one familial occurance of sNCS, chromosomal microarray screening is indicated for patients with
respectively. metopic CS and developmental delays.
Mutation screening of FGFR1, FGFR2, FGFR3, or TWIST1 has Conversely, gene mutations are rare in mNCS. SMAD6 mutations
identified causative mutations in rare cases (Boyadjiev et al., 2002; were identified in 12.8% (10 of 78) of the probands with single suture
Zeiger et al., 2002). Weber et al. (2001) identified a c.943G>A or complex mNCS and seven of the probands also carried the risk C
transition leading to missense A315T mutation in FGFR2 in one out of allele of rs1884302 near BMP2. Seven of the eight unaffected parents
29 patients (3%) with sNCS. A point mutation in the C-terminal domain in the kindreds who carried SMAD6 mutations were homozygous for
of TWIST1 has been found in one of 83 patients (1.2%) with sagittal the common T allele of rs1884302 and the only affected parent carried
synostosis and minor associated dysmorphisms (Seto et al., 2007). The the risk C allele. These facts were interpreted as evidence for a two-
same sequence variant was previously interpreted as a polymorphism, loci inheritance of midline NCS (Timberlake et al., 2016). An unusual
rather than a disease-causing mutation (Kress et al., 2006). Very FGFR1 I300W mutation has been reported in a girl with mNCS and
recently Ye et al. (2016) identified two rare variants in a selected facial skin tags (Kress, Petersen, Collmann, & Grimm, 2000). The
cohort of 93 sNCS cases: a FGFR1 insertion c.730_731insG, which led mutation was not present in the mother DNA (the father could not be
to a premature stop codon, and a c.439C>G variant in TWIST1. The tested) nor in 300 control chromosomes (Kress et al., 2000). Recently,
latter variant was found in the patient‘s mother presenting with jaw FREM1was suggested as a candidate gene for mNCS. FREM1 is
abnormalities. expressed in intrasutural mesenchyme and may play a key role in
Gain-of-function variants in the coding region of ALX4, account maintaining suture patency. FREM1 mutations were identified in eight
for about 1.5% of sNCS patientes (Yagnik et al., 2012). Since these out of 104 (5.8%) patients with metopic CS, including two patients
ALX4 variants were also found in an unaffected parent, they may with mNCS (Vissers et al., 2011). Finally, premature closure of the
represent low penetrance mutations that, as is the case of SMAD6 sagittal and metopic sutures has been found associated with hearing
(Timberlake et al., 2016), require predisposing variants at a second impairment, crumpled ear, widely spaced eyes, and developmental
locus. The incomplete penetrance of MSX2 mutations, leading to an delay in the syndromic phenotype associated with an intronic
extremely variable expression of the Boston-type CS, may also breakpoint in PTH2R (Kim et al., 2015).
explain selected cases of isolated sagittal and/or unicoronal
synostosis (Janssen et al., 2013). By sequencing 27 candidate genes 3.2.3 | Coronal NCS
in 186 patients with sNCS, Cunningham and colleagues found five Coronal NCS (cNCS; incidence 1:10,000 live births) is thought to have
missense mutations in the insulin-like growth factor 1 receptor a stonger genetic component compared to the other forms of NCS
(IGF1R), a gene involved in calvarial suture development (Al-Rekabi (Lajeunie et al., 1995; Wilkie et al., 2010) with specific monogenic
et al., 2016; Cunningham et al., 2011). In particular, distinct private mutations present in about 60% of patients with bicoronal and 30% of
variants were identified in two out of 94 (2%) patients with sagittal patients with unicoronal CS (Twigg et al., 2015). Indeed, diverse genes
involvement, although in one patient the same mutation was found may account for the higher incidence of monogenic variants in cNCS.
in the unaffected mother, indicating possibly incomplete penetrance These include FGFR3, TWIST1, EFNB1, and TCF12, which were also
(Cunningham et al., 2011). More recently, Twigg et al. (2013) found associated with syndromic forms of craniosynostosis (Twigg & Wilkie,
mutations in ERF by exome sequencing of 412 unrelated CS patients. 2015).
Although the mutations were mainly associated with either The P250R substitution in FGFR3 was initially thought to cause
syndromic or complex NCS, a mutation was also described in a cNCS but was later defined as the cause of Muenke syndrome
LATTANZI ET AL.
| 1413

(Muenke et al., 1997). It represents the most frequent genetic anomaly the presence of intrauterine constraint. More recently, new genes
observed in all CS patients (5.6% tested patients; 24% of all causative have been associated with cNCS. Rare IGF1R variants have been found
mutations (Wilkie et al., 2010)). Lajeunie et al. (1999) observed this in three out of 46 (6.5%) cNCS patients. Although considered to be
mutation in 17% of patients with sporadic and 74% of patients with “nonsyndromic,” all patients (with both sagittal and coronal involve-
familial cCS. Due to the phenotype variability of Muenke syndrome, ment) with IGF1R variants in this study had reduced scores
detailed clinical examination and molecular testing is mandatory for all on neuropsychometric testing and/or subtle cognitive delay
patients diagnosed with coronal CS, even in apparently nonsyndromic (Cunningham et al., 2011).
forms. Emphasizing the importance of molecular testing is the fact that Zinc finger protein of cerebellum 1 (ZIC1) belongs to a family of
surgical outcomes and prognosis of patients undergoing surgery for Zn-finger transcription factors that, in vertebrates, play crucial roles in
coronal CS is significantly and specifically affected by the presence of vertebrate neurogenesis, left–right axis formation, myogenesis, and
the P250R mutation (Wilkie et al., 2010). skeletal patterning (Aruga et al., 2006; Merzdorf, 2007). A heterozy-
Seto et al. (2007) found a c.556G>T (p.Ala186Thr) substitution in gous mutation in the last exon of ZIC1 was identified by exome
TWIST1 in a patient diagnosed with unicoronal right cNCS who also sequencing in a proband with severe bicoronal CS, autism and
had mild nonspecific dysmorphic features. The mutation was developmental delay (Twigg et al., 2015). In order to clarify the
considered pathogenic as it falls within the highly evolutionary significance of ZIC1 in the etiology of CS, the authors screened a
conserved TWIST Box, which is needed for inhibition of RUNX2 cohort of 307 patients with various types of syndromic and
transactivation (Seto et al., 2007). The same authors also identified an nonsyndromic CS and identified four additional probands with
18 bp deletion in the polyglycine tract of TWIST1, interpreted as a rare bicoronal CS who had mutations in the last exon of ZIC1. Three of
sequence variant with unclear pathogenetic significance (Seto et al., the affected individuals presented with bony defects of the sutures or
2007). TWIST1 mutations were found in six out of 43 Korean patients the skull, and various brain anomalies were identified in four of the five
(14%) with uni/bicoronal CS, including a single nonsyndromic patient probands. Because developmental disabilities and associated clinical
(2.3%) (Ko, Jeong, Yang, Park, & Yoon, 2012). As a further confirmation features were present, the CS of these patients cannot be defined as
of the somewhat specific role of TWIST1 in coronal suture patterning, nonsyndromic. Given the variable clinical severity in one multiplex
Twist± mutant mice exhibit a brachycephalic skull consistent with the family in this study, ZIC1 testing should be considered in all cCS
involvement of the coronal suture in craniofacial dysmorphism patients in whom mutations in commonly associated genes have been
(Parsons et al., 2014). Based on these facts TWIST1 mutational excluded (Twigg et al., 2015). In the Xenopus embryo, Zic1 expression
analysis should be considered for patients with coronal CS even in the overlaps with that of Engrailed 1 (En1) in the supraorbital region that
absence of clear syndromic traits. directs coronal suture development and patterning, which is plausibly
A recent exome sequencing study identified heterozygous disrupted as a consequence of ZIC1 mutations (Twigg et al., 2015). ZIC
mutations in TCF12, which is functionally related to TWIST1 and orthologues in Drosophila and Xenopus are upstream regulators of the
plays a critical role in coronal suture development (Sharma et al., Wnt pathway (Merzdorf & Sive, 2006), being required for the
2013). In particular, TCF12 mutations were found in 10% of patients activation of expression of the engrailed segment polarity gene
with unicoronal and 37% of patients with bicoronal CS. Although (Benedyk, Mullen, & DiNardo, 1994; Kuo et al., 1998). In mice, En1 is
81% of the individuals with TCF12 mutations presented with expressed in cells that migrate from the paraxial cephalic mesoderm to
apparent cNCS, some of the affected individuals had developmental form and populate the coronal suture (Deckelbaum et al., 2012).
delays and dysmorphic features overlapping with the Saethre– Indeed, mice with homozygous loss of En1 function have abnormal
Chotzen syndrome (Sharma et al., 2013). More recently, the coronal suture fusion and an overall malformed skull (Deckelbaum,
phenotypic spectrum of TCF12 mutations has been extended to Majithia, Booker, Henderson, & Loomis, 2006).
include possible facial and limb anomalies, and intellectual disability
(Di Rocco et al., 2014; Le Tanno et al., 2014; Paumard-Hernández 3.2.4 | Lambdoid NCS
et al., 2015; Piard et al., 2015). Based on these observations, it is Lambdoid CS (lNCS) is the least common type (1 in 33,000 live births),
reasonable to recommend TCF12 testing for all patients with cCS, representing 1% of all CS and 3% of NCS (Boyadjiev & International
with or without associated anomalies. Craniosynostosis Consortium, 2007; Greenwood et al., 2014; Heuzé
As already mentioned above, due to the variable expression and et al., 2014; Wilkie et al., 2010). Posterior plagiocephaly due to true
incomplete penetrance of MSX2 mutations (associated with the lNCS must be differentiated from the much more common positional
Boston-type craniosynostosis syndrome), MSX2 testing should be also molding (Rhodes, Tye, & Fearon, 2014). The synostosis may be
considered for cCS patients with a positive family history, even if the unilateral or bilateral, and occurs more frequently as part of a complex
anomaly appears to be nonsyndromic (Janssen et al., 2013). CS (Rhodes et al., 2014).
A novel FGFR2 A315S mutation was reported in a patient with The genetic basis of simple lNCS is unknown. Familial
right unicoronal NCS born following a pregnancy complicated by recurrence has been reported in two patients (Fryburg, Hwang, &
breech presentation and skull compression (Johnson, Wall, Mann, & Lin, 1995; Kadlub, Persing, da Silva Freitas, & Shin, 2008), and the
Wilkie, 2000). Both the patient‘s mother and maternal grandfather reported incidence of CS in first-degree relatives is 3.9% (Green-
carried the mutation and had mild facial asymmetry but no CS, wood et al., 2014). Chromosomal rearrangements have been rarely
suggesting that the mutation predisposes individuals to synostosis in described (Odell, Siebert, Bradley, & Salk, 1987; Park, Graham, Berg,
1414 | LATTANZI ET AL.

& Wurster-Hill, 1988). More recently, Twigg et al. (2013) identified a this was not confirmed in further studies (Bermejo et al., 2005). Rhodes
c.256C>T (p.Arg86Cys) mutation in ERF, associated with reduced and colleagues reported the occurrence of this complex pattern in 11
gene expression, in one out of 13 (7.6%) patients with either out of 802 CS patients (1.4%), characterized by phenotypic variablity
unilateral, bilateral, or complex lambdoid synostosis. This patient (Rhodes, Kolar, & Fearon, 2010). In particular, the exclusive fusion of
was a member of an affected family in which the mutation was sagittal and lambdoid sutures was observed only in 8 of the 11
associated with different clinical expressions. Rice et al. (2010) patients, reducing the estimated prevalence to less than 1% in the CS
described that mice with a Gli3-null allele exhibited lambdoid CS due cohort while two patients were later diagnosed as having Opitz and
to the increased proliferation and differentiation of osteoprogenitor Potocki–Shaffer syndromes. BLSS with syndromic phenotype has
cells in the calvarial mesenchyme. This study defined the first been associated with chromosome abnormalities, involving the loci of
available animal model of lambdoid CS and confirmed the role of the putative genes regulating skull vault development such as MSX2 on 5q
Hedgehog (HH)-signaling pathway in membranous ossification of (Shiihara et al., 2004) and SOX6 on 11p (Tagariello et al., 2006). Hing
the skull. et al. (2009) described the largest series of 11 patients with BLSS
diagnosed among 701 patients with various CS types. No mutations
3.2.5 | Complex NCS were identified in TWIST1, MSX2, and SOX6 and in the selected hot-
Complex CS (cxCS) presents with more than one fused suture and spot regions of FGFR1, FGFR2, and FGFR3. An Xp11.22 deletion was
causes more severe distortion of the skull. Fusion of multiple sutures is documented in one of five tested individuals, who had spasticity,
more difficult to explain by intra-uterine constraint and is more likely progressive apnea, and seizures and died at 4 months. A patient with
to result from an abnormal genetic background. Although the complex CS involving metopic and bicoronal sutures was reported to
involvement of multiple sutures can occur even in the absence of a have a heterozygous 9.0 Mb deletion in the chromosomal region
clearly recognizable phenotype, the chances of a syndromic diagnosis 7p21.1-p21.3 (Di Rocco et al., 2015).
are indeed higher among this group of patients (Cohen, 2000). CxNCS
accounted for 5.5% in a cohort of 144 patients with NCS (Wilkie et al., 3.3 | Genes and developmental pathways as related
2010), and ranged from 3% to 7.5 %, in more heterogeneous cohorts of
to CS
200 (Bessenyei et al., 2015) and 518 (Greene, Mulliken, Proctor,
Meara, & Rogers, 2008) CS patients, respectively. Within the cohort The following section will provide a more detailed description of the
recruited through the International Craniosynostosis Consortium complex molecular networks integrating the genes that have been

(https://genetics.ucdmc.ucdavis.edu/icc.cfm), 70 out of 660 (11%) associated with CS. The extreme genetic heterogeneity of CS reflects
NCS patients had cxNCS (Greenwood et al., 2014). Mutations in CS- the complex molecular networks underlying head morphogenesis,
associated genes may result in atypical phenotypes, often misdiag- which comprises concerted skull/brain development. A diagram
nosed as unusual cxNCS. In particular, pathogenic FGFR2 mutations schematizing known interactions among the main CS-associated
have been found in patients with apparently cxNCS (Wilkie et al., molecular pathways is proposed in Figure 2.
2010). SMAD6 mutations were identified in two of eight (25%) The bone morphogenetic protein (BMP)/transforming growth
probands with complex CS of both metopic and sagittal sutures from factor beta (TGFβ), fibroblast growth factor (FGF), hedgehog (HH),

multi-generational families. Both probands and one of the affected eph-ephrin, and wingless-type MMTV integration site family (WNT)
parents carried the risk C allele of rs1884302, while an unaffected signaling pathways are the best understood developmental cascades
parent with SMAD6 mutation did not, a fact strengthening the governing the development of the skull. Individual genes within these

proposed two-loci inheritance for at least some patients with NCS. pathways have been implicated in the etiology of CS, both in humans
There have also been reported patients with cxNCS involving sagittal, and animal models (Snider & Mishina, 2014). Interestingly, most of
metopic, and coronal sutures who were found to have TWIST1 these pathways are known to play pivotal roles in neural crest
mutations. Mutations in TWIST1 also cause Saethre–Chotzen syn- development, hence driving the morphogenesis of the craniofacial
drome (Sauerhammer et al., 2014; Tahiri et al., 2015). Therefore, complex (Mishina & Snider, 2014; Twigg & Wilkie, 2015).
genetic testing of SMAD6, FGFR1, 2, 3, and TWIST1 is indicated for
individuals with cxNCS, and TCF12 and ZIC1 sequencing should be 3.3.1 | Homeoproteins and homeotic genes
considered when mutations in other genes have been excluded. The msh homeobox 2 (MSX2) was the first CS causing gene discovered
A characteristic head shape associated with bilateral lambdoid and more than 20 years ago when a gain-of-function missense mutation was
sagittal synostosis (BLSS) known as cranial dyssynostosis (OMIM found in a family with syndromic Boston type CS (Jabs et al., 1993). A
#218350) presents with frontal bossing, turribrachycephaly, biparietal second MSX2 mutation was described in an unrelated family (Florisson
narrowing, occipital concavity, inferior ear displacement, and frequent et al., 2013). MSX2 is a homeodomain transcription factor expressed in
developmental delay (Hing et al., 2009; Moore, Abbott, Netherway, the developing skull. In particular, MSX2 is primarily expressed in the
Menard, & Hanieh, 1998). The resulting posterior vault constraint neural crest which gives rise to frontal and craniofacial bones (Han et al.,
often leads to Chiari I malformation, and requires surgical cranial 2007), acting as a master regulator of gene expression in the embryo
remodeling (Shukla, 2016). In the original description of the syndrome, (Attanasio et al., 2013; Khadka, Luo, & Sargent, 2006). Skull
Neuhauser and colleagues (Neuhäuser, Kaveggia, & Opitz, 1976) development is indeed tightly regulated by MSX2 dosage: gain-of-
suggested an autosomal recessive inheritance within a family, though function (GoF) mutations and copy number increase, due to 5q35
LATTANZI ET AL.
| 1415

Takahashi, 2013; Kayserili et al., 2009). Similarly to MSX2, GoF mutations


of ALX4 cause CS (specifically sagittal NCS) (Yagnik et al., 2012), while LoF
mutations result in parietal foramina (Mavrogiannis et al., 2001).
SIX2, another member of the aristaless gene family, was recently
found deleted in patients with syndromic complex or sagittal
synostosis (Hufnagel et al., 2016). This gene is functionally involved
in specifying the proliferation and migratory properties of neural crest
cells during early embryo development. Along with other SIX genes, it
is required for correct brain development and morphogenesis of the
frontal craniofacial skeleton, counteracting HoxA2 function in mice
(Garcez, Le Douarin, & Creuzet, 2014). Interestingly, Six1-2-4
inactivation affects Bmp signaling through the downregulation of
Bmp antagonists in facial neural crest cells.
ZIC1 is among the latest identified CS-associated genes and is
discussed in Section 3.2.3. This gene acts through the regulation of
engrailed 1 (En1) expression in the embryo (Twigg et al., 2015). This
F I G UR E 2 Craniosynostosis gene network. The diagram shows homeoprotein is expressed in the skeletogenic mesenchyme and
known and predicted interactions among candidate genes drives membranous calvarial ossification by regulating FGF signaling in
implicated in the etiopathogenesis of craniosynostosis, discussed in calvarial osteoblasts (Deckelbaum et al., 2006).
the different sections (see text for details). The network was drawn
using String (version 10.0) license-free software (http://string-db.
3.3.2 | BMP/TGFβ pathway
org/), using the “molecular action” display format. Rounded nodes
refer to gene/protein symbol included in the query; small nodes are Both MSX2 and ALX4 are targets of the BMP/TGFβ pathway (Ishii
for proteins with unknown 3D structure, while large nodes are for et al., 2003; Rice, Olsen, & Thesleff, 2003). This is consistent with
those with known structures. The lines connecting nodes represent our observation of a strong association of BMP2 with sNCS (Justice
the most relevant and best characterized types of protein-protein
et al., 2012) with evidence of increased expression of this gene
association within the networking genes, according to the following
(Justice, 2014). The identification of SMAD6 mutations acting in
color codes: green = activation, red = inhibition, blue = binding, violet
= catalysis, pink = post-translation modification, black = reaction, concert with the risk C allele of rs1884302 further corroborates the
yellow = transcriptional regulation. Grey lines symbolize predicted importance of this pathway in the etiology of NCS. SMURF1 LoF
links based on literature search (i.e., genes/proteins are co- mutation was described in a case of metopic craniosynsotosis
mentioned in PubMed abstracts). Thicker lines represent stronger (Timberlake et al., 2016). Both SMAD6 and SMURF1 mutations
associations between proteins. The line ending shape represents the
appear to enhance osteoblast differentiation through unopposed
effect (whenever applicable) of the molecular action: arrow
end = positive, transverse line end = negative, ball end = unspecified. expression of RUNX2. Interestingly, BMP2 has been also associated
The interactions were automatically integrated in the network by with osteoporosis (Styrkarsdottir et al., 2003), and brachydactyly
the software, with the medium confidence value set at 0.0400 (i.e., type A2 (Dathe et al., 2009) suggesting that, as is the case with MXS2
a 40% probability that a predicted link exists between two enzymes and ALX4, the BMP2 GoF mutation may be a factor in CS while
in the same metabolic map in the KEGG database: http://www.
decreased expression results in the opposite phenotype of skeletal
genome.jp/kegg/pathway.html). Additional interactions among ERF,
under-development. Also, the BMP2-related intracellular osteogenic
RUNX2, FGFR1-3, GLI3, and BMP2, were added manually, given
their functional implication in the molecular background described cascade is activated in calvarial cells isolated from prematurely fused
in this review sutures of affected individuals (Lattanzi et al., 2013). The importance
of the BMP pathway in the pathogenesis of CS is further
corroborated by studies of animal models: A derived F452L
trisomy, result in CS while MSX2 loss of function (LoF) results in parietal missense mutation in BMP3 is nearly fixed among small, brachyce-
foramina (i.e., deficit of skull ossification) (Kariminejad et al., 2009; phalic dog breeds and bmp3 is required for zebrafish craniofacial
Shiihara et al., 2004; Spruijt, Verdyck, Van Hul, Wuyts, & de Die- development (Schoenebeck et al., 2012). Mice with loss of BMP13
Smulders, 2005; Wilkie et al., 2000). MSX2 is thought to act upstream of (also known as growth differentiation factor 6, Gdf6,) have coronal
the FGF pathway and appears to stimulate proliferation while inhibiting CS (Clendenning & Mortlock, 2012), while metopic CS is observed in
differentiation of osteogenic precursors in the suture. It was proposed mice with enhanced BMP signaling through constitutive activation
that GoF mutations in the gene delay osteogenic differentiation and of bmp receptor, type 1A (Bmpr1a) expression in cranial neural crest-
increase the pool of proliferating osteogenic cells that ultimately derived structures (Komatsu et al., 2013). In these models,
orchestrate suture ossification (Liu et al., 1999). craniosynostosis is believed to originate from an upregulation of
Aristaless-like homeobox 4 (ALX4), a homeodomain transcription Smad-dependent BMP signaling in the neural crest, resulting in
factor involved in osteoblast regulation, is closely related to MSX2. It is enhanced ossification of the derived cranial bones (Komatsu et al.,
required for neural crest cell migration and differentiation; hence, playing 2013). Furthermore, there are observations that recombinant human
a critical role in craniofacial development (Dee, Szymoniuk, Mills, & BMP2 accelerates sutural bone formation in rabbits (Liu, Opperman,
1416 | LATTANZI ET AL.

& Buschang, 2009), whereas the BMP-antagonist Noggin, a (Kim et al., 2003; Park et al., 2012; Wang et al., 2012). Importantly,
candidate gene for nonsyndromic cleft lip/palate (Leslie et al., TGFβ-2 also activates the Erk-MAPK (Lee et al., 2006) cascade,
2015), inhibits postoperative resynostosis in a mouse suturectomy suggesting that HH, FGF, and BMP/TGFβ pathways may all converge
model (Cooper et al., 2009). on Erk1/2 or its downstream effectors, including the osteogenic
Deregulation of TGFβ isoforms was observed in the sutural complex master gene runt-related transcription factor (RUNX2). Our group,
of a rabbit CS model (Poisson et al., 2004). These isoforms appear to however, did not find enhanced ERK1/2 phosphorylation in primary
regulate suture patency and fusion through phosphorylation of the osteoblasts from patients with sagittal NCS (Kim, Yagnik, Cunningham,
mitogen-activated protein kinase (Erk1/2, ERK-MAPK cascade) and Kim, & Boyadjiev, 2014) suggesting an alternative causative pathway
SMAD family member two (Smad2) proteins; thus, integrating the action or genetic alteration in MAPK/ERK components downstream of
of both the FGF and BMP signaling pathways (Opperman, Fernandez, So, ERK1/2. ERF encodes the Ets2 repressor transcription factor
& Rawlins, 2006). The importance of BMP/TGFβ signaling in the expressed in migratory cells of the neural crest that act downstream
pathogenesis of CS is also underscored by the frequent occurrence of this of Erk1/2 (Paratore, Brugnoli, Lee, Suter, & Sommer, 2002). Indeed, it
finding in Loeys–Dietz syndrome, caused by TGFβ receptor-1 and -2 was recently documented that ERF mutations cause coronal and
(TGFBR1, TGFBR2), SMAD3, and TGFB2 mutations (Loeys & Dietz, 1993), complex syndromic CS (Twigg et al., 2013).
and in Shprintzen–Goldberg syndrome, caused by mutations in SKI proto-
oncogene (SKI), a repressor of TGFβ (Doyle et al., 2012). 3.3.5 | Ephrin genes
Eph-ephrin is another important developmental pathway that includes
3.3.3 | TWIST1 gene and related signaling
at least 14 tyrosine kinase receptors and eight ligands (Lisabeth,
TWIST1 forms heterodimers with TCF12, a critical process in the
Falivelli, & Pasquale, 2013). This signaling pathway plays a critical role
molecular control of coronal suture development (Connerney et al.,
in the formation of tissue boundaries in vertebrates. Two members of
2006). Indeed, haploinsufficiency of either of these genes has been
the Eph-ephrin signaling pathway have been implicated in CS. LoF
associated with coronal CS and Saethre–Chotzen phenotype (Sharma
mutations of ephrin-B1 (EFNB1) cause craniofrontonasal syndrome, an
et al., 2013). TCF12 mutations may exert their pathogenic effect
X-linked developmental disorder with features of frontonasal dyspla-
through either FGF, BMP, or HH signalling, given the pleiotropic role of
sia and predominantly coronal CS (Twigg et al., 2004). These authors
TWIST1 in these pathways (Hornik, Brand-Saberi, Rudloff, Christ, &
proposed that, at least in females, disturbed tissue boundary formation
Füchtbauer, 2004). TCF12 also functions as a SMAD cofactor, furhter
at the developing coronal suture is responsible for the phenotype.
implicating that the BMP/TGFβ pathway is the etiopathogenesis of CS
Ephrin-A4 (EFNA4) was identified as a potential target for CS through
(Yoon, Wills, Chuong, Gupta, & Baker, 2011).
studies of Twist1± mice strains exhibiting coronal CS due to defective
fronto-parietal boundary. Enhanced Msx2 and reduced Epha4
3.3.4 | The fibroblast growth factor pathway
expression were observed and suppression of the Msx2 in Msx2±;
The fibroblast growth factor (FGF) pathway is one of the best
Twist1± double-mutant mice restored Epha4 levels and rescued the CS
understood signaling systems associated with CS etiopathogenesis. It
phenotype. Indeed, loss-of-function EFNA4 mutations were identified
comprises at least 23 ligands (FGF1-FGF23) and five receptors
in three of 81 individuals with cNCS (Merrill et al., 2006). Importantly,
(FGFR1-FGFR5), essential for the initial stages of skeletal develop-
these studies demonstrate that at least part of the Eph-ephrin signaling
ment. GoF mutations in either FGFR1, FGFR2, and FGFR3 genes or LoF
system is integrated with the developmental pathways discussed
of the TWIST family transcription factor 1 (TWIST1), a repressor of the
above. This is corroborated by the fact that EphA4, through its
FGR signaling, account for approximately 20% of patients with
interaction with Twist1, regulates both Erk1/2 and the BMP pathway
syndromic CS (el Ghouzzi et al., 1997; Howard et al., 1997; Jabs et al.,
complex Smad1/5/8 (Ting et al., 2009).
1994; Meyers, Orlow, Munro, Przylepa, & Jabs, 1995; Muenke et al.,
1994, 1997; Przylepa et al., 1996; Reardon et al., 1994; Schell et al.,
1995; Wilkie et al., 1995). The complex CS syndromes (Saethre–
3.3.6 | Hedgehog and Wnt signaling: the primary cilium
Chotzen, Pfeiffer, Crouzon, Apert, Jackson–Weiss, Beare–Stevenson, environment in CS etiopathogenesis
Muenke, and Crouzon with acanthosis nigricans syndromes), caused HH and BMPs are both members of a complex signaling pathway that
by more than 60 mutations in these genes, are well defined and not the regulates early patterning events in fetal skeletogenesis (Iwasaki, Le, &
subjects of this review. The interested reader is referred to several Helms, 1997). The role of HH signaling in the etiology of CS was
comprehensive reviews of the molecular and clinical characteristics of unveiled by the identification of LoF mutations in member RAS
these syndromes (Jabs, 1998; Johnson & Wilkie, 2011; Passos-Bueno oncogene family 23 (RAB23) gene in patients with Carpenter
et al., 2008; Wilkie, 1997). syndrome (Jenkins et al., 2007). RAB23 is a repressor of HH signaling,
The majority of the FGFR mutations result in constitutive which in turn induces members of the BMP gene family (Bitgood &
activation of the receptor due to ligand-independent dimerization McMahon, 1995). Mutations in GLI family zinc finger 3 (GLI3), a
and activation, and/or changes in ligand specificity. downstream mediator of the Sonic HH, cause several multiple
The common thread of the syndromes caused by perturbations of congenital anomaly syndromes and has been associated with metopic
FGF signaling is the excessive activation of the downstream Ras/ CS with polydactyly (McDonald-McGinn et al., 2010). Indian HH is a
MAPK pathway, with enhanced phosphorylation of Erk1/2 kinases pro-osteogenic factor that positively regulates intramembranous
LATTANZI ET AL.
| 1417

calvarial ossification by modulating BMP signaling (Lenton et al., 2011). and development (Chang, Petersen, Niswander, & Liu, 2015). Indeed,
Interestingly, in a pattern analogous of BMP2 and BMP14 (Plöger et al., a craniofacial phenotype is observed in the ciliopathic Fuz mutant
2008), heterozygous missense mutations in Indian HH also result in mouse due to excessive neural crest, leading to maxillary hyperplasia
brachydactyly (Ma et al., 2011). (Tabler et al., 2013). Fuz is essential for normal trafficking inside
The Wnt pathway promotes osteoblast differentiation and function. vertebrate cilia, and closely interact with hedgehog signaling.
FGFR2-activating mutations downregulate a number of Wnt target genes Interestingly, the authors identified dysregulation of Fgf8 as the
(Mansukhani, Ambrosetti, Holmes, Cornivelli, & Basilico, 2005). Mice with cause of the craniofacial defect observed in this ciliopathic mutant
targeted disruption of the canonical Wnt inhibitor axis inhibition protein model, suggesting a shared pathogenic event between ciliopathies
(Axin2) develop malformations of skull structures, a phenotype resembling and craniofacial syndromes due to overactive FGF signaling (Tabler
craniosynostosis in humans. Axin2 is needed for neural crest-derived et al., 2013).
frontal bone development (Li et al., 2015; Yu et al., 2005). Indeed, studies of
the Axin2−/− mice provided additional evidence for the interactions 3.3.7 | Interleukins
between the Wnt, BMP, and FGF pathways in calvarial morphogenesis.
The interleukins IL-1 and IL-6 are involved in the regulation of bone
Axin2 null mice displayed enhanced β-catenin activation, with expansion
homeostasis and bone cell functions. The initial indications that
of osteoprogenitor cells, and activation of the BMP pathway, suggested by
interleukins (ILs) may be implicated in the pathophysiology of the skull
increased SMAD1/5/8 phosphorylation (Yan et al., 2009; Yu et al., 2005).
malformations came from observations that these molecules modulate
Even though the precise mechanisms integrating these signaling systems
the production of extracellular matrix molecules in periosteal
are not well understood, it appears that β-catenin induces the BMP
fibroblasts of patients with Apert syndrome (Bodo et al., 1997). The
signaling, through FGF, by inhibiting its antagonist Noggin (Warren,
direct evidence that IL defects can cause CS came from the description
Brunet, Harland, Economides, & Longaker, 2003), thus promoting
of a novel autosomal recessive CS syndrome due to homozygous
osteoblast differentiation while canonical Wnt signaling suppresses the
mutations of the interleukin receptor alpha gene (IL11RA) (Nieminen
FGF pathway by inducing TWIST1 (Reinhold, Kapadia, Liao, & Naski, 2006).
et al., 2011). Since IL11RA and its ligand IL11 support both osteoblast
Our group observed strong and reproducible association of
and osteoclast differentiation it remains unclear if the CS occurs due to
sagittal CS with the Bardet–Biedl syndrome associated gene-9
stimulation of osteoblast differentiation or inhibition of osteoclast
(BBS9), named after the eponymous ciliopathy (Justice et al., 2012).
function causing a defect of bone resorption. The integration of IL and
We have also observed significant BBS9 upregulation during in vitro
BMP signaling is supported by their synergistic effect in enhancing
osteogenic induction of calvarial osteoprogenitors, isolated from CS
bone formation in a rat model (Suga et al., 2003). The overlapping
patients, and during the physiologic ossification of rat sutures
phenotype of supernumerary teeth in patients with IL11 and RUNX2
(Lattanzi, unpublished data), suggesting the involvement of this gene
mutations and mice with abnormal Wnt signaling (Wang et al., 2009)
in calvarial suture morphogenesis. Considering the lack of a
suggests that these genes may interact within the Wnt developmental
reproducible craniofacial phenotype occurring in the best known
pathway.
ciliopathies, the view of CS as a manifestation of a ciliopathy may be
While enhanced osteoblast differentiation is the generally
unexpected. Nonetheless, sagittal CS is present in at least 50% of
accepted mechanism of premature sutural fusion, it is plausible
the patients with Sensenbrenner syndrome, a cranioectodermal
that inhibited osteoclast function with deficient bone resorption is at
dyplasia caused by mutations in four genes involved in ciliary
least partially responsible for this phenotype. Interleukin 11
biogenesis and function (Arts et al., 2011). In addition to this
receptor, alpha (Il11ra) deficient mice have a cell-autonomous
example, other syndromes that may include CS phenotypes are
inhibition of osteoclast differentiation resulting in CS (Nieminen
those associated with mutations in ciliary genes: Mainzer–Saldino
et al., 2011); another mice model with increased β-catenin has
(mutations in IFT140), Carpenter (mutations in RAB23), and
massively increased bone mass, and a decreased number of
cranioectodermal dysplasias 2 and 4 (mutations in WDR35 and
osteoclasts, but normal number of osteoblasts (Cohen, 2006); and
WDR19, respectively). Furthermore, the phenotype of mice deficient
EphA4 receptor is a documented suppressor of osteoclast activity
of kinesin family member 3A (Kif3a), a protein needed for cilia
(Stiffel, Amoui, Sheng, Mohan, & Lau, 2014).
formation, includes metopic CS with other midline anomalies (Liu,
Chen, Johnson, & Helms, 2014). In this model, the cranial neural
crest cells in which Kif3a was conditionally deleted showed evidence
3.3.8 | Other signaling pathways involved in CS
of hyper-proliferation and ectopic Wnt responsiveness, linking this etiopathogenesis
ciliopathy to the signaling pathways discussed above. Based on these observations, one can safely predict that variants in
Besides direct inference from human CS phenotypes and animal other members of the signaling pathways discussed above will be
models, it is worth noting that most of the key signaling pathways associated with CS. Nevertheless, genes that are not directly related
involved in craniofacial morphogenesis and CS molecular pathogen- to these pathways have been associated with CS phenotypes.
esis are somehow featured in the intense signaling and trafficking Antley–Bixler syndrome manifests with CS, long bone fractures,
that occurs inside the primary cilium. In particular, Wnt and HH elbow synostosis, and other anomalies. Overlapping clinical features,
pathways are known to play pivotal role in cilium-related signaling. including coronal and lambdoid CS, are found in patients carrying
This is particularly relevant in the processes of neural crest induction null or hypomorphic mutations of CYP26B1 encoding a retinoic acid
1418 | LATTANZI ET AL.

(RA)-metabolizing enzyme (Laue et al., 2011; Morton, Frentz, duplication (Mefford et al., 2010), triplication (Varvagiannis et al.,
Morgan, Sutherland-Smith, & Robertson, 2016). RA signaling has 2013) and quadruplication (Greives et al., 2013) have been reported in
well documented effects in skeletal development; it induces patients with syndromic CS and there appears to be a direct correlation
premature osteoblast-to-preosteocyte transitioning during calvarial between the gene copy number and the severity of the phenotype.
morphogenesis (Jeradi & Hammerschmidt, 2016). In addition, an Our group identified two rare missense variants, M175R and R237C in
autosomal recessive phenotype resembling that of Antley–Bixler two of 137 patients with sNCS. A variant of RUNX2 that has been
syndrome, associated with coronal CS plus genital anomalies due to presumed neutral is an in-frame deletion of 18 base pairs in the QA
steroid biogenesis defects, is caused by cytochrome P450 oxidore- domain located in exon 2, resulting in a shorter stretch of 11 instead of
ductase (POR) mutations (Flück et al., 2004). The POR enzyme is also 17 alanine residues. We documented that the shorter RUNX2 isoform
involved in RA signaling, and POR-dependent cholesterol synthesis is overrepresented 10-fold among the patients with sagittal cranio-
is essential during limb and skeletal development (Ribes et al., 2007; synostosis as compared to controls: 24% versus 2.3%, respectively
Schmidt et al., 2009). It is worth mentioning that a similar phenotype, (Yagnik, Drissi, Stevens, & Boyadjiev, 2010). Importantly however,
without genital defects, has been attributed to heterozygous FGFR2 airorhynchy (dorsal bending of the snout) and midface length in
or FGFR3 mutations (Huang et al., 2005). Additionally, Pfeiffer brachycephalic dog breeds has been shown to correlate with
syndrome can feature elbow synostosis that may resemble that of polyglutamine and polyalanine repeat length of RUNX2 (Fondon &
Antley–Bixler syndrome (Hurley, White, Green, & Kelleher, 2004). Garner 2004). Early expression of Runx2 causes CS and other skeletal
DNA repair and transcription defects are responsible for disorders defects in mice (Maeno et al., 2011) while loss of Runx2 causes a
with significant clinical heterogeneity known as DNA helicase disorders complete lack of ossification of both intramembranous and endo-
(Suhasini & Brosh, 2013). Baller–Gerold syndrome, also known as CS- chondral bone (Komori et al., 1997). Osterix (Osx), a zinc finger-
radial defects syndrome, is caused by homozygous or compound containing transcription factor downstream of Runx2, is also necessary
heterozygous mutations in the recQ helicase-like (RECQL4) gene. for bone formation as no bone formation occurs in Osx null mice
RECQL4 mutations have been also described in patients with (Nakashima et al., 2002). NEL-like 1 (NELL-1) is another downstream
Rothmund–Thompson and RAPADILINO syndrome and all three mediator of RUNX and direct transcriptional target of OSX (Sp7
conditions share some clinical features (e.g., poikiloderma and radial transcription factor) that promotes osteoblastic differentiation
ray defects) (Van Maldergem et al., 2006). This has raised the possibility through activation of MAPK. NELL-1 is upregulated in the fused
that these syndromes are phenotypic variants of the same disorder and coronal suture in humans. Similar to Runx2, MSX2, and ALX4, Nell-1
not distinct disorders. The clinical phenotype of Baller–Gerold deficient mice present with deficient skull ossification (Desai et al.,
syndrome is complex and the differential diagnosis also includes VATER 2006) while over expression of Nell-1 cause CS in mice (Zhang et al.,
association, Roberts syndrome and Fanconi anemia. A patient with 2002).
radial aplasia and bicoronal synostosis without additional malformations Figure 2 synthesizes the network of molecular interactions among
(i.e., narrow definition of Baller–Gerold syndrome) was found to have a the best characterized CS candidate genes discussed in this paper; it is
nonsense TWIST1 mutation (Gripp et al., 1999) which causes Saethre– noteworthy that the main signaling pathways included in the picture
Chotzen syndrome. The contribution and significance of the DNA repair tend to merge on the downstream interaction with RUNX2, as
pathway to the pathogenesis of CS remains unclear at this time. previously discussed.
Finally, mutations in the insulin-like growth factor 1 receptor gene
(IGF1R) were found in three out of 186 patients with cNCS and sNCS
(Cunningham et al., 2011). IGF1 signaling plays an essential role during 4 | NEURODEVELOPMENTAL ASPECTS OF CS
skeletal development and regulation of cellular adhesion and migration.
Specifically, it is involved in cellular contractility and migration of suture Long-term assessment of neurobehavioral outcomes identified
calvarial osteoblasts of CS patients (Al-Rekabi et al., 2016). Interestingly, learning disabilities (most often language or visual perception
among the consequences of the phosphorylation of IGF1R targets, deficits) in 47% of affected school-aged children (Kapp-Simon,
RUNX2 and Wnt signaling are disinhibited, leading to the activation of the 1994) compared to 10% of unaffected children in the general
corresponding osteoblast differentiation cascades (Stamper et al., 2012). population (Altarac & Saroha, 2007). Another study showed that
30–50% of children with NCS may have neurocognitive deficits
(problems with attention and planning, processing speed, visual
3.3.9 | The pivotal role of RUNX2 spatial skills, language, reading, and spelling) that become more
However diverse, the BMP/TGFβ, FGF, HH, eph-ephrin, and WNT apparent as cognitive demands increase during school age
developmental pathways appear to share an important commonality: (Kapp-Simon, Speltz, Cunningham, Patel, & Tomita, 2007). Nearly
they all converge on the same downstream effector RUNX2 through 30% of children with metopic CS have some form of speech and/or
SMAD or ERK1/2 (Hanai et al., 1999; Lenton et al., 2011; Lisabeth language delay, regardless of whether they had surgical repair
et al., 2013; Suga et al., 2003; Xiao, Jiang, Gopalakrishnan, & (Kelleher et al., 2006), and deficits in executive function affect 50%
Franceschi, 2002). This transcription factor acts as a key regulator of those with sagittal CS (Beckett et al., 2014). Longitudinal
of osteogenesis. RUNX2 deficiency results in deficient skull ossifica- evaluation of a large and carefully characterized group of children
tion in the context of cleidocranial dysplasia. Importantly, RUNX2 with single-suture CS documented consistently lower
LATTANZI ET AL.
| 1419

neurodevelopmental scores as compared to controls (Starr et al., (Schweitzer et al., 2012). Given the fact that 3D–CT will remain a
2012). Neuropsychological testing in adolescents who underwent necessity for patients with complex and syndromic CS, significant
early surgery for NCS during infancy (<12 yr) documented visuospa- efforts have been made to reduce the CT radiation load (Badve, K, Iyer,
tial and constructional ability defects with associated visual memory Ishak, & Khanna, 2013). Recent work suggests a radiation dose
recall deficits and attention deficits in 7% and 17% sNCS cases, reduction of 90% is possible without compromising image quality
respectively, while language deficits occurred in 30% of those with (Eckel, Patton, Yu, Keating, & Wetjen, 2013).
cNCS, all otherwise nonsyndromic (Chieffo et al., 2010). It has been Surgery in the first year of life remains the only treatment for CS.
suggested that the cognitive profile of the affected individuals may The main objective is to expand the intracranial volume in order to
result from increased intracranial pressure (Inagaki et al., 2007; prevent the functional consequences of increased intracranial
Tamburrini, Caldarelli, Massimi, Santini, & Di Rocco, 2005). It is pressure, and to restore the cosmetic appearance of the head and
likely, however, that the neurocognitive deficits in this population face. The initial approach to treat CS with strip craniectomy in the
are a reflection of the changes in the brain that cannot be explained 1890‘s evolved to extensive cranial vault remodeling and has come
by skull constraint alone (Aldridge, Marsh, Govier, & Richtsmeier, full-circle back to endoscopic suturectomy and innovative techniques
2002; Aldridge et al., 2005). A recent study has further strengthened as spring-mediated cranioplasty (Okada & Gosain, 2012). Cranial vault
this notion by documenting altered neocortical connectivity in remodeling, while widely accepted as a standard of care in many
patients with sNCS (Beckett et al., 2014). Thus, a primary centers, is associated with significant morbidity, extensive blood loss
abnormality of brain growth and development may be responsible and prolonged anesthesia and hospitalization time. Developed in the
for the long-term developmental outcomes in CS. Aberrant brain late 1990's (Jimenez & Barone, 1998), endoscopic suturectomy with
development may also play a role as an etiologic factor for CS subsequent orthotic helmet therapy is becoming an increasingly
through altered brain-dura-suture homeostasis. attractive option for both single-suture and complex CS (Jimenez &
Barone, 2010; Rivero-Garvía, Marquez-Rivas, Rueda-Torres, & Ollero-
Ortiz, 2012; Rottgers, Lohani, & Proctor, 2016; Sauerhammer et al.,
2014). Despite reports that endoscopic suturectomy requires shorter
5 | I M A G I NG AN D T R E A T M E N T
operative and hospitalization times, less blood transfusion, and costs
A PP R O A C HE S IN CS
about half as much (Abbott, Rogers, Proctor, Busa, & Meara, 2012) as
does cranial vault remodeling, there is no consensus on the best
Often the abnormal head shape is present at birth but the diagnosis is
operative techniques.
not suspected until the first months of life. Plain radiograph, three-
Although non-surgical treatment of CS is not available at this time,
dimensional computed tomography (3D–CT), and ultrasound (US) are
the increased awareness of the molecular mechanisms causing these
the imaging modalities for patients with CS; magnetic resonance
defects has led to identification of potential targets for therapeutic
imaging (MRI) is reserved for complex syndromic cases with possible
interventions. FGFR tyrosine kinase inhibitors are capable of
brain abnormalities. Although it has been suggested that imaging is not
preventing CS in mice with Fgfr2 mutation causing Crouzon syndrome
necessary for patients with typical sNCS (Agrawal, Steinbok, &
(Eswarakumar et al., 2006). Another proof-of-concept study of the
Cochrane, 2005), in practice some imaging could still be necessary for
mouse model of Apert syndrome has demonstrated that prenatal
precise diagnosis and pre-surgical planning. In cases of plagiocephaly,
delivery of small molecule inhibiting the overactivated Ras/MAPK
careful clinical assessment may differentiate positional plagiocephaly
pathway not only completely rescued the skull phenotype but, when
from unilateral coronal or lambdoidal CS. When this is difficult, skull
repeatedly administered postnatally, resulted in viable and fertile
radiography remains a rapid and cost-effective option with low
animals (Shukla et al., 2007).
radiation load and specificity of 95% (Vannier et al., 1989) for children
These observations demonstrate that designing pharmacolog-
with a low probability of CS. However, this method has poor sensitivity
ical compounds for prevention and treatment of CS is a realistic
for more complex and partial sutural synostosis.
goal. Identification of at risk pregnancies through specific
High resolution US of the calvarial sutures has been successfully
biomarkers or fetal DNA analysis of the maternal serum is a
used both for confirmation of positional plagiocephaly (Krimmel et al.,
prerequisite to this task.
2012; Regelsberger et al., 2006) and as a primary diagnostic method
for CS with correct diagnosis in 95% of the patients (Simanovsky,
Hiller, Koplewitz, & Rozovsky, 2009). However, the inter-operator
variability and the longer time required for detailed study have been SUMMARY
deterrents. Since majority of children with positive US findings will still
be examined by 3D–CT, most consider US to be a screening modality.
Traditionally 3D–CT has been the gold standard for diagnosis of 1. The genetic causes of NCS remain largely unknown. Mutations in

CS. However, concerns with the high radiation load and its long-term the genes causing CS syndromes and chromosomal abnormalities
are not a common cause of NCS.
biological effects including cancer have led to revision of this
technique. It has been shown that with a carefully designed diagnostic 2. The causative role of the environment in NCS is yet to be
protocol 3D–CT is rarely necessary for children with single suture CS confirmed.
1420 | LATTANZI ET AL.

3. Recent studies have shown that animal models and some patients NIH-NIDCR grant R01DE16886 to SAB and by funds from the
with presumably NCS have mutations in specific genes (i.e., nonprofit “Federazione GENE” patients’ association to WL. SAB
TCF12, ERF, TWIST, ALX4, RUNX2, FREM1) within interconnected dedicates this work to his mentor Professor Emil Simeonov of the
signaling pathways. This may suggest that CS is a “developmental Medical University Sofia, Bulgaria.
pathway disorder” where mutations in various members of the
same or related pathways result in the same or similar phenotype.
CONFLICT OF INTEREST
Oligogenic inheritance, incomplete penetrance, epigenetic factors
or gene modulators are possible explanation for the general lack of The authors have no conflict of interest to declare.
Mendelian segregation.
4. Linkage, association and targeted candidate gene approaches
REFERENCES
have not yielded the expected results and exome or genome
association and sequencing studies may allow identification of the Abbott, M. M., Rogers, G. F., Proctor, M. R., Busa, K., & Meara, J. G. (2012).
causative genes and loci. Cost of treating sagittal synostosis in the first year of life. Journal of
Craniofacial Surgery, 23(1), 88–93.
5. Early diagnosis and referral to a center with substantial experience
Agrawal, D., Steinbok, P., & Cochrane, D. D. (2005). Diagnosis of isolated
in evaluation and treatment of CS substantially improves sagittal synostosis: Are radiographic studies necessary? Childs Nervous
outcomes. There is a tendency for less radiation-intensive imaging System, 22(4), 375–378.
techniques and less invasive surgical techniques. Alderman, B. W., Lammer, E. J., Joshua, S. C., Cordero, J. F., Ouimette, D. R.,
Wilson, M. J., & Ferguson, S. W. (1988). An epidemiologic study of
6. Neurodevelopmental issues are common and cannot be entirely
craniosynostosis: Risk indicators for the occurrence of craniosynostosis
attributed to the skull phenotype or the surgical trauma. Better in Colorado. American Journal of Epidemiology, 128(2), 431–438.
understanding of the brain-dura-suture axis may allow better Aldridge, K., Kane, A. A., Marsh, J. L., Panchal, J., Boyadjiev, S. A., Yan, P., . . .
treatment approaches. Richtsmeier, J. T. (2005). Brain morphology in nonsyndromic unicoronal
craniosynostosis. The Anatomical Record Part A Discoveries in Molecular
7. Targeted FGFR1, FGFR2, FGFR3, and TWIST1 genetic testing is
Cellular and Evolutionary Biology, 285(2), 690–698.
appropriate as a first-line test for patients with coronal and complex
Aldridge, K., Marsh, J. L., Govier, D., & Richtsmeier, J. T. (2002). Central
NCS as well for those with syndromic CS. Mutation analyses of
nervous system phenotypes in craniosynostosis. Journal of Anatomy,
TCF12 and ERF are justified in all patients with coronal synostosis 201(1), 31–39.
who are negative for FGFR and TWIST1 mutations. The value of these Al-Hertani, W., Hastings, V. A., McGowan-Jordan, J., Hurteau, J., & Graham,
tests for patients with sagittal and lambdoid CS is minimal, if any. G. E. (2013). Severe craniosynostosis in an infant with deletion 22q11.2
syndrome. American Journal of Medical Genetics Part A, 161A(1),
8. Chromosomal microarray has become an important diagnostic
153–157.
tool for patients with CS associated with additional anomalies,
Al-Rekabi, Z., Wheeler, M. M., Leonard, A., Fura, A. M., Juhlin, I., Frazar, C.,
developmental delays, and for those with affected relatives. Based . . . Sniadecki, N. J. (2016). Activation of the IGF1 pathway mediates
on its diagnostic yield it could be justified as a second-tier test changes in cellular contractility and motility in single-suture craniosyn-
after the targeted gene analyses. ostosis. Journal of Cell Science, 129(3), 483–491.
Altarac, M., & Saroha, E. (2007). Lifetime prevalence of learning disability
9. Exome sequencing is clinically available and an appropriate test
among US children. Pediatrics, 119(Suppl 1), S77–S83.
for nonspecific syndromic cases and NCS probands with affected
Alwan, S., Reefhuis, J., Rasmussen, S. A., Olney, R. S., Friedman, J. M., &
relatives when mutations in the genes associated with CS and National Birth Defects Prevention Study. (2007). Use of selective
submicroscopic chromosomal abnormalities have been excluded. serotonin-reuptake inhibitors in pregnancy and the risk of birth defects.
The New England Journal of Medicine, 356(26), 2684–2692.
10. Non-surgical treatment remains unavailable but there are
Ardalan, M., Rafati, A., Nejat, F., Farazmand, B., Majed, M., & El Khashab, M.
indications that it may become a possibility in the future.
(2012). Risk factors associated with craniosynostosis: A case control
11. Extensive collaborations between leading CS experts and sharing study. Pediatric Neurosurgery, 48(3), 152–156.
clinical, imaging, and epidemiological data, as well as pooling Arts, H. H., Bongers, E. M., Mans, D. A., van Beersum, S. E., Oud, M. M.,
biological specimens, are likely to more rapidly advance the Bolat, E., . . . Roepman, R. (2011). C14ORF179 encoding IFT43 is
knowledge of the field of CS. The International Craniosynostosis mutated in Sensenbrenner syndrome. Journal of Medical Genetics, 48(6),
390–395.
Consortium (genetics.ucdavis.edu) is an open collaborative
Aruga, J., Kamiya, A., Takahashi, H., Fujimi, T. J., Shimizu, Y., Ohkawa, K., . . .
network where both clinicians and researchers may contribute
Toyoda, A. (2006). A wide-range phylogenetic analysis of Zic proteins:
data, specimens and expertise related to CS and interested Implications for correlations between protein structure conservation
families may enroll in ongoing studies. and body plan complexity. Genomics, 87(6), 783–792.
Attanasio, C., Nord, A. S., Zhu, Y., Blow, M. J., Li, Z., Liberton, D. K., . . . Visel,
A. (2013). Fine tuning of craniofacial morphology by distant-acting
enhancers. Science, 342(6157), 1241006.

ACKNOWLEDGMENTS Aypar, E., Yildirim, M. S., Sert, A., Ciftci, I., & Odabas, D. (2011). A girl with
metopic synostosis and trisomy 13 mosaicism: Case report and review
We are in debt to all study participants for generously donating their of the literature. American Journal of Medical Genetics Part A, 155A(3),
time and samples for our CS studies. This work is supported by the 638–641.
LATTANZI ET AL.
| 1421

Azimi, C., Kennedy, S. J., Chitayat, D., Chakraborty, P., Clarke, J. T., Forrest, Choucair, N., Mignon-Ravix, C., Cacciagli, P., Abou Ghou, J., Fawaz, A.,
C., & Teebi, A. S. American Journal of Medical Genetics Part A, 117A(2), Mégarbané, A., . . . Chouery, E. (2015). Evidence that homozygous
127–135. PTPRD gene microdeletion causes trigonocephaly, hearing loss, and
Badve, C. A., K, M. M., Iyer, R. S., Ishak, G. E., & Khanna, P. C. (2013). intellectual disability. Molecular Cytogenetics, 8, 39.
Craniosynostosis: Imaging review and primer on computed tomogra- Clendenning, D. E., & Mortlock, D. P. (2012). The BMP ligand Gdf6 prevents
phy. Pediatric Radiology, 43(6), 728–742. differentiation of coronal suture mesenchyme in early cranial develop-
Beckett, J. S., Brooks, E. D., Lacadie, C., Vander Wyk, B., Jou, R. J., ment. PLoS ONE, 7(5), e36789.
Steinbacher, D. M., . . . Persing, J. A. (2014). Altered brain connectivity in Cohen, M. M. Jr, (2006). The new bone biology: Pathologic, molecular, and
sagittal craniosynostosis. Journal of Medical Genetics, 13(6), 690–698. clinical correlates. American Journal of Medical Genetics Part A,
Benedyk, M. J., Mullen, J. R., & DiNardo, S. (1994). Odd-paired: A zinc finger 140A(23), 2646–2706.
pair-rule protein required for the timely activation of engrailed and Cohen, M. M. (2000). Craniosynostosis: Diagnosis, Evaluation, and Manage-
wingless in Drosophila embryos. Genes & Development, 8(1), 105–117. ment. New York: Oxford University Press.
Bermejo, E., Félix, V., Lapunzina, P., Galán, E., Soler, V., Delicado, A., . . . Connerney, J., Andreeva, V., Leshem, Y., Muentener, C., Mercado, M. A., &
Martínez-Frías, M. L. (2005). Craniofacial dyssynostosis: Description of Spicer, D. B. (2006). Twist1 dimer selection regulates cranial suture
the first four Spanish cases and review. American Journal of Medical patterning and fusion. Developmental Dynamics, 235(5), 1345–1357.
Genetics Part A, 132A(1), 41–48.
Cooper, G. M., Usas, A., Olshanski, A., Mooney, M. P., Losee, J. E., & Huard,
Bessenyei, B., Nagy, A., Szakszon, K., Mokánszki, A., Balogh, E., Ujfalusi, A., J. (2009). Ex vivo Noggin gene therapy inhibits bone formation in a
. . . Oláh, (2015). Clinical and genetic characteristics of craniosynostosis mouse model of postoperative resynostosis. Plastic and Reconstructive
in Hungary. American Journal of Medical Genetics Part A, 167A(12), Surgery, 123(2 Suppl), 94S–103S.
2985–2991.
Couly, G. F., Coltey, P. M., & Le Douarin, N. M. (1992). The developmental
Bifari, F., Berton, V., Pino, A., Kusalo, M., Malpeli, G., Di Chio, M., . . . fate of the cephalic mesoderm in quail-chick chimeras. Development,
Decimo, I. (2015). Meninges harbor cells expressing neural precursor 114(1), 1–15.
markers during development and adulthood. Frontiers in Cellular
Couly, G. F., Coltey, P. M., & Le Douarin, N. M. (1993). The triple origin of
Neuroscience, 9, 383.
skull in higher vertebrates: A study in quail-chick chimeras. Develop-
Bitgood, M. J., & McMahon, A. P. (1995). Hedgehog and Bmp genes are ment, 117(2), 409–429.
coexpressed at many diverse sites of cell-cell interaction in the mouse
Cunningham, M. L., Horst, J. A., Rieder, M. J., Hing, A. V., Stanaway, I. B.,
embryo. Developmental Biology, 172(1), 126–138.
Park, S. S., Samudrala, R., & Speltz, M. L. (2011). IGF1R variants
Blaser, S. I. (2008). Abnormal skull shape. Pediatric Radiology, 38(Suppl 3), associated with isolated single suture craniosynostosis. American
S488–S496. Journal of Medical Genetics Part A, 91–97.
Bodo, M., Carinci, F., Baroni, T., Giammarioli, M., Bellucci, C., Bosi, G., . . . Dathe, K., Kjaer, K. W., Brehm, A., Meinecke, P., Nürnberg, P., Neto, J. C., . . .
Carinci, P. (1997). Apert's syndrome: Differential in vitro production of Mundlos, S. (2009). Duplications involving a conserved regulatory
matrix macromolecules and its regulation by interleukins. European element downstream of BMP2 are associated with brachydactyly type
Journal of Clinical Investigation, 27(1), 36–42. A2. The American Journal of Human Genetics, 84(4), 483–492.
Boulet, S. L., Rasmussen, S. A., & Honein, M. A. (2008). A population-based De Marco, P., Raso, A., Beri, S., Gimelli, S., Merello, E., Mascelli, S., . . .
study of craniosynostosis in metropolitan Atlanta, 1989–2003. Gimelli, G. (2011). A de novo balanced translocation t(7;12)(p21.2;
American Journal of Medical Genetics Part A, 146A(8), 984–991. p12.3) in a patient with Saethre-Chotzen-like phenotype down-
Boyadjiev, S. A. & International Craniosynostosis Consortium. (2007). regulates TWIST and an osteoclastic protein-tyrosine phosphatase,
Genetic analysis of nonsyndromic craniosynostosis. Orthodontics & PTP-oc. European Journal of Medical Genetics, 54(5), e478–e483.
Craniofacial Research, 10(3), 129–137. De Silva, D., Duffty, P., Booth, P., Auchterlonie, I., Morrison, N., & Dean, J. C.
Boyadjiev, S. A., Zhang, G., Ingersoll, R., Isaac, N., Kasch, L., Hur, D., . . . Scott, (1995). Family studies in chromosome 22q11 deletion: Further
A. F. (2002). Analysis of candidate genes for nonsyndromic craniosyn- demonstration of phenotypic heterogeneity. Clinical Dysmorphology,
ostosis. The American Journal of Human Genetics, 71, A1795. 4(4), 294–303.
Boyadjiev SA. 2014. Congenital craniofacial abnormalities. Merck Manual Dean, J. C., De Silva, D. C., & Reardon, W. (1998). Craniosynostosis and
2105. Available from: http://www.merckmanuals.com/professional/ chromosome 22q11 deletion. Journal of Medical Genetics, 35(4), 346;
pediatrics/congenital-craniofacial-and-musculoskeletal-abnormalities/ author reply 347.
congenital-craniofacial-abnormalities Deckelbaum, R. A., Holmes, G., Zhao, Z., Tong, C., Basilico, C., & Loomis,
Carmichael, S. L., Ma, C., Rasmussen, S. A., Honein, M. A., & Lammer, E. J. C. A. (2012). Regulation of cranial morphogenesis and cell fate at the
Craniosynostosis and maternal smoking. Birth Defects Research Part A: neural crest-mesoderm boundary by engrailed 1. Development, 139(7),
Clinical and Molecular Teratology, 82(2), 78–85. 1346–1358.
Chang, R., Petersen, J. R., Niswander, L. A., & Liu, A. (2015). A hypomorphic Deckelbaum, R. A., Majithia, A., Booker, T., Henderson, J. E., & Loomis, C. A.
allele reveals an important role of inturned in mouse skeletal (2006). The homeoprotein engrailed 1 has pleiotropic functions in
development. Developmental Dynamics, 244(6), 736–747. calvarial intramembranous bone formation and remodeling. Develop-
ment, 133(1), 63–74.
Chieffo, D., Tamburrini, G., Massimi, L., Di Giovanni, S., Giansanti, C.,
Caldarelli, M., & Di Rocco, C. (2010). Long-term neuropsychological Dee, C. T., Szymoniuk, C. R., Mills, P. E., & Takahashi, T. (2013). Defective
development in single-suture craniosynostosis treated early. Journal of neural crest migration revealed by a Zebrafish model of Alx1-related
Medical Genetics, 5(3), 232–237. frontonasal dysplasia. Human Molecular Genetics, 22(2), 239–251.
Chotai, K. A., Brueton, L. A., van Herwerden, L., Garrett, C., Hinkel, G. K., Desai, J., Shannon, M. E., Johnson, M. D., Ruff, D. W., Hughes, L. A., Kerley,
Schinzel, A., . . . Winter, R. M. (1994). Six cases of 7p deletion: Clinical, M. K., . . . Culiat, C. T. (2006). Nell1-deficient mice have reduced
cytogenetic, and molecular studies. American Journal of Medical expression of extracellular matrix proteins causing cranial and vertebral
Genetics, 51(3), 270–276. defects. Human Molecular Genetics, 15(8), 1329–1341.
1422 | LATTANZI ET AL.

Di Rocco F., Arnaud E., Meyer P., Sainte-Rose C., & Renier D. 2009. Focus Fryssira, H., Makrythanasis, P., Kattamis, A., Stokidis, K., Menten, B., Kosaki,
session on the changing "epidemiology" of craniosynostosis (comparing K., . . . Kanavakis, E. (2011). Severe developmental delay in a patient
two quinquennia: 1985–1989 and 2003–2007) and its impact on the with 7p21.1-p14.3 microdeletion spanning the TWIST gene and the
daily clinical practice: A review from Necker Enfants Malades. Childs HOXA gene cluster. Molecular Syndromology, 2(1), 45–49.
Nervous System 25(7): 807–811. Gajecka, M., Yu, W., Ballif, B. C., Glotzbach, C. D., Bailey, K. A., Shaw, C. A.,
Di Rocco, F., Arnaud, E., & Renier, D. 2009. Evolution in the frequency of . . . Shaffer, L. G. (2005). Delineation of mechanisms and regions of
nonsyndromic craniosynostosis. Neurosurg Pediatrics 4(1), 21–25. dosage imbalance in complex rearrangements of 1p36 leads to a
Di Rocco, F., Baujat, G., Arnaud, E., Rénier, D., Laplanche, J. L., Daire, V. C., & putative gene for regulation of cranial suture closure. European Journal
Collet, C. (2014). Clinical spectrum and outcomes in families with of Human Genetics, 13(2), 139–149.
coronal synostosis and TCF12 mutations. European Journal of Human Garcez, R. C., Le Douarin, N. M., & Creuzet, S. E. (2014). Combinatorial
Genetics, 22(12), 1413–1416. activity of Six1-2-4 genes in cephalic neural crest cells controls
Di Rocco, F., Benoit, A., Vigneron, J., Segura, P. B., Klein, O., Collet, C., & craniofacial and brain development. Cellular and Molecular Life Sciences,
Arnaud, E. (2015). Y-craniosynostosis by premature fusion of the 71(11), 2149–2164.
metopic and coronal sutures: A new nosological entity or a variety of Garcia-Heras, J., Corley, N., Garcia, M. F., Kukolich, M. K., Smith, K. G., &
Saethre-Chotzen syndrome? Birth Defects Research Part A: Clinical and Day, D. W. (1999). De novo partial duplications 1p: Report of two new
Molecular Teratology, 103(4), 306–310. cases and review. American Journal of Medical Genetics, 82(3), 261–264.
Dolk, H., Loane, M., & Garne, E. (2010). The prevalence of congenital Gill, S. K., Broussard, C., Devine, O., Green, R. F., Rasmussen, S. A., Reefhuis,
anomalies in Europe. Advances in Experimental Medicine and Biology, J., & National Birth Defects Prevention Study. (2012). Association
686, 349–364. between maternal age and birth defects of unknown etiology: United
Doyle, A. J., Doyle, J. J., Bessling, S. L., Maragh, S., Lindsay, M. E., Schepers, States, 19972007. Birth Defects Research Part A: Clinical and Molecular
D., . . . Dietz, H. C. (2012). Mutations in the TGF-beta repressor SKI Teratology, 94(12), 1010–1008.
cause Shprintzen-Goldberg syndrome with aortic aneurysm. Nature Gorlin, R. J., Cohen, M. M., & Hennekam, R. C. M. (2001). Syndromes of the
Genetics, 44(11), 1249–1254. Head and Neck, (4th ed.). New York: Oxford University Press Inc.
Eckel, L., Patton, A., Yu, L., Keating, G. F., & Wetjen, N. M. (2013). Radiation Greene, A. K., Mulliken, J. B., Proctor, M. R., Meara, J. G., & Rogers, G. F.
dose reduction and protocol optimization for pediatric head CT in (2008). Phenotypically unusual combined craniosynostoses: Presenta-
evaluation of craniosynostosis. Presented at the 50 th Annual Meeting tion and management. Plastic and Reconstructive Surgery, 122(3),
of the European Society of Paediatric Radiology, June 3–7, Budapest. 853–862.
Hungary Pediatric Radiology, 43(Suppl 3), S475–S656. Greenwood, J., Flodman, P., Osann, K., Boyadjiev, S. A., & Kimonis, V.
el Ghouzzi, V., Le Merrer, M., Perrin-Schmitt, F., Lajeunie, E., Benit, P., (2014). Familial incidence and associated symptoms in a population of
Renier, D., . . . Bonaventure, J. Mutations of the TWIST gene in the individuals with nonsyndromic craniosynostosis. Genetics in Medicine,
Saethre-Chotzen syndrome. Nature Genetics, 15(1), 42–46. 16(4), 302–310.
Eswarakumar, V. P., Ozcan, F., Lew, E. D., Bae, J. H., Tomé, F., Booth, C. J., Greives, M. R., Odessey, E. A., Waggoner, D. J., Shenaq, D. S., Aradhya, S.,
. . . Schlessinger, J. (2006). Attenuation of signaling pathways stimulated Mitchell, A., . . . Reid, R. R. (2013). RUNX2 quadruplication: Additional
by pathologically activated FGF-receptor 2 mutants prevents cranio- evidence toward a new form of syndromic craniosynostosis. Journal of
synostosis. Proceedings of the National Academy of Sciences of the United Craniofacial Surgery, 24(1), 126–129.
States of America, 103(49), 18603–18608. Grillo, L., Greco, D., Pettinato, R., Avola, E., Potenza, N., Castiglia, L., . . .
Fennell, N., Foulds, N., Johnson, D. S., Wilson, L. C., Wyatt, M., Robertson, Fichera, M. (2014). Increased FGF3 and FGF4 gene dosage is a risk
S. P., . . . Wilkie, A. O. (2015). Association of mutations in FLNA with factor for craniosynostosis. Gene, 534(2), 435–439.
craniosynostosis. European Journal of Human Genetics, 23(12), Gripp, K. W., Stolle, C. A., Celle, L., McDonald-McGinn, D. M., Whitaker, L. A., &
1684–1686. Zackai, E. H. (1999). TWIST gene mutation in a patient with radial aplasia
Florisson, J. M., Verkerk, A. J., Huigh, D., Hoogeboom, A. J., Swagemakers, and craniosynostosis: Further evidence for heterogeneity of Baller-Gerold
S., Kremer, A., . . . van der Spek, P. J. (2013). Boston type craniosynos- syndrome. American Journal of Medical Genetics, 82(2), 170–176.
tosis: Report of a second mutation in MSX2. American Journal of Medical Gupta, P. C., Foster, J., Crowe, S., Papay, F. A., Luciano, M., & Traboulsi, E. I.
Genetics Part A, 161A(10), 2626–2633. (2003). Ophthalmologic findings in patients with nonsyndromic
Flück, C. E., Tajima, T., Pandey, A. V., Arlt, W., Okuhara, K., Verge, C. F., . . . plagiocephaly. Journal of Craniofacial Surgery, 14(4), 529–532.
Miller, W. L. (2004). Mutant P450 oxidoreductase causes disordered Hall, J. G., Allanson, J., Gripp, K., & Slavotinek, A. (2013). Handbook of
steroidogenesis with and without Antley-Bixler syndrome. Nature Physical Measurements. (3rd ed). New York: Oxford University Press.
Genetics, 36(3), 228–230.
Han, J., Ishii, M., Bringas, P., Jr, Maas, R. L., Maxson, R. E., Jr, & Chai, Y.
Foley, P., McAuliffe, F., Mullarkey, M., & Reardon, W. (2007). Antenatal (2007). Concerted action of Msx1 and Msx2 in regulating cranial neural
diagnosis of deletion chromosome 11(q23-qter) (Jacobsen syndrome). crest cell differentiation during frontal bone development. Mechanisms
Clinical Dysmorphology, 16(3), 177–179. of Development, 124(9-10), 729–745.
Fondon, J. W., & Garner, H. R. (2004). Molecular origins of rapid and Hanai, J., Chen, L. F., Kanno, T., Ohtani-Fujita, N., Kim, W. Y., Guo, W. H., . . .
continuous morphological evolution. Proceedings of the National Ito, Y. (1999). Interaction and functional cooperation of PEBP2/CBF
Academy of Sciences of the United States of America, 101(52), with Smads. Synergistic induction of the immunoglobulin germline
18058–18063. Calpha promoter. Journal of Biological Chemistry, 274(44),
Foster, K. A., Frim, D. M., & McKinnon, M. (2008). Recurrence of synostosis 31577–31582.
following surgical repair of craniosynostosis. Plastic and Reconstructive Heilstedt, H. A., Shapira, S. K., Gregg, A. R., & Shaffer, L. G. (1999).
Surgery, 121(3), 70e–76e. Molecular and clinical characterization of a patient with duplication of
Fryburg, J. S., Hwang, V., & Lin, K. Y. (1995). Recurrent lambdoid synostosis 1p36.3 and metopic synostosis. Clinical Genetics, 56(2), 123–128.
within two families. American Journal of Medical Genetics, 58(3), Heuzé, Y., Holmes, G., Peter, I., Richtsmeier, J. T., & Jabs, E. W. 2014.
262–266. Closing the gap: genetic and genomic continuum from syndromic to
LATTANZI ET AL.
| 1423

nonsyndromic craniosynostoses. Current Genetic Medicine Reports, 2(3), Jehee, F. S., Johnson, D., Alonso, L. G., Cavalcanti, D. P., de Sá Moreira, E.,
135–145. Moreira, E., . . . Passos-Bueno, M. R. (2005). Molecular screening for
Higashino, T., & Hirabayashi, S. (2013). A secondary craniosynostosis microdeletions at 9p22-p24 and 11q23-q24 in a large cohort of
associated with juvenile hyperthyroidism. Journal of Plastic, Reconstruc- patients with trigonocephaly. Clinical Genetics, 67(6), 503–510.
tive & Aesthetic Surgery, 66(10), e284–e286. Jehee, F. S., Krepischi-Santos, A. C., Rocha, K. M., Cavalcanti, D. P., Kim,
Hing, A. V., Click, E. S., Holder, U., Seto, M. L., Vessey, K., Gruss, J., . . . C. A., Bertola, D. R., . . . Passos-Bueno, M. R. (2008). High frequency of
Cunningham, M. L. (2009). Bilateral lambdoid and sagittal synostosis submicroscopic chromosomal imbalances in patients with syndromic
(BLSS): A unique craniosynostosis syndrome or predictable craniofacial craniosynostosis detected by a combined approach of microsatellite
phenotype? American Journal of Medical Genetics Part A, 149A(5), segregation analysis, multiplex ligation-dependent probe amplification
1024–1032. and array-based comparative genome hybridisation. Journal of Medical
Genetics, 45(7), 447–450.
Hornik, C., Brand-Saberi, B., Rudloff, S., Christ, B., & Füchtbauer, E. M.
(2004). Twist is an integrator of SHH, FGF, and BMP signaling. Anatomy Jenkins, D., Seelow, D., Jehee, F. S., Perlyn, C. A., Alonso, L. G., Bueno, D. F.,
and Embryology (Berl), 209(1), 31–39. . . . Wilkie, A. O. (2007). RAB23 mutations in Carpenter syndrome imply
an unexpected role for hedgehog signaling in cranial-suture develop-
Howard, T. D., Paznekas, W. A., Green, E. D., Chiang, L. C., Ma, N., Ortiz de
ment and obesity. The American Journal of Human Genetics, 80(6),
Luna, R. I., . . . Jabs, E. W. (1997). Nature Genetics, 15(1), 36–41.
1162–1670.
Huang, N., Pandey, A. V., Agrawal, V., Reardon, W., Lapunzina, P. D.,
Jeradi, S., & Hammerschmidt, M. (2016). Retinoic acid-induced premature
Mowat, D., . . . Miller, W. L. (2005). Diversity and function of mutations
osteoblast-to-preosteocyte transitioning has multiple effects on
in p450 oxidoreductase in patients with Antley-Bixler syndrome and
calvarial development. Development, 143(7), 1205–1216.
disordered steroidogenesis. The American Journal of Human Genetics,
76(5), 729–749. Jiang, X., Iseki, S., Maxson, R. E., Sucov, H. M., & Morriss-Kay, G. M. (2002).
Tissue origins and interactions in the mammalian skull vault.
Hufnagel, R. B., Zimmerman, S. L., Krueger, L. A., Bender, P. L., Ahmed, Z. M.,
Developmental Biology, 241(1), 106–116.
& Saal, H. M. (2016). A new frontonasal dysplasia syndrome associated
with deletion of the SIX2 gene. American Journal of Medical Genetics Part Jimenez, D. F., & Barone, C. M. (1998). Endoscopic craniectomy for early
A, 170A(2), 487–491. surgical correction of sagittal craniosynostosis. Journal of Neurosurgery,
88(1), 77–81.
Hurley, M. E., White, M. J., Green, A. J., & Kelleher, J. (2004). Antley-Bixler
syndrome with radioulnar synostosis. Pediatric Radiology, 34(2), Jimenez, D. F., & Barone, C. M. (2010). Multiple-suture nonsyndromic
148–151. craniosynostosis: Early and effective management using endoscopic
techniques. Journal of Medical Genetics, 5(3), 223–231.
Inagaki, T., Kyutoku, S., Seno, T., Kawaguchi, T., Yamahara, T., Oshige, H., . . .
Kawamoto, K. (2007). The intracranial pressure of the patients with mild Johnson, D., Wall, S. A., Mann, S., & Wilkie, A. O. (2000). A novel mutation,
form of craniosynostosis. Childs Nervous System, 23(12), 1455–1459. Ala315Ser, in FGFR2: A gene-environment interaction leading to
craniosynostosis? European Journal of Human Genetics, 8(8), 571–577.
Ishii, M., Merrill, A. E., Chan, Y. S., Gitelman, I., Rice, D. P., Sucov, H. M., &
Maxson, R. E., Jr. (2003). Msx2 and Twist cooperatively control the Johnson, D., & Wilkie, A. O. (2011). Craniosynostosis. European Journal of
development of the neural crest-derived skeletogenic mesenchyme of Human Genetics, 19(4), 369–376.
the murine skull vault. Development, 130(24), 6131–6142. Justice, C. M. (2014). Non-coding variant in the BMP2 locus that is
Iwasaki, M., Le, A. X., & Helms, J. A. (1997). Expression of indian hedgehog, associated with sagittal nonsyndromic craniosynostosis causes differ-
bone morphogenetic protein 6 and gli during skeletal morphogenesis. ential GFP expression in zebrafish. Presented at the 64th Annual
Mechanisms of Development, 69(1–2), 197–202. Meeting of The American Society of Human Genetics meeting, October
18–22, 2014, San Diego, CA.
Jabs, E. W., Li, X., Scott, A. F., Meyers, G., Chen, W., Eccles, M., . . . Jaye, M.
(1994). Jackson-Weiss and Crouzon syndromes are allelic with Justice, C. M., Yagnik, G., Kim, Y., Peter, I., Jabs, E. W., Erazo, M., . . .
mutations in fibroblast growth factor receptor 2. Nature Genetics, Boyadjiev, S. A. (2012). A genome-wide association study identifies
8(3), 275–279. susceptibility loci for nonsyndromic sagittal craniosynostosis near
BMP2 and within B BS9. Nature Genetics, 44(12), 1360–1364.
Jabs, E. W., Müller, U., Li, X., Ma, L., Luo, W., Haworth, I. S., . . . Maxson, R.
(1993). A mutation in the homeodomain of the human MSX2 gene in a Kadlub, N., Persing, J. A., da Silva Freitas, R., & Shin, J. H. (2008). Familial
family affected with autosomal dominant craniosynostosis. Cell, 75, lambdoid craniosynostosis between father and son. Journal of
443–450. Craniofacial Surgery, 19(3), 850–854.

Jabs, E. W. (1998). Toward understanding the pathogenesis of craniosyn- Kallen, K. (1999). Maternal smoking and craniosynostosis. Teratology, 60(3),
ostosis through clinical and molecular correlates. Clinical Genetics, 53(2), 146–150.
79–86. Kapp-Simon, K. A., Speltz, M. L., Cunningham, M. L., Patel, P. K., & Tomita, T.
James, A. W., Levi, B., Xu, Y., Carre, A. L., & Longaker, M. T. (2010). Retinoic (2007). Neurodevelopment of children with single suture craniosynos-
acid enhances osteogenesis in cranial suture-derived mesenchymal tosis: A review. Childs Nervous System, 23(3), 269–281.
cells: Potential mechanisms of retinoid-induced craniosynostosis. Kapp-Simon, K. A. (1994). Mental development in infants with non-
Plastic and Reconstructive Surgery, 125(5), 1352–1361. syndromic craniosynostosis with and without cranial release and
Janssen, A., Hosen, M. J., Jeannin, P., Coucke, P. J., De Paepe, A., & reconstruction. Plastic and Reconstructive Surgery, 94(2), 408–410.
Vanakker, O. M. (2013). Second family with the Boston-type Kariminejad, A., Kariminejad, R., Tzschach, A., Ullmann, R., Ahmed, A.,
craniosynostosis syndrome: Novel mutation and expansion of the Asghari-Roodsari, A., . . . Kariminejad, M. H. (2009). Craniosynostosis in
clinical spectrum. American Journal of Medical Genetics Part A, 161A(9), a patient with 2q37.3 deletion 5q34 duplication: Association of extra
2352–2357. copy of MSX2 with craniosynostosis. American Journal of Medical
Jehee, F. S., Bertola, D. R., Yelavarthi, K. K., Krepischi-Santos, A. C., Kim, C., Genetics Part A, 149A(7), 1544–1549.
Vianna-Morgante, A. M., . . . Passos-Bueno, M. R. (2007). An 11q11- Kawara, H., Yamamoto, T., Harada, N., Yoshiura, K., Niikawa, N., Nishimura,
q13.3 duplication, including FGF3 and FGF4 genes, in a patient with A., . . . Matsumoto, N. (2006). Narrowing candidate region for
syndromic multiple craniosynostoses. American Journal of Medical monosomy 9p syndrome to a 4.7 −Mb segment at 9p22.2-p23.
Genetics Part A, 143A(16), 1912–1918. American Journal of Medical Genetics Part A, 140(4), 373–377.
1424 | LATTANZI ET AL.

Kayserili, H., Uz, E., Niessen, C., Vargel, I., Alanay, Y., Tuncbilek, G., . . . synostosis syndrome. European Journal of Human Genetics, 14(1),
Akarsu, N. A. (2009). ALX4 dysfunction disrupts craniofacial and 39–48.
epidermal development. Human Molecular Genetics, 18(22), Krimmel, M., Will, B., Wolff, M., Kluba, S., Haas-Lude, K., Schaefer, J., . . .
4357–4366. Reinert, S. (2012). Value of high-resolution ultrasound in the differential
Kelleher, M. O., Murray, D. J., McGillivary, A., Kamel, M. H., Allcutt, D., & diagnosis of scaphocephaly and occipital plagiocephaly. International
Earley, M. J. (2006). Behavioral, developmental, and educational Journal of Oral and Maxillofacial Surgery, 41(7), 797–800.
problems in children with nonsyndromic trigonocephaly. Journal of Kuo, J. S., Patel, M., Gamse, J., Merzdorf, C., Liu, X., Apekin, V., & Sive, H.
Neurosurgery, 105(5 Suppl), 382–384. (1998). Opl: A zinc finger protein that regulates neural determination
Khadka, D., Luo, T., & Sargent, T. D. (2006). Msx1 and Msx2 have shared and patterning in Xenopus. Development, 125(15), 2867–2882.
essential functions in neural crest but may be dispensable in epidermis Lajeunie, E., Barcik, U., Thorne, J. A., El Ghouzzi, V., Bourgeois, M., & Renier,
and axis formation in Xenopus. International Journal of Developmental D. (2001). Craniosynostosis and fetal exposure to sodium valproate.
Biology, 50(5), 499–502. Journal of Neurosurgery, 95(5), 778–782.
Khanna, P. C., Thapa, M. M., Iyer, R. S., & Prasad, S. S. (2011). Pictorial essay: Lajeunie, E., Crimmins, D. W., Arnaud, E., & Renier, D. (2005). Genetic
The many faces of craniosynostosis. Indian Journal of Radiology and considerations in nonsyndromic midline craniosynostoses: A study of
Imaging Impact, 21(1), 49–56. twins and their families. Journal of Neurosurgery, 103(4 Suppl),
Kim, H. J., Lee, M. H., Park, H. S., Park, M. H., Lee, S. W., Kim, S. Y., . . . Ryoo, 353–356.
H. M. (2003). Erk pathway and activator protein 1 play crucial roles in Lajeunie, E., El Ghouzzi, V., Le Merrer, M., Munnich, A., Bonaventure, J., &
FGF2-stimulated premature cranial suture closure. Developmental Renier, D. (1999). Sex related expressivity of the phenotype in coronal
Dynamics, 227(3), 335–346. craniosynostosis caused by the recurrent P250R FGFR3 mutation.
Kim, J., Won, H. H., Kim, Y., Choi, J. R., Yu, N., & Lee, K. A. (2015). Breakpoint Journal of Medical Genetics, 36(1), 9–13.
mapping by whole genome sequencing identifies PTH2R gene Lajeunie, E., Le Merrer, M., Bonaïti-Pellie, C., Marchac, D., & Renier, D.
disruption in a patient with midline craniosynostosis and a de novo (1995). Genetic study of nonsyndromic coronal craniosynostosis.
balanced chromosomal rearrangement. Journal of Medical Genetics, American Journal of Medical Genetics, 55(4), 500–504.
52(10), 706–709.
Lajeunie, E., Le Merrer, M., Bonaïti-Pellie, C., Marchac, D., & Renier, D.
Kim, S. D., Yagnik, G., Cunningham, M. L., Kim, J., & Boyadjiev, S. A. (2014). (1996). Genetic study of scaphocephaly. American Journal of Medical
MAPK/ERK signaling pathway analysis in primary osteoblasts from Genetics, 62(3), 282–285.
patients with nonsyndromic sagittal craniosynostosis. Cleft Palate-
Lakin, G. E., Sinkin, J. C., Chen, R., Koltz, P. F., & Girotto, J. A. (2012). Genetic
Craniofacial Journal, 51(1), 115–119.
and epigenetic influences of twins on the pathogenesis of craniosyn-
Kimonis, V., Gold, J. A., Hoffman, T. L., Panchal, J., & Boyadjiev, S. A. (2007). ostosis: A meta-analysis. Plastic and Reconstructive Surgery, 129(4),
Genetics of craniosynostosis. Seminars in Pediatric Neurology, 14(3), 945–954.
150–161.
Lana-Elola E1 Rice, R., Grigoriadis, A. E., & Rice, D. P. (2007). Cell fate
Kini, U., Hurst, J. A., Byren, J. C., Wall, S. A., Johnson, D., & Wilkie, A. O. specification during calvarial bone and suture development. Develop-
(2010). Etiological heterogeneity and clinical characteristics of metopic mental Biology, 311(2), 335–346.
synostosis: Evidence from a tertiary craniofacial unit. American Journal
Lattanzi, W., Barba, M., Novegno, F., Massimi, L., Tesori, V., Tamburrini, G.,
of Medical Genetics Part A, 152A(6), 1383–1389.
. . . Di Rocco, C. (2013). Lim mineralization protein is involved in the
Ko, J. M., Jeong, S. Y., Yang, J. A., Park, D. H., & Yoon, S. H. (2012). Molecular premature calvarial ossification in sporadic craniosynostoses. Bone,
genetic analysis of TWIST1 and FGFR3 genes in Korean patients with 52(1), 474–484.
coronal synostosis: Identification of three novel TWIST1 mutations.
Lattanzi, W., Bukvic, N., Barba, M., Tamburrini, G., Bernardini, C., Michetti,
Plastic and Reconstructive Surgery, 129(5), 814e–821e.
F., & Di Rocco, C. (2012). Genetic basis of single-suture synostoses:
Komatsu, Y., Yu, P. B., Kamiya, N., Pan, H., Fukuda, T., Scott, G. J., . . . Genes, chromosomes and clinical implications. Childs Nervous System,
Mishina, Y. (2013). Augmentation of Smad-dependent BMP signaling in 28(9), 1301–1310.
neural crest cells causes craniosynostosis in mice. Journal of Bone and
Lattanzi, W., Parolisi, R., Barba, M., & Bonfanti, L. (2015). Osteogenic and
Mineral Research, 28(6), 1422–1433.
neurogenic stem cells in their own place: Unraveling differences and
Komori, T., Yagi, H., Nomura, S., Yamaguchi, A., Sasaki, K., Deguchi, K., . . . similarities between niches. Frontiers in Cellular Neuroscience, 9, 455.
Kishimoto, T. (1997). Targeted disruption of Cbfa1 results in a complete
Lattanzi, W. (2016). Molecular Genetics of Craniosynostosis. eLS. John
lack of bone formation owing to maturational arrest of osteoblasts. Cell,
Wiley & Sons, Ltd: Chichester. DOI: 10.1002/9780470015902.
89(5), 755–764.
a0025186.
Komotar, R. J., Zacharia, B. E., Ellis, J. A., Feldstein, N. A., & Anderson, R. C.
Laue, K., Pogoda, H. M., Daniel, P. B., van Haeringen, A., Alanay, Y., von
(2006). Pitfalls for the pediatrician: Positional molding or craniosynos-
Ameln, S., . . . Robertson, S. P. (2011). Craniosynostosis and multiple
tosis? Pediatric Annals, 35(5), 365–375.
skeletal anomalies in humans and zebrafish result from a defect in the
Kosaki, R., Higuchi, M., Mitsui, N., Matsushima, K., Ohashi, H., & Kosaki, K. localized degradation of retinoic acid. The American Journal of Human
(2005). Deletion involving the TWIST locus and the HOXA cluster: A Genetics, 89(5), 595–606.
contiguous gene syndrome on 7p? Congenital Anomalies (Kyoto), 45(1),
Le Tanno, P., Poreau, B., Devillard, F., Vieville, G., Amblard, F., Jouk, P. S., . . .
35–38.
Coutton, C. (2014). Maternal complex chromosomal rearrangement
Kress, W., Petersen, B., Collmann, H., & Grimm, T. (2000). An unusual leads to TCF12 microdeletion in a patient presenting with coronal
FGFR1 mutation (fibroblast growth factor receptor 1 mutation) in a girl craniosynostosis and intellectual disability. American Journal of Medical
with nonsyndromic trigonocephaly. Cytogenetics and Cell Genetics, Genetics Part A, 164A(6), 1530–1536.
91(1–4), 138–140.
Lee, S. W., Choi, K. Y., Cho, J. Y., Jung, S. H., Song, K. B., Park, E. K., . . . Ryoo,
Kress, W., Schropp, C., Lieb, G., Petersen, B., Büsse-Ratzka, M., Kunz, J., . . . H. M. (2006). TGF-beta2 stimulates cranial suture closure through
Collmann, H. (2006). Saethre-Chotzen syndrome caused by TWIST 1 activation of the Erk-MAPK pathway. Journal of Cellular Biochemistry,
gene mutations: Functional differentiation from Muenke coronal 98(4), 981–991.
LATTANZI ET AL.
| 1425

Lenton, K., James, A. W., Manu, A., Brugmann, S. A., Birker, D., Nelson, E. R., Mavrogiannis, L. A., Antonopoulou, I., Baxová, A., Kutílek, S., Kim, C. A.,
. . . Longaker, M. T. (2011). Indian hedgehog positively regulates Sugayama, S. M., . . . Wilkie, A. O. (2001). Haploinsufficiency of the
calvarial ossification and modulates bone morphogenetic protein human homeobox gene ALX4 causes skull ossification defects. Nature
signaling. Genesis, 49(10), 784–796. Genetics, 27(1), 17–18.
Leslie, E. J., Taub, M. A., Liu, H., Steinberg, K. M., Koboldt, D. C., Zhang, Q., McDonald-McGinn, D. M., Feret, H., Nah, H. D., Bartlett, S. P., Whitaker,
. . . Murray, J. C. (2015). Identification of functional variants for cleft lip L. A., & Zackai, E. H. (2010). Metopic craniosynostosis due to mutations
with or without cleft palate in or near PAX7, FGFR2, and NOG by in GLI3: A novel association. Journal of Medical Genetics Part A, 152A(7),
targeted sequencing of GWAS loci. The American Journal of Human 1654–1660.
Genetics, 96(3), 397–411. McDonald-McGinn, D. M., Gripp, K. W., Kirschner, R. E., Maisenbacher,
Li, J., Hu, C., Han, L., Liu, L., Jing, W., Tang, W., . . . Long, J. (2015). MiR-154- M. K., Hustead, V., Schauer, G. M., . . . Zackai, E. H. (2005).
5p regulates osteogenic differentiation of adipose-derived mesenchy- Craniosynostosis: Another feature of the 22q11.2 deletion syndrome.
mal stem cells under tensile stress through the Wnt/PCP pathway by American Journal of Medical Genetics Part A, 136A(4), 358–362.
targeting Wnt11. Bone, 78, 130–141. Mefford, H. C., Shafer, N., Antonacci, F., Tsai, J. M., Park, S. S., Hing, A. V., . . .
Lisabeth, E. M., Falivelli, G., & Pasquale, E. B. (2013). Eph receptor signaling Cunningham, M. L. (2010). Copy number variation analysis in single-
and ephrins. Cold Spring Harbor Perspectives in Biology, 5(9), pii: suture craniosynostosis: Multiple rare variants including RUNX2
a009159. doi: 10.1101/cshperspect.a009159 duplication in two cousins with metopic craniosynostosis. American
Liu, B., Chen, S., Johnson, C., & Helms, J. A. (2014). A ciliopathy with Journal of Medical Genetics Part A, 152A(9), 2203–2210.
hydrocephalus, isolated craniosynostosis, hypertelorism, and clefting Merrill, A. E., Bochukova, E. G., Brugger, S. M., Ishii, M., Pilz, D. T., Wall, S. A.,
caused by deletion of Kif3a. Reproductive Toxicology, 48, 88–97. . . . Maxson, R. E., Jr. (2006). Cell mixing at a neural crest-mesoderm
Liu, S. S., Opperman, L. A., & Buschang, P. H. (2009). Effects of boundary and deficient ephrin-Eph signaling in the pathogenesis of
recombinant human bone morphogenetic protein-2 on midsagittal craniosynostosis. Human Molecular Genetics, 15(8), 1319–1328.
sutural bone formation during expansion. American Journal of Merzdorf, C. S., & Sive, H. L. (2006). The zic1 gene is an activator of Wnt
Orthodontics and Dentofacial Orthopedics, 136(6), 768 e1–8: discus- signaling. International Journal of Developmental Biology, 50(7),
sion 768–769. 611–617.
Liu, Y., Kadlub, N., da Silva Freitas, R., Persing, J. A., Duncan, C., & Shin, J. H. Merzdorf, C. S. (2007). Emerging roles for zic genes in early development.
(2008). The misdiagnosis of craniosynostosis as deformational plagio- Developmental Dynamics, 236(4), 922–940.
cephaly. Journal of Craniofacial Surgery, 19(1), 132–136. Meyers, G. A., Orlow, S. J., Munro, I. R., Przylepa, K. A., & Jabs, E. W. (1995).
Liu, Y. H., Tang, Z., Kundu, R. K., Wu, L., Luo, W., Zhu, D., . . . Maxson, R. E. Fibroblast growth factor receptor 3 (FGFR3) transmembrane mutation
(1999). Msx2 gene dosage influences the number of proliferative in Crouzon syndrome with acanthosis nigricans. Nature Genetics, 11(4),
osteogenic cells in growth centers of the developing murine skull: A 462–464.
possible mechanism for MSX2-mediated craniosynostosis in humans. Miller, D. T., Adam, M. P., Aradhya, S., Biesecker, L. G., Brothman, A. R.,
Developmental Biology, 205(2), 260–274. Carter, N. P., . . . Ledbetter, D. H. (2010). Consensus statement:
Loeys, B. L., & Dietz, H. C., (1993). In R. A. Pagon, M. P. Adam, H. H. Chromosomal microarray is a first-tier clinical diagnostic test for
Ardinger, S. E. Wallace, A. Amemiya, L. J. H. Bean, . . . K. Stephens, (Eds.), individuals with developmental disabilities or congenital anomalies. The
Loeys-Dietz Syndrome. GeneReviews Seattle (WA): University of American Journal of Human Genetics, 86(5), 749–764.
Washington, Seattle. Milne, T. A., Briggs, S. D., Brock, H. W., Martin, M. E., Gibbs, D., Allis, C. D., &
Lupski, J. R. (2012). Digenic inheritance and Mendelian disease. Nature Hess, J. L. (2002). MLL targets SET domain methyltransferase activity to
Genetics, 44(12), 1291–1292. Hox gene promoters. Molecular Cell, 10(5), 1107–1117.
Ma, G., Yu, J., Xiao, Y., Chan, D., Gao, B., Hu, J., . . . He, L. (2011). Indian Mishina, Y., & Snider, T. N. (2014). Neural crest cell signaling pathways
hedgehog mutations causing brachydactyly type A1 impair Hedgehog critical to cranial bone development and pathology. Experimental Cell
signal transduction at multiple levels. Cell Research, 21(9), 1343–1357. Research, 325(2), 138–147.
Maeno, T., Moriishi, T., Yoshida, C. A., Komori, H., Kanatani, N., Izumi, S., . . . Mitsui, N., Shimizu, K., Nishimoto, H., Mochizuki, H., Iida, M., & Ohashi, H.
Komori, T. (2011). Early onset of Runx2 expression caused craniosyn- (2013). Patient with terminal 9 Mb deletion of chromosome 9p:
ostosis, ectopic bone formation, and limb defects. Bone, 49(4), Refining the critical region for 9p monosomy syndrome with
673–682. trigonocephaly. Congenital Anomalies (Kyoto), 53(1), 49–53.
Magge, S. N., Westerveld, M., Pruzinsky, T., & Persing, J. A. (2002). Long- Moore, M. H., Abbott, A. H., Netherway, D. J., Menard, R., & Hanieh, A.
term neuropsychological effects of sagittal craniosynostosis on child (1998). Bilambdoid and posterior sagittal synostosis: The Mercedes
development. Journal of Craniofacial Surgery, 13(1), 99–104. Benz syndrome. Journal of Craniofacial Surgery, 9(5), 417–422.
Mansukhani, A., Ambrosetti, D., Holmes, G., Cornivelli, L., & Basilico, C. Morriss-Kay, G. M., & Wilkie, A. O. (2005). Growth of the normal skull vault
(2005). Sox2 induction by FGF and FGFR2 activating mutations inhibits and its alteration in craniosynostosis: Insights from human genetics and
Wnt signaling and osteoblast differentiation. Journal of Cell Biology, experimental studies. Journal of Anatomy, 207(5), 637–653.
168(7), 1065–1076. Morton, J. E., Frentz, S., Morgan, T., Sutherland-Smith, A. J., & Robertson,
Maruyama, T., Jeong, J., Sheu, T. J., & Hsu, W. (2016). Stem cells of the S. P. (2016). Biallelic mutations in CYP26B1: A differential diagnosis for
suture mesenchyme in craniofacial bone development, repair and Pfeiffer and Antley-Bixler syndromes. American Journal of Medical
regeneration. Nature Communications, 7, 10526. Genetics Part A, 179A, 2706–2710.
Mathijssen, I. M., & Arnaud, E. (2007). Benchmarking for craniosynostosis. Muenke, M., Gripp, K. W., McDonald-McGinn, D. M., Gaudenz, K.,
Journal of Craniofacial Surgery, 18(2), 436–442. Whitaker, L. A., Bartlett, S. P., . . . Zackai, E. H. (1997). A unique point
Mathijssen, I. M., van Splunder, J., Vermeij-Keers, C., Pieterman, H., de mutation in the fibroblast growth factor receptor 3 gene (FGFR3)
Jong, T. H., Mooney, M. P., & Vaandrager, J. M. (1999). Tracing defines a new craniosynostosis syndrome. The American Journal of
craniosynostosis to its developmental stage through bone center Human Genetics, 60(3), 555–564.
displacement. Journal of Craniofacial Genetics and Developmental Muenke, M., Schell, U., Hehr, A., Robin, N. H., Losken, H. W., Schinzel, A., . . .
Biology, 19(2), 57–63. Winter, R. M. (1994). A common mutation in the fibroblast growth
1426 | LATTANZI ET AL.

factor receptor 1 gene in Pfeiffer syndrome. Nature Genetics, 8(3), Passariello, A., De Brasi, D., Defferrari, R., Genesio, R., Tufano, M.,
269–274. Mazzocco, K., . . . Tonini, G. P. (2013). Constitutional 11q14-q22
Nakamura, T., Mori, T., Tada, S., Krajewski, W., Rozovskaia, T., Wassell, R., chromosome deletion syndrome in a child with neuroblastoma MYCN
. . . Canaani, E. (2002). ALL-1 is a histone methyltransferase that single copy. European Journal of Medical Genetics, 56(11), 626–634.
assembles a supercomplex of proteins involved in transcriptional Passos-Bueno, M. R., Serti Eacute, A. E., Jehee, F. S., Fanganiello, R., & Yeh,
regulation. Molecular Cell, 10(5), 1119–1128. E. (2008). Genetics of craniosynostosis: Genes, syndromes, mutations
Nakashima, K., Zhou, X., Kunkel, G., Zhang, Z., Deng, J. M., Behringer, R. R., and genotype-phenotype correlations. Frontiers of Oral Biology, 12,
& de Crombrugghe, B. (2002). The novel zinc finger-containing 107–143.
transcription factor osterix is required for osteoblast differentiation Paumard-Hernández, B., Berges-Soria, J., Barroso, E., Rivera-Pedroza, C. I.,
and bone formation. Cell, 108(1), 17–29. Pérez-Carrizosa, V., Benito-Sanz, S., . . . Heath, K. E. (2015). Expanding
Neuhäuser, G., Kaveggia, E. G., & Opitz, J. M. (1976). Studies of the mutation spectrum in 182 Spanish probands with craniosynostosis:
malformation syndromes of man XXXIX: A craniosynostosis-craniofa- Identification and characterization of novel TCF12 variants. European
cial dysostosis syndrome with mental retardation and other malforma- Journal of Human Genetics, 23(7), 907–914.
tions: “craniofacial dyssynostosis. European Journal of Pediatrics, 123(1), Persing, J. A., Jane, J. A., & Shaffrey, M. (1989). Virchow and the
15–28. pathogenesis of craniosynostosis: Atranslation of his original work.
Nguyen, C., Hernandez-Boussard, T., Khosla, R. K., & Curtin, C. M. (2013). Plastic and Reconstructive Surgery, 83(4), 738–742.
A national study on craniosynostosis surgical repair. Cleft Palate- Petrini, J., Damus, K., Russell, R., Poschman, K., Davidoff, M. J., & Mattison,
Craniofacial Journal, 50(5), 555–560. D. (2002). Contribution of birth defects to infant mortality in the United
Nieminen, P., Morgan, N. V., Fenwick, A. L., Parmanen, S., Veistinen, L., States. Teratology, 66(Suppl 1), S3–S6.
Mikkola, M. L., . . . Thesleff, I. (2011). Inactivation of IL11 signaling Piard, J., Rozé, V., Czorny, A., Lenoir, M., Valduga, M., Fenwick, A. L., . . .
causes craniosynostosis, delayed tooth eruption, and supernumerary Maldergem, L. V. (2015). TCF12 microdeletion in a 72-year-old woman
teeth. The American Journal of Human Genetics, 89(1), 67–81. with intellectual disability. American Journal of Medical Genetics Part A,
Noden, D. M. (1983). The role of the neural crest in patterning of avian 167A(8), 1897–1901.
cranial skeletal, connective, and muscle tissues. Developmental Biology, Plöger, F., Seemann, P., Schmidt-von Kegler, M., Lehmann, K., Seidel, J.,
96(1), 144–165. Kjaer, K. W., . . . Mundlos, S. Brachydactyly type A2 associated with a
Noden, D. M. (1986). Origins and patterning of craniofacial mesenchymal defect in proGDF5 processing. Human Molecular Genetics, 17(9),
tissues. Journal of Craniofacial Genetics and Developmental Biology, 2, 1222–1233.
15–31. Poisson, E., Sciote, J. J., Koepsel, R., Cooper, G. M., Opperman, L. A., &
Odell, J. M., Siebert, J. R., Bradley, C., & Salk, D. (1987). Duplication 7p in a Mooney, M. P. (2004). Transforming growth factor-beta isoform
family with t(7;11): Association with anomalies of the anterior cranial expression in the perisutural tissues of craniosynostotic rabbits. Cleft
base. American Journal of Medical Genetics, 27(3), 687–692. Palate-Craniofacial Journal, 41(4), 392–402.

Okada, H., & Gosain, A. K. (2012). Current approaches to management of Przylepa, K. A., Paznekas, W., Zhang, M., Golabi, M., Bias, W., Bamshad,
nonsyndromic craniosynostosis. Current Opinion in Otolaryngology & M. J., . . . Jabs, E. W. (1996). Fibroblast growth factor receptor 2
Head and Neck Surgery, 20(4), 310–317. mutations in Beare-Stevenson cutis gyrata syndrome. Nature Genetics,
13(4), 492–494.
Opperman, L. A., Fernandez, C. R., So, S., & Rawlins, J. T. (2006). Erk1/2
signaling is required for Tgf-beta 2-induced suture closure. Develop- Rabl, W., Tributsch, W., & Ambach, E. (1990). Premature craniosynostosis-
mental Dynamics, 235(5), 1292–1299. cause of sudden death in children and young adults. Beiträge zur
Gerichtlichen Medizin, 48, 217–221.
Opperman, L. A. (2000). Cranial sutures as intramembranous bone growth
sites. Developmental Dynamics, 219(4), 472–485. Reardon, W., Winter, R. M., Rutland, P., Pulleyn, L. J., Jones, B. M., &
Malcolm, S. (1994). Mutations in the fibroblast growth factor receptor 2
Padmalayam, D., Tubbs, R. S., Loukas, M., & Cohen-Gadol, A. A. (2013).
gene cause Crouzon syndrome. Nature Genetics, 8(1), 98–103.
Absence of the sagittal suture does not result in scaphocephaly. Childs
Nervous System, 29(4), 673–677. Reefhuis, J., Honein, M. A., Shaw, G. M., & Romitti, P. A. (2003). Fertility
treatments and craniosynostosis: California, Georgia, and iowa, 1993–
Paratore, C., Brugnoli, G., Lee, H. Y., Suter, U., & Sommer, L. (2002). The
1997. Pediatrics, 111(5 Part 2), 1163–1166.
role of the Ets domain transcription factor Erm in modulating
differentiation of neural crest stem cells. Developmental Biology, Reefhuis, J., & Honein, M. A. (2004). Maternal age and non-chromosomal
250(1), 168–180. birth defects, Atlanta-1968-2000: Teenager or thirty-something, who
is at risk? Birth Defects Research Part A: Clinical and Molecular Teratology,
Park, J., Park, O. J., Yoon, W. J., Kim, H. J., Choi, K. Y., Cho, T. J., & Ryoo,
70(9), 572–579.
H. M. (2012). Functional characterization of a novel FGFR2 mutation,
E731K, in craniosynostosis. Journal of Cellular Biochemistry, 113(2), Regelsberger, J., Delling, G., Tsokos, M., Helmke, K., Kammler, G., Kränzlein,
457–464. H., & Westphal, M. (2006). High-frequency ultrasound confirmation of
positional plagiocephaly. Journal of Neurosurgery, 105(5 Suppl),
Park, J. P., Graham, J. M., Jr, Berg, S. Z., & Wurster-Hill, D. H. (1988). A de
413–417.
novo interstitial deletion of chromosome 6 (q22.2q23.1). Clinical
Genetics, 33(2), 65–68. Reinhold, M. I., Kapadia, R. M., Liao, Z., & Naski, M. C. (2006). The Wnt-
inducible transcription factor Twist1 inhibits chondrogenesis. Journal of
Parker, S. E., Mai, C. T., Canfield, M. A., Rickard, R., Wang, Y., Meyer, R. E., . . .
Biological Chemistry, 281(3), 1381–1388.
National Birth Defects Prevention Network. (2010). Updated National
Birth Prevalence estimates for selected birth defects in the United Rhodes, J. L., Kolar, J. C., & Fearon, J. A. (2010). Mercedes benz pattern
States, 2004-2006. Birth Defects Research Part A Clinical and Molecular craniosynostosis. Plastic and Reconstructive Surgery, 125(1), 299–304.
Teratology, 88(12), 1008–1016. Rhodes, J. L., Tye, G. W., & Fearon, J. A. (2014). Craniosynostosis of the
Parsons, T. E., Weinberg, S. M., Khaksarfard, K., Howie, R. N., Elsalanty, M., lambdoid suture. Seminars in Plastic Surgery, 28(3), 138–143.
Yu, J. C., & Cray, J. J., Jr. (2014). Craniofacial shape variation in Twist1± Ribes, V., Otto, D. M., Dickmann, L., Schmidt, K., Schuhbaur, B., Henderson,
mutant mice. Anatomical Record (Hoboken), 297(5), 826–833. C., . . . Dollé, P. (2007). Rescue of cytochrome P450 oxidoreductase
LATTANZI ET AL.
| 1427

(Por) mouse mutants reveals functions in vasculogenesis, brain and limb Sharma, V. P., Fenwick, A. L., Brockop, M. S., McGowan, S. J., Goos, J. A.,
patterning linked to retinoic acid homeostasis. Developmental Biology, Hoogeboom, A. J., . . . 500 Whole-Genome Sequences (WGS500)
303(1), 66–81. Consortium. (2013). Mutations in TCF12, encoding a basic helix-loop-
Rice, D. P., Connor, E. C., Veltmaat, J. M., Lana-Elola, E., Veistinen, L., helix partner of TWIST1, are a frequent cause of coronal craniosynos-
Tanimoto, Y., . . . Rice, R. (2010). Gli3Xt-J/Xt-J mice exhibit lambdoid tosis. Nature Genetics, 45(3), 304–307.
suture craniosynostosis which results from altered osteoprogenitor Shetty, A. K., Thomas, T., Rao, J., & Vargas, A. (1998). Rickets and secondary
proliferation and differentiation. Human Molecular Genetics, 19(17), craniosynostosis associated with long-term antacid use in an infant.
3457–3467. Archives of Pediatrics and Adolescent Medicine, 152(12), 1243–1245.
Rice, R., Rice, D. P., Olsen, B. R., & Thesleff, I. (2003). Progression of calvarial Shiihara, T., Kato, M., Kimura, T., Hayasaka, K., Yamamori, S., & Ogata, T.
bone development requires Foxc1 regulation of Msx2 and Alx4. (2004). Craniosynostosis with extra copy of MSX2 in a patient with
Developmental Biology, 262(1), 75–87. partial 5q-trisomy. American Journal of Medical Genetics Part A, 128A(2),
Rivero-Garvía, M., Marquez-Rivas, J., Rueda-Torres, A. B., & Ollero-Ortiz, A. 214–216.
(2012). Early endoscopy-assisted treatment of multiple-suture cranio- Shimoji, T., Shimabukuro, S., Sugama, S., & Ochiai, Y. (2002). Mild
synostosis. Childs Nervous System, 28(3), 427–431. trigonocephaly with clinical symptoms: Analysis of surgical results in
Rogers, G. F. (2011). Deformational plagiocephaly, brachycephaly, and 65 patients. Childs Nervous System, 18(5), 215–224.
scaphocephaly. Part I: Terminology, diagnosis, and etiopathogenesis. Shipster, C., Hearst, D., Somerville, A., Stackhouse, J., Hayward, R., & Wade,
Journal of Craniofacial Surgery, 22(1), 9–16. A. (2003). Speech, language, and cognitive development in children
Rojnueangnit, K., & Robin, N. H. (2013). Craniosynostosis and radial ray with isolated sagittal synostosis. Developmental Medicine and Child
defect: A rare presentation of 22q11.2 deletion syndrome. American Neurology, 45(1), 34–43.
Journal of Medical Genetics Part A, 161A(8), 2024–2026. Shukla, D. (2016). Mercedes benz craniosynostosis. Journal of Neuro-
Rottgers, S. A., Lohani, S., & Proctor, M. R. (2016). Outcomes of endoscopic sciences in Rural Practice, 7(1), 176.
suturectomy with postoperative helmet therapy in bilateral coronal Shukla, V., Coumoul, X., Wang, R. H., Kim, H. S., & Deng, C. X. (2007). RNA
craniosynostosis. Journal of Medical Genetics, 18(3), 281–286. interference and inhibition of MEK-ERK signaling prevent abnormal
Sanchez-Lara, P. A., Carmichael, S. L., Graham, J. M., Jr, Lammer, E. J., Shaw, skeletal phenotypes in a mouse model of craniosynostosis. Nature
G. M., Ma, C., & Rasmussen, S. A. National Birth Defects Prevention Genetics, 39(9), 1145–1150.
Study. (2010). Fetal constraint as a potential risk factor for Simanovsky, N., Hiller, N., Koplewitz, B., & Rozovsky, K. (2009).
craniosynostosis. American Journal of Medical Genetics Part A, 152A(2), Effectiveness of ultrasonographic evaluation of the cranial sutures in
394–400. children with suspected craniosynostosis. European Radiology, 19(3),
Sauerhammer, T. M., Patel, K., Oh, A. K., Proctor, M. R., Mulliken, J. B., & 687–692.
Rogers, G. F. (2014). Combined metopic and unilateral coronal Snider, T. N., & Mishina, Y. (2014). Cranial neural crest cell contribution to
synostoses: A phenotypic conundrum. Journal of Craniofacial Surgery, craniofacial formation, pathology, and future directions in tissue
25(2), 437–440. engineering. Birth Defects Research Part C Embryo Today, 102(3),
Sauerhammer, T. M., Seruya, M., Ropper, A. E., Oh, A. K., Proctor, M. R., & 324–332.
Rogers, G. F. (2014). Craniectomy gap patency and neosuture formation Spazzapan, P., Arnaud, E., Baujat, G., Nizon, M., Malan, V., Brunelle, F., & Di
following endoscopic suturectomy for unilateral coronal craniosynos- Rocco, F. (2016). Clinical and neuroradiological features of the 9p
tosis. Plastic and Reconstructive Surgery, 134(1), 81e–91e. deletion syndrome. Childs Nervous System, 32(2), 327–335.
Schell, U., Hehr, A., Feldman, G. J., Robin, N. H., Zackai, E. H., de Die- Spruijt, L., Verdyck, P., Van Hul, W., Wuyts, W., & de Die-Smulders, C.
Smulders, C., . . . Muenke, M. (1995). Mutations in FGFR1 and FGFR2 (2005). A novel mutation in the MSX2 gene in a family with foramina
cause familial and sporadic Pfeiffer syndrome. Human Molecular parietalia permagna (FPP). American Journal of Medical Genetics Part A,
Genetics, 4(3), 323–328. 139A(1), 45–47.
Schmidt, K., Hughes, C., Chudek, J. A., Goodyear, S. R., Aspden, R. M., Stamper, B. D., Mecham, B., Park, S. S., Wilkerson, H., Farin, F. M., Beyer,
Talbot, R., . . . Tickle, C. (2009). Cholesterol metabolism: The main R. P., . . . Cunningham, M. L. (2012). Transcriptome correlation analysis
pathway acting downstream of cytochrome P450 oxidoreductase in identifies two unique craniosynostosis subtypes associated with IRS1
skeletal development of the limb. Molecular and Cellular Biology, 29(10), activation. Physiological Genomics, 44(23), 1154–1163.
2716–2729. Starr, J. R., Collett, B. R., Gaither, R., Kapp-Simon, K. A., Cradock, M. M.,
Schoenebeck, J. J., Hutchinson, S. A., Byers, A., Beale, H. C., Carrington, B., Cunningham, M. L., & Speltz, M. L. (2012). Multicenter study of
Faden, D. L., . . . Ostrander, E. A. (2012). Variation of BMP3 contributes neurodevelopment in 3-Year-Old children with and without single-
to dog breed skull diversity. PLoS Genetics, 8(8), e1002849. Suture craniosynostosis. Archives of Pediatrics and Adolescent Medicine,
Schweitzer, T., Böhm, H., Meyer-Marcotty, P., Collmann, H., Ernestus, 166(6), 536–542.
R. I., & Krauß, J. (2012). Avoiding CT scans in children with Stiffel, V., Amoui, M., Sheng, M. H., Mohan, S., & Lau, K. H. (2014). EphA4
single-suture craniosynostosis. Childs Nervous System, 28(7), receptor is a novel negative regulator of osteoclast activity. Journal of
1077–1082. Bone and Mineral Research, 29(4), 804–819.
Selber, J., Reid, R. R., Chike-Obi, C. J., Sutton, L. N., Zackai, E. H., McDonald- Styrkarsdottir, U., Cazier, J. B., Kong, A., Rolfsson, O., Larsen, H.,
McGinn, D., . . . Bartlett, S. P. (2008). The changing epidemiologic Bjarnadottir, E., . . . Stefansson, K. (2003). Linkage of osteoporosis to
spectrum of single-suture synostoses. Plastic and Reconstructive chromosome 20p12 and association to BMP2. PLoS Biology, 1(3), E69.
Surgery, 122(2), 527–533. Suga, K., Saitoh, M., Kokubo, S., Fukushima, S., Kaku, S., Yasuda, S., &
Seto, M. L., Hing, A. V., Chang, J., Hu, M., Kapp-Simon, K. A., Patel, P. K., . . . Miyata, K. (2003). Interleukin-11 acts synergistically with bone
Cunningham, M. L. (2007). Isolated sagittal and coronal craniosynosto- morphogenetic protein-2 to accelerate bone formation in a rat ectopic
sis associated with TWIST box mutations. American Journal of Medical model. Journal of Interferon & Cytokine Research, 23(4), 203–207.
Genetics Part A, 143A(7), 678–886. Suhasini, A. N., & Brosh, R. M., Jr, (2013). Disease-causing missense
hapiro, R., & Robinson, F. (1976). Embryogenesis of the human occipital mutations in human DNA helicase disorders. Mutation Research, 752(2),
bone. American Journal of Roentgenology, 126(5), 1063–1068. 138–152.
1428 | LATTANZI ET AL.

Swinkels, M. E., Simons, A., Smeets, D. F., Vissers, L. E., Veltman, J. A., Twigg, S. R., & Wilkie, A. O. (2015). New insights into craniofacial
Pfundt, R., . . . van Ravenswaaij-Arts, C. M. (2008). Clinical and malformations. Human Molecular Genetics, 24(R1), R50–R59.
cytogenetic characterization of 13 Dutch patients with deletion 9p
Ursitti, F., Fadda, T., Papetti, L., Pagnoni, M., Nicita, F., Iannetti, G., & Spalice,
syndrome: Delineation of the critical region for a consensus phenotype.
A. (2011). Evaluation and management of nonsyndromic craniosynos-
American Journal of Medical Genetics Part A, 146A(11), 1430–1438.
tosis. Acta Paediatrica, 100(9), 1185–1194.
Tabler, J. M., Barrell, W. B., Szabo-Rogers, H. L., Healy, C., Yeung, Y.,
van der Meulen, J., van der Hulst, R., van Adrichem, L., Arnaud, E., Chin-
Perdiguero, E. G., . . . Liu, K. J. (2013). Fuz mutant mice reveal shared
Shong, D., Duncan, C., . . . Renier, D. (2009). The increase of metopic
mechanisms between ciliopathies and FGF-related syndromes. Devel-
synostosis: A pan-European observation. Journal of Craniofacial Surgery,
opmental Cell, 25(6), 623-635.
20(2), 283–286.
Tagariello, A., Heller, R., Greven, A., Kalscheuer, V. M., Molter, T., Rauch, A.,
van der Meulen, J. (2012). Metopic synostosis. Childs Nervous System,
. . . Winterpacht, A. (2006). Balanced translocation in a patient with
28(9),1359–1367.
craniosynostosis disrupts the SOX6 gene and an evolutionarily
conserved non-transcribed region. Journal of Medical Genetics, 43(6), Van Maldergem, L., Siitonen, H. A., Jalkh, N., Chouery, E., De Roy, M.,
534–540. Delague, V., . . . Verloes, A. (2006). Revisiting the craniosynostosis-
radial ray hypoplasia association: Baller-Gerold syndrome caused by
Tahiri, Y., Bastidas, N., McDonald-McGinn, D. M., Birgfeld, C., Zackai, E. H.,
mutations in the RECQL4 gene. Journal of Medical Genetics, 43(2),
Taylor, J., & Bartlett, S. P. (2015). New pattern of sutural synostosis
148–152.
associated with TWIST gene mutation and saethre-Chotzen syndrome:
Peace sign synostosis. Journal of Craniofacial Surgery, 26(5), 1564–1567. Vannier, M. W., Hildebolt, C. F., Marsh, J. L., Pilgram, T. K., McAlister, W. H.,
Shackelford, G. D., . . . Knapp, R. H. (1989). Craniosynostosis: Diagnostic
Tamburrini, G., Caldarelli, M., Massimi, L., Santini, P., & Di Rocco, C. (2005).
value of three-dimensional CT reconstruction. Radiology, 173(3),
Intracranial pressure monitoring in children with single suture and
669–673.
complex craniosynostosis: A review. Childs Nervous System, 21(10),
913–921. Varvagiannis, K., Stefanidou, A., Gyftodimou, Y., Lord, H., Williams, L., Sarri,
C., . . . Petersen, M. B. (2013). Pure de novo partial trisomy 6p in a girl
Thompson, D. N., Harkness, W., Jones, B., Gonsalez, S., Andar, U., &
with craniosynostosis. American Journal of Medical Genetics Part A,
Hayward, R. (1995). Subdural intracranial pressure monitoring in
161A(2), 343–351.
craniosynostosis: Its role in surgical management. Childs Nervous
System, 11(5), 269–275. Virchow R. 1851. Ueber den Cretinismus, namentlich in Franken, und ueber
pathologische Schadelformen. Verhandlungen Physikalisch-Medizinische
Thompson, D. N., Malcolm, G. P., Jones, B. M., Harkness, W. J., & Hayward,
Gesellschaft Würzburg, 2 230.
R. D. (1995). Intracranial pressure in single-suture craniosynostosis. The
New England Journal of Medicine, 22(5), 235–240. Vissers, L. E., Cox, T. C., Maga, A. M., Short, K. M., Wiradjaja, F., Janssen,
I. M., . . . Roscioli, T. (2011). Heterozygous mutations of FREM1 are
Timberlake, A. T., Choi, J., Zaidi, S., Lu, Q., Nelson-Williams, C., Brooks, E. D.,
associated with an increased risk of isolated metopic craniosynostosis
. . . Lifton, R. P. (2016). Two locus inheritance of non-syndromic midline
in humans and mice. PLoS Genetics, 7(9), e002278.
craniosynostosis via rare SMAD6 and common BMP2 alleles. Elife, 8(5),
e20125. Wang, X. P., O'Connell, D. J., Lund, J. J., Saadi, I., Kuraguchi, M., Turbe-
Doan, A., . . . Maas, R. L. (2009). Apc inhibition of Wnt signaling
Ting, M. C., Wu, N. L., Roybal, P. G., Sun, J., Liu, L., Yen, Y., & Maxson, R. E., Jr
regulates supernumerary tooth formation during embryogenesis and
(2009). EphA4 as an effector of Twist1 in the guidance of osteogenic
throughout adulthood. Development, 136(11), 1939–1949.
precursor cells during calvarial bone growth and in craniosynostosis.
Development, 136(5), 855–864. Wang, Y., Zhou, X., Oberoi, K., Phelps, R., Couwenhoven, R., Sun, M., . . .
Jabs, E. W. (2012). P38 Inhibition ameliorates skin and skull
Tsuji, K., Narahara, K., Yokoyama, Y., Grzeschik, K. H., & Kunz, J. (1995). The
abnormalities in Fgfr2 Beare-Stevenson mice. Journal of Clinical
breakpoint on 7p in a patient with t(6;7) and craniosynostosis is
Investigation, 122(6), 2153–2164.
spanned by a YAC clone containing the D7S503 Locus. Human Genetics,
95(3), 303–307. Warren, S. M., Brunet, L. J., Harland, R. M., Economides, A. N., & Longaker,
M. T. (2003). The BMP antagonist noggin regulates cranial suture
Tubbs, R. S., Elton, S., Blount, J. P., & Oakes, W. J. (2001). Preliminary
fusion. Nature, 422(6932), 625–629.
observations on the association between simple metopic ridging in
children without trigonocephaly and the Chiari I malformation. The New Weber, I., Ninkovic, M., Janecke, A. R., Utermann, B., Witsch Baumgartner,
England Journal of Medicine, 35(3), 136–139. M., Anderl, H., & Utermann, G. (2001). Molecular analyisis of 74 patients
Twigg, S. R., Forecki, J., Goos, J. A., Richardson, I. C., Hoogeboom, A. J., van with craniosynostosis. European Journal of Human Genetics, 9(Sup. 1),
den Ouweland, A. M., . . . WGS500 Consortium. (2015). Gain-of- P0409.
Function mutations in ZIC1 are associated with Coronal craniosynos- Wehby, G. L., & Cassell, C. H. (2010). The impact of orofacial clefts on
tosis and learning disability. The American Journal of Human Genetics, quality of life and healthcare use and costs. Oral Diseases, 16(1), 3–10.
97(3), 378–388. Weiss, J., Kotelchuck, M., Grosse, S. D., Manning, S. E., Anderka, M.,
Twigg, S. R., Kan, R., Babbs, C., Bochukova, E. G., Robertson, S. P., Wall, S. A., Wyszynski, D. F., . . . Higgins, C. (2009). Hospital use and associated
. . . Wilkie, A. O. (2004). Mutations of ephrin-B1 (EFNB1), a marker of costs of children aged zero-to-two years with craniofacial malforma-
tissue boundary formation, cause craniofrontonasal syndrome. Pro- tions in Massachusetts. Birth Defects Research Part A: Clinical and
ceedings of the National Academy of Sciences of the United States of Molecular Teratology, 85(11), 925–934.
America, 101(23), 8652–8657. Wilkie, A. O., Bochukova, E. G., Hansen, R. M., Taylor, I. B., Rannan-Eliya,
Twigg, S. R., Vorgia, E., McGowan, S. J., Peraki, I., Fenwick, A. L., Sharma, S. V., Byren, J. C., . . . Lester, T. (2007). Clinical dividends from the
V. P., . . . Wilkie, A. O. (2013). Reduced dosage of ERF causes complex molecular genetic diagnosis of craniosynostosis. American Journal of
craniosynostosis in humans and mice and links ERK1/2 signaling to Medical Genetics Part A, 143A(16), 1941–1949.
regulation of osteogenesis. Nature Genetics, 45(3), 308–313. Wilkie, A. O., Byren, J. C., Hurst, J. A., Jayamohan, J., Johnson, D., Knight,
Twigg, S. R., & Wilkie, A. O. (2015). A genetic-pathophysiological S. J., . . . Wall, S. A. (2010). Prevalence and complications of single-gene
framework for craniosynostosis. The American Journal of Human and chromosomal disorders in craniosynostosis. Pediatrics, 126(2),
Genetics, 97(3), 359–377. e391–e400.
LATTANZI ET AL.
| 1429

Wilkie, A. O., Slaney, S. F., Oldridge, M., Poole, M. D., Ashworth, G. J., Yoon, S. J., Wills, A. E., Chuong, E., Gupta, R., & Baker, J. C. (2011). HEB and
Hockley, A. D., . . . Reardon, W. (1995). Apert syndrome results from E2A function as SMAD/FOXH1 cofactors. Genes & Development,
localized mutations of FGFR2 and is allelic with Crouzon syndrome. 25(15), 1654–1661.
Nature Genetics, 9(2), 165–172. Yu, B. D., Hess, J. L., Horning, S. E., Brown, G. A., & Korsmeyer, S. J. (1995).
Wilkie, A. O., Tang, Z., Elanko, N., Walsh, S., Twigg, S. R., Hurst, J. A., . . . Altered Hox expression and segmental identity in Mll-mutant mice.
Maxson, R. E., Jr. (2000). Functional haploinsufficiency of the human Nature, 378(6556), 505–508.
homeobox gene MSX2 causes defects in skull ossification. Nature Yu, H. M., Jerchow, B., Sheu, T. J., Liu, B., Costantini, F., Puzas, J. E., . . . Hsu,
Genetics, 24(4), 330–331. W. (2005). The role of Axin2 in calvarial morphogenesis and
Wilkie, A. O. (1997). Craniosynostosis: Genes and mechanisms. Human craniosynostosis. Development, 132(8), 1995–2005.
Molecular Genetics, 6(10), 1647–1656. Yu, J. C., Lucas, J. H., Fryberg, K., & Borke, J. L. (2001). Extrinsic tension
Williams, H. (2008). Lumps, bumps and funny shaped heads. Archives of results in FGF-2 release, membrane permeability change, and
Disease in Childhood. Education and Practice, 93(4), 120–128. intracellular Ca + + increase in immature cranial sutures. Journal of
Xiao, G., Jiang, D., Gopalakrishnan, R., & Franceschi, R. T. (2002). Fibroblast Craniofacial Surgery, 12(4), 391–398.
growth factor 2 induction of the osteocalcin gene requires MAPK Zeiger, J. S., Beaty, T. H., Hetmanski, J. B., Wang, H., Scott, A. F., Kasch, L.,
activity and phosphorylation of the osteoblast transcription factor, . . . VanderKolk, C. (2002). Genetic and environmental risk factors for
Cbfa1/Runx2. Journal of Biological Chemistry, 277(39), 36181–36187. sagittal craniosynostosis. Journal of Craniofacial Surgery, 13(5),
Yagnik, G., Drissi, H., Stevens, C., & Boyadjiev, S. (2010). RUNX2 602–606.
Polymorphisms in Patients with Nonsyndromic Sagittal Craniosynos- Zhang, X., Kuroda, S., Carpenter, D., Nishimura, I., Soo, C., Moats, R., . . .
tosis. Proceedings of the 60th Annual Meeting of The American Society Ting, K. (2002). Craniosynostosis in transgenic mice overexpressing
of Human Genetics, November 6, Washington DC. Nell-1. Journal of Clinical Investigation, 110(6), 861–870.
Yagnik, G., Ghuman, A., Kim, S., Stevens, C. G., Kimonis, V., Stoler, J., . . . Zhao, H., & Chai, Y. (2015). Stem cells in teeth and craniofacial bones. J Dent
Boyadjiev, S. A. (2012). ALX4 gain-of-function mutations in Res, 94(11), 1495–1501.
nonsyndromic craniosynostosis. Human Mutation, 33(12), 1626–1629. Zneimer, S. M., Cotter, P. D., & Stewart, S. D. (2000). Telomere-
Yamamoto, T., Sameshima, K., Sekido, K., Aida, N., Matsumoto, N., telomere (end to end) fusion of chromosomes 7 and 22 with an
Naritomi, K., & Kurosawa, K. (2006). Trigonocephaly in a boy with interstitial deletion of chromosome 7p11. 2–>p15. 1: Phenotypic
paternally inherited deletion 22q11. 2 syndrome. American Journal of consequences and possible mechanisms. Clinical Genetics, 58(2),
Medical Genetics Part A, 140A(12), 1302–1304. 129–133.
Yan, Y., Tang, D., Chen, M., Huang, J., Xie, R., Jonason, J. H., . . . Chen, D.
(2009). Axin2 controls bone remodeling through the beta-catenin-BMP
signaling pathway in adult mice. Journal of Cell Science, 122(Pt 19), How to cite this article: Lattanzi W, Barba M, Pietro LD,
3566–3578.
Boyadjiev SA. Genetic advances in craniosynostosis. Am J
Ye, X., Guilmatre, A., Reva, B., Peter, I., Heuzé, Y., Richtsmeier, J. T., . . .
Med Genet Part A. 2017;173A:1406–1429. https://doi.org/
Romitti, P. A. (2016). Mutation screening of candidate genes in patients
with nonsyndromic sagittal craniosynostosis. Plastic and Reconstructive
10.1002/ajmg.a.38159
Surgery, 137(3), 952–961.

You might also like