Skip to main content
Erschienen in: Italian Journal of Pediatrics 1/2017

Open Access 01.12.2017 | Review

Fragile X syndrome: a review of clinical and molecular diagnoses

verfasst von: Claudia Ciaccio, Laura Fontana, Donatella Milani, Silvia Tabano, Monica Miozzo, Susanna Esposito

Erschienen in: Italian Journal of Pediatrics | Ausgabe 1/2017

Abstract

Background

Fragile X Syndrome (FXS) is the second cause of intellectual disability after Down syndrome and the most prevalent cause of intellectual disability in males, affecting 1:5000–7000 men and 1:4000–6000 women. It is caused by an alteration of the FMR1 gene, which maps at the Xq27.3 band: more than 99% of individuals have a CGG expansion (>200 triplets) in the 5′ UTR of the gene, and FMR1 mutations and duplication/deletion are responsible for the remaining (<1%) molecular diagnoses of FXS. The aim of this review was to gather the current clinical and molecular knowledge about FXS to provide clinicians with a tool to guide the initial assessment and follow-up of FXS and to offer to laboratory workers and researchers an update about the current diagnostic procedures.

Discussion

FXS is a well-known condition; however, most of the studies thus far have focused on neuropsychiatric features. Unfortunately, some of the available studies have limitations, such as the paucity of patients enrolled or bias due to the collection of the data in a single-country population, which may be not representative of the average global FXS population. In recent years, insight into the adult presentation of the disease has progressively increased. Pharmacological treatment of FXS is essentially symptom based, but the growing understanding of the molecular and biological mechanisms of the disease are paving the way to targeted therapy, which may reverse the effects of FMRP deficiency and be a real cure for the disease itself, not just its symptoms.

Conclusions

The clinical spectrum of FXS is wide, presenting not only as an isolated intellectual disability but as a multi-systemic condition, involving predominantly the central nervous system but potentially affecting any apparatus. Given the relative high frequency of the condition and its complex clinical management, FXS appears to have an important economic and social burden.
Abkürzungen
ACMG
American College of Medical Genetics
ACOG
American Congress of Obstetricians and Gynecologists
ADHD
Anxiety disorder/hyperactivity disorder
ASD
Autism spectrum disorder
CTDs
Connective tissue disorders
FM
Full mutation
FXS
Fragile X syndrome
FXTAS
Fragile X-associated tremor/ataxia syndrome
GZ
Grey-zone
ID
Intellectual disability
IQ
Intellective quotient
mGluRs
Metabotropic glutamate receptors
MRI
Magnetic resonance imaging
MS-LR-PCR
Methylation-sensitive long-range PCR
PBLs
Peripheral blood lymphocytes
PM
Permutation
POI
Primary ovarian failure
SSRIs
Selective serotonin reuptake inhibitors
XCI
X chromosome inactivation

Background

Fragile X Syndrome (FXS, OMIM #300624), also known as Martin-Bell Syndrome, was first described in 1943 by Martin and Bell as a form of intellectual disability (ID) following an X-linked inheritance pattern [1]. In 1969, Lubs first reported a distinct fragile site on the X chromosome that segregated with ID in 3 generations of a family, and in 1991, the association of the Xq27.3 fragile site with X-linked ID was confirmed [2, 3]. Therefore, FXS was defined as a clinical and cytogenetic entity and acquired its current name. Currently, it is known to be the second cause of ID after Down Syndrome (2.4% of all IDs), the first cause of inherited ID and the most prevalent cause of ID in males [4, 5]. The actual worldwide prevalence is estimated to range between 1:5000–7000 men and 1:4000–6000 women [5].
The diagnosis of FXS is based on the detection of an alteration of the Fragile X Mental Retardation-1 gene (FMR1), which maps at the Xq27.3 band [2]. More than 99% of individuals with FXS have an FMR1 loss-of-function caused by an increased number of CGG trinucleotide repeats in the 5′ untranslated (5′ UTR) region (typically >200 triplets). This allelic constitution is called a full mutation (FM) and produces the expression of the cytogenetic fragile site (FRAXA). Its result is a hypermethylated state of the FMR1 promoter, with consequent inhibition of FMR1 transcription and loss or heavy reduction of the protein product (FMRP). Therefore, the FXS phenotype is a direct consequence of the absence of FMRP; different types of FMR1 alterations (deletions encompassing the gene, intragenic deletions/duplications, single-nucleotide variants) are responsible for the remaining (<1%) molecular diagnoses of FXS [6].
The normal number of repeats within the FMR1 gene ranges from 5 to 44; a repeat number of 45–54 is considered to be a grey zone. A repeat number of 55–200 is called pre-mutation (PM), and it is associated with pathological conditions that differ from FXS : premature ovarian failure (POI) in females and fragile X - associated tremor/ataxia syndrome (FXTAS) in males (less frequently also in females) [6]. Given the phenotype breadth of the FMR variations, typical of trinucleotide expansion disease, we chose here to provide a review that is limited to the complete FXS phenotype, which affects individuals carrying a FM allele.
FXS inheritance does not follow a Mendelian pattern, but it depends on the number of trinucleotide repeats within the promoter of the FMR1 gene [6]: a transition from the PM to FM allele can occur because of the expansion phenomenon during the transmission of the maternal (very rarely of the paternal) X chromosome carrying a PM to her children [3, 4]. The frequency of individuals with the PM allele in the total population is approximately 1:850 for males and 1:257–300 for females [5, 7] - i.e., one in 300 females randomly chosen among the general population can potentially generate an affected male child.
Affected men have a typical phenotype, characterized by ID, long face, large and protruding ears, and macroorchidism [4, 6, 8]. Females heterozygous for the FM allele have a 30% chance of having a normal intelligence quotient and a 25% chance of having ID with an IQ < 70; nonetheless, they can present learning deficits and emotional difficulties [4]. The phenotype in females is strongly connected to the X inactivation (XCI) pattern.
However, FXS is much more than a simple ID; it is a multi-systemic condition that can potentially affect any apparatus because FMRP is widely expressed. The aim of this review was to gather the current clinical and molecular knowledge about FXS to provide clinicians with a tool to guide the initial assessment and follow-up of FXS and to offer laboratory workers and researchers an update regarding the current diagnostic procedures.

Discussion

General clinical features

Generally, prenatal and neonatal diagnoses are not possible with a negative family history because of the lack of ultrasound and clinical findings. At birth, the height, weight, and head circumference of FXS children are within the normal range [9]. The most prominent clinical features of the condition are summarized in Table 1. The height and weight seem to follow the normal growth curves; otherwise, the head circumference tends to reach the higher centiles: in prepubertal age, the majority of FXS children develop macrocephaly, with a head circumference larger than the 50th percentile [4, 6, 9]. The facial characteristics become more distinctive in early childhood, when the patients start showing a long narrow face and prominent ears [4, 9, 10]. This latter sign is one of the hallmarks of FXS, but it is often a relative parameter, as the narrowness of the faces of some affected males exaggerates their ear prominence [10]. Other reported facial features are the prominence of the jaw, a high-arched palate, puffiness around the eyes, long palpebral fissures, closely spaced eyes, epicanthal folds, strabismus, flat nasal bridge, broad nose, broad philtrum, and facial hypotonia (demonstrated by slackness of the lower jaw) [4, 6, 810] (Fig. 1). Notably, not all facial features are recognizable at a young age (most have been recorded only after puberty), and approximately 30% of young children with FXS will not have obvious dysmorphic features [6, 10].
Table 1
Clinical features of FXS males [4, 7, 9, 11, 13, 17, 18, 24, 26, 27, 32, 34, 36, 37, 51]
Features
Frequency
Face
Long/Narrow face
83%
Macrocephaly
81%
Prominent ears
72–78%
High-arched palate
94%
Prominent jaw
80%
Facial hypotonia
NA
Eye puffiness
NA
Closely spaced eyes
NA
Long palpebral fissures
NA
Epicanthal folds
NA
Flat nasal bridge
NA
Broad nose
NA
Broad philtrum
NA
Central nervous system
EEG anomalies
74%
Epilepsy
10–20%
Brain MRI anomalies
Up to 50% of patients with neurologic comorbidities
Neuropsychiatric involvement
Psychomotor delay
~100%
ID
~100%
Aggressiveness
90%
Attention problems
74–84%
Hyperactivity
50–66%
Anxiety Disorder
58–86%
ASD
30–50%
Sleep problems
30%
ADHD
12–23%
Depression
8–12%
Musculoskeletal system
Joint hypermobility
50%
Pectus excavatum
50%
Flat feet
29–69%
Spine deformity
6–9%
Cardiovascular
Mitral valve anomalies
3–12%
Aortic root dilatation
25%
Eye
Refractive errors
17–59%
Strabismus
8–40%
Nystagmus
5–13%
Other
Macroorchidism
63–95%
Obesity/overweight
53–61%
Recurrent otitis media
47–97%
Gastrointestinal complaints
31%
Soft skin
NA
Abbreviations: NA not available, ID intellectual disability, ASD autism spectrum disorder, ADHD anxiety disorder/hyperactivity disorder
The most important clinical abnormality associated with defects of the FMR1 gene is global developmental delay/ID. The psychomotor delay involves both walking age (mean = 2,12 years) and age at first words (mean = 2,43 years) [11]. Both males and females with FXS present a wide range of learning disabilities in the context of normal, borderline IQ or mild to severe ID [12]. The IQ of males with FM varies with studies, with a mean value of 40–51 [1113]; 68% of FM males have an IQ score lower than 50, while 18% have a score above 70 [11]. The IQ score directly correlates with the level of FMRP production: higher levels of FMRP are found in individuals with an IQ above 70, showing only moderate emotional and learning difficulties [4, 14, 15]. Similarly, those individuals with “size-mosaicism” (full mutation plus premutation, grey zone or normal alleles) have higher IQs than those without mosaicism [15]. Females with FM present a wider range of phenotypic characteristics than men, depending on the XCI pattern: 70% of FM women present with some degree of cognitive impairment [4].

Neurological features

An important comorbidity in FXS is epilepsy. Reports have suggested a prevalence of seizures among FXS children, present in 10–20% in boys and 5–10% in girls [13, 16, 17]. Complex partial seizures have been reported to be the most frequent among FXS patients with epilepsy (89.3%) [13, 16], followed by generalized tonic-clonic seizures (46.4%), and simple partial seizures (25%). The latter type is always associated with another type of epilepsy; febrile convulsions have been reported in 7.1% of patients with epileptic seizures [13]. The age of onset is usually between 2 and 10 years, and this comorbidity typically disappears with growth, although 25% of FXS patients continue to have epilepsy into their adult years [13, 16]. Seizures have usually a low frequency of recurrence and sometimes manifest themselves following intercurrent infections or exposure to other environmental factors [13]. Epilepsy usually has a good response to therapy [13, 16]. Most patients control their seizures with antiepileptic drugs (AEDs); only 7% of the patients need more than one drug, and 10% of the patients do not need any therapy [13].
Independently from epilepsy, patients with FXS also have a higher prevalence of EEG abnormalities (74%) [13, 18]; these abnormal EEG findings, however, may not always manifest with seizures and/or a subsequent diagnosis of epilepsy. In a study by Hear et al., 47% of FXS patients exhibited slowing of the posterior dominant rhythm for age, and 42% had focal spikes from various anatomic regions [18]. Nevertheless, consistent with seizure remission with age, 35% of children showed normalization of the EEG background after the age of 8 years, and when present, they are more often nonspecific and limited to only one location [13, 18].
MRI is usually normal [13, 18]. When anomalies are found, these are more frequently diffuse atrophy and cortical thickness, increased whole hemispheric and lobar cortical volume, and increased cortical complexity [13, 19]. These aspects are consistent with the decreased pruning and increased spine density and length and with the presence of an immature spine, as reported in FXS patients and mice [1921].
Further reported findings are atrophy of the cerebellar vermis, thinning of the corpus callosum, hippocampal anomalies, enlarged fourth ventricle, lacunar infarction of the basal ganglia, and mesial temporal sclerosis; the latter describes only cases of refractory seizures due to recurrent prolonged episodes of status epilepticus [13, 18]. A recent study by Hall et al. discovered increased fractional anisotropy in patients with FXS in the left and right inferior longitudinal fasciculus, right uncinate fasciculus and left cingulum hippocampus compared with that in controls; additionally, this aspect could be attributed to the aberrant pruning and axon growth dysregulation, resulting from FMRP reduction [22]. All of these MRI anomalies in brain morphology correlate negatively with cognitive performance in FXS children [19].

Neuropsychiatric features

Over the years, FXS has been associated with several neuropsychiatric and neuropsychological phenotypes, showing that ID is rarely presented alone in this disease. Psychomotor delay is the first sign of an upcoming ID in the scholar age, and it is quite an early finding. The developmental profile of infants with FXS deviate from that of the general population by 6 months of age, involving all domains of development (fine motor, visual reception, expressive communication, and receptive communication) [23]. The average functional level of male patients shows an improving trend until the age of 25, even remaining below that of the general population. Then, patients enter a relatively stable phase until the age of 50; at that time, the skills of FXS males begin to worsen [24].
FXS patients are considered to be at a high risk of developing one or more neuropsychiatric disorders. An association between autism and FXS was first noted in the early 1980s, and a growing number of reports of further neuropsychiatric conditions emerged in the following years. Among those, the associations FXS/autism and FXS/Anxiety Disorder Hyperactivity Disorder (ADHD) are the most studied [11, 17, 2533].
The autism-like presentation of many FXS males is known since the earliest study on the psychological characterization of the syndrome [25]. It is estimated that 30–50% males and 25% females with FXS have an Autism Spectrum Disorder (ASD) as a comorbidity [26, 27, 30]. Some studies suggested that there is an age-related improvement in some but not all ASD symptoms across adulthood for FXS men [27, 28]: autism is diagnosed in approximately 49% of children but 41% of adolescents/adults [27]. Nevertheless, the ASD-related impairment seems to be less severe in FXS individuals than in those with non-syndromic ASD [29]. In recent years, the change in the diagnostic criteria for ASD, due to the transition from DSM-IV TR to DSM-5 in 2013, is modifying this rate of incidence because the prevalence of ASD diagnoses is lower using the DSM-5 criteria across all age and sex groups [28, 30]. Only 50% of males diagnosed with ASD using the DSM-IV-TR parameter still fulfil the ASD criteria using the DSM-5. This gap is even broader for females; only 30% of FXS girls meet an ASD diagnosis changing from DSM-IV-TR to DSM-5 [30]. Anyway, given the high prevalence of autistic features in FXS and the fact that sometimes it represents the only sign of the syndrome, all children affected by ASD, especially boys, should be tested for FMR1 [6].
ADHD is considered one of the most common comorbidities in FXS, with more than one-half of male patients fulfilling the diagnostic criteria at some point in their lives. The prevalence of ADHD spectrum symptoms is 54–59%, a higher rate than that in individuals with isolated ID or different neuropsychiatric disorders [31]. The complete diagnostic criteria of ADHD are fulfilled by 12–23% of the FXS subjects [11, 32]. Preschool boys seem not to differ from typically developing controls in the mean level of ADHD symptoms and reach their peak at school age (5–6 years) [32].
As part of the ADHD spectrum, isolated hyperactivity also has a high incidence, with 50–66% of FXS children being affected [11, 17, 27, 33]; attention problems are well represented too, with an overall prevalence of 74–84% [17, 27]. Anxiety disorder is another frequent trait of FXS subjects, with a prevalence that largely varies with studies in the range of 58–86% [17, 27, 34]. Cordeiro et al. demonstrated that in a group of 58 males and 39 females with FXS aged 5–33 years, 86.2% of males and 76.9% of females met the criteria for at least one anxiety disorder [34]. Both anxiety and attention problems seem to follow an increasing trend with age [27].
The same trend also characterizes depression, which is prevalent in adolescents/adults rather than in children and has an overall prevalence of 8–12% in FXS individuals [17, 27]. This may not always manifest, but it can be revealed by withdrawal or increased aggression rather than by sadness, anhedonia, or irritability [26].
Other common neuropsychiatric conditions are the following: pervasive developmental disorder [11, 23], stereotypies (mostly hand/finger mannerisms) [11, 35], sleep problems [36], specific or social phobias [34], selective mutism [34], restricted interests [35], compulsive and ritualistic/sameness behaviours [35], self-injurious behaviour [35], and aggressiveness [27]. This latter aspect has been fully examined recently by Wheeler et al., who demonstrate that 90% of individuals with FXS, both males and females, were reported to be engaged in at least one aggressive act in the previous 12 months [37]. Sleep disorders affect approximately 30% of FXS children of both sexes and manifest themselves as difficulties falling asleep, frequent night-time awakenings, and early awakening in the morning [36].
It is then clear that aside from ID, the neuropsychiatric/neuropsychological profile of FXS is complex, and it has been demonstrated that overall, when using DSM-IV-TR criteria, 73% of FXS patients can be diagnosed with at least one axis I psychiatric disorder [11]. This incidence can indeed be underestimated because the assessment of psychiatric symptoms in patients with FXS is often complicated by limitations in the accuracy of self-reporting and insight, atypical manifestation of some symptoms in the context of ID, and the relative lack of validated assessment tools. For example, limited expressive language and social reciprocity impairments often prevent a reliable communication of symptoms. Diagnostic overshadowing occurs when psychiatric symptoms are not appreciated as a co-morbid problem in a patient with ID but are attributed only to the disability itself [26].

A real syndrome: multi-systemic involvement

The neurologic/neuropsychiatric presentation certainly is the hallmark of the syndrome, but FXS also shows an association of various medical problems that may or may not be present; however, when manifested, it can worsen the phenotype and complicate the clinical management of these patients.
Since the first reports of FXS, it has been clear that the condition shares some features with the connective tissue disorder (CTD) spectrum. Although a specific abnormality of the connective tissue has not yet been shown, the prevalence of connective tissue signs has suggested that there is an underlying connective tissue anomaly, perhaps similar to that observed in CTDs (in particular, Marfan Syndrome and Ehlers-Danlos Syndrome); the precise association between FXS and signs of connective tissue abnormality still awaits biochemical and molecular explanation [38]. The skin can be soft [24, 39, 40], and joint hypermobility is present in about half of the patients, affecting predominantly the small joints (mostly metacarpal-phalangeal joints) [6, 24, 39, 40]. Skeletal signs may include a high-arched palate, scoliosis, pectus excavatum, and flat feet [6, 9, 24, 39, 40].
Connective tissue fragility also involves the heart because FXS patients can develop cardiac defects similar to those observed in CTDs. Recurrent findings are aortic root dilatation (approximately 25% of the patients) and mitral valve prolapse (3–50%) [9, 3840]; this latter feature is frequent in the general population but has a higher prevalence in CTDs. Hyperarousal (i.e., faster heart rate) and reduced parasympathetic vagal tone have also been documented [41]. In adult age (>40 years), FXS patients tend to develop the common cardiovascular problems shared by the age-matched general population, such as hypertension (24.2%) and heart rhythm disorders (24.2%) [42].
The function of the gastrointestinal system in individuals with FXS has not been well studied yet. Given the presence of connective tissue signs, hypotonia and connective tissue anomalies could contribute to some of the gastrointestinal problems reported in this condition, such as gastro-oesophageal reflux, constipation, and loose bowel movements. In a study by Utari et al. including FXS males and females aged 40–71 years, a prevalence of gastrointestinal problems of 30.6% was reported [42]. Nonetheless, the literature lacks studies documenting an effective increased incidence of gastrointestinal involvement compared with that in the general population.
The genitourinary system seems to be affected only in males, where pubertal macroorchidism is considered a hallmark of the condition, shared by 80-95% of adults, but it is less common in prepubertal boys [4, 6, 8, 9, 39, 40]. In adult FXS men, the mean testicular volume is approximately 50 mL (normal mean testicular volume: <25 mL) [8]. Given the presence of ID, most FXS men do not have any real fertility complaints, even if 1% of them have been reported to reproduce [4].
Ocular anomalies are known since the first reports of the condition and affect at least 25% of FXS children and a greater number of FXS adults [43, 44]. Strabismus and refractive errors have a higher prevalence in FXS than in the general population [6, 8, 24, 45]: that prevalence, considerably variable with studies, is 8–40% for strabismus and 17–59% for refractive errors (primarily hyperopia and astigmatisms, but myopia is also reported) [8, 24, 43, 44, 46]. Nystagmus has been identified as a less rare finding (5–13%) [43, 46]; other observable ocular features include palpebral ptosis and convergence insufficiency [46].
FXS children tend to have recurrent otitis media, which may lead to conductive hearing loss [6, 8, 9, 11]. These patients already have poor expressive language skills, so it becomes fundamental that any possible otologic problem is promptly treated to avoid interference with speech improvement [47]. It has been reported that language skills are better among children who did not have recurrence of this complication [48].
Metabolic problems are common and well reported, with obesity and overweight being quite frequent in both sexes [6, 8, 42]. Studies conducted in FXS adults reported an incidence of 53–61% for obesity/overweight [42, 49]. Males are more frequently overweight, while women tend to reach obesity [42]. Furthermore, in males with FXS, the serum levels of HDL are shifted to lower numbers across all age ranges, but conversely, their triacylglycerol levels are higher than those of the general population [49, 50].

Differential diagnosis

The differential diagnosis of FXS includes syndromic forms of ID but also non-syndromic psychomotor delays/ID. The differential diagnosis includes Sotos Syndrome, Prader-Willi Syndrome, Klinefelter Syndrome, and FRAXE [6, 51, 52]. These conditions share the following phenotypic features with FXS:
  • Sotos Syndrome: ID, macrocephaly, behavioural problems, and epilepsy [6, 51, 53]
  • Prader-Willi Syndrome: developmental delay, elements of facial appearance, sucking problems in neonatal age, obesity, and genital anomalies [6, 51, 54]
  • Klinefelter Syndrome: ID (20%) and genital anomalies [51]
  • FRAXE: ID (generally milder than FXS), language impairment, hyperactivity, and autistic behaviour [6, 51].
Angelman and Rett Syndromes may also be considered for the differential diagnosis, even if their typical presentation differs from that of FXS children. Shared features are ID, language impairment, and autistic behaviour. An Array-CGH can be performed to exclude cytogenetic rearrangements responsible for ID. When genetic testing is not helpful, isolated ID, autism, or ADHD must be considered [52].

The FMR1 gene and FMRP: from triplet expansion to pathology

FXS is associated with a rare fragile and unsteady site on Xq27. This site, named FRAXA, was originally observed as a non-staining gap, break or constriction in the metaphase chromosomes placed under selective culture conditions, such as folic acid or thymidine deprivation [2, 55, 56]. In 1991, an association between FXS and alterations of the FMR1 gene was identified, located at the FRAXA locus [3]. The FMR1 gene product, FMRP, is involved in the regulation of post-transcriptional RNA metabolism, playing an important role in synaptic plasticity, dendrite and axon development, and underlying learning and memory. FMRP acts as an organizer of both mRNA transport (shuttle protein), targets mRNA translation (RNA-binding protein) and is involved in a feedback loop by controlling its own local protein levels [57].
The absence of FMRP derives, in most cases, from a dynamic mutation consisting of variable expansion of a trinucleotide (CGG) repeat in the 5′ UTR of the FMR1 gene. The size of the CGG repeat in normal individuals ranges between 5 and 44, and it is usually stably transmitted throughout generations. Alleles with 45–54 repeats are defined as intermediate, borderline or “grey-zone” (GZ). Carriers of GZ alleles do not show an FXS phenotype but can present with peripheral neuropathy, ataxia, anxiety and/or depression, and clinical symptoms similar to those of Parkinson patients, including bradykinesia, rigidity, memory complaints, and a positive response to dopaminergic medications [58]. PM carriers have a number of repeats that ranges from 55 to 200. PM disorders were first identified in 1991 with the discovery of an increased incidence of early menopause (prior to the age of 40 years) in female carriers [59]. Twenty percent of PM females manifest an FMR1-related POI [6], while FXTAS occurs in PM males (rarely in females) and is characterized by late-onset, progressive cerebellar ataxia and intention tremor [6, 60]. The necessary findings to confirm an FXTAS diagnosis are an FMR1 PM associated with an MRI showing white matter lesions in the middle cerebellar peduncles and/or brain stem (the major neuroradiologic sign) and intention tremor or gait ataxia (the two major clinical signs) [6]. The prevalence of FXTAS is estimated to be 40–45% overall for males and 16% for females with PM older than 50 years [60, 61]. The incomplete penetrance of POI and FXTAS phenotypes makes it difficult to predict whether a carrier would develop one of the PM-associated conditions. A major role in determining this penetrance is certainly due to environmental and genetic/epigenetic factors that may influence the susceptibility to phenotype expression. These latter include CGG repeat length, FMR1 mRNA concentration, XCI, translation of the repeat sequence, and any genomic asset able to influence FMR1 expression [62]. In recent years, it has emerged that a low-normal repeat number may also have important clinical implications: a CGG number below 26 can be considered a risk factor for cognition disability and mental health problems [63]. Therefore, there is growing awareness that there is a fine homeostatic equilibrium of FMRP expression levels, so that both high and low numbers of CGG repeats could alter brain function and alleles previously considered benign (<26 or 45–54) may lead to neuropsychiatric manifestations; anyway those are preliminary data that still need to be confirmed by additional studies. In FXS patients, the CGG trait is expanded (FM), with a dimension greater than 200 repeats. This expansion results in transcription silencing and the consequent absence of FMRP, due to hypermethylation of the CpG islands adjacent to the expanded trinucleotide repeats and heterochromatin conformation of the FMR1 promoter region. Conversely, GZ and PM alleles are unmethylated.
The risk of a PM allele becoming an FM allele is correlated with the number of CGG trinucleotide repeats, with nearly all alleles with ≥100 repeats expanding to FM in the next generation when transmitted by the mother; when the PM is carried by the father, small increases in the trinucleotide repeats may occur in meiosis but typically do not result in FM [6, 64, 65]. It has been estimated that 66% of maternal and paternal PM alleles change by one or more repeats in the offspring, even though with a significant difference in number of repeat expansions among maternal and paternal transmission: for alleles with <70 repeats, paternal alleles have a greater likelihood for instability; however, for larger alleles, maternal transmissions are more often unstable [64]. Approximately 17% of intermediate alleles are unstable, and maternal GZ alleles may evolve in PM (but not in FM) in future generations [6, 65]. Predictably, GZ alleles ranging from 50 to 54 repeats are less stable than those with <50 repeats and can more often evolve into PM ones [65]. Regarding normal-range alleles, 0.2% of maternal and 1.5% of paternal alleles exhibit an increasing size upon transmission [65].
Beyond CGG repeat size, one of the major factors influencing FMR1 stability is the presence of AGG triplets interspersed within the FMR1 repeated region. One or two AGG interruptions are usually interposed with CGG repeats in FMR1 (most commonly on the 5′ end of the repeat tract), and this occurs in 94% of the general population alleles [66]. Conversely, FXS alleles containing long stretches of uninterrupted CGG triplets at the 3′ end usually do not show any AGG at the 5′ end [66]. Maternal alleles with no AGGs have the greatest risk for FM expansion, while the presence of even a single AGG significantly reduces this risk, especially for alleles with <70 repeats. When the repeat number exceeds 70 triplets, the allele shows high instability even when AGG interruptions are present, and when the CGGs expand beyond 90, AGG interruptions do not have any ability to block the triplet growth [64].
A decrease in CGG repeat number through generations, although rare, is also possible: retractions from FM to PM and from PM to normal size have both been widely reported, while retraction from FM to normal size appears to be sporadic [6769]. In a study by Nolin et al., by screening 1040 FMR1 pedigrees, repeat contraction had been observed in 2.3% of maternal and 5.7% of paternal transmissions [64]; previous work of the same research group had already shown a similar rate of contraction for maternal PM alleles (3,1%) [65]. Retraction occurs post zygotically because of an excision of a variable number of trinucleotides, resulting in a mosaic normal size/GZ/PM/FM [68, 69]. Generally, mosaics have a higher percentage of the larger allele [69].
Mosaicism is a source of phenotypic variability, and it can be observed in both sexes, with a higher incidence in males. The prevalence of mosaicism in FXS males largely varies among studies, in the range of 12–41% [6, 67]. Both repeat-size mosaicism (e.g., FM/PM, FM/GZ) and methylation mosaicism have been described; in the latter, FM has varying degrees of methylation from tissue to tissue [6, 6870]. Somatic FM/deletion mosaicism has also been reported because deletion can occur mitotically during embryonic cell divisions, usually before the 11th week, with the result of two distinct subpopulations of cells carrying the deletion and FM alleles [71].
In addition to the FMR1 promoter expansion, a small number of FXS cases (<1%) are caused by mutation in the coding region or deletion of the FMR1 gene [6]. Mutations account for approximately 4% of FXS patients meeting the clinical criteria for FXS but with a normal range of CGG repeats [72]. Point, missense, nonsense, frameshift, and UTR region mutations have all been described [7275]. Some of the variants lately identified still require investigation to definitively classify them as pathological because the FMRP is sometimes still present to some degree in carriers [73]. In line with this genotype-phenotype correlation, a recent study by Teckan et al. showed that approximately 30–50% of all FMR1 missense SNPs could be associated with diseases using an in silico approach [75].
Another important issue to consider, when approaching a diagnosis in females, is X chromosome inactivation (XCI), consisting of the silencing of one of the two X chromosomes in mammalian females. Normally, XCI occurs randomly; however, under particular conditions, preferential XCI can be observed. In the case of FMR1 mutation carriers, if the mutated chromosome is preferentially inactivated, FMRP is produced by the normal allele, and the resulting phenotype would be less severe [76].

Diagnostic procedures

Initially, the diagnosis of FXS was based on the cytogenetic evaluation of the presence of FRAXA in peripheral blood lymphocytes (PBLs). However, this procedure was time consuming and difficult to interpret, required specific technical skills and was also unable to distinguish between FRAXA and the other neighbouring fragile sites on Xq [55]. To overcome some of these limitations and improve the detection rate of FRAXA, fluorescence in situ hybridization (FISH) with DNA probes was then introduced [77].
Cytogenetic analyses were replaced by Southern blot analysis of DNA from peripheral blood after digestion with specific restriction endonucleases [78], and finally by PCR. Southern blot analysis can detect all FMR1 alleles, including normal, PM, and FM, and can determine the methylation status of the FMR1 promoter region; however, it is time consuming, relatively expensive, and similarly to the previously used approaches, difficult to interpret. Standard PCR plus Southern blot analysis has been considered the gold standard for FMR1 molecular diagnosis for a long time, even if it provides a low-resolution estimation of the repeat number [66, 79, 80]. Standard PCR, based on the direct amplification of the CGG-repeat using flanking primers, is faster and highly sensitive to detect FMR1 repeats in the normal and PM range; however, it could only reveal alleles with up to ~300 repeats in males and up to ~160 repeats in females and it therefore fails to identify the large CGG expansions (e.g. more than 300 CGGs) [81].
The limitations of the PCR plus southern Blot technique lead to the development of new PCR-based procedures able to detect all FMR1 alleles. Triplet primed PCR (TP-PCR) was designed: it is a procedure in which the forward PCR primer is located upstream the CGG region and the other overlaps the CGG repeat and the adjacent unique sequence; after PCR cycles, the CGG repeat number can be determined by fragment sizing of PCR amplicons using capillary electrophoresis [66, 82]. TP-PCR is the evolution of previous PCR protocols; this procedure allows the simultaneous amplification of both the full-length FMR1 alleles (using PCR primers flanking the repeated region) and CGG triplets (using a third primer, complementary to the FMR1 triplet repeat region) in the same PCR reaction. TP-PCR-based procedures became the gold standard for the first level assessment of FXS and can detect the expanded allele even in mosaic fashion. This PCR based kit also allows the detection of AGG interruptions.
As the second-level analysis in the diagnostic flow-chart, CGG methylation testing can be performed to evaluate FMRP silencing. Dedicated kits, such as the Methylation-Sensitive Long-Range PCR (MS-LR-PCR) kit, have been developed to measure the methylation fraction of each FMR1 allele, using DNA after digestion with methylation-sensitive restriction enzymes [83]. This approach is also very useful to identify those rare FM unmethylated males that are asymptomatic carriers of a pathologic allele [83].
Finally, the finding of the loss of function mutations of FMR1 as causative of FXS prompted the development of specific molecular techniques. FMR1 sequence analysis and MLPA must therefore be offered to patients with a clinical phenotype highly suggestive of FXS but with a normal range of CGG repeats. Additional testing to identify intragenic deletions or duplications is required when PCR amplification fails, suggesting a possible exonic or whole-gene anomaly [6].

Prenatal FMR1 testing

FXS molecular tests are usually performed postnatally on PBLs in the presence of the appropriate clinical criteria described in the clinical section. Moreover, it is also possible to perform a prenatal test of FXS using LR-PCR-based protocols on DNA from either chorionic villi or amniocytes. Currently, according to ACMG (American College of Medical Genetics) and ACOG (American Congress of Obstetricians and Gynaecologists) guidelines, FMR1 prenatal testing should be offered to couples with a personal or familial history of the following:
1.
FXS- or FX-related disorders
 
2.
Unexplained ID or developmental delay
 
3.
Isolated cognitive impairment
 
4.
Autism
 
5.
Idiopathic familiar POI or elevated FSH at age <40 years
 
6.
Isolated cerebellar ataxia with tremor
 
In addition, given the high incidence of FXS in the general population, a consistent number of genetic health professionals have supported prenatal testing for all women who request the analysis, regardless of their personal/familial history [84]. However, it should be taken into consideration that FMR1 test interpretation could be complicated by different problems; in particular, the presence of GZ alleles is very difficult to be interpreted and counselled because the risk of expansion, although very low, cannot be excluded [76]. The prenatal findings of FM females are also complex because the XCI pattern can modify the clinical phenotype. Moreover, the presence of post-zygotic mosaicism can complicate the diagnosis. This phenomenon can generate false results, especially in prenatal screening, because the clinical phenotype of the proband would not be available until birth.
Finally, it has been suggested that GZ and PM carriers with a positive family history of FX-associated disorders are at higher risk of expansion [65]. This is at least partly due to the presence and number of AGG interruptions in the parental allele. AGG trinucleotide repeats genotyping can be therefore offered to determine the number and location of AGG trinucleotide interruptions within the tract of CGG repeats of FMR1, particularly in female carriers of a GZ or a small PM allele [6, 65, 85].
Considering all of these issues, for couples who request FXS screening, the ideal test should be proposed in the preconception period: this could be advantageous because the couple will be allowed to make conscious reproductive decisions. In addition, the parents would also receive useful information for their personal health. Indeed, the identification of premutated females (which are at risk for POI) may allow a more effective reproductive intervention in those desiring a pregnancy.
Newborn screening for FXS has been proposed [86, 87], even if its application remains controversial. Although FXS is a relatively frequent disorder, and there would be the possibility to apply a quite sensitive and specific test, a real benefit of testing is lacking because there is no specific therapy currently to treat children, and treatment is essentially symptom based. In 2014, Godler et al. proposed a screening protocol where FMR1 methylation analysis would be used as the first-line test to selectively identify only FM carriers, followed by a triple-primed CGG-based test to confirm the positive results [87]. Nonetheless, to date, newborn screening for FXS is not performed in any country. The quest for a blood-based biomarker of FXS is currently underway [74]. Finding a simple and low-cost biochemical test would pave the way to include an FXS test in routine newborn screening.

Patient management: therapeutic strategies and social issues

FXS emerges as a complex disease with a primary neuropsychiatric involvement but potentially affects more than one apparatus, therefore needing a large-scale intervention able to address all the physical, psychological and social implications of the disease. Treatment plans should be individualized based on the symptoms and age-related comorbidities of each individual. Speech and language therapy must be recommended to children, especially in that with early diagnosis. Most males show in fact moderate to severe delay in communication skills, while the communication skills of females are considerably less affected [48]. Behavioural therapy is another useful tool that helps normalize some of the symptoms, and it has been reported to be an effective approach for aggression in 71% of patients [37]. Therapy techniques that have been validated for autism are often helpful for FXS but must be modified based on the particular ASD features in FXS phenotype [88]. Physical therapy is needed in some cases, and occupational therapy must be offered to adults. A study by Martin et al., collecting males and females with FXS aged 0–63, showed that 72% of males and 47% of females receive one or more non-pharmacological therapy service [89]. The most common service for both males and females is speech–language therapy (68% males, 42% females), followed by occupational therapy (59% males, 30% females), physical therapy (25% males, 10% females), and behaviour management therapy (21% males, 6% females). Overall, males are more likely to receive therapy services as well as a greater number of services than females. All these strategies are mostly adopted during childhood and an age-related decline of therapies occurs for both males and females, with the use of services peaking by 11 years and essentially non-existent after the age of 20 [89].
Pharmacologic therapy may be recommended to improve behaviour problems (such as aggression, anxiety, hyperactivity, problems with impulse control, and poor attention span) and also to treat more severe disorders, such as ADHD or depression [8, 52]. The most common classes of medication used by FXS patients are selective serotonin reuptake inhibitors (SSRIs) and atypical anti-psychotics, with SSRIs use being more common among females than males [90]. In approximately 60% of patients, drug treatment for psychological issues helped the remission of some symptoms; in particular, stimulants can be used to improve attention and hyperactivity, SSRIs for anxiety, alpha-agonists for hyperactivity and overarousal, and antipsychotics for irritable and aggressive behaviours [91]. Drugs should anyway always be combined with non-pharmacological strategy to reach the best outcome. For ADHD in particular, it has been demonstrated that the early diagnosis and treatment play a role in improving both concurrent and long-term social functioning [33].
Lithium has been proposed as possible treatment, and in 2008, a pilot add-on trial has been conducted to evaluate the safety and efficacy of this drug in FXS patients. The results showed a significant improvement in hyperactivity, inappropriate speech, aggression, abnormal vocalizations, self-abuse, work refusal, outbursts, over-emotionality, anxiety, mood swings, tantrums, perseveration, crying, and maladaptive behaviour after 2 months of treatment [92]. Scores on the Clinical Global Improvement Scale were also significantly enhanced, and positive responses were distributed across all the age ranges of the study cohort, suggesting that both children and young adults with FXS can benefit from lithium treatment [92].
New targeted treatments for FXS (mGluR5 antagonists, GABA A and B agonists, minocycline) are now being studied. In Fmr1 knockout (KO) mice, the glutamatergic receptors signalling and/or localization is enhanced. New studies that try to reduce excitatory neurotransmission by antagonism of group I metabotropic glutamate receptors (mGluRs), particularly mGluR5, are still underway [93]. By now, studies conducted on Fmr1 KO mice demonstrated that the reduction of mGluR5 levels can normalize protein synthesis, dendritic spines, and some behaviour [94]. In FXS, an insufficient inhibitory GABAergic function has also been reported. A positive modulation of GABA(A) receptors has been demonstrated to improve some behavioural and neurophysiological alterations in Fmr1 KO mice [95]. Drugs modulating GABA signalling could therefore be an effective therapy. Minocycline is an FDA-approved treatment for acne and is known to have inhibitory effects on matrix metalloproteinase-9 activity. It is currently under study for FXS treatment and seems to have its greatest effect in young children, where it strengthens synaptic connections and enhances cognitive development [96].
Moreover, specific pharmacological intervention is needed to address common complications, such as epilepsy, metabolic disorders and hypertension.
Going beyond treatment, an important issue to consider in the management of FXS patients is the psychological health of their parents and caregivers; both have to take care of these complicated patients in different ways, and both have been proven to suffer psychological consequences.
Although a diagnosis of FXS is beneficial to the family for establishing the reason why a child has cognitive deficits and/or behavioural problems, all the family members have to cope with the disease and the derived stress. Mental and physical health problems especially affect families with higher stress levels, and these families often have an impaired ability to manage the difficult behaviours and physical problems of their children. This leads to an increased need for intervention with caregivers at the family level [37]. On the other hand, caregivers of patients with FXS are more likely to develop comorbidities such as anxiety, depression, stress, and sleep disorders than the caregivers of patients without FXS and are also more likely to receive medications for these conditions [97, 98].
Despite the relatively low incidence of FXS, this disease has a significant impact on parents, caregivers and entire society within which the patient lives. The poor social functioning of FXS individuals, together with the need for medication and non-pharmacological intervention, has important consequences in terms of the costs and resources employed. The percentage of working FXS patients varies largely with studies, in the range of 20–70%, with females being more frequently employed than males [97, 98]. Predictably, the higher functioning employed group of FXS patients also has lower direct healthcare costs [98]. It has been estimated that the economic burden of FXS in Europe is significant, with a mean annual cost per patient reaching up to €58,862 [97]. The patient cost is even higher in the USA, where it has been estimated to be as high as $14,677 every month [98].

Conclusions

FXS is a well-known condition; however, most of the studies thus far have focused on its neuropsychiatric features. This review was aimed to provide an extensive overview of both the clinical and molecular features of this syndrome. Unfortunately, some of the studies cited in this work have limitations, such as the paucity of patients enrolled and the bias due to the collection of data in a single-country population, which may not be representative of the average global FXS population. Most of the studies collected focus on FXS children; however, in recent years, insight into the adult presentation of the disease has progressively increased. Furthermore, only a few studies have been conducted including non-Caucasian populations. Pharmacological treatment in FXS is essentially symptom based, but the growing understanding of the molecular and biological mechanisms of the disease are paving the way to targeted therapy, which might reverse the effects of FMRP deficiency and be a real cure for the disease itself, not just its symptoms.

Acknowledgements

We thank the patient and the parents of the patient for their kind consent for image publication. We also thank Dr. Arianna Tucci and Dr. Giulietta Scuvera for their support during the writing of the manuscript.

Funding

This review was partially supported by a grant received from the Italian Ministry of Health (Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico – Fonndi Ricerca Corrente 2016 850/01).

Availability of data and materials

The data and materials used are included in the review.

Authors’ contributions

CC, LF and ST wrote the first draft of the manuscript. DM, MM and SE supervised and critically revised the manuscript. All authors have read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.
Informed consent has been obtained from the parents of the patient for image publication.
All the studies mentioned in this review have been approved by the Ethics Committee, and written informed consent has been obtained by all the participants.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
3.
Zurück zum Zitat Verkerk AJ, Pieretti M, Sutcliffe JS, Fu YH, Kuhl DP, Pizzuti A, Reiner O, Richards S, Victoria MF, Zhang F, Eussen BE, van Ommen GJB, Blonden LAJ, Riggins GJ, Chastain JL, Kunst CB, Galjaard H, Caskey CT, Nelson DL, Oostra BA, Warren ST. Identification of a gene (FMR-1) containing a CGG repeat coincident with a breakpoint cluster region exhibiting length variation in fragile X syndrome. Cell. 1991;65(5):905–14.PubMedCrossRef Verkerk AJ, Pieretti M, Sutcliffe JS, Fu YH, Kuhl DP, Pizzuti A, Reiner O, Richards S, Victoria MF, Zhang F, Eussen BE, van Ommen GJB, Blonden LAJ, Riggins GJ, Chastain JL, Kunst CB, Galjaard H, Caskey CT, Nelson DL, Oostra BA, Warren ST. Identification of a gene (FMR-1) containing a CGG repeat coincident with a breakpoint cluster region exhibiting length variation in fragile X syndrome. Cell. 1991;65(5):905–14.PubMedCrossRef
4.
Zurück zum Zitat Hagerman RJ, Hagerman PJ. Fragile X Syndrome: diagnosis, treatment and research. Baltimore: The John Hopkins University Press; 2002.CrossRef Hagerman RJ, Hagerman PJ. Fragile X Syndrome: diagnosis, treatment and research. Baltimore: The John Hopkins University Press; 2002.CrossRef
5.
Zurück zum Zitat Hunter J, Rivero-Arias O, Angelov A, Kim E, Fotheringham I, Leal J. Epidemiology of fragile X syndrome: a systematic review and meta-analysis. Am J Med Genet A. 2014;164A(7):1648–58.PubMedCrossRef Hunter J, Rivero-Arias O, Angelov A, Kim E, Fotheringham I, Leal J. Epidemiology of fragile X syndrome: a systematic review and meta-analysis. Am J Med Genet A. 2014;164A(7):1648–58.PubMedCrossRef
7.
Zurück zum Zitat Cronister A, Teicher J, Rohlfs EM, Donnenfeld A, Hallam S. Prevalence and instability of fragile X alleles: implications for offering fragile X prenatal diagnosis. Obstet Gynecol. 2008;111(3):596–601.PubMedCrossRef Cronister A, Teicher J, Rohlfs EM, Donnenfeld A, Hallam S. Prevalence and instability of fragile X alleles: implications for offering fragile X prenatal diagnosis. Obstet Gynecol. 2008;111(3):596–601.PubMedCrossRef
8.
Zurück zum Zitat Hersh JH, Saul RA. Committee on Genetics. Health supervision for children with fragile X syndrome. Pediatrics. 2011;127(5):994–1006.PubMedCrossRef Hersh JH, Saul RA. Committee on Genetics. Health supervision for children with fragile X syndrome. Pediatrics. 2011;127(5):994–1006.PubMedCrossRef
9.
Zurück zum Zitat Lachiewicz AM, Dawson DV, Spiridigliozzi GA. Physical characteristics of young boys with fragile X syndrome: reasons for difficulties in making a diagnosis in young males. Am J Med Genet. 2000;92(4):229–36.PubMedCrossRef Lachiewicz AM, Dawson DV, Spiridigliozzi GA. Physical characteristics of young boys with fragile X syndrome: reasons for difficulties in making a diagnosis in young males. Am J Med Genet. 2000;92(4):229–36.PubMedCrossRef
10.
Zurück zum Zitat Heulens I, Suttie M, Postnov A, De Clerck N, Perrotta CS, Mattina T, Faravelli F, Forzano F, Kooy RF, Hammond P. Craniofacial characteristics of fragile X syndrome in mouse and man. Eur J Hum Genet. 2013;21(8):816–23.PubMedCrossRef Heulens I, Suttie M, Postnov A, De Clerck N, Perrotta CS, Mattina T, Faravelli F, Forzano F, Kooy RF, Hammond P. Craniofacial characteristics of fragile X syndrome in mouse and man. Eur J Hum Genet. 2013;21(8):816–23.PubMedCrossRef
11.
Zurück zum Zitat Alanay Y, Unal F, Turanli G, Alikaşifoğlu M, Alehan D, Akyol U, Belgin E, Sener C, Aktaş D, Boduroğlu K, Utine E, Volkan-Salanci B, Ozusta S, Genç A, Başar F, Sevinç S, Tunçbilek E. A multidisciplinary approach to the management of individuals with fragile X syndrome. J Intellect Disabil Res. 2007;51(Pt2):151–61.PubMedCrossRef Alanay Y, Unal F, Turanli G, Alikaşifoğlu M, Alehan D, Akyol U, Belgin E, Sener C, Aktaş D, Boduroğlu K, Utine E, Volkan-Salanci B, Ozusta S, Genç A, Başar F, Sevinç S, Tunçbilek E. A multidisciplinary approach to the management of individuals with fragile X syndrome. J Intellect Disabil Res. 2007;51(Pt2):151–61.PubMedCrossRef
13.
Zurück zum Zitat Musumeci SA, Hagerman RJ, Ferri R, Bosco P, Dalla Bernardina B, Tassinari CA, De Sarro GB, Elia M. Epilepsy and EEG findings in males with fragile X syndrome. Epilepsia. 1999;40(8):1092–9.PubMedCrossRef Musumeci SA, Hagerman RJ, Ferri R, Bosco P, Dalla Bernardina B, Tassinari CA, De Sarro GB, Elia M. Epilepsy and EEG findings in males with fragile X syndrome. Epilepsia. 1999;40(8):1092–9.PubMedCrossRef
14.
Zurück zum Zitat Kaufmann WE, Abrams MT, Chen W, Reiss AL. Genotype, molecular phenotype, and cognitive phenotype: correlations in fragile X syndrome. Am J Med Genet. 1999;83:286–95.PubMedCrossRef Kaufmann WE, Abrams MT, Chen W, Reiss AL. Genotype, molecular phenotype, and cognitive phenotype: correlations in fragile X syndrome. Am J Med Genet. 1999;83:286–95.PubMedCrossRef
15.
Zurück zum Zitat Schneider A, Seritan A, Tassone F, Rivera SM, Hagerman R, Hessl D. Psychiatric features in high-functioning adult brothers with fragile X spectrum disorders. Prim Care Companion CNS Disord. 2013;15:2. Schneider A, Seritan A, Tassone F, Rivera SM, Hagerman R, Hessl D. Psychiatric features in high-functioning adult brothers with fragile X spectrum disorders. Prim Care Companion CNS Disord. 2013;15:2.
16.
Zurück zum Zitat Berry-Kravis E, Raspa M, Loggin-Hester L, Bishop E, Holiday D, Bailey DB. Seizures in fragile X syndrome: characteristics and comorbid diagnoses. Am J Intellect Dev Disabil. 2010;115(6):461–72.PubMedCrossRef Berry-Kravis E, Raspa M, Loggin-Hester L, Bishop E, Holiday D, Bailey DB. Seizures in fragile X syndrome: characteristics and comorbid diagnoses. Am J Intellect Dev Disabil. 2010;115(6):461–72.PubMedCrossRef
17.
Zurück zum Zitat Bailey Jr DB, Raspa M, Olmsted M, Holiday DB. Co-occurring conditions associated with FMR1 gene variations: findings from a national parent survey. Am J Med Genet A. 2008;146A(16):2060–9.PubMedCrossRef Bailey Jr DB, Raspa M, Olmsted M, Holiday DB. Co-occurring conditions associated with FMR1 gene variations: findings from a national parent survey. Am J Med Genet A. 2008;146A(16):2060–9.PubMedCrossRef
18.
Zurück zum Zitat Heard TT, Ramgopal S, Picker J, Lincoln SA, Rotenberg A, Kothare SV. EEG abnormalities and seizures in genetically diagnosed Fragile X syndrome. Int J Dev Neurosci. 2014;38:155–60.PubMedCrossRef Heard TT, Ramgopal S, Picker J, Lincoln SA, Rotenberg A, Kothare SV. EEG abnormalities and seizures in genetically diagnosed Fragile X syndrome. Int J Dev Neurosci. 2014;38:155–60.PubMedCrossRef
19.
Zurück zum Zitat Meguid NA, Fahim C, Sami R, Nashaat NH, Yoon U, Anwar M, El-Dessouky HM, Shahine EA, Ibrahim AS, Mancini-Marie A, Evans AC. Cognition and lobar morphology in full mutation boys with fragile X syndrome. Brain Cogn. 2012;78(1):74–84.PubMed Meguid NA, Fahim C, Sami R, Nashaat NH, Yoon U, Anwar M, El-Dessouky HM, Shahine EA, Ibrahim AS, Mancini-Marie A, Evans AC. Cognition and lobar morphology in full mutation boys with fragile X syndrome. Brain Cogn. 2012;78(1):74–84.PubMed
20.
Zurück zum Zitat Hinton VJ, Brown WT, Wisniewski K, Rudelli RD. Analysis of neocortex in three males with the fragile X syndrome. Am J Med Genet. 1991;41(3):289–94.PubMedCrossRef Hinton VJ, Brown WT, Wisniewski K, Rudelli RD. Analysis of neocortex in three males with the fragile X syndrome. Am J Med Genet. 1991;41(3):289–94.PubMedCrossRef
21.
Zurück zum Zitat Galvez R, Greenough WT. Sequence of abnormal dendritic spine development in primary somatosensory cortex of a mouse model of the fragile X mental retardation syndrome. Am J Med Genet A. 2005;135(2):155–60.PubMedCrossRef Galvez R, Greenough WT. Sequence of abnormal dendritic spine development in primary somatosensory cortex of a mouse model of the fragile X mental retardation syndrome. Am J Med Genet A. 2005;135(2):155–60.PubMedCrossRef
22.
23.
Zurück zum Zitat Roberts JE, McCary LM, Shinkareva SV, Bailey Jr DB. Infant development in fragile X syndrome: cross-syndrome comparisons. J Autism Dev Disord. 2016;46(6):2088–99.PubMedCrossRefPubMedCentral Roberts JE, McCary LM, Shinkareva SV, Bailey Jr DB. Infant development in fragile X syndrome: cross-syndrome comparisons. J Autism Dev Disord. 2016;46(6):2088–99.PubMedCrossRefPubMedCentral
24.
Zurück zum Zitat Arvio M. Fragile-X syndrome-a 20-year follow-up study of male patients. Clin Genet. 2016;89(1):55–9.PubMedCrossRef Arvio M. Fragile-X syndrome-a 20-year follow-up study of male patients. Clin Genet. 2016;89(1):55–9.PubMedCrossRef
25.
Zurück zum Zitat Brown WT, Friedman E, Jenkins EC, Brooks J, Wisniewski K, Raguthu S, French JH. Association of fragile X syndrome with autism. Lancet. 1982;1(8263):100.PubMedCrossRef Brown WT, Friedman E, Jenkins EC, Brooks J, Wisniewski K, Raguthu S, French JH. Association of fragile X syndrome with autism. Lancet. 1982;1(8263):100.PubMedCrossRef
26.
Zurück zum Zitat Wadell PM, Hagerman RJ, Hessl DR. Fragile X syndrome: psychiatric manifestations, assessment and emerging therapies. Curr Psychiatry Rev. 2013;9(1):53–8.PubMedPubMedCentral Wadell PM, Hagerman RJ, Hessl DR. Fragile X syndrome: psychiatric manifestations, assessment and emerging therapies. Curr Psychiatry Rev. 2013;9(1):53–8.PubMedPubMedCentral
27.
Zurück zum Zitat Talisa VB, Boyle L, Crafa D, Kaufmann WE. Autism and anxiety in males with fragile X syndrome: an exploratory analysis of neurobehavioral profiles from a parent survey. Am J Med Genet A. 2014;164A(5):1198–203.PubMedCrossRef Talisa VB, Boyle L, Crafa D, Kaufmann WE. Autism and anxiety in males with fragile X syndrome: an exploratory analysis of neurobehavioral profiles from a parent survey. Am J Med Genet A. 2014;164A(5):1198–203.PubMedCrossRef
28.
Zurück zum Zitat Hartley SL, Wheeler AC, Mailick MR, Raspa M, Mihaila I, Bishop E, Bailey DB. Autism symptoms across adulthood in Men with fragile X syndrome: a cross-sectional analysis. J Autism Dev Disord. 2015;45(11):3668–79.PubMedPubMedCentralCrossRef Hartley SL, Wheeler AC, Mailick MR, Raspa M, Mihaila I, Bishop E, Bailey DB. Autism symptoms across adulthood in Men with fragile X syndrome: a cross-sectional analysis. J Autism Dev Disord. 2015;45(11):3668–79.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Wheeler AC, Mussey J, Villagomez A, Bishop E, Raspa M, Edwards A, Bodfish J, Bann C, Bailey Jr DB. DSM-5 changes and the prevalence of parent-reported autism spectrum symptoms in Fragile X syndrome. J Autism Dev Disord. 2015;45(3):816–29.PubMedCrossRef Wheeler AC, Mussey J, Villagomez A, Bishop E, Raspa M, Edwards A, Bodfish J, Bann C, Bailey Jr DB. DSM-5 changes and the prevalence of parent-reported autism spectrum symptoms in Fragile X syndrome. J Autism Dev Disord. 2015;45(3):816–29.PubMedCrossRef
31.
Zurück zum Zitat Sullivan K, Hatton D, Hammer J, Sideris J, Hooper S, Ornstein P, Bailey Jr D. ADHD symptoms in children with FXS. Am J Med Genet A. 2006;140:2275–88.PubMedCrossRef Sullivan K, Hatton D, Hammer J, Sideris J, Hooper S, Ornstein P, Bailey Jr D. ADHD symptoms in children with FXS. Am J Med Genet A. 2006;140:2275–88.PubMedCrossRef
32.
Zurück zum Zitat Grefer M, Flory K, Cornish K, Hatton D, Roberts J. The emergence and stability of attention deficit hyperactivity disorder in boys with fragile X syndrome. J Intellect Disabil Res. 2016;60(2):167–78.PubMedCrossRef Grefer M, Flory K, Cornish K, Hatton D, Roberts J. The emergence and stability of attention deficit hyperactivity disorder in boys with fragile X syndrome. J Intellect Disabil Res. 2016;60(2):167–78.PubMedCrossRef
33.
Zurück zum Zitat Chromik LC, Quintin EM, Lepage JF, Hustyi KM, Lightbody AA, Reiss AL. The influence of hyperactivity, impulsivity, and attention problems on social functioning in adolescents and young adults with Fragile X syndrome. J Atten Disord. 2015, PMID: 25731183 [Epub ahead of print]. Chromik LC, Quintin EM, Lepage JF, Hustyi KM, Lightbody AA, Reiss AL. The influence of hyperactivity, impulsivity, and attention problems on social functioning in adolescents and young adults with Fragile X syndrome. J Atten Disord. 2015, PMID: 25731183 [Epub ahead of print].
34.
Zurück zum Zitat Cordeiro L, Ballinger E, Hagerman R, Hessl D. Clinical assessment of DSM-IV anxiety disorders in fragile X syndrome: prevalence and characterization. J Neurodev Disord. 2011;3(1):57–67.PubMedCrossRef Cordeiro L, Ballinger E, Hagerman R, Hessl D. Clinical assessment of DSM-IV anxiety disorders in fragile X syndrome: prevalence and characterization. J Neurodev Disord. 2011;3(1):57–67.PubMedCrossRef
35.
Zurück zum Zitat Oakes A, Thurman AJ, McDuffie A, Bullard LM, Hagerman RJ, Abbeduto L. Characterising repetitive behaviours in young boys with fragile X syndrome. J Intellect Disabil Res. 2016;60(1):54–67.PubMedCrossRef Oakes A, Thurman AJ, McDuffie A, Bullard LM, Hagerman RJ, Abbeduto L. Characterising repetitive behaviours in young boys with fragile X syndrome. J Intellect Disabil Res. 2016;60(1):54–67.PubMedCrossRef
36.
Zurück zum Zitat Kronk R, Bishop EE, Raspa M, Bickel JO, Mandel DA, Bailey Jr DB. Prevalence, nature, and correlates of sleep problems among children with fragile X syndrome based on a large scale parent survey. Sleep. 2010;33(5):679–87.PubMedPubMedCentralCrossRef Kronk R, Bishop EE, Raspa M, Bickel JO, Mandel DA, Bailey Jr DB. Prevalence, nature, and correlates of sleep problems among children with fragile X syndrome based on a large scale parent survey. Sleep. 2010;33(5):679–87.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Wheeler AC, Raspa M, Bishop E, Bailey Jr DB. Aggression in fragile X syndrome. J Intellect Disabil Res. 2016;60(2):113–25.PubMedCrossRef Wheeler AC, Raspa M, Bishop E, Bailey Jr DB. Aggression in fragile X syndrome. J Intellect Disabil Res. 2016;60(2):113–25.PubMedCrossRef
39.
Zurück zum Zitat Davids JR, Hagerman RJ, Eilert RE. Orthopaedic aspects of fragile-X syndrome. J Bone Joint Surg Am. 1990;72:889–96.PubMedCrossRef Davids JR, Hagerman RJ, Eilert RE. Orthopaedic aspects of fragile-X syndrome. J Bone Joint Surg Am. 1990;72:889–96.PubMedCrossRef
40.
Zurück zum Zitat Pyeritz RE, Stamberg J, Thomas GH, Bell BB, Zahka KG, Bernhardt BA. The marker Xq28 syndrome (“fragile-X SYndrome”) in a retarded man with mitral valve prolapse. Johns Hopkins Med J. 1982;151:231–7.PubMed Pyeritz RE, Stamberg J, Thomas GH, Bell BB, Zahka KG, Bernhardt BA. The marker Xq28 syndrome (“fragile-X SYndrome”) in a retarded man with mitral valve prolapse. Johns Hopkins Med J. 1982;151:231–7.PubMed
41.
Zurück zum Zitat Klusek J, Roberts JE, Losh M. Cardiac autonomic regulation in autism and Fragile X syndrome: a review. Psychol Bull. 2015;41(1):141–75.CrossRef Klusek J, Roberts JE, Losh M. Cardiac autonomic regulation in autism and Fragile X syndrome: a review. Psychol Bull. 2015;41(1):141–75.CrossRef
42.
Zurück zum Zitat Utari A, Adams E, Berry-Kravis E, Chavez A, Scaggs F, Ngotran L, Boyd A, Hessl D, Gane LW, Tassone F, Tartaglia N, Leehey MA, Hagerman RJ. Aging in fragile X syndrome. J Neurodev Disord. 2010;2(2):70–6.PubMedPubMedCentralCrossRef Utari A, Adams E, Berry-Kravis E, Chavez A, Scaggs F, Ngotran L, Boyd A, Hessl D, Gane LW, Tassone F, Tartaglia N, Leehey MA, Hagerman RJ. Aging in fragile X syndrome. J Neurodev Disord. 2010;2(2):70–6.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Maino DM, Wesson M, Schlange D, Cibis G, Maino JH. Optometric findings in the fragile X syndrome. Optom Vis Sci. 1991;68(8):634–40.PubMedCrossRef Maino DM, Wesson M, Schlange D, Cibis G, Maino JH. Optometric findings in the fragile X syndrome. Optom Vis Sci. 1991;68(8):634–40.PubMedCrossRef
44.
Zurück zum Zitat Hatton DD, Buckley E, Lachiewicz A, Roberts J. Ocular status of boys with fragile X syndrome: a prospective study. J AAPOS. 1998;2(5):298–302.PubMedCrossRef Hatton DD, Buckley E, Lachiewicz A, Roberts J. Ocular status of boys with fragile X syndrome: a prospective study. J AAPOS. 1998;2(5):298–302.PubMedCrossRef
45.
Zurück zum Zitat Van Splunder J, Stilma JS, Evenhuis HM. Visual performance in specific syndromes associated with intellectual disability. Eur J Ophthalmol. 2003;13(6):566–74.PubMed Van Splunder J, Stilma JS, Evenhuis HM. Visual performance in specific syndromes associated with intellectual disability. Eur J Ophthalmol. 2003;13(6):566–74.PubMed
46.
Zurück zum Zitat King RA, Hagerman RJ, Houghton M. Ocular findings in fragile X syndrome. Dev Brain Dysfunct. 1995;8(4–6):223–9. King RA, Hagerman RJ, Houghton M. Ocular findings in fragile X syndrome. Dev Brain Dysfunct. 1995;8(4–6):223–9.
47.
Zurück zum Zitat Kidd SA, Lachiewicz A, Barbouth D, Blitz RK, Delahunty C, McBrien D, Visootsak J, Berry-Kravis E. Fragile X syndrome: a review of associated medical problems. Pediatrics. 2014;134(5):995–1005.PubMedCrossRef Kidd SA, Lachiewicz A, Barbouth D, Blitz RK, Delahunty C, McBrien D, Visootsak J, Berry-Kravis E. Fragile X syndrome: a review of associated medical problems. Pediatrics. 2014;134(5):995–1005.PubMedCrossRef
48.
Zurück zum Zitat Badran HS, Abulnasr KM, Abd El Hameed Nasser S. Effect of recurrent otitis media on language profile in children with fragile x syndrome. Clin Med Insights Ear Nose Throat. 2013;6:1–7.PubMedPubMedCentralCrossRef Badran HS, Abulnasr KM, Abd El Hameed Nasser S. Effect of recurrent otitis media on language profile in children with fragile x syndrome. Clin Med Insights Ear Nose Throat. 2013;6:1–7.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Lisik MZ, Gutmajster E, Sieroń AL. Low levels of HDL in fragile X syndrome patients. Lipids. 2016;51(2):189–92.PubMedCrossRef Lisik MZ, Gutmajster E, Sieroń AL. Low levels of HDL in fragile X syndrome patients. Lipids. 2016;51(2):189–92.PubMedCrossRef
50.
Zurück zum Zitat Berry-Kravis E, Levin R, Shah H, Mathur S, Darnell JC, Ouyang B. Cholesterol levels in fragile X syndrome. Am J Med Genet A. 2015;167A(2):379–84.PubMedCrossRef Berry-Kravis E, Levin R, Shah H, Mathur S, Darnell JC, Ouyang B. Cholesterol levels in fragile X syndrome. Am J Med Genet A. 2015;167A(2):379–84.PubMedCrossRef
51.
Zurück zum Zitat Saldarriaga W, Tassone F, González-Teshima LY, Forero-Forero JV, Ayala-Zapata S, Hagerman R. Fragile X syndrome. Colomb Med (Cali). 2014;45(4):190–8. Saldarriaga W, Tassone F, González-Teshima LY, Forero-Forero JV, Ayala-Zapata S, Hagerman R. Fragile X syndrome. Colomb Med (Cali). 2014;45(4):190–8.
52.
Zurück zum Zitat Wiesner GL, Cassidy SB, Grimes SJ, Matthews AL, Acheson LS. Clinical consult: developmental delay/fragile X syndrome. Prim Care. 2004;31(3):621–5. x. Review.PubMedCrossRef Wiesner GL, Cassidy SB, Grimes SJ, Matthews AL, Acheson LS. Clinical consult: developmental delay/fragile X syndrome. Prim Care. 2004;31(3):621–5. x. Review.PubMedCrossRef
53.
Zurück zum Zitat Tatton-Brown K, Cole TRP, Rahman N. Sotos syndrome. Gene reviews. 2004 Dec 17 [Updated 2015 Nov 19]. In: Pagon RA, Adam MP, Ardinger HH, et al., editors. GeneReviews® [Internet]. Seattle: University of Washington; 1993–2016. Available from: http://www.ncbi.nlm.nih.gov/books/NBK1479. Accessed 17 Aug 2016. Tatton-Brown K, Cole TRP, Rahman N. Sotos syndrome. Gene reviews. 2004 Dec 17 [Updated 2015 Nov 19]. In: Pagon RA, Adam MP, Ardinger HH, et al., editors. GeneReviews® [Internet]. Seattle: University of Washington; 1993–2016. Available from: http://​www.​ncbi.​nlm.​nih.​gov/​books/​NBK1479. Accessed 17 Aug 2016.
54.
Zurück zum Zitat Driscoll DJ, Miller JL, Schwartz S, Cassidy SB. Prader-Willi syndrome. Gene reviews. 1998 Oct 6 [Updated 2016 Feb 4]. In: Pagon RA, Adam MP, Ardinger HH, et al., editors. GeneReviews® [Internet]. Seattle: University of Washington; 1993–2016. Available from: http://www.ncbi.nlm.nih.gov/books/NBK1330. Accessed 17 Aug 2016. Driscoll DJ, Miller JL, Schwartz S, Cassidy SB. Prader-Willi syndrome. Gene reviews. 1998 Oct 6 [Updated 2016 Feb 4]. In: Pagon RA, Adam MP, Ardinger HH, et al., editors. GeneReviews® [Internet]. Seattle: University of Washington; 1993–2016. Available from: http://​www.​ncbi.​nlm.​nih.​gov/​books/​NBK1330. Accessed 17 Aug 2016.
55.
Zurück zum Zitat Sutherland GR. Fragile sites on human chromosomes: demonstration of their dependence on the type of tissue culture medium. Science. 1977;197(4300):265–6.PubMedCrossRef Sutherland GR. Fragile sites on human chromosomes: demonstration of their dependence on the type of tissue culture medium. Science. 1977;197(4300):265–6.PubMedCrossRef
56.
Zurück zum Zitat Hecht F, Sutherland GR. Detection of fragile sites on human chromosomes. Clin Genet. 1985;28(1):95–6.PubMedCrossRef Hecht F, Sutherland GR. Detection of fragile sites on human chromosomes. Clin Genet. 1985;28(1):95–6.PubMedCrossRef
57.
Zurück zum Zitat Noto V, Harrity C, Walsh D, Marron K. The impact of FMR1 gene mutations on human reproduction and development: a systematic review. J Assist Reprod Genet. 2016. [Epub ahead of print] PMID: 27432256. doi:10.1007/s10815-016-0765-6. Noto V, Harrity C, Walsh D, Marron K. The impact of FMR1 gene mutations on human reproduction and development: a systematic review. J Assist Reprod Genet. 2016. [Epub ahead of print] PMID: 27432256. doi:10.​1007/​s10815-016-0765-6.
58.
Zurück zum Zitat Debrey SM, Leehey MA, Klepitskaya O, Filley CM, Shah RC, Kluger B, Berry-Kravis E, Spector E, Tassone F, Hall DA. Clinical phenotype of adult fragile X gray zone allele carriers: a case series. Cerebellum. 2016. [Epub ahead of print] PMID: 27372099. Debrey SM, Leehey MA, Klepitskaya O, Filley CM, Shah RC, Kluger B, Berry-Kravis E, Spector E, Tassone F, Hall DA. Clinical phenotype of adult fragile X gray zone allele carriers: a case series. Cerebellum. 2016. [Epub ahead of print] PMID: 27372099.
59.
Zurück zum Zitat Cronister A, Schreiner R, Wittenberger M, Amiri K, Harris K, Hagerman RJ. Heterozygous fragile X female: historical, physical, cognitive, and cytogenetic features. Am J Med Genet. 1991;38:269–74.PubMedCrossRef Cronister A, Schreiner R, Wittenberger M, Amiri K, Harris K, Hagerman RJ. Heterozygous fragile X female: historical, physical, cognitive, and cytogenetic features. Am J Med Genet. 1991;38:269–74.PubMedCrossRef
60.
Zurück zum Zitat Jacquemont S, Hagerman RJ, Leehey MA, Hall DA, Levine RA, Brunberg JA, Zhang L, Jardini T, Gane LW, Harris SW, Herman K, Grigsby J, Greco CM, Berry-Kravis E, Tassone F, Hagerman PJ. Penetrance of the fragile X-associated tremor/ataxia syndrome in a premutation carrier population. JAMA. 2004;291:460–9.PubMedCrossRef Jacquemont S, Hagerman RJ, Leehey MA, Hall DA, Levine RA, Brunberg JA, Zhang L, Jardini T, Gane LW, Harris SW, Herman K, Grigsby J, Greco CM, Berry-Kravis E, Tassone F, Hagerman PJ. Penetrance of the fragile X-associated tremor/ataxia syndrome in a premutation carrier population. JAMA. 2004;291:460–9.PubMedCrossRef
61.
Zurück zum Zitat Rodriguez-Revenga L, Madrigal I, Pagonabarraga J, Xunclà M, Badenas C, Kulisevsky J, Gomez B, Milà M. Penetrance of FMR1 premutation associated pathologies in fragile X syndrome families. Eur J Hum Genet. 2009;17:1359–62.PubMedPubMedCentralCrossRef Rodriguez-Revenga L, Madrigal I, Pagonabarraga J, Xunclà M, Badenas C, Kulisevsky J, Gomez B, Milà M. Penetrance of FMR1 premutation associated pathologies in fragile X syndrome families. Eur J Hum Genet. 2009;17:1359–62.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Milà M, Rodriguez-Revenga L, Matilla-Dueñas A. FMR1 premutation: basic mechanisms and clinical involvement. Cerebellum. 2016. [Epub ahead of print]. PMID: 27338822. Milà M, Rodriguez-Revenga L, Matilla-Dueñas A. FMR1 premutation: basic mechanisms and clinical involvement. Cerebellum. 2016. [Epub ahead of print]. PMID: 27338822.
63.
Zurück zum Zitat Mailick MR, Hong J, Rathouz P, Baker MW, Greenberg JS, Smith L, Maenner M. Low-normal FMR1 CGG repeat length: phenotypic associations. Front Genet. 2014;5:309.PubMedPubMedCentralCrossRef Mailick MR, Hong J, Rathouz P, Baker MW, Greenberg JS, Smith L, Maenner M. Low-normal FMR1 CGG repeat length: phenotypic associations. Front Genet. 2014;5:309.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Nolin SL, Glicksman A, Ersalesi N, Dobkin C, Brown WT, Cao R, Blatt E, Sah S, Latham GJ, Hadd AG. Fragile X full mutation expansions are inhibited by one or more AGG interruptions in premutation carriers. Genet Med. 2015;17(5):358–64.PubMedCrossRef Nolin SL, Glicksman A, Ersalesi N, Dobkin C, Brown WT, Cao R, Blatt E, Sah S, Latham GJ, Hadd AG. Fragile X full mutation expansions are inhibited by one or more AGG interruptions in premutation carriers. Genet Med. 2015;17(5):358–64.PubMedCrossRef
65.
Zurück zum Zitat Nolin SL, Glicksman A, Ding X, Ersalesi N, Brown WT, Sherman SL, Dobkin C. Fragile X analysis of 1112 prenatal samples from 1991 to 2010. Prenat Diagn. 2011;31(10):925–31.PubMedCrossRef Nolin SL, Glicksman A, Ding X, Ersalesi N, Brown WT, Sherman SL, Dobkin C. Fragile X analysis of 1112 prenatal samples from 1991 to 2010. Prenat Diagn. 2011;31(10):925–31.PubMedCrossRef
66.
Zurück zum Zitat Chen L, Hadd A, Sah S, Filipovic-Sadic S, Krosting J, Sekinger E, Pan R, Hagerman PJ, Stenzel TT, Tassone F, Latham GJ. An information-rich CGG repeat primed PCR that detects the full range of fragile X expanded alleles and minimizes the need for Southern blot analysis. J Mol Diagn. 2010;12:589–600.PubMedPubMedCentralCrossRef Chen L, Hadd A, Sah S, Filipovic-Sadic S, Krosting J, Sekinger E, Pan R, Hagerman PJ, Stenzel TT, Tassone F, Latham GJ. An information-rich CGG repeat primed PCR that detects the full range of fragile X expanded alleles and minimizes the need for Southern blot analysis. J Mol Diagn. 2010;12:589–600.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Nolin SL, Glicksman A, Houck Jr GE, Brown WT, Dobkin CS. Mosaicism in fragile X affected males. Am J Med Genet. 1994;51(4):509–12.PubMedCrossRef Nolin SL, Glicksman A, Houck Jr GE, Brown WT, Dobkin CS. Mosaicism in fragile X affected males. Am J Med Genet. 1994;51(4):509–12.PubMedCrossRef
68.
Zurück zum Zitat Stark Z, Francis D, Gaffney L, Greenberg J, Hills L, Li X, Godler DE, Slater HR. Prenatal diagnosis of fragile X syndrome complicated by full mutation retraction. Am J Med Genet A. 2015;167A(10):2485–7.PubMedCrossRef Stark Z, Francis D, Gaffney L, Greenberg J, Hills L, Li X, Godler DE, Slater HR. Prenatal diagnosis of fragile X syndrome complicated by full mutation retraction. Am J Med Genet A. 2015;167A(10):2485–7.PubMedCrossRef
69.
Zurück zum Zitat Schmucker B, Seidel J. Mosaicism for a full mutation and a normal size allele in two fragile X males. Am J Med Genet. 1999;84:221–5.PubMedCrossRef Schmucker B, Seidel J. Mosaicism for a full mutation and a normal size allele in two fragile X males. Am J Med Genet. 1999;84:221–5.PubMedCrossRef
70.
Zurück zum Zitat Bonarrigo FA, Russo S, Vizziello P, Menni F, Cogliati F, Giorgini V, Monti F, Milani D. Think about it: FMR1 gene mosaicism. J Child Neurol. 2014;29(9):NP74–7.PubMedCrossRef Bonarrigo FA, Russo S, Vizziello P, Menni F, Cogliati F, Giorgini V, Monti F, Milani D. Think about it: FMR1 gene mosaicism. J Child Neurol. 2014;29(9):NP74–7.PubMedCrossRef
71.
Zurück zum Zitat Gonçalves TF, dos Santos JM, Gonçalves AP, Tassone F, Mendoza-Morales G, Ribeiro MG, Kahn E, Boy R, Pimentel MM, Santos-Rebouças CB. Finding FMR1 mosaicism in Fragile X syndrome. Expert Rev Mol Diagn. 2016;16(4):501–17.PubMedPubMedCentralCrossRef Gonçalves TF, dos Santos JM, Gonçalves AP, Tassone F, Mendoza-Morales G, Ribeiro MG, Kahn E, Boy R, Pimentel MM, Santos-Rebouças CB. Finding FMR1 mosaicism in Fragile X syndrome. Expert Rev Mol Diagn. 2016;16(4):501–17.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Handt M, Epplen A, Hoffjan S, Mese K, Epplen JT, Dekomien G. Point mutation frequency in the FMR1 gene as revealed by fragile X syndrome screening. Mol Cell Probes. 2014;28(5–6):279–83.PubMedCrossRef Handt M, Epplen A, Hoffjan S, Mese K, Epplen JT, Dekomien G. Point mutation frequency in the FMR1 gene as revealed by fragile X syndrome screening. Mol Cell Probes. 2014;28(5–6):279–83.PubMedCrossRef
73.
Zurück zum Zitat Suhl JA, Warren ST. Single-nucleotide mutations in FMR1 reveal novel functions and regulatory mechanisms of the fragile X syndrome protein FMRP. J Exp Neurosci. 2015;9 Suppl 2:35–41.PubMedPubMedCentralCrossRef Suhl JA, Warren ST. Single-nucleotide mutations in FMR1 reveal novel functions and regulatory mechanisms of the fragile X syndrome protein FMRP. J Exp Neurosci. 2015;9 Suppl 2:35–41.PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Tekcan A. In silico analysis of FMR1 gene missense SNPs. Cell Biochem Biophys. 2016;74(2):109–27.PubMedCrossRef Tekcan A. In silico analysis of FMR1 gene missense SNPs. Cell Biochem Biophys. 2016;74(2):109–27.PubMedCrossRef
76.
Zurück zum Zitat Gutiérrez JF, Bajaj K, Klugman SD. Prenatal screening for fragile x: carriers, controversies, and counseling. Rev Obstet Gynecol. 2013;6(1):e1–7.PubMedPubMedCentral Gutiérrez JF, Bajaj K, Klugman SD. Prenatal screening for fragile x: carriers, controversies, and counseling. Rev Obstet Gynecol. 2013;6(1):e1–7.PubMedPubMedCentral
77.
Zurück zum Zitat Hirst MC, Knight SJ, Christodoulou Z, Grewal PK, Fryns JP, Davies KE. Origins of the fragile X syndrome mutation. J Med Genet. 1993;30(8):647–50.PubMedPubMedCentralCrossRef Hirst MC, Knight SJ, Christodoulou Z, Grewal PK, Fryns JP, Davies KE. Origins of the fragile X syndrome mutation. J Med Genet. 1993;30(8):647–50.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Rousseau F, Heitz D, Biancalana V, Blumenfeld S, Kretz C, Boue J, Tommerup N, Van Der Hagen C, DeLozier- Blanchet C, Croquette MF, Gilgenkrantz S, Jalbert P, Voelckel MA, Oberlé I, Mandel JL. Direct diagnosis by DNA analysis of the fragile X syndrome of mental retardation. N Engl J Med. 1991;325(24):1673–81.PubMedCrossRef Rousseau F, Heitz D, Biancalana V, Blumenfeld S, Kretz C, Boue J, Tommerup N, Van Der Hagen C, DeLozier- Blanchet C, Croquette MF, Gilgenkrantz S, Jalbert P, Voelckel MA, Oberlé I, Mandel JL. Direct diagnosis by DNA analysis of the fragile X syndrome of mental retardation. N Engl J Med. 1991;325(24):1673–81.PubMedCrossRef
79.
Zurück zum Zitat Chen L, Hadd A, Sah S, Houghton J, Filipovic-Sadic S, Zhang W, Hagerman PJ, Tassone F, Latham GJ. High-resolution methylation polymerase chain reaction for fragile X analysis: evidence for novel FMR1 methylation patterns undetected in Southern blot analyses. Genet Med. 2011;13:528–38.PubMedPubMedCentralCrossRef Chen L, Hadd A, Sah S, Houghton J, Filipovic-Sadic S, Zhang W, Hagerman PJ, Tassone F, Latham GJ. High-resolution methylation polymerase chain reaction for fragile X analysis: evidence for novel FMR1 methylation patterns undetected in Southern blot analyses. Genet Med. 2011;13:528–38.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Filipovic-Sadic S, Sah S, Chen L, Krosting J, Sekinger E, Zhang W, Hagerman PJ, Stenzel TT, Hadd AG, Latham GJ, Tassone F. A novel FMR1 PCR method for the routine detection of low abundance expanded alleles and full mutations in fragile X syndrome. Clin Chem. 2010;56:399–408.PubMedPubMedCentral Filipovic-Sadic S, Sah S, Chen L, Krosting J, Sekinger E, Zhang W, Hagerman PJ, Stenzel TT, Hadd AG, Latham GJ, Tassone F. A novel FMR1 PCR method for the routine detection of low abundance expanded alleles and full mutations in fragile X syndrome. Clin Chem. 2010;56:399–408.PubMedPubMedCentral
81.
Zurück zum Zitat Saluto A, Brussino A, Tassone F, Arduino C, Cagnoli C, Pappi P, Hagerman P, Migone N, Brusco A. An enhanced polymerase chain reaction assay to detect pre- and full mutation alleles of the fragile X mental retardation 1 gene. J Mol Diagn. 2005;7(5):605–12.PubMedPubMedCentralCrossRef Saluto A, Brussino A, Tassone F, Arduino C, Cagnoli C, Pappi P, Hagerman P, Migone N, Brusco A. An enhanced polymerase chain reaction assay to detect pre- and full mutation alleles of the fragile X mental retardation 1 gene. J Mol Diagn. 2005;7(5):605–12.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Lyon E, Laver T, Yu P, Jama M, Young K, Zoccoli M, Marlowe N. A simple, high-throughput assay for Fragile X expanded alleles using triple repeat primed PCR and capillary electrophoresis. J Mol Diagn. 2010;12(4):505–11.PubMedPubMedCentralCrossRef Lyon E, Laver T, Yu P, Jama M, Young K, Zoccoli M, Marlowe N. A simple, high-throughput assay for Fragile X expanded alleles using triple repeat primed PCR and capillary electrophoresis. J Mol Diagn. 2010;12(4):505–11.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Weinhaeusel A, Haas OA. Evaluation of the fragile X (FRAXA) syndrome with methylation-sensitive PCR. Hum Genet. 2001;108:450–8.CrossRef Weinhaeusel A, Haas OA. Evaluation of the fragile X (FRAXA) syndrome with methylation-sensitive PCR. Hum Genet. 2001;108:450–8.CrossRef
84.
Zurück zum Zitat Acharya K, Ross LF. Fragile X screening: attitudes of genetic health professionals. Am J Med Genet A. 2009;149A(4):626–32.PubMedCrossRef Acharya K, Ross LF. Fragile X screening: attitudes of genetic health professionals. Am J Med Genet A. 2009;149A(4):626–32.PubMedCrossRef
85.
Zurück zum Zitat Eichler EE, Holden JJ, Popovich BW, Reiss AL, Snow K, Thibodeau SN, Richards CS, Ward PA, Nelson DL. Length of uninterrupted CGG repeats determines instability in the FMR1 gene. Nat Genet. 1994;8:88–94.PubMedCrossRef Eichler EE, Holden JJ, Popovich BW, Reiss AL, Snow K, Thibodeau SN, Richards CS, Ward PA, Nelson DL. Length of uninterrupted CGG repeats determines instability in the FMR1 gene. Nat Genet. 1994;8:88–94.PubMedCrossRef
86.
Zurück zum Zitat Famula J, Basuta K, Gane LW, Hagerman RJ, Tassone F. Identification of a male with fragile X syndrome through newborn screening. Intractable Rare Dis Res. 2015;4(4):198–202.PubMedPubMedCentralCrossRef Famula J, Basuta K, Gane LW, Hagerman RJ, Tassone F. Identification of a male with fragile X syndrome through newborn screening. Intractable Rare Dis Res. 2015;4(4):198–202.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Godler DE, Amor DJ, Slater HR. Methylation analysis in newborn screening for fragile X syndrome. JAMA Neurol. 2014;71(6):800.PubMedCrossRef Godler DE, Amor DJ, Slater HR. Methylation analysis in newborn screening for fragile X syndrome. JAMA Neurol. 2014;71(6):800.PubMedCrossRef
88.
Zurück zum Zitat Gross C, Hoffmann A, Bassell GJ, Berry-Kravis EM. Therapeutic strategies in fragile X syndrome: from bench to bedside and back. Neurotherapeutics. 2015;12(3):584–608.PubMedPubMedCentralCrossRef Gross C, Hoffmann A, Bassell GJ, Berry-Kravis EM. Therapeutic strategies in fragile X syndrome: from bench to bedside and back. Neurotherapeutics. 2015;12(3):584–608.PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat Martin GE, Ausderau KK, Raspa M, Bishop E, Mallya U, Bailey Jr DB. Therapy service use among individuals with fragile X syndrome: findings from a US parent survey. J Intellect Disabil Res. 2013;57(9):837–49.PubMedCrossRef Martin GE, Ausderau KK, Raspa M, Bishop E, Mallya U, Bailey Jr DB. Therapy service use among individuals with fragile X syndrome: findings from a US parent survey. J Intellect Disabil Res. 2013;57(9):837–49.PubMedCrossRef
91.
Zurück zum Zitat Berry-Kravis E, Sumis A, Hervey C, Mathur S. Clinic-based retrospective analysis of psychopharmacology for behavior in fragile X syndrome. Int J Pediatr. 2012;2012:843016.PubMedPubMedCentralCrossRef Berry-Kravis E, Sumis A, Hervey C, Mathur S. Clinic-based retrospective analysis of psychopharmacology for behavior in fragile X syndrome. Int J Pediatr. 2012;2012:843016.PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat Berry-Kravis EM, Sumis A, Hervey C, Nelson M, Porges SW, Weng N, Weiler IJ, Greenough WT. Open-label treatment trial of lithium to target the underlying defect in fragile X syndrome. J Dev Behav Pediatr. 2008;29(4):293–302.PubMedCrossRef Berry-Kravis EM, Sumis A, Hervey C, Nelson M, Porges SW, Weng N, Weiler IJ, Greenough WT. Open-label treatment trial of lithium to target the underlying defect in fragile X syndrome. J Dev Behav Pediatr. 2008;29(4):293–302.PubMedCrossRef
93.
Zurück zum Zitat Davenport MH, Schaefer TL, Friedmann KJ, Fitzpatrick SE, Erickson CA. Pharmacotherapy for fragile X syndrome: progress to date. Drugs. 2016;76(4):431–45.PubMedCrossRef Davenport MH, Schaefer TL, Friedmann KJ, Fitzpatrick SE, Erickson CA. Pharmacotherapy for fragile X syndrome: progress to date. Drugs. 2016;76(4):431–45.PubMedCrossRef
94.
Zurück zum Zitat Dolen G, Osterweil E, Rao BS, Smith GB, Auerbach BD, Chattarji S, Bear MF. Correction of fragile X syndrome in mice. Neuron. 2007;56(6):955–62.PubMedPubMedCentralCrossRef Dolen G, Osterweil E, Rao BS, Smith GB, Auerbach BD, Chattarji S, Bear MF. Correction of fragile X syndrome in mice. Neuron. 2007;56(6):955–62.PubMedPubMedCentralCrossRef
95.
Zurück zum Zitat Heulens I, D’Hulst C, Van Dam D, De Deyn PP, Kooy RF. Pharmacological treatment of fragile X syndrome with GABAergic drugs in a knockout mouse model. Behav Brain Res. 2012;229(1):244–9.PubMedCrossRef Heulens I, D’Hulst C, Van Dam D, De Deyn PP, Kooy RF. Pharmacological treatment of fragile X syndrome with GABAergic drugs in a knockout mouse model. Behav Brain Res. 2012;229(1):244–9.PubMedCrossRef
96.
Zurück zum Zitat Leigh MJS, Nguyen DV, Mu Y, Winarni TI, Schneider A, Chechi T, Polussa J, Doucet P, Tassone F, Rivera SM, Hessl D, Hagerman RJ. A randomized double-blind, placebo-controlled trial of minocycline in children and adolescents with fragile x syndrome. J Dev Behav Pediatr. 2013;34(3):147–55.PubMedPubMedCentralCrossRef Leigh MJS, Nguyen DV, Mu Y, Winarni TI, Schneider A, Chechi T, Polussa J, Doucet P, Tassone F, Rivera SM, Hessl D, Hagerman RJ. A randomized double-blind, placebo-controlled trial of minocycline in children and adolescents with fragile x syndrome. J Dev Behav Pediatr. 2013;34(3):147–55.PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Chevreul K, Gandré C, Brigham KB, López-Bastida J, Linertová R, Oliva-Moreno J, Serrano-Aguilar P, Posada-de-la-Paz M, Taruscio D, Schieppati A, Iskrov G, Gulácsi L, von der Schulenburg JM, Kanavos P, Persson U, Fattore G, BURQOL-RD Research Network. Social/economic costs and health-related quality of life in patients with fragile X syndrome in Europe. Eur J Health Econ. 2016;(Suppl 1):43–52.CrossRef Chevreul K, Gandré C, Brigham KB, López-Bastida J, Linertová R, Oliva-Moreno J, Serrano-Aguilar P, Posada-de-la-Paz M, Taruscio D, Schieppati A, Iskrov G, Gulácsi L, von der Schulenburg JM, Kanavos P, Persson U, Fattore G, BURQOL-RD Research Network. Social/economic costs and health-related quality of life in patients with fragile X syndrome in Europe. Eur J Health Econ. 2016;(Suppl 1):43–52.CrossRef
98.
Zurück zum Zitat Vekeman F, Gauthier-Loiselle M, Faust E, Lefebvre P, Lahoz R, Duh MS, Sacco P. Patient and caregiver burden associated with fragile X syndrome in the United States. Am J Intellect Dev Disabil. 2015;120(5):444–59.PubMedCrossRef Vekeman F, Gauthier-Loiselle M, Faust E, Lefebvre P, Lahoz R, Duh MS, Sacco P. Patient and caregiver burden associated with fragile X syndrome in the United States. Am J Intellect Dev Disabil. 2015;120(5):444–59.PubMedCrossRef
Metadaten
Titel
Fragile X syndrome: a review of clinical and molecular diagnoses
verfasst von
Claudia Ciaccio
Laura Fontana
Donatella Milani
Silvia Tabano
Monica Miozzo
Susanna Esposito
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Italian Journal of Pediatrics / Ausgabe 1/2017
Elektronische ISSN: 1824-7288
DOI
https://doi.org/10.1186/s13052-017-0355-y

Weitere Artikel der Ausgabe 1/2017

Italian Journal of Pediatrics 1/2017 Zur Ausgabe

Update Pädiatrie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.