ArticlePDF AvailableLiterature Review

Fragile X syndrome and fragile X-associated disorders

F1000
F1000Research
Authors:

Abstract and Figures

Fragile X syndrome (FXS) is caused by a full mutation on the FMR1 gene and a subsequent lack of FMRP, the protein product of FMR1. FMRP plays a key role in regulating the translation of many proteins involved in maintaining neuronal synaptic connections; its deficiency may result in a range of intellectual disabilities, social deficits, psychiatric problems, and dysmorphic physical features. A range of clinical involvement is also associated with the FMR1 premutation, including fragile X-associated tremor ataxia syndrome, fragile X-associated primary ovarian insufficiency, psychiatric problems, hypertension, migraines, and autoimmune problems. Over the past few years, there have been a number of advances in our knowledge of FXS and fragile X-associated disorders, and each of these advances offers significant clinical implications. Among these developments are a better understanding of the clinical impact of the phenomenon known as mosaicism, the revelation that various types of mutations can cause FXS, and improvements in treatment for FXS.
Content may be subject to copyright.
Open Peer Review
F1000FacultyReviewsarecommissioned
frommembersoftheprestigiousF1000
.InordertomakethesereviewsasFaculty
comprehensiveandaccessibleaspossible,
peerreviewtakesplacebeforepublication;the
refereesarelistedbelow,buttheirreportsare
notformallypublished.
Discuss this article
(0)Comments
REVIEW
Fragile X syndrome and fragile X-associated disorders [version
1; referees: 2 approved]
AkashRajaratnam , JasdeepShergill , MariaSalcedo-Arellano ,
WilmarSaldarriaga , XianlaiDuan , RandiHagerman 1,4
MINDInstitute,UCDavisHealth,Sacramento,CA,USA
DepartmentofMorphologyandObstetrics&Gynecology,UniversidaddelValle,SchoolofMedicine,Cali,ValledelCauca,Colombia
DepartmentofNeurology,TheThirdHospitalofChangsha,HunanSheng,China
DepartmentofPediatrics,UniversityofCalifornia,Davis,SchoolofMedicine,Sacramento,CA,USA
Abstract
FragileXsyndrome(FXS)iscausedbyafullmutationonthe geneandaFMR1
subsequentlackofFMRP,theproteinproductof .FMRPplaysakeyroleFMR1
inregulatingthetranslationofmanyproteinsinvolvedinmaintainingneuronal
synapticconnections;itsdeficiencymayresultinarangeofintellectual
disabilities,socialdeficits,psychiatricproblems,anddysmorphicphysical
features.ArangeofclinicalinvolvementisalsoassociatedwiththeFMR1
premutation,includingfragileX-associatedtremorataxiasyndrome,fragile
X-associatedprimaryovarianinsufficiency,psychiatricproblems,hypertension,
migraines,andautoimmuneproblems.Overthepastfewyears,therehave
beenanumberofadvancesinourknowledgeofFXSandfragileX-associated
disorders,andeachoftheseadvancesofferssignificantclinicalimplications.
Amongthesedevelopmentsareabetterunderstandingoftheclinicalimpactof
thephenomenonknownasmosaicism,therevelationthatvarioustypesof
mutationscancauseFXS,andimprovementsintreatmentforFXS.
1 1 1
1,2 1,3 1,4
1
2
3
4
 
Referee Status:
 InvitedReferees
version 1
published
08Dec2017
1 2
,UniversityofAntwerp,R Frank Kooy
Belgium
1
,NationalInstitutesofHealth,Karen Usdin
USA
2
08Dec2017, (F1000FacultyRev):2112(doi:First published: 6
)10.12688/f1000research.11885.1
08Dec2017, (F1000FacultyRev):2112(doi:Latest published: 6
)10.12688/f1000research.11885.1
v1
Page 1 of 11
F1000Research 2017, 6(F1000 Faculty Rev):2112 Last updated: 08 DEC 2017
AkashRajaratnam( ),RandiHagerman( )Corresponding authors: arajaratnam@ucdavis.edu rjhagerman@ucdavis.edu
 :Conceptualization,Software,Supervision,Writing–OriginalDraftPreparation,Writing–Review&Editing;Author roles: Rajaratnam A Shergill
:Software,Writing–OriginalDraftPreparation; :Conceptualization,Writing–OriginalDraftPreparation,Writing–Review&J Salcedo-Arellano M
Editing; :Writing–OriginalDraftPreparation; :Writing–OriginalDraftPreparation; :Conceptualization,Saldarriaga W Duan X Hagerman R
FundingAcquisition,ProjectAdministration,Resources,Supervision,Writing–Review&Editing
RandiHagermanhasreceivedfundingfromNovartis,Neuren,Marinus,andAlcobraforcarryingouttreatmentstudiesinCompeting interests:
patientswithFXSandhasalsoconsultedwithRoche,Novartis,FulcrumandZynerbaregardingtreatmentstudiesinindividualswithFXS.
RajaratnamA,ShergillJ,Salcedo-ArellanoM How to cite this article: et al. Fragile X syndrome and fragile X-associated disorders [version
2017, (F1000FacultyRev):2112(doi: )1; referees: 2 approved] F1000Research 610.12688/f1000research.11885.1
©2017RajaratnamA .Thisisanopenaccessarticledistributedunderthetermsofthe ,Copyright: et al CreativeCommonsAttributionLicence
whichpermitsunrestricteduse,distribution,andreproductioninanymedium,providedtheoriginalworkisproperlycited.
Thisworkwassupportedthroughthefollowinggrants:NationalInstituteofChildHealthandHumanDevelopmentgrantGrant information:
(NICHD)HD036071;theMINDInstituteIntellectualandDevelopmentalDisabilitiesResearchCenter(NICHDU54HD079125),HRSA
R40MC26641,andHHS-ADD90DD05969fortheCenterforExcellenceinDevelopmentalDisabilitiessupportandtheInternationalTraining
ProgramforNeurodevelopmentalDisordersattheMINDUCDavisMedicalCenter.
The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
08Dec2017, (F1000FacultyRev):2112(doi: )First published: 6 10.12688/f1000research.11885.1
Page 2 of 11
F1000Research 2017, 6(F1000 Faculty Rev):2112 Last updated: 08 DEC 2017
Introduction
Fragile X syndrome (FXS) is the most common inherited cause
of intellectual disability (ID) and the most common single-gene
cause of autism spectrum disorder (ASD). FXS arises from a
full mutation repeat expansion (>200 CGG repeats in the
5 untranslated region) in the FMR1 gene on the X chromosome;
this results in the methylation and subsequent silencing of the
gene. Although FXS is the most well-known disorder caused by an
FMR1 mutation, there is also a spectrum of disorders associ-
ated with the FMR1 premutation (55–200 CGG repeats)1. Those
with the full mutation have symptoms related to the absence
or deficiency of FMRP, the protein encoded by the FMR1 gene.
These individuals are susceptible to global developmental delay,
learning disabilities, and social and behavioral deficits. In con-
trast, those with the premutation allele have symptoms related
to the elevated production of FMR1 mRNA, leading to mRNA
toxicity2. Individuals with the premutation, especially males,
are at risk for developing fragile X-associated tremor/ataxia
syndrome (FXTAS), whereas females with the premutation
have an increased likelihood of developing fragile X-associated
primary ovarian insufficiency (FXPOI) before age 403. Although
global estimates for the frequency of both the full mutation and
premutation exist, recent research has indicated that founder
effects, as well as racial and ethnic differences, can significantly
affect the risk of individuals in certain regions of the world. Thus,
prevalence estimates may be more useful on a smaller, more
regional scale46.
A robust clinical picture of full mutation FXS, as well as the
variety of manifestations associated with the premutation, has
existed for years; however, the improved understanding of
mosaicism has begun to blur this picture. Mosaicism can refer to
a condition in individuals who express both full mutation cells
and premutation cells (size mosaicism) or individuals who have
the full mutation but in whom only a portion of the full mutation
alleles are methylated (methylation mosaicism). This may lead
to unique cases, such as individuals with features of both FXS
and FXTAS7. Another discovery that has challenged previous
understandings of FXS is that mutations other than full muta-
tion repeat expansions have the ability to cause the disorder.
The advent of more frequent whole exome sequencing (WES),
whole genome sequencing (WGS) and microarray testing has led
to the identification of different mutations such as point muta-
tions, deletions, and duplications as causes of FXS. Moreover,
these other mutations not only can cause FXS but also can result
in partial FMRP functionality and lead to many subtly different
phenotypes8. There have also been rapid advances in the devel-
opment of targeted treatments for patients with FXS over
the past few years, as many animal studies as well as clinical
trials have provided researchers with encouraging results. Certain
treatments have been shown to reverse aspects of the neurobio-
logical dysfunction in FXS; if used early in development, these
treatments are likely to significantly improve the outcome of
patients9,10.
Clinical presentation of fragile X syndrome
Individuals with FXS present with varying degrees of cognitive
impairment depending on sex and the level of FMRP produced11.
Patients producing higher levels of FMRP are typically less
cognitively affected. Females with FXS, therefore, may present
with a very wide range of clinical involvement due to differences
in the activation ratio (AR). The AR refers to the proportion of
normal FMR1 alleles on the active X chromosome, which sig-
nificantly impacts the amount of FMRP a female will produce12.
FMRP production also depends on CGG repeat number as well
as the proportion of methylated full mutation alleles; there-
fore, individuals presenting with mosaicism are also likely to
produce more FMRP. In turn, these individuals are typically less
cognitively affected than non-mosaic patients with FXS13.
When FMRP levels are not significantly diminished, affected
individuals may experience only modest socioemotional and
learning deficits with relatively normal IQ levels. However,
if FMRP production is severely decreased or fully silenced,
moderate to severe cognitive dysfunction is likely to occur14. In
addition to ID, there are several physical features associated with
FXS, such as a long face, broad forehead, high-arched palate,
prominent ears, macrocephaly, and macroorchidism (Figure 1
and Table 1)11. Although not all individuals with FXS will have
Figure 1. This 7-year-old boy with fragile X syndrome demonstrates
a broad forehead (a) and a high arched palate (b). However, he does
not have a long face or prominent ears. He is a high-functioning
individual with mosaicism, and DNA testing displays a band at 300
CGG repeats that is methylated as well as bands between 100
and 790 repeats that are unmethylated; overall, 30% of his alleles
are unmethylated. He presents with a sequential IQ of 71 and a
simultaneous IQ of 83. He has done well with treatment; sertraline has
improved his anxiety symptoms, and a long-acting methylphenidate
preparation has improved his attention-deficit hyperactivity disorder
symptoms.
Page 3 of 11
F1000Research 2017, 6(F1000 Faculty Rev):2112 Last updated: 08 DEC 2017
Table 1. Clinical features of fragile X syndrome.
Clinical characteristics Prevalence
Physical Long face
Macrocephaly
Prominent ears
Prominent jaw
Flat feet
Joint hypermobility
Macroorchidism
83%; occurs more commonly in adults
50–81%
75%
80%; occurs in adults only
29–69%
50–70%; occurs less commonly in adults
95%; occurs in adolescents and adults
Psychological Attention-deficit
hyperactivity disorder
Anxiety
Autism spectrum disorder
80% of boys and 40% of girls
58–86%
30–60%
Developmental Intellectual disability
Language deficits
85% of boys and 25–30% of girls
~100% of boys and 60–75% of girls
Other Strabismus
Recurrent otitis
Gastrointestinal complaints
Obesity
Seizures
8–30%
47–75%; occurs in the first 5 years of life
31%
30–61%
15–20%
Clinical features of fragile X syndrome11,14,15
obviously dysmorphic features, roughly 80% of patients with FXS
will present with at least one of these common characteristics14.
A significant overlap exists between FXS and ASD. Monogenic
disorders account for nearly one fifth of all diagnosed cases of
ASD, and the most common of these monogenic causes is
FXS16. Many of the behavioral characteristics seen in individu-
als with FXS, such as impaired social communication, social
anxiety, social gaze avoidance, and stereotypic behaviors, are
the same traits seen in other causes of ASD. Moreover, a large
portion of both individuals with FXS and individuals with
ASD meet criteria for attention-deficit hyperactivity disorder
(Figure 2). Just as ASD is seen more often in males than females,
males with FXS meet ASD criteria more frequently (60%)
than do females (20%)17. The overlap between FXS and ASD
also has a molecular basis, as FMRP controls the translation of
approximately 30% of the genes associated with ASD18. These
observations demonstrate that the underlying molecular etiolo-
gies of these disorders are intertwined, and targeted treatments
of FXS may be helpful for other types of ASD.
Clinical presentation of premutation disorders
Premutation expansions are associated with a variety of clinical
manifestations, including psychiatric, developmental, and neuro-
logical problems. The premutation causes psychiatric problems,
such as depression and anxiety, in approximately 50% of carri-
ers. It also causes premature ovarian failure (menopause occurring
before the age of 40) in a significant number of female carriers.
This condition, known as FXPOI, occurs in 16% to 20% of female
carriers; moreover, an additional 20% of carriers will experience
menopause before the age of 45. Some clinical manifestations
seen in those with the premutation are relatively common, even
in individuals with normal FMR1 alleles; however, their preva-
lence in premutation carriers is typically higher than their preva-
lence in the general population (Table 2). The greatest clinical
involvement associated with the premutation expansion results
from the neurodegenerative phenotype known as FXTAS, which
occurs in 40% of aging males and 16% of aging females with
the premutation19. FXTAS is a late-onset condition characterized
by intention tremor, cerebellar ataxia leading to frequent falling,
neuropathy, parkinsonian features, autonomic dysfunction, and
cognitive decline20. It is the most severe phenotype of premu-
tation-associated disorders. FXTAS is also characterized by
generalized brain atrophy and white matter disease in the middle
cerebellar peduncle and the splenium of the corpus callosum. In
addition, FXTAS is accompanied by the presence of intranuclear
inclusions in both neurons and astrocytes in the central nervous
system (CNS) as well as neurons in the peripheral nervous
system19,20.
Premutation disorders in extended family members are often
identified when a proband is diagnosed with FXS, after which
the proband’s mother is typically found to have the premutation.
Moreover, if the grandfather of the proband also has the premu-
tation, this means that all of the mother’s sisters will be obligate
carriers as well. The FMR1 premutation is known to be the most
common genetic cause of primary ovarian insufficiency2; thus,
premutation disorders are now also being identified through
OB-GYN offices, when fragile X DNA testing is ordered after
seeing ovarian dysfunction. If a female is diagnosed with the
premutation, her offspring have a 50% chance of inheriting a frag-
ile X mutation; whether offspring inherit a premutation or a full
Page 4 of 11
F1000Research 2017, 6(F1000 Faculty Rev):2112 Last updated: 08 DEC 2017
Table 2. Clinical involvement associated with the premutation.
Phenotype Prevalence (premutation carriers versus general population)
Male carriers Female
carriers
Male
non-carriers
Female
non-carriers
FXTAS 40% 16% N/A N/A
Fragile X-associated
primary ovarian
insufficiency
N/A 16–20% N/A ~1% (primary
ovarian insufficiency)
Hypertension 57% 22% ~30% ~30%
Migraine 27% 54% ~12% ~20%
Neuropathy 62% 17% <5% <5%
Sleep apnea 32% with FXTAS 32% with FXTAS ~15% ~5%
Psychiatric problems ~50% ~50% ~ 3.6%
(>45 years old)
~10.3%
(>45 years old)
19,20,2328 Abbreviations: FXTAS, fragile X-associated tremor ataxia syndrome; N/A, not applicable.
Figure 2. There is significant overlap between fragile X syndrome (FXS), autism spectrum disorder (ASD), and attention-deficit
hyperactivity disorder (ADHD). Approximately 60% of all patients with FXS also meet criteria for ASD, although FXS accounts for only 2% to
6% of all cases of ASD. Furthermore, nearly 80% of children with FXS and 50% of children with ASD have co-occurring ADHD21,22.
mutation depends on the number of CGG repeats in the mother
as well as on the number of AGG “anchors” present29. AGG
triplets, which typically interrupt CGG triplets after every nine or
ten repeats, have been shown to decrease the likelihood of expan-
sion to the full mutation when passed on to the next generation.
Therefore, higher numbers of these AGG anchors in the maternal
FMR1 gene lower the risk of the full mutation in the offspring29.
If a male has the premutation, all of his daughters will also carry
the premutation and are at risk of having children affected with
FXS20.
Mosaicism
Two types of mosaicism exist: size mosaicism and methylation
mosaicism. Size mosaicism refers to a condition in individuals
carrying both cells with the premutation and cells with the full
mutation in their blood. Furthermore, the ratio of premutation
cells versus full mutation cells in the blood may differ when com-
pared with other tissues, such as fibroblasts and brain tissue13,30.
Methylation mosaicism refers to a condition in individuals who
have the full mutation but only a portion of the cells containing
full mutation alleles are methylated; methylation status may vary
Page 5 of 11
F1000Research 2017, 6(F1000 Faculty Rev):2112 Last updated: 08 DEC 2017
from tissue to tissue as well30. The presence of either type (or
both types) of mosaicism can blur the boundaries between the
phenotypes of the premutation and the full mutation (FXS).
Individuals with methylation mosaicism produce more FMRP
than individuals with fully methylated full mutation alleles31.
Additionally, CGG repeat numbers in the premutation range and
FMRP expression are inversely related32; therefore, individu-
als with size mosaicism who carry premutation alleles in addi-
tion to full mutation alleles will likely also produce more FMRP
than non-mosaic individuals. Increased FMRP levels in FXS are
associated with fewer clinical symptoms and correlate directly
with IQ30; thus, individuals with mosaicism tend to be higher-
functioning. For example, one postmortem case study looked at
multiple tissues of a high-functioning patient displaying both size
mosaicism and methylation mosaicism. Only one region of the
brain, the parietal lobe, had a methylated full mutation and silenced
FMRP expression. However, FMRP was expressed in other parts
of the brain, including the superior temporal cortex, frontal cor-
tex, and hippocampus, which likely explains why this patient had
only mild cognitive and behavioral deficits33. Although mosaic
individuals with FXS may be less cognitively impaired than non-
mosaic individuals with FXS, they are more likely to develop
FXTAS if their mRNA levels are elevated1,20. This can lead to an
intriguing scenario known as the “double hit” phenomenon, in
which an individual is affected by both lowered FMRP levels and
elevated FMR1 mRNA levels34.
In 2013, Schneider and colleagues34 documented two broth-
ers in their 40s who both displayed this double hit of elevated
FMR1 mRNA levels and moderately decreased FMRP expres-
sion. The first brother, an individual with methylation mosai-
cism, presented with multiple physical features characteristic of
FXS, such as macroorchidism, flat feet, and a prominent jaw. He
was cognitively high-functioning but also displayed psychotic
symptoms, including a history of bipolar 1 disorder. The second
brother carried the premutation (118 CGG repeats); he experienced
typical development but presented with a few physical features
of FXS. Additionally, he presented with psychotic features asso-
ciated with major depressive disorder. Another case report docu-
mented a 58-year-old male with size and methylation mosaicism
and CGG repeats ranging from 20 to 80030. He presented with a
slightly below average IQ and physical features of FXS. At age
50, he experienced neurodegenerative symptoms characteristic
of FXTAS, including a worsening tremor and severe ataxia; he
eventually experienced the cognitive decline typical of males with
FXTAS. However, he was also an alcoholic, which may have
increased CNS toxicity and exacerbated his FXTAS progression.
Interestingly, he also met criteria for bipolar I disorder; taken
together, these reports indicate that individuals with a double hit
may be at an increased psychopathological risk. Additionally,
these case studies challenge the traditionally clear distinction
between FXS and premutation disorders and support the notion
of a spectrum-based nature of disorders associated with FMR1
mutations.
Epidemiology
The reported rates of prevalence of FXS in the general popula-
tion have been estimated to be approximately 1:5,000 males and
1:4,000 to 1:8,000 females3537. However, the estimated preva-
lence of the mutation has varied because of the differing testing
methodologies that have been used over time. Since FMR1 test-
ing began, the primary means of diagnosis has moved from cytoge-
netic testing for the presence of a folate-sensitive fragile site, to
Southern blot analyses, to polymerase chain reaction (PCR)-based
techniques. Additionally, wide variations in prevalence that are
seen in different populations make global prevalence estimates
difficult38,39.
To attempt to address these issues, Hunter and colleagues car-
ried out a systematic literature review and meta-analysis of the
prevalence of expanded FMR1 alleles by using a random effects
statistical model to analyze 54 epidemiological studies39. After
accounting for characteristics of the populations (that is, sub-
jects with or without ID) and including only studies that used
PCR and Southern blot techniques, obtaining screening data on
over 90,000 females and 50,000 males; the determined rates of
prevalence of the full mutation were 1:7,143 males and 1:11,111
females39. Additionally, meta-analyses investigating the preva-
lence of premutation alleles among the general population have
determined estimated frequencies of 1:150–300 females and
1:400–850 males35,39,40. Although we are gaining a better under-
standing of the prevalence of repeat expansions, this is not the only
type of mutation that can cause the disorder; a growing number
of deletions and point mutations on FMR1 have been identi-
fied in patients with FXS41,42. However, because repeat expan-
sions are tested far more frequently than these other mutations,
it is likely that the number of patients with FXS with non-repeat
mutations has been underestimated. Ideally, this epidemiologi-
cal shortcoming will be ameliorated through the increased use of
WES and microarray testing going forward.
The frequency of expanded FMR1 alleles varies globally because
of both founder effects and racial differences in haplotypes that
may predispose individuals in certain regions of the world to
CGG expansions43. Recently, the highest prevalence of expanded
alleles has been reported in Ricuarte, a small town in Colombia4.
In Ricuarte, the rates of prevalence of the full mutation are 1:21
males and 1:49 females, whereas the frequencies of the premu-
tation are 1:71 males and 1:28 females. This genetic cluster is
likely a consequence of a strong founder effect from founding
families that migrated from Spain. In sharp contrast to Ricuarte
is Ireland, which has extremely low rates of prevalence of the full
mutation: 1:10,619 males and 1:43,540 females. Researchers in
Ireland speculate that a lineage-specific haplotype is responsible
for this low incidence43. China also has a relatively low reported
incidence of FXS; however, owing to a gap between China and
Western countries regarding FXS awareness, it is likely that a
significant number of potential patients with FXS in China have
been misdiagnosed or underdiagnosed6. Other genetic clusters of
fragile X mutations can be found in various parts of the
Page 6 of 11
F1000Research 2017, 6(F1000 Faculty Rev):2112 Last updated: 08 DEC 2017
world, including Indonesia, Finland, and the Spanish island of
Mallorca5,44,45. These observations suggest that the prevalence
of FXS should be estimated separately in different countries or
continental regions, as certain shared predispositions or strong
instances of founder effects can lead to significantly different
prevalence rates.
Non-traditional ways of developing fragile X syndrome
FXS is almost always diagnosed through molecular testing for
a CGG repeat expansion in the triplet repeat sequence of FMR1,
as this is by far the most frequent cause of FXS8. However, recent
studies have implicated other mutations, such as point mutations
and deletions, in those with FXS41,42,46. There is nothing inher-
ent about repeat expansions in the full mutation that cause the
disorder; rather, it is the reduced FMRP production that leads to
FXS8. Thus, it is likely that any FMR1 mutation that adversely
affects the production of functional FMRP will lead to FXS.
Indeed, a review of non-expansion FMR1 mutations documented
various deletions causing FXS; over 20 case reports documented
FXS in patients who had normal CGG repeat numbers but had
deletions of varying sizes in the triplet repeat sequence or flanking
regions42.
Though less common, numerous point mutations in FMR1 have
also been discovered in individuals with FXS and other develop-
mental delays. In 2014, researchers documented FXS in a patient
with a loss-of-function missense mutation (p.(Gly266Glu)) and
a CGG repeat number of only 238. A separate case study attrib-
uted a patient’s FXS to a loss-of-function nonsense mutation
(p.(Ser27X); this patient’s CGG repeat number was just 2946. These
case studies corroborate the notion that any mutation that inter-
feres with the function of FMRP, not just full repeat expansions,
can lead to FXS. Furthermore, two related studies sequenced the
FMR1 gene of hundreds of developmentally delayed males who
did not meet diagnostic criteria for FXS; these mass sequenc-
ings revealed a total of three novel FMR1 missense mutations41,47.
Usually, developmentally delayed individuals with point muta-
tions on FMR1 have some of the features of FXS, although their
phenotype may vary in some ways from that of the typical patient
with FXS8,48.
Targeted treatments
Significant advances have been made in the development of
treatments for FXS. Animal models, including the FMR1 knock-
out (KO) mouse and the Drosophila fly model, have provided
researchers with several promising leads regarding effective
pharmacological interventions for patients with FXS. In addi-
tion, many clinical trials have been carried out in the past few
years and have been summarized in multiple comprehensive
reviews49,50. Moreover, a number of these trials have led to medi-
cations that are now available for use by treating physicians.
Minocycline, for example, is a semisynthetic tetracycline deriva-
tive that was first proven to be effective in improving anxiety
and cognition in KO mice51. A 3-month, double-blind control-
led trial of minocycline in children and adolescents with FXS
resulted in an improvement in global functioning as well as sig-
nificant improvements in anxiety and mood-related behaviors52.
Lovastatin, a specific inhibitor of the cholesterol biosynthesis
enzyme 3HMG-CoA reductase, has also shown promising results
in KO mice; in these mice, lovastatin decreased excessive pro-
tein production and blocked epileptiform activity in the mice
hippocampi9. A controlled trial combining lovastatin treatment
with parent-implemented language intervention (PILI) is currently
being carried out at the UC Davis MIND Institute, investigating
whether this combination of pharmacological and behavioral
interventions will improve spoken language and behavior in
children with FXS.
Another targeted treatment for FXS informed by animal studies
is metformin, a drug that is currently approved by the US Food
and Drug Administration for both obesity and type 2 diabetes.
Drosophila fly models of FXS, as well as KO mice, have dis-
played metformin’s ability to improve defects in circadian rhythm,
memory deficits, and social novelty5355. Clinically, a recent
open-label study saw anecdotal reports of improved behav-
ior and language in children and adults with FXS56,57. Although
initially metformin was hypothesized to be helpful in only those
with the Prader-Willi phenotype (PWP) of FXS, associated with
hyperphagia and severe obesity56, this study suggests that patients
without PWP or obesity may also benefit. This study demon-
strates the need for a large controlled trial of metformin to see
whether improvements in behavior, cognition, and language can
be seen in children and adults with FXS.
The neurobiological overlap between ASD and FXS has led to
the trial of a selective serotonin reuptake inhibitor (SSRI), spe-
cifically sertraline, to help alleviate the low serotonin levels seen
in the CNS of those with ASD10,58. Metabolomic studies in vari-
ous cases of ASD have demonstrated depressed levels of enzymes
with the ability to metabolize tryptophan into serotonin, indicat-
ing that an SSRI may be helpful59. An initial retrospective study of
young children with FXS demonstrated a significantly improved
receptive and expressive language trajectory in those on sertraline
compared with those not on sertraline60. A subsequent double-
blind controlled trial of low-dose sertraline in young children with
FXS found significant improvements in fine motor skills, visual
reception, and the composite T score on the Mullen Scales of
Early Development (MSEL). In a post-hoc assessment, the chil-
dren with FXS together with ASD (60% of the study population)
demonstrated significant improvement in expressive language on
the MSEL when on sertraline61.
Mavoglurant, an mGluR5 antagonist, has also been helpful in
animal models, but efficacy has not been demonstrated in ado-
lescents and adults with FXS62. Similarly, arbaclofen, a GABAB
agonist, has not shown efficacy in adults; however, outcome
data for a limited number of measures in children look more
promising63. There may be a number of reasons for the failure
of some trials to demonstrate efficacy. The outcome measures in
many previous studies included only behavioral checklists, which
are subject to parental bias62,63. Additionally, studies of arbaclofen
and sertraline suggest that young children may respond bet-
ter to targeted treatments than adults with FXS21,61. Addressing
some of these concerns, a current trial of mavoglurant combined
with PILI in young children with FXS is using novel outcome
measures such as event-related potentials (ERPs), eye tracking
Page 7 of 11
F1000Research 2017, 6(F1000 Faculty Rev):2112 Last updated: 08 DEC 2017
methodology, and language sampling to better detect cognitive
benefits and improvement in CNS function. Overall, a multitude
of targeted treatments for FXS have provided researchers with
encouraging results; however, much more research is needed
before we can establish which interventions, or combinations of
interventions, are the most effective for this population.
Whereas a significant amount of research has been dedicated
toward treatments for FXS, research into targeted treatments for
FXTAS and other premutation disorders is just beginning. A con-
trolled trial of memantine, an N-methyl-D-aspartate (NMDA)
receptor antagonist commonly used to treat Alzheimer’s disease,
did not demonstrate improvements in tremor, ataxia, or execu-
tive function deficits in individuals with FXTAS64. However, ERP
studies demonstrated improvements in brain processing and
attention in individuals with FXTAS when treatment was meman-
tine versus placebo65,66. A more recent open-label study dem-
onstrated that a weekly dose of intravenous allopregnanolone,
a GABAA agonist, over the course of 12 weeks in patients with
FXTAS resulted in improvements in both neuropsychological
testing and neuropathy symptoms67. As mentioned earlier,
researchers are still in the early stages of finding effective targeted
treatments for FXTAS and other premutation disorders, and there
are many more clinical trials expected to come.
Conclusions
Our understanding of FXS and other fragile X-associated
disorders has grown significantly in recent years. It has become
clear that disorders related to FMR1 mutations are associated with
a wide range of clinical presentations; there is a continuum of
clinical involvement from premutation disorders into full muta-
tion disorders due to both low FMRP and high FMR1 mRNA.
Some studies, such as reports indicating that high-functioning
individuals with mosaicism are at risk for FXTAS, illustrate the
ability of mosaicism to cloud the clinical picture of FXS and
premutation-related phenotypes. Additionally, the number of
detected deletions and point mutations in FMR1 will continue
to increase with the more widespread use of WES, WGS and
the identification of these non-repeat mutations is further widen-
ing the spectrum of clinical involvement in FXS. Lastly, there is
a promising outlook for effective targeted treatments; several
medications have shown encouraging results in both animal
models and clinical settings, and there are likely many more
effective interventions to be found.
Ethics
Written informed consent for the publication of the image found
in Figure 1 was obtained from the child’s parent.
Competing interests
Randi Hagerman has received funding from Novartis, Neuren,
Marinus, and Alcobra for carrying out treatment studies in
patients with FXS and has also consulted with Roche, Novartis,
Fulcrum and Zynerba regarding treatment studies in individuals
with FXS. The other authors declare that they have no competing
interests.
Grant information
This work was supported through the following grants: National
Institute of Child Health and Human Development grant
(NICHD) HD036071; the MIND Institute Intellectual and
Developmental Disabilities Research Center (NICHD U54
HD079125), HRSA R40MC26641, and HHS-ADD 90DD05969
for the Center for Excellence in Developmental Disabilities
support and the International Training Program for Neurodevelop-
mental Disorders at the MIND UC Davis Medical Center.
The funders had no role in study design, data collection and analysis,
decision to publish, or preparation of the manuscript.
References
1. Lozano R, Rosero CA, Hagerman RJ: Fragile X spectrum disorders. Intractable
Rare Dis Res. 2014; 3(4): 134–46.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
2. Hagerman R, Hagerman P: Advances in clinical and molecular understanding
of the FMR1 premutation and fragile X-associated tremor/ataxia syndrome.
Lancet Neurol. 2013; 12(8): 786–98.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
3. Sullivan AK, Marcus M, Epstein MP, et al.: Association of FMR1 repeat size with
ovarian dysfunction. Hum Reprod. 2005; 20(2): 402–12.
PubMed Abstract
|
Publisher Full Text
4. Saldarriaga W, et al.: Genetic Cluster of Fragile X Syndrome In A Colombian
District. Journal of Medical Genetics. 2017, (in press).
5. Oudet C, von Koskull H, Nordström AM, et al.: Striking founder effect for the
fragile X syndrome in Finland. Eur J Hum Genet. 1993; 1(3): 181–9.
PubMed Abstract
|
Publisher Full Text
6. Jin X, Chen L: Fragile X syndrome as a rare disease in China - Therapeutic
challenges and opportunities. Intractable Rare Dis Res. 2015; 4(1): 39–48.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
|
F1000 Recommendation
7. Basuta K, Schneider A, Gane L, et al.: High functioning male with fragile X
syndrome and fragile X-associated tremor/ataxia syndrome. Am J Med Genet
A. 2015; 167A(9): 2154–61.
PubMed Abstract
|
Publisher Full Text
8. Myrick LK, Nakamoto-Kinoshita M, Lindor NM, et al.: Fragile X syndrome due to a
missense mutation. Eur J Hum Genet. 2014; 22(10): 1185–9.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
9. Osterweil EK, Chuang S, Chubykin AA, et al.: Lovastatin corrects excess
protein synthesis and prevents epileptogenesis in a mouse model of fragile X
syndrome. Neuron. 2013; 77(2): 243–50.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
10. Hanson AC, Hagerman RJ: Serotonin dysregulation in Fragile X Syndrome:
implications for treatment. Intractable Rare Dis Res. 2014; 3(4): 110–7.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
11. Ciaccio C, Fontana L, Milani D, et al.: Fragile X syndrome: a review of clinical
and molecular diagnoses. Ital J Pediatr. 2017; 43(1): 39.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
|
F1000 Recommendation
12. Sobesky WE, Taylor AK, Pennington BF, et al.: Molecular/clinical correlations in
females with fragile X. Am J Med Genet. 1996; 64(2): 340–5.
PubMed Abstract
|
Publisher Full Text
Page 8 of 11
F1000Research 2017, 6(F1000 Faculty Rev):2112 Last updated: 08 DEC 2017
13. Pretto DI, Mendoza-Morales G, Lo J, et al.: CGG allele size somatic mosaicism
and methylation in FMR1 premutation alleles. J Med Genet. 2014; 51(5): 309–18.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
14. Saldarriaga W, Tassone F, González-Teshima LY, et al.: Fragile X syndrome.
Colomb Med. 2014; 45(4): 190–8.
PubMed Abstract
|
Free Full Text
15. Hagerman RJ, Hagerman PJ: Fragile X Syndrome. Baltimore: The John Hopkins
University Press. 2002.
Reference Source
16. Zafeiriou DI, Ververi A, Dafoulis V, et al.: Autism spectrum disorders: the quest
for genetic syndromes. Am J Med Genet B Neuropsychiatr Genet. 2013; 162B(4):
327–66.
PubMed Abstract
|
Publisher Full Text
17. Lee M, Martin GE, Berry-Kravis E, et al.: A developmental, longitudinal
investigation of autism phenotypic profiles in fragile X syndrome. J Neurodev
Disord. 2016; 8: 47.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
|
F1000 Recommendation
18. Iossifov I, O'Roak BJ, Sanders SJ, et al.: The contribution of de novo coding
mutations to autism spectrum disorder. Nature. 2014; 515(7526): 216–21.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
19. Polussa J, Schneider A, Hagerman R: Molecular Advances Leading to Treatment
Implications for Fragile X Premutation Carriers. Brain Disord Ther. 2014; 3:
pii: 1000119.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
20. Hagerman RJ, Hagerman P: Fragile X-associated tremor/ataxia syndrome -
features, mechanisms and management. Nat Rev Neurol. 2016; 12(7): 403–12.
PubMed Abstract
|
Publisher Full Text
21. Raspa M, Wheeler AC, Riley C: Public Health Literature Review of Fragile X
Syndrome. Pediatrics. 2017; 139(Suppl 3): S153–S171.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
|
F1000 Recommendation
22. Kaufmann WE, Kidd SA, Andrews HF, et al.: Autism Spectrum Disorder
in Fragile X Syndrome: Cooccurring Conditions and Current Treatment.
Pediatrics. 2017; 139(Suppl 3): S194–S206.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
|
F1000 Recommendation
23. Coffey SM, Cook K, Tartaglia N, et al.: Expanded clinical phenotype of women
with the FMR1 premutation. Am J Med Genet A. 2008; 146A(8): 1009–16.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
24. Sherman SL: Premature ovarian failure in the fragile X syndrome. Am J Med
Genet. 2000; 97(3): 189–94.
PubMed Abstract
|
Publisher Full Text
25. McDonald M, Hertz RP, Unger AN, et al.: Prevalence , awareness, and
management of hypertension, dyslipidemia, and diabetes among United States
adults aged 65 and older. J Gerontol A Biol Sci Med Sci. 2009; 64(2): 256–63.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
26. Bolay H, Ozge A, Saginc P, et al.: Gender influences headache characteristics
with increasing age in migraine patients. Cephalalgia. 2015; 35(9): 792–800.
PubMed Abstract
|
Publisher Full Text
27. Berry-Kravis E, Goetz CG, Leehey MA, et al.: Neuropathic features in fragile X
premutation carriers. Am J Med Genet A. 2007; 143A(1): 19–26.
PubMed Abstract
|
Publisher Full Text
28. Jonas DE, Amick HR, Feltner C, et al.: Screening for Obstructive Sleep Apnea
in Adults: An Evidence Review for the U.S. Preventive Services Task Force
[Internet]. U.S Preventie Services Task Force Evidence Syntheses, Evidence
Synthesis No. 146(AHRQ Publication 14-05216-EF-1). 2017.
PubMed Abstract
29. Yrigollen CM, Durbin-Johnson B, Gane L, et al.: AGG interruptions within the
maternal FMR1 gene reduce the risk of offspring with fragile X syndrome.
Genet Med. 2012; 14(8): 729–36.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
30. Pretto D, Yrigollen CM, Tang HT, et al.: Clinical and molecular implications of
mosaicism in FMR1 full mutations. Front Genet. 2014; 5: 318.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
31. Loesch DZ, Huggins RM, Hagerman RJ: Phenotypic variation and FMRP levels
in fragile X. Ment Retard Dev Disabil Res Rev. 2004; 10(1): 31–41.
PubMed Abstract
|
Publisher Full Text
32. Ludwig AL, Espinal GM, Pretto DI, et al.: CNS expression of murine fragile X
protein (FMRP) as a function of CGG-repeat size. Hum Mol Genet. 2014; 23(23):
3228–38.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
33. Taylor AK, Tassone F, Dyer PN, et al.: Tissue heterogeneity of the FMR1
mutation in a high-functioning male with fragile X syndrome. Am J Med Genet.
1999; 84(3): 233–9.
PubMed Abstract
|
Publisher Full Text
34. Schneider A, Seritan A, Tassone F, et al.: Psychiatric features in high-functioning
adult brothers with fragile x spectrum disorders. Prim Care Companion CNS
Disord. 2013; 15(2): pii: PCC.12l01492.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
35. Tassone F, Iong KP, Tong TH, et al.: FMR1 CGG allele size and prevalence
ascertained through newborn screening in the United States. Genome Med.
2012; 4(12): 100.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
36. Coffee B, Keith K, Albizua I, et al.: Incidence of fragile X syndrome by newborn
screening for methylated FMR1 DNA. Am J Hum Genet. 2009; 85(4):
503–14.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
37. Tassone F: Advanced technologies for the molecular diagnosis of fragile X
syndrome. Expert Rev Mol Diagn. 2015; 15(11): 1465–73.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
38. Hagerman PJ: The fragile X prevalence paradox. J Med Genet. 2008; 45(8):
498–9.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
39. Hunter J, Rivero-Arias O, Angelov A, et al.: Epidemiology of fragile X syndrome:
a systematic review and meta-analysis. Am J Med Genet A. 2014; 164A(7):
1648–58.
PubMed Abstract
|
Publisher Full Text
40. Hall D, Mailick M: The epidemiology of FXTAS. In FXTAS, FXPOI, and Other
Premutation Disorders, F. Tassone, Editor. Springer: Switzerland. 2016; 25–38.
Publisher Full Text
41. Handt M, Epplen A, Hoffjan S, et al.: Point mutation frequency in the FMR1 gene
as revealed by fragile X syndrome screening. Mol Cell Probes. 2014; 28(5–6):
279–83.
PubMed Abstract
|
Publisher Full Text
42. Wells RD: Mutation spectra in fragile X syndrome induced by deletions of
CGG*CCG repeats. J Biol Chem. 2009; 284(12): 7407–11.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
43. O'Byrne JJ, Sweeney M, Donnelly DE, et al.: Incidence of Fragile X syndrome
in Ireland. Am J Med Genet A. 2017; 173(3): 678–83.
PubMed Abstract
|
Publisher Full Text
|
F1000 Recommendation
44. Mundhofir FEP, Winarni TI, Nillesen W, et al.: Prevalence of fragile X syndrome in
males and females in Indonesia. WJMG. 2012; 2(3): 15–22.
Publisher Full Text
45. Alfaro Arenas R, Rosell Andreo J, Heine Suñer D: Fragile X syndrome
screening in pregnant women and women planning pregnancy shows a
remarkably high FMR1 premutation prevalence in the Balearic Islands. Am J
Med Genet B Neuropsychiatr Genet. 2016; 171(8): 1023–31.
PubMed Abstract
|
Publisher Full Text
|
F1000 Recommendation
46. Grønskov K, Brøndum-Nielsen K, Dedic A, et al.: A nonsense mutation in FMR1
causing fragile X syndrome. Eur J Hum Genet. 2011; 19(4): 489–91.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
47. Collins SC, Bray SM, Suhl JA, et al.: Identification of novel FMR1 variants by
massively parallel sequencing in developmentally delayed males. Am J Med
Genet A. 2010; 152A(10): 2512–20.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
48. Quartier A, Poquet H, Gilbert-Dussardier B, et al.: Intragenic FMR1 disease-
causing variants: a significant mutational mechanism leading to Fragile-X
syndrome. Eur J Hum Genet. 2017; 25(4): 423–31.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
|
F1000 Recommendation
49. Berry-Kravis E: Mechanism-based treatments in neurodevelopmental disorders:
fragile X syndrome. Pediatr Neurol. 2014; 50(4): 297–302.
PubMed Abstract
|
Publisher Full Text
50. Ligsay A, Hagerman RJ: Review of targeted treatments in fragile X syndrome.
Intractable Rare Dis Res. 2016; 5(3): 158–67.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
51. Bilousova TV, Dansie L, Ngo M, et al.: Minocycline promotes dendritic spine
maturation and improves behavioural performance in the fragile X mouse
model. J Med Genet. 2009; 46(2): 94–102.
PubMed Abstract
|
Publisher Full Text
52. Leigh MJ, Nguyen DV, Mu Y, et al.: 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.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
53. Monyak RE, Emerson D, Schoenfeld BP, et al.: Insulin signaling
misregulation underlies circadian and cognitive deficits in a Drosophila fragile
X model. Mol Psychiatry. 2017; 22(8): 1140–8.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
|
F1000 Recommendation
54. Weisz ED, Monyak RE, Jongens TA: Deciphering discord: How Drosophila
research has enhanced our understanding of the importance of FMRP in
different spatial and temporal contexts. Exp Neurol. 2015; 274(Pt A): 14–24.
PubMed Abstract
|
Publisher Full Text
|
F1000 Recommendation
55. Gantois I, Khoutorsky A, Popic J, et al.: Metformin ameliorates core deficits
in a mouse model of fragile X syndrome. Nat Med. 2017; 23(6): 674–7.
PubMed Abstract
|
Publisher Full Text
|
F1000 Recommendation
56. Muzar Z, Lozano R, Kolevzon A, et al.: The neurobiology of the Prader-Willi
phenotype of fragile X syndrome. Intractable Rare Dis Res. 2016; 5(4):
255–61.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
57. Dy ABC, Tassone F, Eldeeb M, et al.: Metformin as targeted treatment in fragile
X syndrome. Clin Genet. 2017.
PubMed Abstract
|
Publisher Full Text
58. Chandana SR, Behen ME, Juhász C, et al.: Significance of abnormalities in
developmental trajectory and asymmetry of cortical serotonin synthesis in
Page 9 of 11
F1000Research 2017, 6(F1000 Faculty Rev):2112 Last updated: 08 DEC 2017
autism. Int J Dev Neurosci. 2005; 23(2–3): 171–82.
PubMed Abstract
|
Publisher Full Text
59. Boccuto L, Chen CF, Pittman AR, et al.: Decreased tryptophan metabolism in
patients with autism spectrum disorders. Mol Autism. 2013; 4(1): 16.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
60. Indah Winarni T, Chonchaiya W, Adams E, et al.: Sertraline may improve
language developmental trajectory in young children with fragile x syndrome:
a retrospective chart review. Autism Res Treat. 2012; 2012: 104317.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
61. Greiss Hess L, Fitzpatrick SE, Nguyen DV, et al.: A Randomized, Double-Blind,
Placebo-Controlled Trial of Low-Dose Sertraline in Young Children With Fragile
X Syndrome. J Dev Behav Pediatr. 2016; 37(8): 619–28.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
62. Berry-Kravis E, Des Portes V, Hagerman R, et al.: Mavoglurant in fragile X
syndrome: Results of two randomized, double-blind, placebo-controlled trials.
Sci Transl Med. 2016; 8(321): 321ra5.
PubMed Abstract
|
Publisher Full Text
|
F1000 Recommendation
63. Berry-Kravis E, Hagerman R, Visootsak J, et al.: Arbaclofen in fragile X
syndrome: results of phase 3 trials. J Neurodev Disord. 2017; 9: 3.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
64. Seritan AL, Nguyen DV, Mu Y, et al.: Memantine for fragile X-associated tremor/
ataxia syndrome: a randomized, double-blind, placebo-controlled trial. J Clin
Psychiatry. 2014; 75(3): 264–71.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
65. Yang JC, Niu YQ, Simon C, et al.: Memantine effects on verbal memory in fragile
X-associated tremor/ataxia syndrome (FXTAS): a double-blind brain potential
study. Neuropsychopharmacology. 2014; 39(12): 2760–8.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
66. Yang JC, Rodriguez A, Royston A, et al.: Memantine Improves Attentional
Processes in Fragile X-Associated Tremor/Ataxia Syndrome:
Electrophysiological Evidence from a Randomized Controlled Trial. Sci Rep.
2016; 6: 21719.
PubMed Abstract
|
Publisher Full Text
|
Free Full Text
67. Wang JY, Trivedi AM, Carrillo NR, et al.: Open-Label Allopregnanolone Treatment
of Men with Fragile X-Associated Tremor/Ataxia Syndrome. Neurotherapeutics.
2017; 1–11.
PubMed Abstract
|
Publisher Full Text
Page 10 of 11
F1000Research 2017, 6(F1000 Faculty Rev):2112 Last updated: 08 DEC 2017
Open Peer Review
Current Referee Status:
Editorial Note on the Review Process
arecommissionedfrommembersoftheprestigious andareeditedasaF1000FacultyReviews F1000Faculty
servicetoreaders.Inordertomakethesereviewsascomprehensiveandaccessibleaspossible,thereferees
provideinputbeforepublicationandonlythefinal,revisedversionispublished.Therefereeswhoapprovedthe
finalversionarelistedwiththeirnamesandaffiliationsbutwithouttheirreportsonearlierversions(anycomments
willalreadyhavebeenaddressedinthepublishedversion).
The referees who approved this article are:
Version 1
ThebenefitsofpublishingwithF1000Research:
Yourarticleispublishedwithindays,withnoeditorialbias
Youcanpublishtraditionalarticles,null/negativeresults,casereports,datanotesandmore
Thepeerreviewprocessistransparentandcollaborative
YourarticleisindexedinPubMedafterpassingpeerreview
Dedicatedcustomersupportateverystage
Forpre-submissionenquiries,contact research@f1000.com
SectiononGeneStructureandDisease,LaboratoryofMolecularandCellularBiology,NationalKaren Usdin
InstituteofDiabetes,DigestiveandKidneyDiseases,NationalInstitutesofHealth,Bethesda,MD,USA
Nocompetinginterestsweredisclosed.Competing Interests:
1
DepartmentofMedicalGenetics,UniversityofAntwerp,Antwerp,BelgiumR Frank Kooy
Nocompetinginterestsweredisclosed.Competing Interests:
1
Page 11 of 11
F1000Research 2017, 6(F1000 Faculty Rev):2112 Last updated: 08 DEC 2017
... The 5' untranslated region (UTR) of the human FMR1 gene has polymorphic CGG trinucleotide repeats, with 30 repeats as the mode in the human population [7,8]. The expansion of 55 to 200 repeats is associated with pathological conditions, including neurodegenerative disorder fragile X-associated tremor/ataxia syndrome (FXTAS) in males and fragile-X-associated primary ovarian insufficiency (FXPOI) in females [9]. The expansion of CGG repeats to over 200 leads to DNA methylation and silencing of the FMR1 gene, which is the main cause of FXS. ...
... FXS patients may exhibit a wide range of symptoms, with the most common being moderate to severe intellectual disability, language impairment, anxiety, hyperactivity, aggression, and increased seizure risk [1,22]. They also have physical attributes such as elongated face, large ears, joint hypermobility, and macroorchidism [9]. Most patients have many behavioral symptoms that overlap with autism such as attention deficit disorder, repetitive behaviors, sleep problems, and sensory overload [9,23]. ...
... They also have physical attributes such as elongated face, large ears, joint hypermobility, and macroorchidism [9]. Most patients have many behavioral symptoms that overlap with autism such as attention deficit disorder, repetitive behaviors, sleep problems, and sensory overload [9,23]. To understand the pathophysiology of FXS, researchers have used genetic techniques to create animal models lacking FMRP expression. ...
Article
Full-text available
Fragile X syndrome (FXS) is caused by epigenetic silencing of the X-linked fragile X messenger ribonucleoprotein 1 (FMR1) gene located on chromosome Xq27.3, which leads to the loss of its protein product, fragile X messenger ribonucleoprotein (FMRP). It is the most prevalent inherited form of intellectual disability and the highest single genetic cause of autism. Since the discovery of the genetic basis of FXS, extensive studies using animal models and human pluripotent stem cells have unveiled the functions of FMRP and mechanisms underlying FXS. However, clinical trials have not yielded successful treatment. Here we review what we have learned from commonly used models for FXS, potential limitations of these models, and recommendations for future steps. Supplementary Information The online version contains supplementary material available at 10.1186/s11689-024-09545-w.
... [1] It is the most common cause of single-gene autism spectrum disorder (ASD) and is believed to be responsible for 2-6% of all autism cases. The disease primarily affects the nervous system resulting in mental retardation, lowered socio-behavioral adaptation, and intellectual disability (ID) [2] . The dysmorphic features present in the FXS include a long face with a broad forehead, prominent jaw and ears, and a high-arched palate. ...
... 18] Most common features of FXS noticeable upon physical examination and history taking.[2,3,19] ...
Article
Full-text available
Fragile X syndrome is a dominantly inherited genetic disease and is a consequence of the FMR1 gene mutation located on the X chromosome. The number of patients affected by this disease with full mutation is estimated at 1 in 4000 men and 1 in 8000 women, however, the number of carriers with premutation is far greater. Depending on the mutation of the FMR1 gene and levels of its products FMRP a variety of symptoms can be observed including- intellectual disability, mental retardation, lowered behavioral adaptation, autism, and dysmorphic features such as a long face with a broad forehead and prominent ears. The treatment is mostly symptomatic- managing comorbidities and an emphasis on psychological therapy. The objective of this paper is to sum up the most up-to-date knowledge regarding the pathogenesis, treatment- current and clinically tested, and novelties in the Fragile X syndrome
... In addition, premutation carriers need to know the risk of premutation disorders. FXPOI occurs in about 20% of female carriers [35] ; FXTAS occurs in about 40% of aging male carriers and 16% of aging female carriers [6] , and FXAND, which encompasses depression, anxiety, insomnia, chronic pain, and chronic fatigue and social deficits, occurs in up to 50% of carriers, typically in adulthood, but sometimes also in childhood [7,36,37] . ...
Article
Full-text available
Fragile X syndrome (FXS) is caused by a full mutation (> 200 repeats) in the 5’untranslated region of the Fragile X Messenger Ribonucleoprotein 1 gene (FMR1 gene), which leads to methylation and silencing of expression, generating the total or partial absence of its product, FMR1 protein (FMRP). When the repetitions are between 55 and 200 cytosine-guanine-guanine (CGG) repeats, it is called a premutation, which is related to a wide spectrum of conditions such as Fragile X-associated tremor ataxia syndrome (FXTAS), fragile X-associated primary ovarian insufficiency (FXPOI), and fragile X-associated neuropsychiatric disorders (FXAND). High levels of FMR1 mRNA are implicated in premutation pathophysiology, which differs from the deficiency or absence of FMRP in FXS. In recent years, numerous attempts have been made to find treatments that can counteract the effects of the absence of FMRP and improve symptoms associated with the condition such as intellectual disability, anxiety, autism, stereotypies, language delay, and aggressive behavior. Here, we review current treatments in addition to targeted treatments that can reverse some of the neurobiological abnormalities in those with FXS. We also review molecular interventions that will hopefully lead to a promising future for those affected by FXS and their families.
... In addition, in preschool children, symptoms within the autism spectrum disorder phenotype, such as impairment of social communication, social anxiety, absence of eye contact, and stereotypic behavior, including hand flapping and hand biting, are often observed. Parents, caregivers, or teachers may detect irritable behavior, hyperarousal and learning problems 51 . In early to middle childhood and adolescence, neuropsychiatric disorders such as attention deficit and hyperactivity disorder and anxiety appear, and physical characteristics typical of Fragile X syndrome become more evident as they age 49 . ...
Article
Full-text available
Fragile X syndrome is caused by the expansion of CGG triplets in the FMR1 gene, which generates epigenetic changes that silence its expression. The absence of the protein coded by this gene, FMRP, causes cellular dysfunction, leading to impaired brain development and functional abnormalities. The physical and neurologic manifestations of the disease appear early in life and may suggest the diagnosis. However, it must be confirmed by molecular tests. It affects multiple areas of daily living and greatly burdens the affected individuals and their families. Fragile X syndrome is the most common monogenic cause of intellectual disability and autism spectrum disorder; the diagnosis should be suspected in every patient with neurodevelopmental delay. Early interventions could improve the functional prognosis of patients with Fragile X syndrome, significantly impacting their quality of life and daily functioning. Therefore, healthcare for children with Fragile X syndrome should include a multidisciplinary approach.
... Microscopic analysis of the excised specimen confirmed the diagnosis of OM as the excised specimen revealed typical features such as focal areas of odontogenic lining epithelium, Some reports have been published on the diagnosis and treatment of malignant neoplasms in FXS. [12][13][14] Another study described an increased risk for the development of lip cancer. 15 Bone tumors are extremely rare in FXS patients and have seldom been reported. ...
... Both girls and boys diagnosed with FXS tend to have connective tissue anomalies that can lead to heart disease (e.g., mitral valve prolapses) and are atypically short but otherwise have a normal physical appearance. Overall, the clinical presentation of FXS is less observable and more variable in female individuals than in their male counterparts [58]. ...
Article
Full-text available
This review discusses the discovery, epidemiology, pathophysiology, genetic etiology, molecular diagnosis, and medication-based management of fragile X syndrome (FXS). It also highlights the syndrome’s variable expressivity and common comorbid and overlapping conditions. FXS is an X-linked dominant disorder associated with a wide spectrum of clinical features, including but not limited to intellectual disability, autism spectrum disorder, language deficits, macroorchidism, seizures, and anxiety. Its prevalence in the general population is approximately 1 in 5000–7000 men and 1 in 4000–6000 women worldwide. FXS is associated with the fragile X messenger ribonucleoprotein 1 (FMR1) gene located at locus Xq27.3 and encodes the fragile X messenger ribonucleoprotein (FMRP). Most individuals with FXS have an FMR1 allele with > 200 CGG repeats (full mutation) and hypermethylation of the CpG island proximal to the repeats, which silences the gene’s promoter. Some individuals have mosaicism in the size of the CGG repeats or in hypermethylation of the CpG island, both produce some FMRP and give rise to milder cognitive and behavioral deficits than in non-mosaic individuals with FXS. As in several monogenic disorders, modifier genes influence the penetrance of FMR1 mutations and FXS’s variable expressivity by regulating the pathophysiological mechanisms related to the syndrome’s behavioral features. Although there is no cure for FXS, prenatal molecular diagnostic testing is recommended to facilitate early diagnosis. Pharmacologic agents can reduce some behavioral features of FXS, and researchers are investigating whether gene editing can be used to demethylate the FMR1 promoter region to improve patient outcomes. Moreover, clustered regularly interspaced palindromic repeats (CRISPR)/Cas9 and developed nuclease defective Cas9 (dCas9) strategies have promised options of genome editing in gain-of-function mutations to rewrite new genetic information into a specified DNA site, are also being studied.
... Elevated FXS FMRP levels are associated with fewer clinical symptoms and positively correlate with I.Q. (Rajaratnam et al.,2017). ...
Article
Full-text available
This analysis aimed to clarify the molecular basis of fragile X syndrome and explain the role of genetic material in the genetic disease's development and treatment. Fragile X syndrome is an X-linked mutation inheritance disorder. The mutated gene is called FMR-1. This is important for normal brain development and synaptic plasticity, which was verified and recognized in 1991, and this has become a hope for more clarification. FMR1 influences the translation of messenger RNA (mRNA), but identifying functional targets was complex and directly related to translational control and showed that dysregulated translation initiation signaling was observed for the FMR1 gene in the FMR1 knockout mouse model of FXS. Because of the epigenetic alteration, such as hypermethylated at the DNA promoter region, and chromatin modification, such as H3K9 methylation, the FMR1 gene can be imprinted. Still, their mechanisms of aberrant epigenetic marks play a role in the etiology of many neurodevelopmental disorders, some of which we still do not fully understand and need to show more. The opportunities for epigenetic markers to map and alter epigenetic marks and the potential for therapies based on epigenetics and noncoding manipulation. For neurodevelopmental and behavioral conditions, including mental retardation, autism, anxiety, and mood disturbance, FMR1 loss of control is a model. Most studies have focused on the new and effective approach for Fragile X syndrome, which is Gene therapy is unarguably the definitive way to treat and possibly cure genetic diseases. Many of them are under clinical trial, but more studies, such as the CRISPR/Cas9-based method, should be approved. Adeno-associated viral (AAV) vectors are highly effective for generating models. Most research is used in the mouse model of fragile X syndrome, where AAVs have been used to express fragile X mental retardation protein (FMRP), which is missing or highly reduced in the disorder. The vast expansions need southern blotting in myotonic dystrophy. Fragile X is diagnosed by a form of Southern blotting that relies on the size and the FRAXA gene's methylation status. Almost always, genetic testing is performed by PCR. The few Southern blotting uses include checking for significant destructive gene rearrangements and complete mutations of fragile X and myotonic dystrophy. Expansions suppress the expression of closely adjacent genes, causing loss of function. A named FMRI (fragile-X mental retardation syndrome) gene cDNA probe. Some unique molecular mechanisms, such as CGG expansion in Fragile-X syndrome, can make a particular sequence change in a gene far more probable than any other change.
... The European Fragile X Network (EFXN), in 2019, had a symposium on recent research on conditions which affect fragile X premutation carriers; they agreed to use the term fragile X-associated neuropsychiatric disorder (FXAND), including auto-immune conditions to describe all conditions that might be associated with premutation syndrome [16]. The present study enrolled 92 Egyptian male children age ranged from 6 to 14 years (mean ± SD; 7.7521 ± 1.017) manifesting various clinical features similar to those typical of FXS cases described in previous studies [17]. They suffered from intellectual disability (n: 86/92; 94%), autistic were discovered within the current study; about 14% of intermediate alleles are unstable and are considered to be precursors of premutation cases when transmitted from mothers to their offspring. ...
Article
Full-text available
Background Fragile X syndrome (FXS) is the most common form of inherited intellectual disability, caused by CGG-repeats expansion (> 200 repeats). Premutation alleles (PM) (55–200 CGG repeats) are associated with tremor ataxia syndrome (FXTAS), fragile X-associated primary ovarian insufficiency (FXPOI), and autistic problems. Aim To screen the frequency of premutation carriers using molecular diagnostic assays, in a cohort of Egyptian males with suspected clinical features of (FXS) checking for the presence of premutation alleles. Methods The current study comprised 192 Egyptian male children, 92 participants presented with intellectual disability, delayed language development, autistic-like features, behavioral difficulties, anxiety, seizures, and depression compared to 100 healthy males. All cases were subjected to clinical and neuroimaging assessments, when indicated as well as molecular analysis using methylation-specific PCR (MS-PCR) and quantitative real-time PCR (qRT-PCR). Results Thirty-four premutation carriers out of 92 Egyptian males (37%) of CGG repeats (55 to 200) were illustrated with elevated FMR1 mRNA expression level ( p -value < 0.001). Additionally, 2 intermediate (IM) cases (0.03%) (45–55 CGG repeats) showed poor increase in expression level ( p -value = 0.02838) plus 6 full mutation (FM) patients (0.07%) with (> 200 CGG repeats) ( p -value < 0.001) resulted in FMR1 gene silence. Conclusion Molecular diagnostic assay including (MS-PCR) and (qRT-PCR) proved to be a sensitive and rapid screening tool for the detection of premutation cases. Furthermore, the presence of positive correlation between FMR1 mRNA expression levels with CGG repeats in premutation cases could serve as a potential diagnostic marker. Application of these diagnostic tools on larger number clinically suspected cases is recommended.
... Psychiatric manifestations are also more prevalent in males than females. 12 The typical clinical features include a large, long head and ears, short stature, hyperextensible joints, post-pubertal macroorchidism, and mild to severe mental retardation. Attention deficit hyperkinetic disorder (ADHD), learning disorders, and pervasive developmental disorders such as autism are more common. ...
Article
Full-text available
Neuropsychiatry is an area of medicine that deals with behavioural issues caused by brain dysfunction. It is found at the intersection of neurology and psychiatry. Individuals with congenital and genetic diseases are more likely to experience neuropsychiatric symptoms (particularly mental retardation), which can lead to considerable disability and a lower quality of life. Developmental delay, intellectual disability, autism spectrum disorders (ASDs), and cognitive dysfunction are the most common symptoms of neuropsychiatric illnesses. Many intellectual developmental abnormalities are caused by complex genetic components (such as attention deficit hyperactivity disorder), pregnancy or birth complications, or environmental variables, among other things. Patients with congenital and genetic diseases with neuropsychiatric indications benefit from a multidisciplinary approach to management. Interdepartmental liaisoning may be advantageous in the absence of a multidisciplinary team.
Article
The class X-associated spectrum disorders (FXSD) combines the following clinical syndromes: fragile X chromosome-linked mental retardation syndrome (fragile X syndrome, FXS), fragile X-associated tremor/ataxia syndrome (FXTAS), fragile X-associated primary ovarian insufficiency (FXPOI), and a group of neuropsychiatric disorders (fragile X-associated neuropsychiatric disorders, FXAND). These syndromes occur as a result of dynamic mutations of the FMR1 gene caused by expansion of trinucleotide repeats in the gene promoter. FXS syndrome (mental retardation, autism spectrum disorder (ASD), attention deficit hyperactivity disorder (ADHD)) occurs when the FMR1 gene is completely mutated (an increase in the number of CGG repeats to more than 200), which is accompanied by full or partial suppression of FMRP protein expression. The clinical manifestations of FXTAS, FXPOI, and FXAND can occur in individuals carrying the FMR1 premutation (CGG repeats in the range 55–200). The main pathogenetic mechanisms of these diseases include a decrease in FMRP expression and accumulation of FMR1 mRNA containing an increased number of repeats. This review analyzes genophenotypic relationships within the spectrum of conditions associated with FMR1 dynamic mutations. An analysis of the relationship between the molecular mechanisms (FMRP defi ciency, translation of atypical isoforms, increased function of FMR1 mRNA) and clinical manifestations (level of cognitive development, severity of ASD, severity of symptoms of FXTAS, FXPOI and FXAND)is presented.
Article
Full-text available
Background: Fragile X syndrome (FXS) is the most common cause of inherited intellectual disabilities and autism. The reported prevalence of the full mutation (FM) gene FMR1 in the general population is 0.2-0.4 per 1000 males and 0.125-0.4 per 1000 females. Population screening for FMR1 expanded alleles has been performed in newborns and in an adult population. However, it has never been carried out in an entire town. Ricaurte is a Colombian district with 1186 habitants, with a high prevalence of FXS, which was first described by cytogenetic techniques in 1999. Methods: Using a PCR-based approach, screening for FXS was performed on blood spot samples obtained from 926 (502 males and 424 females) inhabitants from Ricaurte, accounting for 78% of total population. Results: A high prevalence of carriers of the expanded allele was observed in all FXS mutation categories. Using the Bayesian methods the carrier frequency of FM was 48.2 (95% Credibility Region CR: 36.3-61.5) per 1000 males and 20.5 (95% CR:13.5-28.6) per 1000 females; the frequency of premutation carrier was 14.1 (95% RC: 8.0-21.7) per 1000 males (95% RC: 8.0-21.7 per 1000 males) and 35.9 (95% RC: 26.5-46.2) per 1000 for females (95% RC: 26.5-46.2 per 1000 females), and gray zone carrier was 13.4 (95% RC: 7.4-20.7) per 1000 males (95% RC: 7.4-20.7 per 1000 males) and 42.2 (95% RC: 32.2-53.8) per 1000 for females (95% RC: 32.2-53.8 per 1000 females). Differences in carrier frequencies were observed for premutation and FM alleles between natives and non-natives. Conclusions: This study shows that in Ricaurte the carrier frequencies of FMR1 expanded alleles (premutations and FMs) are higher than those reported in the literature, suggesting that Ricaurte constitutes a genetic cluster of FXS.
Article
Full-text available
Background Arbaclofen improved multiple abnormal phenotypes in animal models of fragile X syndrome (FXS) and showed promising results in a phase 2 clinical study. The objective of the study is to determine safety and efficacy of arbaclofen for social avoidance in FXS. Methods Two phase 3 placebo-controlled trials were conducted, a flexible dose trial in subjects age 12–50 (209FX301, adolescent/adult study) and a fixed dose trial in subjects age 5–11 (209FX302, child study). The primary endpoint for both trials was the Social Avoidance subscale of the Aberrant Behavior Checklist-Community Edition, FXS-specific (ABC-CFX). Secondary outcomes included other ABC-CFX subscale scores, Clinical Global Impression-Improvement (CGI-I), Clinical Global Impression-Severity (CGI-S), and Vineland Adaptive Behavior Scales, Second Edition (Vineland-II) Socialization domain score. Results A total 119 of 125 randomized subjects completed the adolescent/adult study (n = 57 arbaclofen, 62 placebo) and 159/172 completed the child study (arbaclofen 5 BID n = 38; 10 BID n = 39; 10 TID n = 38; placebo n = 44). There were no serious adverse events (AEs); the most common AEs included somatic (headache, vomiting, nausea), neurobehavioral (irritability/agitation, anxiety, hyperactivity), decreased appetite, and infectious conditions, many of which were also common on placebo. In the combined studies, there were 13 discontinuations (n = 12 arbaclofen, 1 placebo) due to AEs (all neurobehavioral). The adolescent/adult study did not show benefit for arbaclofen over placebo for any measure. In the child study, the highest dose group showed benefit over placebo on the ABC-CFX Irritability subscale (p = 0.03) and Parenting Stress Index (PSI, p = 0.03) and trends toward benefit on the ABC-CFX Social Avoidance and Hyperactivity subscales (both p < 0.1) and CGI-I (p = 0.119). Effect size in the highest dose group was similar to effect sizes for FDA-approved serotonin reuptake inhibitors (SSRIs). Conclusions Arbaclofen did not meet the primary outcome of improved social avoidance in FXS in either study. Data from secondary measures in the child study suggests younger patients may derive benefit, but additional studies with a larger cohort on higher doses would be required to confirm this finding. The reported studies illustrate the challenges but represent a significant step forward in translating targeted treatments from preclinical models to clinical trials in humans with FXS.
Article
Full-text available
Background and objective: Individuals with fragile X syndrome (FXS) are frequently codiagnosed with autism spectrum disorder (ASD). Most of our current knowledge about ASD in FXS comes from family surveys and small studies. The objective of this study was to examine the impact of the ASD diagnosis in a large clinic-based FXS population to better inform the care of people with FXS. Methods: The study employed a data set populated by data from individuals with FXS seen at specialty clinics across the country. The data were collected by clinicians at the patient visit and by parent report for nonclinical and behavioral outcomes from September 7, 2012 through August 31, 2014. Data analyses were performed by using χ(2) tests for association, t tests, and multiple logistic regression to examine the association between clinical and other factors with ASD status. Results: Half of the males and nearly 20% of females met Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition criteria for current ASD. Relative to the FXS-only group, the FXS with ASD (FXS+ASD) group had a higher prevalence of seizures (20.7% vs 7.6%, P < .001), persistence of sleep problems later in childhood, increased behavior problems, especially aggressive/disruptive behavior, and higher use of α-agonists and antipsychotics. Behavioral services, including applied behavior analysis, appeared to be underused in children with FXS+ASD (only 26% and 16% in prekindergarten and school-age periods, respectively) relative to other populations with idiopathic ASD. Conclusions: These findings confirm among individuals with FXS an association of an ASD diagnosis with important cooccurring conditions and identify gaps between expected and observed treatments among individuals with FXS+ASD.
Article
Full-text available
Fragile X syndrome (FXS) is the leading monogenic cause of autism spectrum disorders (ASD). Trinucleotide repeat expansions in FMR1 abolish FMRP expression, leading to hyperactivation of ERK and mTOR signaling upstream of mRNA translation. Here we show that metformin, the most widely used drug for type 2 diabetes, rescues core phenotypes in Fmr1-/y mice and selectively normalizes ERK signaling, eIF4E phosphorylation and the expression of MMP-9. Thus, metformin is a potential FXS therapeutic. © 2017 Nature America, Inc., part of Springer Nature. All rights reserved.
Article
Full-text available
Background: Individuals with fragile X syndrome (FXS) have both behavioral and medical comorbidities and the latter include obesity in approximately 30% and the Prader-Willi Phenotype (PWP) characterized by severe hyperphagia and morbid obesity in less than 10%. Metformin is a drug used in individuals with type 2 diabetes, obesity or impaired glucose tolerance and it has a strong safety profile in children and adults. Recently published studies in the Drosophila model and the knock out mouse model of FXS treated with metformin demonstrate the rescue of multiple phenotypes of FXS. Materials and methods: We present 7 cases of individuals with FXS who have been treated with metformin clinically. One case with type 2 diabetes, 3 cases with the PWP, 2 adults with obesity and/or behavioral problems and, a young child with FXS. These individuals were clinically treated with metformin and monitored for behavioral changes with the Aberrant Behavior Checklist and metabolic changes with a fasting glucose and HgbA1c. Results: We found consistent improvements in irritability, social responsiveness, hyperactivity, and social avoidance, in addition to comments from the family regarding improvements in language and conversational skills. No significant side-effects were noted and most patients with obesity lost weight. Conclusion: We recommend a controlled trial of metformin in those with FXS. Metformin appears to be an effective treatment of obesity including those with the PWP in FXS. Our study suggests that metformin may also be a targeted treatment for improving behavior and language in children and adults with FXS.
Article
Full-text available
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. DiscussionFXS 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.
Article
Fragile X-associated tremor/ataxia syndrome (FXTAS) is a late-onset neurodegenerative disorder affecting approximately 45% of male and 16% of female carriers of the FMR1 premutation over the age of 50 years. Currently, no effective treatment is available. We performed an open-label intervention study to assess whether allopregnanolone, a neurosteroid promoting regeneration and repair, can improve clinical symptoms, brain activity, and magnetic resonance imaging (MRI) measurements in patients with FXTAS. Six patients underwent weekly intravenous infusions of allopregnanolone (2–6 mg over 30 min) for 12 weeks. All patients completed baseline and follow-up studies, though MRI scans were not collected from 1 patient because of MRI contraindications. The MRI scans from previous visits, along with scans from 8 age-matched male controls, were also included to establish patients’ baseline condition as a reference. Functional outcomes included quantitative measurements of tremor and ataxia and neuropsychological evaluations. Brain activity consisted of event-related potential N400 word repetition effect during a semantic memory processing task. Structural MRI outcomes comprised volumes of the hippocampus, amygdala, and fluid-attenuated inversion recovery hyperintensities, and microstructural integrity of the corpus callosum. The results of the study showed that allopregnanolone infusions were well tolerated in all subjects. Before treatment, the patients disclosed impairment in executive function, verbal fluency and learning, and progressive deterioration of all MRI measurements. After treatment, the patients demonstrated improvement in executive functioning, episodic memory and learning, and increased N400 repetition effect amplitude. Although MRI changes were not significant as a group, both improved and deteriorated MRI measurements occurred in individual patients in contrast to uniform deterioration before the treatment. Significant correlations between baseline MRI measurements and changes in neuropsychological test scores indicated the effects of allopregnanolone on improving executive function, learning, and memory for patients with relatively preserved hippocampus and corpus callosum, while reducing psychological symptoms for patients with small hippocampi and amygdalae. The findings show the promise of allopregnanolone in improving cognitive functioning in patients with FXTAS and in partially alleviating some aspects of neurodegeneration. Further studies are needed to verify the efficacy of allopregnanolone for treating FXTAS.
Article
Objectives: The purpose of this systematic literature review is to describe what is known about fragile X syndrome (FXS) and to identify research gaps. The results can be used to help inform future public health research and provide pediatricians with up-to-date information about the implications of the condition for individuals and their families. Methods: An electronic literature search was conducted, guided by a variety of key words. The search focused on 4 areas of both clinical and public health importance: (1) the full mutation phenotype, (2) developmental trajectories across the life span, (3) available interventions and treatments, and (4) impact on the family. A total of 661 articles were examined and 203 were included in the review. Results: The information is presented in the following categories: developmental profile (cognition, language, functional skills, and transition to adulthood), social-emotional profile (cooccurring psychiatric conditions and behavior problems), medical profile (physical features, seizures, sleep, health problems, and physiologic features), treatment and interventions (educational/behavioral, allied health services, and pharmacologic), and impact on the family (family environment and financial impact). Research gaps also are presented. Conclusions: The identification and treatment of FXS remains an important public health and clinical concern. The information presented in this article provides a more robust understanding of FXS and the impact of this complex condition for pediatricians. Despite a wealth of information about the condition, much work remains to fully support affected individuals and their families.
Article
Fragile-X syndrome (FXS) is a frequent genetic form of intellectual disability (ID). The main recurrent mutagenic mechanism causing FXS is the expansion of a CGG repeat sequence in the 5′-UTR of the FMR1 gene, therefore, routinely tested in ID patients. We report here three FMR1 intragenic pathogenic variants not affecting this sequence, identified using high-throughput sequencing (HTS): a previously reported hemizygous deletion encompassing the last exon of FMR1, too small to be detected by array-CGH and inducing decreased expression of a truncated form of FMRP protein, in three brothers with ID (family 1) and two splice variants in boys with sporadic ID: a de novo variant c.990+1G>A (family 2) and a maternally inherited c.420-8A>G variant (family 3). After clinical reevaluation, the five patients presented features consistent with FXS (mean Hagerman’s scores=15). We conducted a systematic review of all rare non-synonymous variants previously reported in FMR1 in ID patients and showed that six of them are convincing pathogenic variants. This study suggests that intragenic FMR1 variants, although much less frequent than CGG expansions, are a significant mutational mechanism leading to FXS and demonstrates the interest of HTS approaches to detect them in ID patients with a negative standard work-up.