Fragile X syndrome

Last updated
Fragile X syndrome
Other namesMartin–Bell syndrome, [1]
Escalante syndrome
Fragx-2.jpg
Boy with protruding ears characteristic of fragile X syndrome
Specialty Medical genetics, pediatrics, psychiatry
Symptoms Intellectual disability, long and narrow face, large ears, flexible fingers, large testicles [1]
Complications Seizures [1]
Usual onsetNoticeable by age 2 [1]
DurationLifelong [2]
Causes Genetic (X-linked dominant) [1]
Diagnostic method Genetic testing [2]
Treatment Supportive care, early interventions [2]
Frequency1 in 4,000 (males), 1 in 8,000 (females) [1]

Fragile X syndrome (FXS) is a genetic disorder characterized by mild-to-moderate intellectual disability. [1] The average IQ in males with FXS is under 55, while about two thirds of affected females are intellectually disabled. [3] [4] Physical features may include a long and narrow face, large ears, flexible fingers, and large testicles. [1] About a third of those affected have features of autism such as problems with social interactions and delayed speech. [1] Hyperactivity is common, and seizures occur in about 10%. [1] Males are usually more affected than females. [1]

Contents

This disorder and finding of fragile X syndrome has an X-linked dominant inheritance. [1] It is typically caused by an expansion of the CGG triplet repeat within the FMR1 (fragile X messenger ribonucleoprotein 1) gene on the X chromosome. [1] This results in silencing (methylation) of this part of the gene and a deficiency of the resultant protein (FMRP), which is required for the normal development of connections between neurons. [1] Diagnosis requires genetic testing to determine the number of CGG repeats in the FMR1 gene. [5] Normally, there are between 5 and 40 repeats; fragile X syndrome occurs with more than 200. [1] A premutation is said to be present when the gene has between 55 and 200 repeats; females with a premutation have an increased risk of having an affected child. [1] Testing for premutation carriers may allow for genetic counseling. [5]

There is no cure. [2] Early intervention is recommended, as it provides the most opportunity for developing a full range of skills. [6] These interventions may include special education, speech therapy, physical therapy, or behavioral therapy. [2] [7] Medications may be used to treat associated seizures, mood problems, aggressive behavior, or ADHD. [8] Fragile X syndrome tends to show more symptoms on affected males since females have another X chromosome which can compensate for the damaged one. [4] [9]

Signs and symptoms

Prominent characteristics of the syndrome include an elongated face and large or protruding ears. Fragx-1.jpg
Prominent characteristics of the syndrome include an elongated face and large or protruding ears.

Most young children do not show any physical signs of FXS. [10] It is not until puberty that physical features of FXS begin to develop. [10] Aside from intellectual disability, prominent characteristics of the syndrome may include an elongated face, large or protruding ears, flat feet, larger testes (macroorchidism), and low muscle tone. [11] [12] Recurrent otitis media (middle ear infection) and sinusitis is common during early childhood. Speech may be cluttered or nervous. Behavioral characteristics may include stereotypic movements (e.g., hand-flapping) and atypical social development, particularly shyness, limited eye contact, memory problems, and difficulty with face encoding. Some individuals with fragile X syndrome also meet the diagnostic criteria for autism. [13]

Males with a full mutation display virtually complete penetrance and will therefore almost always display symptoms of FXS, while females with a full mutation generally display a penetrance of about 50% as a result of having a second, normal X chromosome. [14] Females with FXS may have symptoms ranging from mild to severe, although they are generally less affected than males due to variability in X-inactivation. [15] [16]

Physical phenotype

Intellectual development

Individuals with FXS may present anywhere on a continuum from learning disabilities in the context of a normal intelligence quotient (IQ) to severe intellectual disability, with an average IQ of 40 in males who have complete silencing of the FMR1 gene. [12] Females, who tend to be less affected, generally have an IQ which is normal or borderline with learning difficulties. The main difficulties in individuals with FXS are with working and short-term memory, executive function, visual memory, visual-spatial relationships, and mathematics, with verbal abilities being relatively unaffected. [12] [20]

Data on intellectual development in FXS are limited. However, there is some evidence that standardized IQ decreases over time in the majority of cases, apparently as a result of slowed intellectual development. A longitudinal study looking at pairs of siblings where one child was affected and the other was not found that affected children had an intellectual learning rate which was 55% slower than unaffected children. [20]

Individuals with FXS often demonstrated language and communicative problems. [21] This may be related to muscle function of the mouth and frontal-lobe deficits. [21]

Autism

Fragile X syndrome co-occurs with autism in many cases and is a suspected genetic cause of the autism in these cases. [11] [22] This finding has resulted in screening for FMR1 mutation to be considered mandatory in children diagnosed with autism. [11] Of those with fragile X syndrome, prevalence of concurrent autism spectrum disorder (ASD) has been estimated to be between 15 and 60%, with the variation due to differences in diagnostic methods and the high frequency of autistic features in individuals with fragile X syndrome not meeting the DSM criteria for an ASD. [22]

Although individuals with FXS have difficulties in forming friendships, those with FXS and ASD characteristically also have difficulties with reciprocal conversation with their peers. Social withdrawal behaviors, including avoidance and indifference, appear to be the best predictors of ASD in FXS, with avoidance appearing to be correlated more with social anxiety while indifference was more strongly correlated to ASD. [22] When both autism and FXS are present, a greater language deficit and lower IQ is observed as compared to children with only FXS. [23]

Genetic mouse models of FXS have also been shown to have autistic-like behaviors. [24] [25] [26] [27] [28]

Social interaction

FXS is characterized by social anxiety, including poor eye contact, gaze aversion, prolonged time to commence social interaction, and challenges forming peer relationships. [29] Social anxiety is one of the most common features associated with FXS, with up to 75% of males in one series characterized as having excessive shyness and 50% having panic attacks. [22] Social anxiety in individuals with FXS is related to challenges with face encoding, the ability to recognize a face that one has seen before. [30]

It appears that individuals with FXS are interested in social interaction and display greater empathy than groups with other causes of intellectual disability, but display anxiety and withdrawal when placed in unfamiliar situations with unfamiliar people. [22] [29] This may range from mild social withdrawal, which is predominantly associated with shyness, to severe social withdrawal, which may be associated with co-existing autism spectrum disorder. [22]

Females with FXS frequently display shyness, social anxiety and social avoidance or withdrawal. [12] In addition, premutation in females has been found to be associated with social anxiety.

Female individuals with FXS show decreased activation in the prefrontal regions of the brain. [31] [ citation needed ]

Mental health

Attention deficit hyperactivity disorder (ADHD) is found in the majority of males with FXS and 30% of females, making it the most common psychiatric diagnosis in those with FXS. [11] [29] Children with fragile X have very short attention spans, are hyperactive, and show hypersensitivity to visual, auditory, tactile, and olfactory stimuli. These children have difficulty in large crowds due to the loud noises and this can lead to tantrums due to hyperarousal. Hyperactivity and disruptive behavior peak in the preschool years and then gradually decline with age, although inattentive symptoms are generally lifelong. [29]

Aside from the characteristic social phobia features, a range of other anxiety symptoms are very commonly associated with FXS, with symptoms typically spanning a number of psychiatric diagnoses but not fulfilling any of the criteria in full. [29] Children with FXS pull away from light touch and can find textures of materials to be irritating. Transitions from one location to another can be difficult for children with FXS. Behavioral therapy can be used to decrease the child's sensitivity in some cases. [19] Behaviors such as hand flapping and biting, as well as aggression, can be an expression of anxiety.[ citation needed ]

Perseveration is a common communicative and behavioral characteristic in FXS. Children with FXS may repeat a certain ordinary activity over and over. In speech, the trend is not only in repeating the same phrase but also talking about the same subject continually. Cluttered speech and self-talk are commonly seen. Self-talk includes talking with oneself using different tones and pitches. [19] Although only a minority of FXS cases will meet the criteria for obsessive–compulsive disorder (OCD), a significant majority will have symptoms of obsession. However, as individuals with FXS generally find these behaviors pleasurable, unlike individuals with OCD, they are more frequently referred to as stereotypic behaviors.[ citation needed ]

Mood symptoms in individuals with FXS rarely meet diagnostic criteria for a major mood disorder as they are typically not of sustained duration. [29] Instead, these are usually transient and related to stressors, and may involve labile (fluctuating) mood, irritability, self-injury and aggression.[ citation needed ]

Individuals with fragile X-associated tremor/ataxia syndrome (FXTAS) are likely to experience combinations of dementia, mood, and anxiety disorders. Males with the FMR1 premutation and clinical evidence of FXTAS were found to have increased occurrence of somatization, obsessive–compulsive disorder, interpersonal sensitivity, depression, phobic anxiety, and psychoticism. [32]

Vision

Ophthalmologic problems include strabismus. This requires early identification to avoid amblyopia. Surgery or patching are usually necessary to treat strabismus if diagnosed early. Refractive errors in patients with FXS are also common. [23]

Neurology

Individuals with FXS are at a higher risk of developing seizures, with rates between 10% and 40% reported in the literature. [33] In larger study populations the frequency varies between 13% and 18%, [12] [33] consistent with a recent survey of caregivers which found that 14% of males and 6% of females experienced seizures. [33] The seizures tend to be partial, are generally not frequent, and are amenable to treatment with medication.[ citation needed ]

Individuals who are carriers of premutation alleles are at risk for developing fragile X-associated tremor/ataxia syndrome (FXTAS), a progressive neurodegenerative disease. [14] [34] It is seen in approximately half of male carriers over the age of 70, while penetrance in females is lower. Typically, onset of tremor occurs in the sixth decade of life, with subsequent progression to ataxia (loss of coordination) and gradual cognitive decline. [34]

Working memory

From their 40s onward, males with FXS begin developing progressively more severe problems in performing tasks that require the central executive of working memory. Working memory involves the temporary storage of information 'in mind', while processing the same or other information. Phonological memory (or verbal working memory) deteriorates with age in males, while visual-spatial memory is not found to be directly related to age. Males often experience an impairment in the functioning of the phonological loop. The CGG length is significantly correlated with central executive and the visual–spatial memory. However, in a premutation individual, CGG length is only significantly correlated with the central executive, not with either phonological memory or visual–spatial memory. [35]

Fertility

About 20% of women who are carriers for the fragile X premutation are affected by fragile X-related primary ovarian insufficiency (FXPOI), which is defined as premature menopause, which is menopause occurring before 40 years of age (average age at menopause is 51 years old in the US). [14] [34] The number of CGG repeats correlates with penetrance and age of onset, but it is not a linear relationship. [36] [14] However premature menopause is more common in premutation carriers than in women with the full mutation, and the highest risk for FXPOI is observed in women with between 70-100 repeats the risk of FXPOI. [37] [38] Fragile X-associated primary ovarian insufficiency (FXPOI) is one of three Fragile X-associated Disorders (FXD) caused by changes in the FMR1 gene. FXPOI affects female premutation carriers, of which is caused by the FMR1 gene, when their ovaries are not functioning properly. Women with FXPOI may exhibit changes in menstrual cycles and have changes in hormone levels but not be considered menopausal. Women with FXPOI still have the chance to get pregnant in about 10% of cases, because their ovaries occasionally release viable eggs through "escape" ovulation. [39] [40]

FMRP is a chromatin-binding protein that functions in the DNA damage response. [41] [42] FMRP also occupies sites on meiotic chromosomes and regulates the dynamics of the DNA damage response machinery during spermatogenesis. [41]

Causes

Location of the FMR1 gene on the X chromosome Fmr1.jpeg
Location of the FMR1 gene on the X chromosome

Fragile X syndrome is a genetic disorder which occurs as a result of a mutation of the Fragile X Messenger Ribonucleoprotein 1 (FMR1) gene on the X chromosome, most commonly an increase in the number of CGG trinucleotide repeats in the 5' untranslated region of FMR1. [14] [34] Mutation at that site is found in 1 out of about every 2000 males and 1 out of about every 259 females. Incidence of the disorder itself is about 1 in every 3600 males and 1 in 4000–6000 females. [43] Although this accounts for over 98% of cases, FXS can also occur as a result of point mutations affecting FMR1. [14] [34]

In unaffected individuals, the FMR1 gene contains 5–44 repeats of the sequence CGG, most commonly 29 or 30 repeats. [14] [34] [44] Between 45 and 54 repeats is considered a "grey zone", with a premutation allele generally considered to be between 55 and 200 repeats in length. Individuals with fragile X syndrome have a full mutation of the FMR1 allele, with over 200 CGG repeats. [11] [44] [45] In these individuals with a repeat expansion greater than 200, there is methylation of the CGG repeat expansion and FMR1 promoter, leading to the silencing of the FMR1 gene and a lack of its product.

This methylation of FMR1 in chromosome band Xq27.3 is believed to result in constriction of the X chromosome which appears 'fragile' under the microscope at that point, a phenomenon that gave the syndrome its name. One study found that FMR1 silencing is mediated by the FMR1 mRNA. The FMR1 mRNA contains the transcribed CGG-repeat tract as part of the 5' untranslated region, which hybridizes to the complementary CGG-repeat portion of the FMR1 gene to form an RNA·DNA duplex. [46]

A subset of people with intellectual disability and symptoms resembling fragile X syndrome are found to have point mutations in FMR1. This subset lacked the CGG repeat expansion in FMR1 traditionally associated with fragile x syndrome. [47] The first complete DNA sequence of the repeat expansion in someone with the full mutation was generated by scientists in 2012 using SMRT sequencing. [48]

Inheritance

Fragile X syndrome has traditionally been considered an X-linked dominant condition with variable expressivity and possibly reduced penetrance. [12] The likelihood of transmission depends on the parent's gender, the X chromosome carrying the mutation, and the number of CGG repeats in the premutation.

Due to genetic anticipation and X-inactivation in females, the inheritance of Fragile X syndrome does not follow the usual pattern of X-linked dominant inheritance, and scholars from The University of Chicago Medical Center and Groningen University Hospital have had an abstract published in the American Journal of Medical Genetics that proposes discontinuing labeling X-linked disorders as dominant or recessive. [49] Males with a full mutation are usually affected and infertile, while carrier females have a 50% chance of passing the mutation.

Before the FMR1 gene was discovered, analysis of pedigrees showed the presence of male carriers who were asymptomatic, with their grandchildren affected by the condition at a higher rate than their siblings suggesting that genetic anticipation was occurring. [14] This tendency for future generations to be affected at a higher frequency became known as the Sherman paradox after its description in 1985. [14] [50] Due to this, male children often have a greater degree of symptoms than their mothers. [51]

The explanation for this phenomenon is that male carriers pass on their premutation to all of their daughters, with the length of the FMR1 CGG repeat typically not increasing during meiosis, the cell division that is required to produce sperm. [14] [34] Incidentally, males with a full mutation only pass on premutations to their daughters. [34] However, females with a full mutation are able to pass this full mutation on, so theoretically there is a 50% chance that a child will be affected. [34] [44] In addition, the length of the CGG repeat frequently does increase during meiosis in female premutation carriers due to instability and so, depending on the length of their premutation, they may pass on a full mutation to their children who will then be affected. Repeat expansion is considered to be a consequence of strand slippage either during DNA replication or DNA repair synthesis. [52]

Mosaicism

Mosaicism refers to cases where individuals have both full mutation and premutation copies. Mosaicism can result from instability in the CGG repeats, and affected individuals may show classic symptoms, although some evidence suggests higher intellectual abilities compared to those with a full mutation. [53]

Pathophysiology

FMRP is found throughout the body, but in highest concentrations within the brain and testes. [11] [14] It appears to be primarily responsible for selectively binding to around 4% of mRNA in mammalian brains and transporting it out of the cell nucleus and to the synapses of neurons. Most of these mRNA targets have been found to be located in the dendrites of neurons, and brain tissue from humans with FXS and mouse models shows abnormal dendritic spines, which are required to increase contact with other neurons. The subsequent abnormalities in the formation and function of synapses and development of neural circuits result in impaired neuroplasticity, an integral part of memory and learning. [11] [14] [54] Connectome changes have long been suspected to be involved in the sensory pathophysiology [55] and most recently a range of circuit alterations have been shown, involving structurally increased local connectivity and functionally decreased long-range connectivity. [56]

In addition, FMRP has been implicated in several signalling pathways that are being targeted by a number of drugs undergoing clinical trials. The group 1 metabotropic glutamate receptor (mGluR) pathway, which includes mGluR1 and mGluR5, is involved in mGluR-dependent long term depression (LTD) and long term potentiation (LTP), both of which are important mechanisms in learning. [11] [14] The lack of FMRP, which represses mRNA production and thereby protein synthesis, leads to exaggerated LTD. FMRP also appears to affect dopamine pathways in the prefrontal cortex which is believed to result in the attention deficit, hyperactivity and impulse control problems associated with FXS. [11] [14] [29] The downregulation of GABA pathways, which serve an inhibitory function and are involved in learning and memory, may be a factor in the anxiety symptoms which are commonly seen in FXS.[ citation needed ]

Research in a mouse model of FSX shows that cortical neurons receive reduced sensory information (hyposensitivity), contrary to the common assumption that these neurons are hypersensitive, accompanied by enhanced contextual information, accumulated from previous experiences. Therefore, these results suggest that the hypersensitive phenotype of affected individuals might arise from mismatched contextual input onto these neurons. [57]

Diagnosis

Clinical diagnosis relies on identifying a variant of FMR1 associated with decreased function alongside moderate to severe intellectual impairment, particularly in males or moderate in females. Diagnostic tests include PCR to analyze the number of CGG repeats, Southern blot analysis, and examination of AGG trinucleotides in the FMR1 gene region.

Cytogenetic analysis for fragile X syndrome was first available in the late 1970s when diagnosis of the syndrome and carrier status could be determined by culturing cells in a folate deficient medium and then assessing for "fragile sites" (discontinuity of staining in the region of the trinucleotide repeat) on the long arm of the X chromosome. [58] This technique proved unreliable, however, as the fragile site was often seen in less than 40% of an individual's cells. This was not as much of a problem in males, but in female carriers, where the fragile site could generally only be seen in 10% of cells, the mutation often could not be visualised.[ citation needed ]

Since the 1990s, more sensitive molecular techniques have been used to determine carrier status. [58] The fragile X abnormality is now directly determined by analysis of the number of CGG repeats using polymerase chain reaction (PCR) and methylation status using Southern blot analysis. [12] By determining the number of CGG repeats on the X chromosome, this method allows for more accurate assessment of risk for premutation carriers in terms of their own risk of fragile X associated syndromes, as well as their risk of having affected children. Because this method only tests for expansion of the CGG repeat, individuals with FXS due to missense mutations or deletions involving FMR1 will not be diagnosed using this test and should therefore undergo sequencing of the FMR1 gene if there is clinical suspicion of FXS.[ citation needed ]

Prenatal testing with chorionic villus sampling or amniocentesis allows diagnosis of FMR1 mutation while the fetus is in utero and appears to be reliable. [12]

Early diagnosis of fragile X syndrome or carrier status is important for providing early intervention in children or fetuses with the syndrome, and allowing genetic counselling with regards to the potential for a couple's future children to be affected. Most parents notice delays in speech and language skills, difficulties in social and emotional domains as well as sensitivity levels in certain situations with their children. [59]

Management

There is no cure for the underlying defects of FXS. [2] Management of FXS may include speech therapy, behavioral therapy, occupational therapy, special education, or individualised educational plans, and, when necessary, treatment of physical abnormalities. Persons with fragile X syndrome in their family histories are advised to seek genetic counseling to assess the likelihood of having children who are affected, and how severe any impairments may be in affected descendants. [60]

Medication

Current trends in treating the disorder include medications for symptom-based treatments that aim to minimize the secondary characteristics associated with the disorder. If an individual is diagnosed with FXS, genetic counseling for testing family members at risk for carrying the full mutation or premutation is a critical first-step. Due to a higher prevalence of FXS in boys, the most commonly used medications are stimulants that target hyperactivity, impulsivity, and attentional problems. [12] For co-morbid disorders with FXS, antidepressants such as selective serotonin reuptake inhibitors (SSRIs) are utilized to treat the underlying anxiety, obsessive-compulsive behaviors, and mood disorders. Following antidepressants, antipsychotics such as risperidone and quetiapine are used to treat high rates of self-injurious, aggressive and aberrant behaviors in this population (Bailey Jr et al., 2012). Anticonvulsants are another set of pharmacological treatments used to control seizures as well as mood swings in 13%–18% of individuals with FXS. Drugs targeting the mGluR5 (metabotropic glutamate receptors) that are linked with synaptic plasticity are especially beneficial for targeted symptoms of FXS. [12] Lithium is also currently being used in clinical trials with humans, showing significant improvements in behavioral functioning, adaptive behavior, and verbal memory. Few studies suggested using folic acid, but more researches are needed due to the low quality of that evidence. [61] Alongside pharmacological treatments, environmental influences such as home environment and parental abilities as well as behavioral interventions such as speech therapy, sensory integration, etc. all factor in together to promote adaptive functioning for individuals with FXS. [60] While metformin may reduce body weight in persons with fragile X syndrome, it is uncertain whether it improves neurological or psychiatric symptoms. [62]

Current pharmacological treatment centers on managing problem behaviors and psychiatric symptoms associated with FXS. However, as there has been very little research done in this specific population, the evidence to support the use of these medications in individuals with FXS is poor. [63]

ADHD, which affects the majority of boys and 30% of girls with FXS, is frequently treated using stimulants. [11] However, the use of stimulants in the fragile X population is associated with a greater frequency of adverse events including increased anxiety, irritability and mood lability. [29] Anxiety, as well as mood and obsessive-compulsive symptoms, may be treated using SSRIs, although these can also aggravate hyperactivity and cause disinhibited behavior. [12] [29] Atypical antipsychotics can be used to stabilise mood and control aggression, especially in those with comorbid ASD. However, monitoring is required for metabolic side effects including weight gain and diabetes, as well as movement disorders related to extrapyramidal side effects such as tardive dyskinesia. Individuals with coexisting seizure disorder may require treatment with anticonvulsants.

Prognosis

A 2013 review stated that life expectancy for FXS was 12 years lower than the general population and that the causes of death were similar to those found for the general population. [64]

Pharmacological therapy

Fragile X syndrome is the most "translated" human neurodevelopmental disorder under study. Hence, research into the etiology of FXS has given rise to many attempts at drug discovery. [65] The increased understanding of the molecular mechanisms of disease in FXS has led to the development of therapies targeting the affected pathways. Evidence from mouse models shows that mGluR5 antagonists (blockers) can rescue dendritic spine abnormalities and seizures, as well as cognitive and behavioral problems, and may show promise in the treatment of FXS. [11] [66] [67] Two new drugs, AFQ-056 (mavoglurant) and dipraglurant, as well as the repurposed drug fenobam are currently undergoing human trials for the treatment of FXS. [11] [68] There is also early evidence for the efficacy of arbaclofen, a GABAB agonist, in improving social withdrawal in individuals with FXS and ASD. [11] [22] In addition, there is evidence from mouse models that minocycline, an antibiotic used for the treatment of acne, rescues abnormalities of the dendrites. An open trial in humans has shown promising results, although there is currently no evidence from controlled trials to support its use. [11]

History

In 1943, British neurologist James Purdon Martin and British geneticist Julia Bell described a pedigree of X-linked intellectual disability, without considering the macroorchidism (larger testicles). [69] In 1969, Herbert Lubs first sighted an unusual "marker X chromosome" in association with intellectual disability. [70] In 1970, Frederick Hecht coined the term "fragile site". And, in 1985, Felix F. de la Cruz outlined extensively the physical, psychological, and cytogenetic characteristics of those with the condition in addition to prospects for therapy. [71] Continued advocacy later won him an honour through the FRAXA Research Foundation in December 1998. [72]

See also

Related Research Articles

<span class="mw-page-title-main">Lesch–Nyhan syndrome</span> Rare genetic disorder

Lesch–Nyhan syndrome (LNS) is a rare inherited disorder caused by a deficiency of the enzyme hypoxanthine-guanine phosphoribosyltransferase (HGPRT). This deficiency occurs due to mutations in the HPRT1 gene located on the X chromosome. LNS affects about 1 in 380,000 live births. The disorder was first recognized and clinically characterized by American medical student Michael Lesch and his mentor, pediatrician William Nyhan, at Johns Hopkins.

In genetics, anticipation is a phenomenon whereby as a genetic disorder is passed on to the next generation, the symptoms of the genetic disorder become apparent at an earlier age with each generation. In most cases, an increase in the severity of symptoms is also noted. Anticipation is common in trinucleotide repeat disorders, such as Huntington's disease and myotonic dystrophy, where a dynamic mutation in DNA occurs. All of these diseases have neurological symptoms. Prior to the understanding of the genetic mechanism for anticipation, it was debated whether anticipation was a true biological phenomenon or whether the earlier age of diagnosis was related to heightened awareness of disease symptoms within a family.

Norrie disease is a rare disease and genetic disorder that primarily affects the eyes and almost always leads to blindness. It is caused by mutations in the Norrin cystine knot growth factor (NDP) gene, which is located on the X chromosome. In addition to the congenital ocular symptoms, the majority of patients experience a progressive hearing loss starting mostly in their 2nd decade of life, and some may have learning difficulties among other additional characteristics.

Macroorchidism is a disorder found in males, specifically in children, where a subject has abnormally large testes. The condition is commonly inherited in connection with fragile X syndrome (FXS), which is also the second most common genetic cause of intellectual disability. The condition is also a rare sign of the McCune-Albright syndrome. The opposite of macroorchidism is called microorchidism, which is the condition of abnormally small testes.

Trinucleotide repeat disorders, a subset of microsatellite expansion diseases, are a set of over 30 genetic disorders caused by trinucleotide repeat expansion, a kind of mutation in which repeats of three nucleotides increase in copy numbers until they cross a threshold above which they cause developmental, neurological or neuromuscular disorders. Depending on its location, the unstable trinucleotide repeat may cause defects in a protein encoded by a gene; change the regulation of gene expression; produce a toxic RNA, or lead to production of a toxic protein. In general, the larger the expansion the faster the onset of disease, and the more severe the disease becomes.

The Sherman paradox was a term used to describe the anomalous pattern of inheritance found in fragile X syndrome. The phenomenon is also referred to as anticipation or dynamic mutation.

<span class="mw-page-title-main">Heritability of autism</span>

The heritability of autism is the proportion of differences in expression of autism that can be explained by genetic variation; if the heritability of a condition is high, then the condition is considered to be primarily genetic. Autism has a strong genetic basis. Although the genetics of autism are complex, autism spectrum disorder (ASD) is explained more by multigene effects than by rare mutations with large effects.

<span class="mw-page-title-main">FMR1</span> Human protein and coding gene

FMR1 is a human gene that codes for a protein called fragile X messenger ribonucleoprotein, or FMRP. This protein, most commonly found in the brain, is essential for normal cognitive development and female reproductive function. Mutations of this gene can lead to fragile X syndrome, intellectual disability, premature ovarian failure, autism, Parkinson's disease, developmental delays and other cognitive deficits. The FMR1 premutation is associated with a wide spectrum of clinical phenotypes that affect more than two million people worldwide.

<span class="mw-page-title-main">XXYY syndrome</span> Extra X and Y chromosome in males

XXYY syndrome is a sex chromosome anomaly in which males have 2 extra chromosomes, one X and one Y chromosome. Human cells usually contain two sex chromosomes, one from the mother and one from the father. Usually, females have two X chromosomes (XX) and males have one X and one Y chromosome (XY). The appearance of at least one Y chromosome with a properly functioning SRY gene makes a male. Therefore, humans with XXYY are genotypically male. Males with XXYY syndrome have 48 chromosomes instead of the typical 46. This is why XXYY syndrome is sometimes written as 48, XXYY syndrome or 48, XXYY. It affects an estimated one in every 18,000–40,000 male births.

A trinucleotide repeat expansion, also known as a triplet repeat expansion, is the DNA mutation responsible for causing any type of disorder categorized as a trinucleotide repeat disorder. These are labelled in dynamical genetics as dynamic mutations. Triplet expansion is caused by slippage during DNA replication, also known as "copy choice" DNA replication. Due to the repetitive nature of the DNA sequence in these regions, 'loop out' structures may form during DNA replication while maintaining complementary base pairing between the parent strand and daughter strand being synthesized. If the loop out structure is formed from the sequence on the daughter strand this will result in an increase in the number of repeats. However, if the loop out structure is formed on the parent strand, a decrease in the number of repeats occurs. It appears that expansion of these repeats is more common than reduction. Generally, the larger the expansion the more likely they are to cause disease or increase the severity of disease. Other proposed mechanisms for expansion and reduction involve the interaction of RNA and DNA molecules.

<span class="mw-page-title-main">Fragile X-associated tremor/ataxia syndrome</span>

Fragile X-associated tremor/ataxia syndrome (FXTAS) is a late-onset neurodegenerative disorder most frequently seen in male premutation carriers of Fragile X syndrome (FXS) over the age of 50. The main clinical features of FXTAS include problems of movement with cerebellar gait ataxia and action tremor. Associated features include parkinsonism, cognitive decline, and dysfunction of the autonomic nervous system. FXTAS is found in Fragile X "premutation" carriers, which is defined as a trinucleotide repeat expansion of 55-200 CGG repeats in the Fragile X mental retardation-1 (FMR1) gene. 4-40 CGG repeats in this gene is considered normal, while individual with >200 repeats have full Fragile X Syndrome.

<span class="mw-page-title-main">FMR1-AS1 gene</span> Non-coding RNA in the species Homo sapiens

In molecular biology, FMR1 antisense RNA 1 (FMR1-AS1), also known as ASFMR1 or FMR4, is a long non-coding RNA. The FMR1-AS1 gene overlaps, and is antisense to, the CGG repeat region of the FMR1 gene. Its expression is upregulated in fragile X syndrome premutation carriers, and silenced in patients with fragile X syndrome. FMR1-AS1 has an anti-apoptotic function.

Autism spectrum disorder (ASD) refers to a variety of conditions typically identified by challenges with social skills, communication, speech, and repetitive sensory-motor behaviors. The 11th International Classification of Diseases (ICD-11), released in January 2021, characterizes ASD by the associated deficits in the ability to initiate and sustain two-way social communication and restricted or repetitive behavior unusual for the individual's age or situation. Although linked with early childhood, the symptoms can appear later as well. Symptoms can be detected before the age of two and experienced practitioners can give a reliable diagnosis by that age. However, official diagnosis may not occur until much older, even well into adulthood. There is a large degree of variation in how much support a person with ASD needs in day-to-day life. This can be classified by a further diagnosis of ASD level 1, level 2, or level 3. Of these, ASD level 3 describes people requiring very substantial support and who experience more severe symptoms. ASD-related deficits in nonverbal and verbal social skills can result in impediments in personal, family, social, educational, and occupational situations. This disorder tends to have a strong correlation with genetics along with other factors. More research is identifying ways in which epigenetics is linked to autism. Epigenetics generally refers to the ways in which chromatin structure is altered to affect gene expression. Mechanisms such as cytosine regulation and post-translational modifications of histones. Of the 215 genes contributing, to some extent in ASD, 42 have been found to be involved in epigenetic modification of gene expression. Some examples of ASD signs are specific or repeated behaviors, enhanced sensitivity to materials, being upset by changes in routine, appearing to show reduced interest in others, avoiding eye contact and limitations in social situations, as well as verbal communication. When social interaction becomes more important, some whose condition might have been overlooked suffer social and other exclusion and are more likely to have coexisting mental and physical conditions. Long-term problems include difficulties in daily living such as managing schedules, hypersensitivities, initiating and sustaining relationships, and maintaining jobs.

Fragile X-associated Primary Ovarian Insufficiency (FXPOI) is the most common genetic cause of premature ovarian failure in women with a normal karyotype 46, XX. The expansion of a CGG repeat in the 5' untranslated region of the FMR1 gene from the normal range of 5-45 repeats to the premutation range of 55-199 CGGs leads to risk of FXPOI for ovary-bearing individuals. About 1:150-1:200 women in the US population carry a premutation. Women who carry an FMR1 premutation have a roughly 20% risk of being diagnosed with FXPOI, compared to 1% for the general population, and an 8-15% risk of developing the neurogenerative tremor/ataxia disorder (FXTAS). FMR1 premutation women are also at increased risk of having a child with a CGG repeat that is expanded to >200 repeats. Individuals with a full mutation, unlike the premutation, produce little to no mRNA or protein from the FMR1 gene and are affected with Fragile X syndrome.

<span class="mw-page-title-main">Metadoxine</span> Medication used for alcohol intoxication

Metadoxine, also known as pyridoxine-pyrrolidone carboxylate, is a drug used to treat chronic and acute alcohol intoxication. Metadoxine accelerates alcohol clearance from the blood.

Nagwa Abdel Meguid is an Egyptian geneticist and 2002 winner of the L’Oreal UNESCO Award for Women in Science for Africa and the Middle East. Her research has "identified several genetic mutations that cause common syndromes such as the fragile X syndrome and Autism".

<span class="mw-page-title-main">MECP2 duplication syndrome</span> Medical condition

MECP2 duplication syndrome (M2DS) is a rare disease that is characterized by severe intellectual disability and impaired motor function. It is an X-linked genetic disorder caused by the overexpression of MeCP2 protein.

Randi J. Hagerman is an American physician who is the medical director of MIND Institute at the University of California, Davis. She works for the pediatrics department under the division of child development and behavior. She is an internationally recognized researcher in the field of genetics of autism spectrum disorder with special focus on genomic instability. Along with her husband Paul Hagerman, she discovered the Fragile X-associated tremor/ataxia syndrome (FXTAS), a neurological disorder that affects older male and rare female carriers of fragile X.

ADNP syndrome, also known as Helsmoortel-Van der Aa syndrome (HVDAS), is a non-inherited neurodevelopmental disorder caused by mutations in the activity-dependent neuroprotector homeobox (ADNP) gene.

David L. Nelson is an American human geneticist, currently an associate director at the Intellectual and Developmental Disabilities Research Center (1995), and professor at the Department of Molecular and Human Genetics at Baylor College of Medicine BCM since 1999. Since 2018, he is the director at the Cancer and Cell Biology Ph.D program, and the director of Integrative Molecular and Biomedical Sciences Ph.D since 2015 at BCM.

References

  1. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 "fragile X syndrome". Genetics Home Reference. April 2012. Archived from the original on 9 October 2016. Retrieved 7 October 2016.
  2. 1 2 3 4 5 6 "What is Fragile X Syndrome?". National Center on Birth Defects and Developmental Disabilities. US: Centers for Disease Control and Prevention. 3 June 2022. Archived from the original on 10 May 2017. Retrieved 10 May 2017.
  3. Raspa M, Wheeler AC, Riley C (June 2017). "Public Health Literature Review of Fragile X Syndrome". Pediatrics. 139 (Suppl 3): S153–S171. doi:10.1542/peds.2016-1159C. PMC   5621610 . PMID   28814537.
  4. 1 2 "Data and Statistics Fragile X Syndrome (FXS)". Centers for Disease Control and Prevention. 2018-08-09. Archived from the original on 2019-10-13. Retrieved 2018-11-05.
  5. 1 2 "Technical Standards and Guidelines for Fragile X". www.acmg.net. 2006. Archived from the original on 12 October 2016. Retrieved 10 May 2017.
  6. "What are the treatments for Fragile X syndrome?". www.nichd.nih.gov. Archived from the original on 2016-11-21. Retrieved 2016-11-21.
  7. "Therapy Treatments". NICHD. Archived from the original on 5 May 2017. Retrieved 10 May 2017.
  8. "Medication Treatments". NICHD. Archived from the original on 5 May 2017. Retrieved 10 May 2017.
  9. "What is Fragile-X syndrome?". @yourgenome · Science website. Retrieved 2023-07-15.
  10. 1 2 "What are the symptoms of Fragile X syndrome?". www.nichd.nih.gov. Archived from the original on 2016-11-21. Retrieved 2016-11-21.
  11. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 McLennan Y, Polussa J, Tassone F, Hagerman R (May 2011). "Fragile x syndrome". Current Genomics. 12 (3): 216–224. doi:10.2174/138920211795677886. PMC   3137006 . PMID   22043169.
  12. 1 2 3 4 5 6 7 8 9 10 11 Garber KB, Visootsak J, Warren ST (June 2008). "Fragile X syndrome". European Journal of Human Genetics. 16 (6): 666–672. doi:10.1038/ejhg.2008.61. PMC   4369150 . PMID   18398441.
  13. "Fragile X and Autism | Fragile X Society | UK". fragilex. Retrieved 2022-09-07.
  14. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 Santoro MR, Bray SM, Warren ST (2012). "Molecular mechanisms of fragile X syndrome: a twenty-year perspective". Annual Review of Pathology. 7: 219–245. doi:10.1146/annurev-pathol-011811-132457. PMID   22017584.
  15. Bartholomay KL, Lee CH, Bruno JL, Lightbody AA, Reiss AL (January 2019). "Closing the Gender Gap in Fragile X Syndrome: Review on Females with FXS and Preliminary Research Findings". Brain Sciences. 9 (1): 11. doi: 10.3390/brainsci9010011 . PMC   6356553 . PMID   30642066.
  16. CDC (2022-06-03). "What is Fragile X Syndrome (FXS)? | CDC". Centers for Disease Control and Prevention. Retrieved 2022-11-04.
  17. Jordan JA, Regezi JJ, Sciubba RC (2008). "15". Oral pathology : clinical pathologic correlations (5th ed.). St. Louis, Mo.: Saunders/Elsevier. ISBN   978-1-4160-4570-0. Section on fragile X syndrome[ page needed ]
  18. Nichols C, Nichols D (1 June 2019). "Fragile X Syndrome: Implications for Physical Education". Palaestra. 33 (3).
  19. 1 2 3 Goldstein S, Reynolds CR (1999). Handbook of neurodevelopmental and genetic disorders in children. New York: Guilford Press. ISBN   978-1-57230-448-2.[ page needed ]
  20. 1 2 Hall SS, Burns DD, Lightbody AA, Reiss AL (August 2008). "Longitudinal changes in intellectual development in children with Fragile X syndrome". Journal of Abnormal Child Psychology. 36 (6): 927–939. doi:10.1007/s10802-008-9223-y. PMC   4820329 . PMID   18347972.
  21. 1 2 Abbeduto L, Hagerman RJ (1997). "Language and communication in fragile X syndrome". Mental Retardation and Developmental Disabilities Research Reviews. 3 (4): 313–322. doi:10.1002/(SICI)1098-2779(1997)3:4<313::AID-MRDD6>3.0.CO;2-O. ISSN   1098-2779.
  22. 1 2 3 4 5 6 7 Budimirovic DB, Kaufmann WE (2011). "What can we learn about autism from studying fragile X syndrome?". Developmental Neuroscience. 33 (5): 379–394. doi:10.1159/000330213. PMC   3254037 . PMID   21893949.
  23. 1 2 Hagerman, Randi J., and Paul J. Hagerman. Fragile X syndrome: diagnosis, treatment, and research. 3, illustrated ed. Baltimore, MD: JHU P, 2002.[ page needed ]
  24. Pietropaolo S, Guilleminot A, Martin B, D'Amato FR, Crusio WE (February 2011). "Genetic-background modulation of core and variable autistic-like symptoms in Fmr1 knock-out mice". PLOS ONE. 6 (2): e17073. Bibcode:2011PLoSO...617073P. doi: 10.1371/journal.pone.0017073 . PMC   3043074 . PMID   21364941.
  25. Bernardet M, Crusio WE (September 2006). "Fmr1 KO mice as a possible model of autistic features". TheScientificWorldJournal. 6: 1164–1176. doi: 10.1100/tsw.2006.220 . PMC   5917219 . PMID   16998604.
  26. Mineur YS, Huynh LX, Crusio WE (March 2006). "Social behavior deficits in the Fmr1 mutant mouse". Behavioural Brain Research. 168 (1): 172–175. doi:10.1016/j.bbr.2005.11.004. PMID   16343653. S2CID   45731129.
  27. Spencer CM, Alekseyenko O, Hamilton SM, Thomas AM, Serysheva E, Yuva-Paylor LA, Paylor R (February 2011). "Modifying behavioral phenotypes in Fmr1KO mice: genetic background differences reveal autistic-like responses". Autism Research. 4 (1): 40–56. doi:10.1002/aur.168. PMC   3059810 . PMID   21268289.
  28. Spencer CM, Graham DF, Yuva-Paylor LA, Nelson DL, Paylor R (June 2008). "Social behavior in Fmr1 knockout mice carrying a human FMR1 transgene". Behavioral Neuroscience. 122 (3): 710–715. doi:10.1037/0735-7044.122.3.710. PMID   18513141.
  29. 1 2 3 4 5 6 7 8 9 Tranfaglia MR (2011). "The psychiatric presentation of fragile x: evolution of the diagnosis and treatment of the psychiatric comorbidities of fragile X syndrome". Developmental Neuroscience. 33 (5): 337–348. doi: 10.1159/000329421 . PMID   21893938. S2CID   207554509.
  30. Holsen LM, Dalton KM, Johnstone T, Davidson RJ (November 2008). "Prefrontal social cognition network dysfunction underlying face encoding and social anxiety in fragile X syndrome". NeuroImage. 43 (3): 592–604. doi:10.1016/j.neuroimage.2008.08.009. PMC   2598775 . PMID   18778781.
  31. Lightbody AA, Reiss AL (2009). "Gene, brain, and behavior relationships in fragile X syndrome: evidence from neuroimaging studies". Developmental Disabilities Research Reviews. Wiley. 15 (4): 343–352. doi:10.1002/ddrr.77. OCLC   175296472. PMC   4354896 . PMID   20014368.
  32. Bourgeois JA, Cogswell JB, Hessl D, Zhang L, Ono MY, Tassone F, et al. (2007). "Cognitive, anxiety and mood disorders in the fragile X-associated tremor/ataxia syndrome". General Hospital Psychiatry. 29 (4): 349–356. doi:10.1016/j.genhosppsych.2007.03.003. PMC   3991490 . PMID   17591512.
  33. 1 2 3 Berry-Kravis E, Raspa M, Loggin-Hester L, Bishop E, Holiday D, Bailey DB (November 2010). "Seizures in fragile X syndrome: characteristics and comorbid diagnoses". American Journal on Intellectual and Developmental Disabilities. 115 (6): 461–472. doi:10.1352/1944-7558-115.6.461. PMID   20945999.
  34. 1 2 3 4 5 6 7 8 9 Peprah E (March 2012). "Fragile X syndrome: the FMR1 CGG repeat distribution among world populations". Annals of Human Genetics. 76 (2): 178–191. doi:10.1111/j.1469-1809.2011.00694.x. PMC   3288311 . PMID   22188182.
  35. Cornish KM, Kogan CS, Li L, Turk J, Jacquemont S, Hagerman RJ (April 2009). "Lifespan changes in working memory in fragile X premutation males". Brain and Cognition. 69 (3): 551–558. doi:10.1016/j.bandc.2008.11.006. PMC   4158922 . PMID   19114290.
  36. Sullivan AK, Marcus M, Epstein MP, Allen EG, Anido AE, Paquin JJ, et al. (February 2005). "Association of FMR1 repeat size with ovarian dysfunction". Human Reproduction. 20 (2): 402–412. doi: 10.1093/humrep/deh635 . PMID   15608041.
  37. Allen EG, Charen K, Hipp HS, Shubeck L, Amin A, He W, et al. (September 2021). "Refining the risk for fragile X-associated primary ovarian insufficiency (FXPOI) by FMR1 CGG repeat size". Genetics in Medicine. 23 (9): 1648–1655. doi:10.1038/s41436-021-01177-y. PMC   8460441 . PMID   33927378.
  38. Bibi G, Malcov M, Yuval Y, Reches A, Ben-Yosef D, Almog B, et al. (August 2010). "The effect of CGG repeat number on ovarian response among fragile X premutation carriers undergoing preimplantation genetic diagnosis". Fertility and Sterility. 94 (3): 869–874. doi: 10.1016/j.fertnstert.2009.04.047 . PMID   19481741.
  39. Sherman SL, Curnow EC, Easley CA, Jin P, Hukema RK, Tejada MI, et al. (2014). "Use of model systems to understand the etiology of fragile X-associated primary ovarian insufficiency (FXPOI)". Journal of Neurodevelopmental Disorders. 6 (1): 26. doi: 10.1186/1866-1955-6-26 . PMC   4139715 . PMID   25147583.
  40. Hipp HS, Charen KH, Spencer JB, Allen EG, Sherman SL (September 2016). "Reproductive and gynecologic care of women with fragile X primary ovarian insufficiency (FXPOI)". Menopause. 23 (9): 993–999. doi:10.1097/GME.0000000000000658. PMC   4998843 . PMID   27552334.
  41. 1 2 Alpatov R, Lesch BJ, Nakamoto-Kinoshita M, Blanco A, Chen S, Stützer A, et al. (May 2014). "A chromatin-dependent role of the fragile X mental retardation protein FMRP in the DNA damage response". Cell. 157 (4): 869–881. doi:10.1016/j.cell.2014.03.040. PMC   4038154 . PMID   24813610.
  42. Dockendorff TC, Labrador M (January 2019). "The Fragile X Protein and Genome Function". Molecular Neurobiology. 56 (1): 711–721. doi:10.1007/s12035-018-1122-9. PMID   29796988. S2CID   44159474.
  43. "Monogenic diseases". Human Genomics in Global Health. World Health Organization. Archived from the original on 2012-10-20. Retrieved 2020-10-04.
  44. 1 2 3 Maddalena A, Richards CS, McGinniss MJ, Brothman A, Desnick RJ, Grier RE, et al. (2001). "Technical standards and guidelines for fragile X: the first of a series of disease-specific supplements to the Standards and Guidelines for Clinical Genetics Laboratories of the American College of Medical Genetics. Quality Assurance Subcommittee of the Laboratory Practice Committee". Genetics in Medicine. 3 (3): 200–205. doi:10.1097/00125817-200105000-00010. PMC   3110344 . PMID   11388762.
  45. Nolin SL, Brown WT, Glicksman A, Houck GE, Gargano AD, Sullivan A, et al. (February 2003). "Expansion of the fragile X CGG repeat in females with premutation or intermediate alleles". American Journal of Human Genetics. 72 (2): 454–464. doi:10.1086/367713. PMC   379237 . PMID   12529854.
  46. Colak D, Zaninovic N, Cohen MS, Rosenwaks Z, Yang WY, Gerhardt J, et al. (February 2014). "Promoter-bound trinucleotide repeat mRNA drives epigenetic silencing in fragile X syndrome". Science. 343 (6174): 1002–1005. Bibcode:2014Sci...343.1002C. doi:10.1126/science.1245831. PMC   4357282 . PMID   24578575.
  47. Sitzmann AF, Hagelstrom RT, Tassone F, Hagerman RJ, Butler MG (January 2018). "Rare FMR1 gene mutations causing fragile X syndrome: A review". American Journal of Medical Genetics. Part A. 176 (1): 11–18. doi:10.1002/ajmg.a.38504. PMC   6697153 . PMID   29178241.
  48. Loomis EW, Eid JS, Peluso P, Yin J, Hickey L, Rank D, et al. (January 2013). "Sequencing the unsequenceable: expanded CGG-repeat alleles of the fragile X gene". Genome Research. 23 (1): 121–128. doi:10.1101/gr.141705.112. PMC   3530672 . PMID   23064752.
  49. Dobyns WB, Filauro A, Tomson BN, Chan AS, Ho AW, Ting NT, et al. (August 2004). "Inheritance of most X-linked traits is not dominant or recessive, just X-linked". American Journal of Medical Genetics. Part A. 129A (2): 136–143. doi:10.1002/ajmg.a.30123. PMID   15316978. S2CID   42108591.
  50. Sherman SL, Jacobs PA, Morton NE, Froster-Iskenius U, Howard-Peebles PN, Nielsen KB, et al. (1985). "Further segregation analysis of the fragile X syndrome with special reference to transmitting males". Human Genetics. 69 (4): 289–299. doi:10.1007/BF00291644. PMID   3838733. S2CID   23299935.
  51. Marco EJ, Skuse DH (December 2006). "Autism-lessons from the X chromosome". Social Cognitive and Affective Neuroscience. 1 (3): 183–193. doi:10.1093/scan/nsl028. PMC   2555419 . PMID   18985105.
  52. Usdin K, House NC, Freudenreich CH (2015). "Repeat instability during DNA repair: Insights from model systems". Critical Reviews in Biochemistry and Molecular Biology. 50 (2): 142–167. doi:10.3109/10409238.2014.999192. PMC   4454471 . PMID   25608779.
  53. Mineur YS, Sluyter F, de Wit S, Oostra BA, Crusio WE (2002). "Behavioral and neuroanatomical characterization of the Fmr1 knockout mouse". Hippocampus. 12 (1): 39–46. doi:10.1002/hipo.10005. ISSN   1050-9631. PMID   11918286. S2CID   35472224.
  54. Bassell GJ, Warren ST (October 2008). "Fragile X syndrome: loss of local mRNA regulation alters synaptic development and function". Neuron. 60 (2): 201–214. doi:10.1016/j.neuron.2008.10.004. PMC   3691995 . PMID   18957214.
  55. Bureau I, Shepherd GM, Svoboda K (May 2008). "Circuit and plasticity defects in the developing somatosensory cortex of FMR1 knock-out mice". The Journal of Neuroscience. 28 (20): 5178–5188. doi:10.1523/JNEUROSCI.1076-08.2008. PMC   2696604 . PMID   18480274.
  56. Haberl MG, Zerbi V, Veltien A, Ginger M, Heerschap A, Frick A (November 2015). "Structural-functional connectivity deficits of neocortical circuits in the Fmr1 (-/y) mouse model of autism". Science Advances. 1 (10): e1500775. Bibcode:2015SciA....1E0775H. doi:10.1126/sciadv.1500775. PMC   4681325 . PMID   26702437.
  57. Mitchell DE, Miranda-Rottmann S, Blanchard MG, Araya R (January 2023). "Altered integration of excitatory inputs onto the basal dendrites of layer 5 pyramidal neurons in a mouse model of Fragile X Syndrome". PNAS. 120 (2): e2208963120. Bibcode:2023PNAS..12008963M. doi:10.1073/pnas.2208963120. PMC   9926222 . PMID   36595706.
  58. 1 2 Hogan AJ (June 2012). "Visualizing carrier status: fragile X syndrome and genetic diagnosis since the 1940s". Endeavour. 36 (2): 77–84. doi:10.1016/j.endeavour.2011.12.002. PMID   22257912. Archived from the original on 2019-11-05. Retrieved 2018-04-20.
  59. "How do health care providers diagnose Fragile X syndrome?". National Institute of Child Health and Human Development. US: NIH. 2021-08-25. Archived from the original on 2016-11-21. Retrieved 2016-11-21.
  60. 1 2 Hagerman RJ, Berry-Kravis E, Kaufmann WE, Ono MY, Tartaglia N, Lachiewicz A, et al. (January 2009). "Advances in the treatment of fragile X syndrome". Pediatrics. 123 (1): 378–390. doi:10.1542/peds.2008-0317. PMC   2888470 . PMID   19117905.
  61. Rueda JR, Ballesteros J, Guillen V, Tejada MI, Solà I (May 2011). "Folic acid for fragile X syndrome". The Cochrane Database of Systematic Reviews (5): CD008476. doi:10.1002/14651858.CD008476.pub2. PMID   21563169.
  62. Gantois I, Popic J, Khoutorsky A, Sonenberg N (January 2019). "Metformin for Treatment of Fragile X Syndrome and Other Neurological Disorders". Annual Review of Medicine. 70: 167–181. doi: 10.1146/annurev-med-081117-041238 . PMID   30365357. S2CID   53093694.
  63. Rueda JR, Ballesteros J, Tejada MI (October 2009). "Systematic review of pharmacological treatments in fragile X syndrome". BMC Neurology. 9: 53. doi: 10.1186/1471-2377-9-53 . PMC   2770029 . PMID   19822023.
  64. Coppus AM (2013-08-01). "People with intellectual disability: what do we know about adulthood and life expectancy?". Developmental Disabilities Research Reviews. 18 (1): 6–16. doi:10.1002/ddrr.1123. PMID   23949824.
  65. Duy PQ, Budimirovic DB (January 2017). "Fragile X Syndrome: Lessons Learned from the Most Translated Neurodevelopmental Disorder in Clinical Trials". Translational Neuroscience. 8 (1): 7–8. doi:10.1515/tnsci-2017-0002. PMC   5382936 . PMID   28400977.
  66. Dölen G, Osterweil E, Rao BS, Smith GB, Auerbach BD, Chattarji S, Bear MF (December 2007). "Correction of fragile X syndrome in mice". Neuron. 56 (6): 955–962. doi:10.1016/j.neuron.2007.12.001. PMC   2199268 . PMID   18093519.
  67. Dölen G, Carpenter RL, Ocain TD, Bear MF (July 2010). "Mechanism-based approaches to treating fragile X". Pharmacology & Therapeutics. 127 (1): 78–93. doi:10.1016/j.pharmthera.2010.02.008. PMID   20303363.
  68. Cole P (2012). "Mavoglurant". Drugs of the Future. 37 (1): 7–12. doi:10.1358/dof.2012.037.01.1772147. S2CID   258330291.
  69. Martin JP, Bell J (July 1943). "A Pedigree of Mental Defect Showing Sex-Linkage". Journal of Neurology and Psychiatry. 6 (3–4): 154–157. doi:10.1136/jnnp.6.3-4.154. PMC   1090429 . PMID   21611430.
  70. Lubs HA (May 1969). "A marker X chromosome". American Journal of Human Genetics. 21 (3): 231–244. PMC   1706424 . PMID   5794013.
  71. de la Cruz FF (September 1985). "Fragile X syndrome". American Journal of Mental Deficiency. 90 (2): 119–123. PMID   3901755.
  72. "FRAXA Member Update (three issues: Spring 1999, Summer 1999, Fall 1999)" (PDF) (newsletter). Newburyport, Massachusetts: FRAXA Research Foundation. Archived (PDF) from the original on 2010-12-15. Retrieved 2017-12-14.