Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
PLoS Biol ; 22(2): e3002502, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38421949

ABSTRACT

Peer review is an important part of the scientific process, but traditional peer review at journals is coming under increased scrutiny for its inefficiency and lack of transparency. As preprints become more widely used and accepted, they raise the possibility of rethinking the peer-review process. Preprints are enabling new forms of peer review that have the potential to be more thorough, inclusive, and collegial than traditional journal peer review, and to thus fundamentally shift the culture of peer review toward constructive collaboration. In this Consensus View, we make a call to action to stakeholders in the community to accelerate the growing momentum of preprint sharing and provide recommendations to empower researchers to provide open and constructive peer review for preprints.


Subject(s)
Peer Review , Research Personnel , Humans , Motion
2.
Neurol Genet ; 8(5): e200018, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36531974

ABSTRACT

Background and Objectives: Deletions and duplications at 16p11.2 (BP4 to BP5; 29.5-30.1 Mb) have been associated with several neurodevelopmental and neuropsychiatric disorders including autism spectrum disorder, intellectual disability (ID), and schizophrenia. Seizures have also been reported in individuals with these particular copy number variants, but the epilepsy phenotypes have not been well-delineated. We aimed to systematically characterize the seizure types, epilepsy syndromes, and epilepsy severity in a large cohort of individuals with these 16p11.2 deletions and duplications. Methods: The cohort of ascertained participants with the recurrent 16p11.2 copy number variant was assembled through the multicenter Simons Variation in Individuals Project. Detailed data on individuals identified as having a history of seizures were obtained using a semistructured phone interview and review of medical records, EEG, and MRI studies obtained clinically or as part of the Simons Variation in Individuals Project. Results: Among 129 individuals with the 16p11.2 deletion, 31 (24%) had at least one seizure, including 23 (18%) who met criteria for epilepsy; 42% of them fit the phenotype of classic or atypical Self-limited (Familial) Infantile Epilepsy (Se(F)IE). Among 106 individuals with 16p11.2 duplications, 16 (15%) had at least one seizure, including 11 (10%) who met criteria for epilepsy. The seizure types and epilepsy syndromes were heterogeneous in this group. Most of the individuals in both the deletion and duplication groups had well-controlled seizures with subsequent remission. Pharmacoresistant epilepsy was uncommon. Seizures responded favorably to phenobarbital, carbamazepine, and oxcarbazepine in the deletion group, specifically in the Se(F)IE, and to various antiseizure medications in the duplication group. Discussion: These findings delineate the spectrum of seizures and epilepsies in the recurrent 16p11.2 deletions and duplications and provide potential diagnostic, therapeutic, and prognostic information.

3.
Curr Opin Genet Dev ; 68: 49-56, 2021 06.
Article in English | MEDLINE | ID: mdl-33667823

ABSTRACT

The 16p11.2 BP4 and BP5 region, is a recurrent ∼600kb copy number variant (CNV), and deletions are one of the most frequent etiologies of neurodevelopmental disorders and autism spectrum disorder with an incidence of approximately 1/2000. Deletion carriers have delays in early neurodevelopment that most specifically impair speech, phonology and language in 70%. Intelligence quotient is shifted 1.8 standard deviations lower than family controls without the deletion. Other common neurobehavioral conditions include motor coordination difficulties (60%) and autism (20-25%). Unprovoked seizures are common (24%) and readily treated and resolve with age in many. Obesity evolves throughout childhood and by adulthood 75% are obese. Congenital anomalies are more common than the general population. The deletion is associated with an increase in brain volumes across all areas of the brain, changes in the white matter microstructural properties, and early electrophysiological cortical responses from auditory cortex. Studies of genetically defined conditions, particularly CNVs that are not associated with profound disabilities, provide homogeneity to study genetic impact on brain development, structure, and function to better understand complex neurobehavioral phenotypes such as autism.


Subject(s)
Autistic Disorder/genetics , Chromosome Disorders/genetics , DNA Copy Number Variations , Intellectual Disability/genetics , Neurodevelopmental Disorders/genetics , Chromosome Deletion , Chromosomes, Human, Pair 16/genetics , Gene Deletion , Genetic Association Studies , Humans , Language Development Disorders/genetics , Neuroimaging , Obesity/genetics , Seizures/genetics , White Matter/physiopathology
4.
Biol Psychiatry ; 84(4): 253-264, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29778275

ABSTRACT

BACKGROUND: 16p11.2 breakpoint 4 to 5 copy number variants (CNVs) increase the risk for developing autism spectrum disorder, schizophrenia, and language and cognitive impairment. In this multisite study, we aimed to quantify the effect of 16p11.2 CNVs on brain structure. METHODS: Using voxel- and surface-based brain morphometric methods, we analyzed structural magnetic resonance imaging collected at seven sites from 78 individuals with a deletion, 71 individuals with a duplication, and 212 individuals without a CNV. RESULTS: Beyond the 16p11.2-related mirror effect on global brain morphometry, we observe regional mirror differences in the insula (deletion > control > duplication). Other regions are preferentially affected by either the deletion or the duplication: the calcarine cortex and transverse temporal gyrus (deletion > control; Cohen's d > 1), the superior and middle temporal gyri (deletion < control; Cohen's d < -1), and the caudate and hippocampus (control > duplication; -0.5 > Cohen's d > -1). Measures of cognition, language, and social responsiveness and the presence of psychiatric diagnoses do not influence these results. CONCLUSIONS: The global and regional effects on brain morphometry due to 16p11.2 CNVs generalize across site, computational method, age, and sex. Effect sizes on neuroimaging and cognitive traits are comparable. Findings partially overlap with results of meta-analyses performed across psychiatric disorders. However, the lack of correlation between morphometric and clinical measures suggests that CNV-associated brain changes contribute to clinical manifestations but require additional factors for the development of the disorder. These findings highlight the power of genetic risk factors as a complement to studying groups defined by behavioral criteria.


Subject(s)
Brain/pathology , Chromosome Deletion , Chromosome Duplication , Chromosomes, Human, Pair 16/genetics , DNA Copy Number Variations , Adolescent , Adult , Autism Spectrum Disorder/diagnostic imaging , Autism Spectrum Disorder/genetics , Child , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/genetics , Female , Humans , Intellectual Disability/diagnostic imaging , Intellectual Disability/genetics , Language , Magnetic Resonance Imaging , Male , Middle Aged , Neurodevelopmental Disorders/diagnostic imaging , Neurodevelopmental Disorders/genetics , Schizophrenia/diagnostic imaging , Schizophrenia/genetics , Young Adult
5.
Trends Neurosci ; 41(7): 442-456, 2018 07.
Article in English | MEDLINE | ID: mdl-29691040

ABSTRACT

Advances in gene discovery for neurodevelopmental disorders have identified SCN2A dysfunction as a leading cause of infantile seizures, autism spectrum disorder, and intellectual disability. SCN2A encodes the neuronal sodium channel NaV1.2. Functional assays demonstrate strong correlation between genotype and phenotype. This insight can help guide therapeutic decisions and raises the possibility that ligands that selectively enhance or diminish channel function may improve symptoms. The well-defined function of sodium channels makes SCN2A an important test case for investigating the neurobiology of neurodevelopmental disorders more generally. Here, we discuss the progress made, through the concerted efforts of a diverse group of academic and industry scientists as well as policy advocates, in understanding and treating SCN2A-related disorders.


Subject(s)
NAV1.2 Voltage-Gated Sodium Channel/genetics , NAV1.2 Voltage-Gated Sodium Channel/metabolism , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/metabolism , Animals , Humans , Neurodevelopmental Disorders/drug therapy
6.
Article in English | MEDLINE | ID: mdl-29196435

ABSTRACT

Scientists who leave the laboratory bench to work for biomedical foundations mobilize and focus resources on the most promising research behind a foundation's mission. They acquire a broad view of a field, interact closely with research scientists at meetings and laboratory visits, and often manage proposal review boards and monitor grant progress. Increasingly, scientists at foundations also have a more active role in catalyzing research: They are involved in organizing targeted workshops, setting research priorities, and directly creating and managing resources for a scientific community. They often work closely with patient advocacy groups, contract research organizations, government funders and regulators, and biotech and pharmaceutical companies.Job opportunities in foundations are highly varied, reflecting the diversity of foundations and missions. For example, some foundations have general missions to support broad basic scientific research, whereas others focus specifically on a particular disease and may require different skills. Most scientific positions at foundations require a PhD or MD and often some postdoctoral or other experience, although not all do. Foundations often lack the clear career path that an academic position offers (various levels of professorship, tenure, etc.), although there is often significant potential for growth in responsibilities in terms of managing science, people, and budgets. Jobs in foundations can be challenging yet extremely rewarding, especially for those with broad interests and goal-oriented personalities. Many at foundations feel that they have the potential to have an even greater impact than if they had stayed at the bench.


Subject(s)
Biomedical Research/trends , Career Choice , Foundations , Humans , Organizational Objectives , Research Personnel , Research Support as Topic
8.
Hum Brain Mapp ; 37(8): 2833-48, 2016 08.
Article in English | MEDLINE | ID: mdl-27219475

ABSTRACT

Copy number variants at the 16p11.2 chromosomal locus are associated with several neuropsychiatric disorders, including autism, schizophrenia, bipolar disorder, attention-deficit hyperactivity disorder, and speech and language disorders. A gene dosage dependence has been suggested, with 16p11.2 deletion carriers demonstrating higher body mass index and head circumference, and 16p11.2 duplication carriers demonstrating lower body mass index and head circumference. Here, we use diffusion tensor imaging to elucidate this reciprocal relationship in white matter organization, showing widespread increases of fractional anisotropy throughout the supratentorial white matter in pediatric deletion carriers and, in contrast, extensive decreases of white matter fractional anisotropy in pediatric and adult duplication carriers. We find associations of these white matter alterations with cognitive and behavioral impairments. We further demonstrate the value of imaging metrics for characterizing the copy number variant phenotype by employing linear discriminant analysis to predict the gene dosage status of the study subjects. These results show an effect of 16p11.2 gene dosage on white matter microstructure, and further suggest that opposite changes in diffusion tensor imaging metrics can lead to similar cognitive and behavioral deficits. Given the large effect sizes found in this study, our results support the view that specific genetic variations are more strongly associated with specific brain alterations than are shared neuropsychiatric diagnoses. Hum Brain Mapp 37:2833-2848, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Brain/pathology , Chromosomes, Human, Pair 16/genetics , White Matter/pathology , Adolescent , Adult , Child , Chromosome Deletion , Chromosome Duplication , Diffusion Tensor Imaging , Female , Gene Dosage , Heterozygote , Humans , Male , Middle Aged , Young Adult
9.
J Autism Dev Disord ; 46(8): 2734-2748, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27207092

ABSTRACT

The 16p11.2 duplication (BP4-BP5) is associated with Autism Spectrum Disorder (ASD), although significant heterogeneity exists. Quantitative ASD, behavioral and neuropsychological measures and DSM-IV diagnoses in child and adult carriers were compared with familial non-carrier controls, and to published results from deletion carriers. The 16p11.2 duplication phenotype ranges widely from asymptomatic presentation to significant disability. The most common diagnoses were intellectual disability, motor delays and Attention Deficit Hyperactivity Disorder in children, and anxiety in adults. ASD occurred in nearly 20 % of child cases, but a majority of carriers did not show the unique social features of ASD. The 16p11.2 duplication phenotype is characterized by wider variability than the reciprocal deletion, likely reflecting contributions from additional risk factors.


Subject(s)
Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/psychology , Chromosomes, Human, Pair 16/genetics , Gene Duplication/genetics , Adolescent , Adult , Autism Spectrum Disorder/diagnosis , Autistic Disorder/diagnosis , Autistic Disorder/genetics , Autistic Disorder/psychology , Child , Child, Preschool , Chromosome Deletion , Chromosome Disorders/diagnosis , Chromosome Disorders/genetics , Chromosome Disorders/psychology , Female , Humans , Infant , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Intellectual Disability/psychology , Male , Phenotype
10.
Cereb Cortex ; 26(5): 1957-64, 2016 May.
Article in English | MEDLINE | ID: mdl-25678630

ABSTRACT

Individuals with the 16p11.2 BP4-BP5 copy number variant (CNV) exhibit a range of behavioral phenotypes that may include mild impairment in cognition and clinical diagnoses of autism spectrum disorder (ASD). To better understand auditory processing impairments in populations with this chromosomal variation, auditory evoked responses were examined in children with the 16p11.2 deletion, 16p11.2 duplication, and age-matched controls. Stimuli consisted of sinusoidal binaural tones presented passively while children underwent recording with magnetoencephalography (MEG). The primary indicator of auditory processing impairment was the latency of the ∼100-ms "M100" auditory response detected by MEG, with the 16p11.2 deletion population exhibiting profoundly delayed M100 latencies relative to controls. This delay remained even after controlling for potential confounds such as age and cognitive ability. No significant difference in M100 latency was observed between 16p11.2 duplication carriers and controls. Additionally, children meeting diagnostic criteria for ASD (16p11.2 deletion carriers) exhibited nonsignificant latency delays when compared with the corresponding CNV carriers not meeting criteria for ASD. Present results indicate that 16p11.2 deletion is associated with auditory processing delays analogous to (but substantially more pronounced than) those previously reported in "idiopathic" ASD.


Subject(s)
Auditory Cortex/physiopathology , Autistic Disorder/physiopathology , Chromosome Disorders/physiopathology , Chromosome Duplication , Evoked Potentials, Auditory/genetics , Intellectual Disability/physiopathology , Acoustic Stimulation , Adolescent , Child , Chromosome Deletion , Chromosomes, Human, Pair 16 , Female , Genotype , Humans , Magnetoencephalography , Male , Neuropsychological Tests
11.
Genet Med ; 18(4): 341-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26066539

ABSTRACT

PURPOSE: To characterize the clinical phenotype of the recurrent copy-number variation (CNV) at 1q21.1, we assessed the psychiatric and medical phenotypes of 1q21.1 deletion and duplication carriers ascertained through clinical genetic testing and family member cascade testing, with particular emphasis on dimensional assessment across multiple functional domains. METHODS: Nineteen individuals with 1q21.1 deletion, 19 individuals with the duplication, and 23 familial controls (noncarrier siblings and parents) spanning early childhood through adulthood were evaluated for psychiatric, neurologic, and other medical diagnoses, and their cognitive, adaptive, language, motor, and neurologic domains were also assessed. Twenty-eight individuals with 1q21.1 CNVs (15 deletion, 13 duplication) underwent structural magnetic resonance brain imaging. RESULTS: Probands with 1q21.1 CNVs presented with a range of psychiatric, neurologic, and medical disorders. Deletion and duplication carriers shared several features, including borderline cognitive functioning, impaired fine and gross motor functioning, articulation abnormalities, and hypotonia. Increased frequency of Autism Spectrum Disorder (ASD) diagnosis, increased ASD symptom severity, and increased prevalence of macrocephaly were observed in the duplication relative to deletion carriers, whereas reciprocally increased prevalence of microcephaly was observed in the deletion carriers. CONCLUSIONS: Individuals with 1q21.1 deletions or duplications exhibit consistent deficits on motor and cognitive functioning and abnormalities in head circumference.Genet Med 18 4, 341-349.


Subject(s)
Chromosome Disorders/diagnosis , Chromosome Disorders/genetics , Chromosomes, Human, Pair 1 , DNA Copy Number Variations , Phenotype , Adult , Child , Child, Preschool , Chromosome Deletion , Chromosome Duplication , Female , Humans , Male , Middle Aged , Neuropsychological Tests , Registries , Young Adult
12.
Neuroimage Clin ; 9: 50-7, 2015.
Article in English | MEDLINE | ID: mdl-26413471

ABSTRACT

Copy number variations at chromosome 16p11.2 contribute to neurodevelopmental disorders, including autism spectrum disorder (ASD). This study seeks to improve our understanding of the biological basis of behavioral phenotypes common in ASD, in particular the prominent and prevalent disruption of spoken language seen in children with the 16p11.2 BP4-BP5 deletion. We examined the auditory and language white matter pathways with diffusion MRI in a cohort of 36 pediatric deletion carriers and 45 age-matched controls. Diffusion MR tractography of the auditory radiations and the arcuate fasciculus was performed to generate tract specific measures of white matter microstructure. In both tracts, deletion carriers exhibited significantly higher diffusivity than that of controls. Cross-sectional diffusion parameters in these tracts changed with age with no group difference in the rate of maturation. Within deletion carriers, the left-hemisphere arcuate fasciculus mean and radial diffusivities were significantly negatively correlated with clinical language ability, but not non-verbal cognitive ability. Diffusion metrics in the right-hemisphere arcuate fasciculus were not predictive of language ability. These results provide insight into the link between the 16p11.2 deletion, abnormal auditory and language pathway structures, and the specific behavioral deficits that may contribute to neurodevelopmental disorders such as ASD.


Subject(s)
Auditory Pathways/pathology , Autistic Disorder/pathology , Chromosome Disorders/pathology , Intellectual Disability/pathology , Language , Adolescent , Brain/pathology , Child , Chromosome Deletion , Chromosomes, Human, Pair 16 , Diffusion Magnetic Resonance Imaging , Female , Humans , Male , White Matter/pathology
13.
JAMA Psychiatry ; 72(2): 119-26, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25493922

ABSTRACT

IMPORTANCE: Most disorders caused by copy number variants (CNVs) display significant clinical variability, often referred to as incomplete penetrance and variable expressivity. Genetic and environmental sources of this variability are not well understood. OBJECTIVES: To investigate the contributors to phenotypic variability in probands with CNVs involving the same genomic region; to measure the effect size for de novo mutation events; and to explore the contribution of familial background to resulting cognitive, behavioral, and motor performance outcomes in probands with de novo CNVs. DESIGN, SETTING, AND PARTICIPANTS: Family-based study design with a volunteer sample of 56 individuals with de novo 16p11.2 deletions and their noncarrier parents and siblings from the Simons Variation in Individuals Project. MAIN OUTCOMES AND MEASURES: We used linear mixed-model analysis to measure effect size and intraclass correlation to determine the influence of family background for a de novo CNV on quantitative traits representing the following 3 neurodevelopmental domains: cognitive ability (Full-Scale IQ), social behavior (Social Responsiveness Scale), and neuromotor performance (Purdue Pegboard Test). We included an anthropometric trait, body mass index, for comparison. RESULTS: A significant deleterious effect of the 16p11.2 deletion was demonstrated across all domains. Relative to the biparental mean, the effect sizes were -1.7 SD for cognitive ability, 2.2 SD for social behavior, and -1.3 SD for neuromotor performance (P < .001). Despite large deleterious effects, significant positive correlations between parents and probands were preserved for the Full-Scale IQ (0.42 [P = .03]), the verbal IQ (0.53 [P = .004]), and the Social Responsiveness Scale (0.52 [P = .009]) scores. We also observed a 1-SD increase in the body mass index of probands compared with siblings, with an intraclass correlation of 0.40 (P = .07). CONCLUSIONS AND RELEVANCE: Analysis of families with de novo CNVs provides the least confounded estimate of the effect size of the 16p11.2 deletion on heritable, quantitative traits and demonstrates a 1- to 2-SD effect across all neurodevelopmental dimensions. Significant parent-proband correlations indicate that family background contributes to the phenotypic variability seen in this and perhaps other CNV disorders and may have implications for counseling families regarding their children's developmental and psychiatric prognoses. Use of biparental mean scores rather than general population mean scores may be more relevant to examine the effect of a mutation or any other cause of trait variation on a neurodevelopmental outcome and possibly on systems of diagnosis and trait ascertainment for developmental disorders.


Subject(s)
Autistic Disorder/physiopathology , Chromosome Disorders/physiopathology , Intellectual Disability/physiopathology , Intelligence/genetics , Parents , Phenotype , Psychomotor Performance/physiology , Social Behavior , Adult , Autistic Disorder/genetics , Chromosome Deletion , Chromosome Disorders/genetics , Chromosomes, Human, Pair 16/genetics , DNA Copy Number Variations/genetics , Female , Humans , Intellectual Disability/genetics , Male , Siblings
14.
Biol Psychiatry ; 77(9): 785-93, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25064419

ABSTRACT

BACKGROUND: Deletion of the recurrent ~600 kb BP4-BP5 chromosomal region 16p11.2 has been associated with a wide range of neurodevelopmental outcomes. METHODS: To clarify the phenotype of 16p11.2 deletion, we examined the psychiatric and developmental presentation of predominantly clinically referred individuals, with a particular emphasis on broader autism phenotype characteristics in individuals with recurrent ~600 kb chromosome 16p11.2 deletions. Using an extensive standardized assessment battery across three clinical sites, 85 individuals with the 16p11.2 deletion and 153 familial control subjects were evaluated for symptom presentation and clinical diagnosis. RESULTS: Individuals with the 16p11.2 deletion presented with a high frequency of psychiatric and developmental disorders (>90%). The most commonly diagnosed conditions were developmental coordination disorder, phonologic processing disorder, expressive and receptive language disorders (71% of individuals >3 years old with a speech and language-related disorder), and autism spectrum disorder. Individuals with the 16p11.2 deletion not meeting diagnostic criteria for autism spectrum disorder had a significantly higher prevalence of autism-related characteristics compared with the familial noncarrier control group. Individuals with the 16p11.2 deletion had a range of intellectual ability, but IQ scores were 26 points lower than noncarrier family members on average. CONCLUSIONS: Clinically referred individuals with the 16p11.2 deletion have high rates of psychiatric and developmental disorders and provide a genetically well-defined group to study the emergence of developmental difficulties, particularly associated with the broader autism phenotype.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 16 , Cognition , Developmental Disabilities/genetics , Mental Disorders/genetics , Phenotype , Adolescent , Child , Child, Preschool , Developmental Disabilities/physiopathology , Developmental Disabilities/psychology , Heterozygote , Humans , Intelligence/genetics , Mental Disorders/physiopathology , Mental Disorders/psychology , Young Adult
15.
J Neurosci ; 34(34): 11199-211, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-25143601

ABSTRACT

Deletions and duplications of the recurrent ~600 kb chromosomal BP4-BP5 region of 16p11.2 are associated with a broad variety of neurodevelopmental outcomes including autism spectrum disorder. A clue to the pathogenesis of the copy number variant (CNV)'s effect on the brain is that the deletion is associated with a head size increase, whereas the duplication is associated with a decrease. Here we analyzed brain structure in a clinically ascertained group of human deletion (N = 25) and duplication (N = 17) carriers from the Simons Variation in Individuals Project compared with age-matched controls (N = 29 and 33, respectively). Multiple brain measures showed increased size in deletion carriers and reduced size in duplication carriers. The effects spanned global measures of intracranial volume, brain size, compartmental measures of gray matter and white matter, subcortical structures, and the cerebellum. Quantitatively, the largest effect was on the thalamus, but the collective results suggest a pervasive rather than a selective effect on the brain. Detailed analysis of cortical gray matter revealed that cortical surface area displays a strong dose-dependent effect of CNV (deletion > control > duplication), whereas average cortical thickness is less affected. These results suggest that the CNV may exert its opposing influences through mechanisms that influence early stages of embryonic brain development.


Subject(s)
Brain/pathology , Child Development Disorders, Pervasive/pathology , Chromosome Deletion , Chromosome Duplication , Chromosomes, Human, Pair 16/genetics , Adolescent , Adult , Age Factors , Brain/growth & development , Child , Child Development Disorders, Pervasive/genetics , Cognition Disorders/genetics , Female , Humans , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Middle Aged , Neuropsychological Tests , Psychiatric Status Rating Scales , Social Behavior Disorders/genetics , Young Adult
16.
J Neurosci ; 34(18): 6214-23, 2014 Apr 30.
Article in English | MEDLINE | ID: mdl-24790192

ABSTRACT

Copy number variants (CNVs) of the chromosomal locus 16p11.2, consisting of either deletions or duplications, have been implicated in autism, schizophrenia, epilepsy, and other neuropsychiatric disorders. Since abnormal white matter microstructure can be seen in these more broadly defined clinical disorders, we used diffusion magnetic resonance imaging and tract-based spatial statistics to investigate white matter microstructural integrity in human children with 16p11.2 deletions. We show that deletion carriers, compared with typically developing matched controls, have increased axial diffusivity (AD) in many major central white matter tracts, including the anterior corpus callosum as well as bilateral internal and external capsules. Higher AD correlated with lower nonverbal IQ in the deletion carriers, but not controls. Increases in fractional anisotropy and mean diffusivity were also found in some of the same tracts with elevated AD. Closer examination with neurite orientation dispersion and density imaging revealed that fiber orientation dispersion was decreased in some central white matter tracts. Notably, these alterations of white matter are unlike microstructural differences reported for any other neurodevelopmental disorders, including autism spectrum disorders that have phenotypic overlap with the deletion carriers. These findings suggest that deletion of the 16p11.2 locus is associated with a unique widespread pattern of aberrant white matter microstructure that may underlie the impaired cognition characteristic of this CNV.


Subject(s)
Autistic Disorder , Brain/pathology , Chromosome Deletion , Chromosome Disorders , Intellectual Disability , Leukoencephalopathies/etiology , Nerve Fibers, Myelinated/pathology , Adolescent , Anisotropy , Autistic Disorder/complications , Autistic Disorder/genetics , Autistic Disorder/pathology , Biophysics , Case-Control Studies , Child , Chromosome Disorders/complications , Chromosome Disorders/genetics , Chromosome Disorders/pathology , Chromosomes, Human, Pair 16/genetics , Cognition Disorders/etiology , Cognition Disorders/genetics , Diffusion Magnetic Resonance Imaging , Female , Humans , Image Processing, Computer-Assisted , Intellectual Disability/complications , Intellectual Disability/genetics , Intellectual Disability/pathology , Leukoencephalopathies/genetics , Male , Models, Neurological , Statistics as Topic
17.
J Med Genet ; 49(10): 660-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23054248

ABSTRACT

BACKGROUND: The recurrent ~600 kb 16p11.2 BP4-BP5 deletion is among the most frequent known genetic aetiologies of autism spectrum disorder (ASD) and related neurodevelopmental disorders. OBJECTIVE: To define the medical, neuropsychological, and behavioural phenotypes in carriers of this deletion. METHODS: We collected clinical data on 285 deletion carriers and performed detailed evaluations on 72 carriers and 68 intrafamilial non-carrier controls. RESULTS: When compared to intrafamilial controls, full scale intelligence quotient (FSIQ) is two standard deviations lower in carriers, and there is no difference between carriers referred for neurodevelopmental disorders and carriers identified through cascade family testing. Verbal IQ (mean 74) is lower than non-verbal IQ (mean 83) and a majority of carriers require speech therapy. Over 80% of individuals exhibit psychiatric disorders including ASD, which is present in 15% of the paediatric carriers. Increase in head circumference (HC) during infancy is similar to the HC and brain growth patterns observed in idiopathic ASD. Obesity, a major comorbidity present in 50% of the carriers by the age of 7 years, does not correlate with FSIQ or any behavioural trait. Seizures are present in 24% of carriers and occur independently of other symptoms. Malformations are infrequently found, confirming only a few of the previously reported associations. CONCLUSIONS: The 16p11.2 deletion impacts in a quantitative and independent manner FSIQ, behaviour and body mass index, possibly through direct influences on neural circuitry. Although non-specific, these features are clinically significant and reproducible. Lastly, this study demonstrates the necessity of studying large patient cohorts ascertained through multiple methods to characterise the clinical consequences of rare variants involved in common diseases.


Subject(s)
Child Development Disorders, Pervasive/genetics , Chromosome Deletion , Chromosomes, Human, Pair 16 , Developmental Disabilities/genetics , Phenotype , Adolescent , Adult , Body Mass Index , Child , Child Development Disorders, Pervasive/diagnosis , Developmental Disabilities/diagnosis , Female , Gene Order , Heterozygote , Humans , Intelligence Tests , Male , Syndrome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL