Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 213
1.
Cells ; 12(3)2023 01 21.
Article En | MEDLINE | ID: mdl-36766733

Williams-Beuren syndrome (WBS) is a neurodevelopmental disorder caused by a chromosomic microdeletion (7q11.23). WBS has been modeled by a mouse line having a complete deletion (CD) of the equivalent mouse locus. This model has been largely used to investigate the etiopathological mechanisms of WBS, although pharmacological therapies have not been identified yet. Surprisingly, CD mice were so far mainly tested in adulthood, despite the developmental nature of WBS and the critical relevance of early timing for potential treatments. Here we provide for the first time a phenotypic characterization of CD mice of both sexes during infancy and adolescence, i.e., between birth and 7 weeks of age. CD pups of both sexes showed reduced body growth, delayed sensory development, and altered patterns of ultrasonic vocalizations and exploratory behaviors. Adolescent CD mice showed reduced locomotion and acoustic startle response, and altered social interaction and communication, the latter being more pronounced in female mice. Juvenile CD mutants of both sexes also displayed reduced brain weight, cortical and hippocampal dendritic length, and spine density. Our findings highlight the critical relevance of early neurobehavioral alterations as biomarkers of WBS pathology, underlying the importance of adolescence for identifying novel therapeutic targets for this neurological disorder.


Williams Syndrome , Male , Mice , Female , Animals , Williams Syndrome/genetics , Williams Syndrome/pathology , Reflex, Startle , Disease Models, Animal , Hippocampus/pathology
2.
Mol Psychiatry ; 28(3): 1112-1127, 2023 03.
Article En | MEDLINE | ID: mdl-36577841

Williams syndrome (WS) is a neurodevelopmental disorder caused by a heterozygous micro-deletion in the WS critical region (WSCR) and is characterized by hyper-sociability and neurocognitive abnormalities. Nonetheless, whether and to what extent WSCR deletion leads to epigenetic modifications in the brain and induces pathological outcomes remains largely unknown. By examining DNA methylation in frontal cortex, we revealed genome-wide disruption in the methylome of individuals with WS, as compared to typically developed (TD) controls. Surprisingly, differentially methylated sites were predominantly annotated as introns and intergenic loci and were found to be highly enriched around binding sites for transcription factors that regulate neuronal development, plasticity and cognition. Moreover, by utilizing enhancer-promoter interactome data, we confirmed that most of these loci function as active enhancers in the human brain or as target genes of transcriptional networks associated with myelination, oligodendrocyte (OL) differentiation, cognition and social behavior. Cell type-specific methylation analysis revealed aberrant patterns in the methylation of active enhancers in neurons and OLs, and important neuron-glia interactions that might be impaired in individuals with WS. Finally, comparison of methylation profiles from blood samples of individuals with WS and healthy controls, along with other data collected in this study, identified putative targets of endophenotypes associated with WS, which can be used to define brain-risk loci for WS outside the WSCR locus, as well as for other associated pathologies. In conclusion, our study illuminates the brain methylome landscape of individuals with WS and sheds light on how these aberrations might be involved in social behavior and physiological abnormalities. By extension, these results may lead to better diagnostics and more refined therapeutic targets for WS.


Williams Syndrome , Humans , Williams Syndrome/genetics , Williams Syndrome/pathology , Neurons/metabolism , DNA Methylation , Oligodendroglia/pathology , DNA
3.
Cells ; 11(1)2022 01 04.
Article En | MEDLINE | ID: mdl-35011720

Williams syndrome (WS) is a multisystem neurodevelopmental disorder caused by a de novo hemizygous deletion of ~26 genes from chromosome 7q11.23, among them the general transcription factor II-I (GTF2I). By studying a novel murine model for the hypersociability phenotype associated with WS, we previously revealed surprising aberrations in myelination and cell differentiation properties in the cortices of mutant mice compared to controls. These mutant mice had selective deletion of Gtf2i in the excitatory neurons of the forebrain. Here, we applied diffusion magnetic resonance imaging and fiber tracking, which showed a reduction in the number of streamlines in limbic outputs such as the fimbria/fornix fibers and the stria terminalis, as well as the corpus callosum of these mutant mice compared to controls. Furthermore, we utilized next-generation sequencing (NGS) analysis of cortical small RNAs' expression (RNA-Seq) levels to identify altered expression of microRNAs (miRNAs), including two from the miR-34 cluster, known to be involved in prominent processes in the developing nervous system. Luciferase reporter assay confirmed the direct binding of miR-34c-5p to the 3'UTR of PTPRU-a gene involved in neural development that was elevated in the cortices of mutant mice relative to controls. Moreover, we found an age-dependent variation in the expression levels of doublecortin (Dcx)-a verified miR-34 target. Thus, we demonstrate the substantial effect a single gene deletion can exert on miRNA regulation and brain structure, and advance our understanding and, hopefully, treatment of WS.


Brain/growth & development , Doublecortin Protein/metabolism , MicroRNAs/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , White Matter/physiopathology , Williams Syndrome/genetics , Animals , Disease Models, Animal , Female , Humans , Mice , Williams Syndrome/pathology
4.
Am J Med Genet A ; 185(12): 3821-3824, 2021 12.
Article En | MEDLINE | ID: mdl-34235868

We present an 18-month-old male with Tetralogy of Fallot, retrognathia, short stature, global developmental delay, and dysmorphic features who was found to have dual diagnoses of both Williams syndrome and 22q11.2 deletion syndrome (22q11.2DS). To our knowledge, this is the second case of such a co-occurrence documented in the medical literature. Our patient presents with a blended physical phenotype of these two conditions and a behavioral phenotype that is distinct from what is typically observed in either disorder alone. We compare our patient's phenotype to the previously reported case and to the typical phenotypes for each individual condition. Additionally, we discuss why the occurrence of these two disorders together seems to be so rare, and the benefit of a genetics evaluation to an inpatient service team and the patient.


Developmental Disabilities/genetics , DiGeorge Syndrome/genetics , Tetralogy of Fallot/genetics , Williams Syndrome/genetics , Developmental Disabilities/complications , Developmental Disabilities/pathology , DiGeorge Syndrome/complications , DiGeorge Syndrome/pathology , Humans , Infant , Male , Phenotype , Tetralogy of Fallot/complications , Tetralogy of Fallot/pathology , Williams Syndrome/complications , Williams Syndrome/pathology
5.
Pediatr Res ; 90(5): 1065-1072, 2021 11.
Article En | MEDLINE | ID: mdl-33531674

BACKGROUND: Williams-Beuren syndrome (WS) is characterized by cardiovascular abnormalities associated with a multigene deletion on 7q11.23, in particular elastin (ELN). Peripheral pulmonary artery stenosis (PPAS) frequently affects pediatric patients with WS. Molecular investigation of WS pulmonary arterial (PA) tissue is limited by tissue scarcity. METHODS: We compared transcriptomes, tissue architecture, and localized changes in protein expression in PA tissue from patients with WS (n = 8) and donors (n = 5). RESULTS: Over 100 genes were differentially expressed at the ≥4-fold level, including genes related to the serotonin signaling pathway: >60-fold downregulation of serotonin transporter SLC6A4 and >3-fold upregulation of serotonin receptor HTR2A. Histologic examination revealed abnormal elastin distribution and smooth muscle cell morphology in WS PA, with markedly shorter, disorganized elastin fibers, and expanded proteoglycan-rich extracellular matrix between muscle layers. CONCLUSIONS: There were significant abnormalities in the PA expression of genes regulating serotonin signaling, metabolism, and receptors in WS. Those changes were associated with distinct changes in the arterial structure and may play a role in the stenosis-promoting effects of elevated shear stress at PA bifurcations in WS. IMPACT: Serotonin pathway signaling is significantly altered in the pulmonary arteries of patients with Williams syndrome and severe peripheral arterial stenosis. The present study compares the histological and biochemical characteristics of pulmonary arteries from patients with Williams syndrome to those of controls, something that has not, to our knowledge, been done previously. It demonstrates marked abnormalities in the pulmonary arteries of patients with Williams syndrome, especially significant pathologic alterations in the signaling of the serotonin pathway. The findings of this study provide direction for the development of potential therapies to treat pulmonary artery stenosis in patients with Williams syndrome.


Pulmonary Artery/metabolism , Serotonin/metabolism , Williams Syndrome/pathology , Actins/metabolism , Adolescent , Adult , Case-Control Studies , Child , Elastin/metabolism , Female , Gene Expression Regulation , Humans , Male , Receptors, Serotonin/genetics , Receptors, Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Williams Syndrome/genetics , Young Adult
6.
Am J Med Genet A ; 185(1): 242-249, 2021 01.
Article En | MEDLINE | ID: mdl-33098373

Williams-Beurens syndrome (WBS) is a rare genetic disorder caused by a recurrent 7q11.23 microdeletion. Clinical characteristics include typical facial dysmorphisms, weakness of connective tissue, short stature, mild to moderate intellectual disability and distinct behavioral phenotype. Cardiovascular diseases are common due to haploinsufficiency of ELN gene. A few cases of larger or smaller deletions have been reported spanning towards the centromeric or the telomeric regions, most of which included ELN gene. We report on three patients from two unrelated families, presenting with distinctive WBS features, harboring an atypical distal deletion excluding ELN gene. Our study supports a critical role of CLIP2, GTF2IRD1, and GTF2I gene in the WBS neurobehavioral profile and in craniofacial features, highlights a possible role of HIP1 in the autism spectrum disorder, and delineates a subgroup of WBS individuals with an atypical distal deletion not associated to an increased risk of cardiovascular defects.


Celiac Disease/genetics , Elastin/genetics , Neurocognitive Disorders/genetics , Williams Syndrome/genetics , Adolescent , Adult , Celiac Disease/complications , Celiac Disease/pathology , Child , Chromosome Deletion , Chromosomes, Human, Pair 7/genetics , Female , Genetic Predisposition to Disease , Haploinsufficiency/genetics , Humans , Neurocognitive Disorders/complications , Neurocognitive Disorders/pathology , Phenotype , Williams Syndrome/complications , Williams Syndrome/pathology
7.
Mol Genet Genomic Med ; 9(1): e1560, 2021 01.
Article En | MEDLINE | ID: mdl-33269527

BACKGROUND: Dual diagnoses in genetics practice are not uncommon and patients with dual diagnosis often present with complex and challenging phenotypes. A combination of meticulous phenotyping and molecular genetic techniques are essential in solving these diagnostic odysseys. METHODS: Clinical features and genetic workup of a patient presenting with incidental schwannomatosis. RESULTS: A 19-year-old male presented with incidental painless schwannomatosis in the background of macrocephaly, distinctive facies, and learning disability. Comprehensive genetic testing with gene panel and chromosomal microarray led to a dual diagnosis of LZTR1-related schwannomatosis and 7q11.23 duplication syndrome. CONCLUSION: We emphasize the need for high index of suspicion and comprehensive genetic testing in complex phenotypes. Interrogation of the interplay between the pathogenic variants in multiple genes could improve our understanding of the pathophysiologic pathways and contribute to therapeutic discoveries.


Neurilemmoma/genetics , Neurofibromatoses/genetics , Phenotype , Skin Neoplasms/genetics , Transcription Factors/genetics , Williams Syndrome/genetics , Humans , Male , Neurilemmoma/complications , Neurilemmoma/pathology , Neurofibromatoses/complications , Neurofibromatoses/pathology , Skin Neoplasms/complications , Skin Neoplasms/pathology , Spinal Nerves/pathology , Williams Syndrome/complications , Williams Syndrome/pathology , Young Adult
8.
Eur J Med Genet ; 63(12): 104084, 2020 Dec.
Article En | MEDLINE | ID: mdl-33045407

Williams-Beuren syndrome (WBS) is a rare neurodevelopmental disorder characterized by supravalvular aortic stenosis (SVAS), intellectual disability, overfriendliness and dysmorphic features. It is typically caused by 1.5-1.8 Mb deletions on 7q11.23. The 22q11.21 microduplication syndrome has a variable phenotype and is frequently associated with congenital heart disease. Here we present a unique patient, carrying aberrations within both of the above syndrome regions, referred for possible diagnosis of WBS because of SVAS. The patient was a boy who died suddenly 47 days after birth, possibly due to cardiac complications. Genetic testing was carried out, including array Comparative Genomic Hybridization (aCGH), Fluorescence In situ Hybridization (FISH) and Multiplex Ligation-Dependent Probe Amplification (MLPA) showing that the proband was heterozygous for a novel and atypical 0.3 Mb deletion in WBS region (7q11.23) encompassing the ELN gene. In addition, he was found heterozygous for a 22q11.21 microduplication. Parental studies revealed that the 7q11.23 deletion was inherited from the mother who also exhibited a cardiovascular phenotype, however very mild. The same maternally inherited deletion was detected in one of the proband's siblings, born two years later with a less severe SVAS. The 22q11.2 microduplication was de novo in origin. Detection and investigation of atypical deletions within known syndrome regions are crucial for better genotype-phenotype correlations and more accurate characterization of critical regions. The combined effect of two different genetic defects - one in a known syndrome region and one with variable clinical significance, is valuable for revealing gene interactions and enabling more accurate predictions, especially in prenatal diagnosis.


Abnormalities, Multiple/genetics , Aortic Stenosis, Supravalvular/genetics , Chromosome Duplication/genetics , DiGeorge Syndrome/genetics , Williams Syndrome/genetics , Abnormalities, Multiple/pathology , Adult , Aortic Stenosis, Supravalvular/pathology , Chromosomes, Human, Pair 22/genetics , DiGeorge Syndrome/pathology , Female , Humans , Infant , Inheritance Patterns , Male , Williams Syndrome/pathology
9.
Exp Hematol ; 89: 80-86, 2020 09.
Article En | MEDLINE | ID: mdl-32739457

In the elderly with cytopenia, the diagnosis of myelodysplastic syndrome (MDS) may be missed. Cytokine levels contribute to the pathology of MDS. Hence, the objectives were to evaluate cytogenetic profile as a prognostic indicator in risk stratification and cytokine levels as a screening tool in patients with cytopenia for diagnosis. Over 2 years (2016-2018), 150 elderly patients were screened. MDS diagnosis was confirmed by morphology. Interleukin-2 (IL-2) and IL-6 levels were assessed in 50 patients, and karyotyping was performed in 20 confirmed cases of MDS. Age-matched healthy controls were used for comparison of cytokine levels. Among 150 patients, 88.6% had anemia, including nutritional anemia (51.2%). MDS diagnosis was confirmed in 35 patients. In 15 patients, unexplained cytopenia (UC) was present. Karyotyping in 20 MDS patients was normal in 15 (75%) patients and revealed a complex karyotype in four (20%) patients and double chromosomal abnormality in one (5%) patient. The Revised International Prognostic Scoring System (IPSS-R) scored 91% in the low-risk group and 9% (n = 3) in the high-risk group; the latter three developed acute myeloid leukemia (AML) and two of them had a 7q deletion. Among the 15 cases of UC, one patient died from refractory anemia. No significant difference in levels of IL-2 and IL-6 were found between MDS and UC patients when compared with healthy controls, as well as between different risk groups and karyotypes. A significant difference in IL-2 levels was found in MDS patients with disease progression and with stable disease. On the basis of the findings, it is suggested that IL-2 levels will help in predicting disease progression.


Anemia/genetics , Biomarkers, Tumor/genetics , Interleukin-2/genetics , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/genetics , Williams Syndrome/genetics , Aged , Aged, 80 and over , Anemia/blood , Anemia/diagnosis , Anemia/pathology , Biomarkers, Tumor/blood , Case-Control Studies , Cross-Sectional Studies , Disease Progression , Female , Gene Expression , Humans , Interleukin-2/blood , Interleukin-6/blood , Interleukin-6/genetics , Karyotype , Karyotyping , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/pathology , Williams Syndrome/blood , Williams Syndrome/diagnosis , Williams Syndrome/pathology
10.
Am J Med Genet A ; 182(9): 2102-2109, 2020 09.
Article En | MEDLINE | ID: mdl-32706523

To describe special facial features of children with Williams syndrome in China by using method of three-dimensional craniofacial anthropometry. Using three-dimensional stereo photogrammetric device, 14 craniofacial anthropometric measurements were performed and five indices were calculated in 52 children with Williams syndrome and 208 age and sex matched controls of Han Chinese ethnicity. Except intercanthal width, mouth breadth, morphological face height, nasal height-breadth index, nasal breadth-depth index, morphological ear index, the Williams syndrome group under 3 years old were smaller than the control group in the other 12 variables. Compared with the control group, the Williams syndrome group aged 3-5 years old had smaller biocular breadth, nasal length, nasorostral angle, bitragal breadth, ear width, morphological ear index and face depth. The Williams syndrome group aged above 6 years old had smaller biocular breadth, nasal breadth, bitragal breadth, ear width, ear length and face depth than the control group. The craniofacial variability index of the Williams syndrome group was greater than the control group. Greater variation was found among children with Williams syndrome than normal in China, specifically at eye, nose, ear and face shape, which demonstrate the usefulness of three-dimensional stereo photogrammetric analysis in supporting accurate diagnose of the patient with Williams syndrome.


Anthropometry , Face/abnormalities , Skull/abnormalities , Williams Syndrome/genetics , Asian People/genetics , Cephalometry , Child , Child, Preschool , China/epidemiology , Ear/abnormalities , Ear/pathology , Face/pathology , Female , Humans , Male , Nose/abnormalities , Nose/pathology , Skull/pathology , Williams Syndrome/pathology
11.
Pediatr Cardiol ; 41(6): 1199-1205, 2020 Aug.
Article En | MEDLINE | ID: mdl-32474735

The aim of this study was to compare the size and geometry of the aorta in patients with 7q11.23 duplication (Dup7) to healthy controls. We retrospectively reviewed all echocardiograms in all patients with Dup7 evaluated at our institutions from June 2017 through September 2019. All standard aortic diameter measurements were made and recorded. Z-scores for the measurements were calculated. For comparison, a set of control echocardiograms was developed by randomly selecting 24 normal echocardiograms in age-matched patients who had undergone echocardiograms for an indication of either chest pain or syncope. In 58 echocardiograms from 21 Dup7 patients, all aortic measurements were increased compared to controls (p < 0.0001). Effacement of the sinotubular junction (STJ) of the aorta was present in all Dup7 patients. Our novel STJ-to-aortic annulus ratio of ≥ 1.15 had a 98.28% sensitivity (95% CI 90.76-99.96) and 100% specificity (95% CI 85.75-100) for distinguishing Dup7 from controls with a positive predictive value of 100% and a negative predictive value of 96.00% (95% CI 77.47-99.41). All patients in our study with Dup7 had echocardiographic evidence of aortopathy. Effacement of the STJ was present in all Dup7 patients. The STJ-to-annulus ratio is a better indicator of aortopathy in Dup7 than the aortic Z-score.


Aorta/pathology , Williams Syndrome/pathology , Adolescent , Aorta/diagnostic imaging , Child , Child, Preschool , Echocardiography , Female , Humans , Male , Retrospective Studies , Sensitivity and Specificity , Sinus of Valsalva/diagnostic imaging , Sinus of Valsalva/pathology , Williams Syndrome/diagnostic imaging
12.
Hum Mol Genet ; 29(9): 1498-1519, 2020 06 03.
Article En | MEDLINE | ID: mdl-32313931

Gtf2ird1 and Gtf2i are two transcription factors (TFs) among the 28 genes deleted in Williams syndrome, and prior mouse models of each TF show behavioral phenotypes. Here we identify their genomic binding sites in the developing brain and test for additive effects of their mutation on transcription and behavior. GTF2IRD1 binding targets were enriched for transcriptional and chromatin regulators and mediators of ubiquitination. GTF2I targets were enriched for signal transduction proteins, including regulators of phosphorylation and WNT. Both TFs are highly enriched at promoters, strongly overlap CTCF binding and topological associating domain boundaries and moderately overlap each other, suggesting epistatic effects. Shared TF targets are enriched for reactive oxygen species-responsive genes, synaptic proteins and transcription regulators such as chromatin modifiers, including a significant number of highly constrained genes and known ASD genes. We next used single and double mutants to test whether mutating both TFs will modify transcriptional and behavioral phenotypes of single Gtf2ird1 mutants, though with the caveat that our Gtf2ird1 mutants, like others previously reported, do produce low levels of a truncated protein product. Despite little difference in DNA binding and transcriptome-wide expression, homozygous Gtf2ird1 mutation caused balance, marble burying and conditioned fear phenotypes. However, mutating Gtf2i in addition to Gtf2ird1 did not further modify transcriptomic or most behavioral phenotypes, suggesting Gtf2ird1 mutation alone was sufficient for the observed phenotypes.


CCCTC-Binding Factor/genetics , Muscle Proteins/genetics , Trans-Activators/genetics , Transcription Factors, TFII/genetics , Williams Syndrome/genetics , Animals , Brain/metabolism , Brain/pathology , CRISPR-Cas Systems/genetics , DNA-Binding Proteins/genetics , Disease Models, Animal , Embryonic Development/genetics , Gene Editing , Gene Expression Regulation, Developmental/genetics , Humans , Mice , Transcription, Genetic/genetics , Williams Syndrome/pathology
14.
Rheumatol Int ; 40(8): 1309-1316, 2020 Aug.
Article En | MEDLINE | ID: mdl-32239321

The differential diagnosis in children with the systemic vasculopathy is still a challenge for clinicians. The progress in vascular imaging and the latest recommendations improve the diagnostic process, but only single reports describe the use of new imaging tests in children. The publication aims to demonstrate the important role of 18F-fluoro-2-deoxy-D-glucose (FDG) positron emission tomography combined with anatomical computed tomography angiography (PET/CTA) imaging in the case of a 15-year-old boy with chest pain, intermittent claudication, hypertension and features of middle aortic syndrome in computed tomography angiography (CTA). The patient was suspected to have Takayasu arteritis, but was finally diagnosed with Williams-Beuren syndrome. The case indicates that the FDG PET/CT imaging might be essential in the diagnostic process of middle aortic syndrome in children. We suggest that this imaging technique should be considered in the diagnostic process of systemic vasculopathy particularly in children.


Positron Emission Tomography Computed Tomography/methods , Williams Syndrome/diagnostic imaging , Adolescent , Diagnosis, Differential , Fluorodeoxyglucose F18/administration & dosage , Humans , Male , Radiopharmaceuticals/administration & dosage , Takayasu Arteritis/diagnosis , Williams Syndrome/pathology
15.
Brain Struct Funct ; 225(3): 1019-1032, 2020 Apr.
Article En | MEDLINE | ID: mdl-32189114

Williams syndrome (WS) is a rare neurodevelopmental disorder caused by the hemideletion of approximately 25-28 genes at 7q11.23. Its unusual social and cognitive phenotype is most strikingly characterized by the disinhibition of social behavior, in addition to reduced global IQ, with a relative sparing of language ability. Hypersociality and increased social approach behavior in WS may represent a unique inability to inhibit responses to specific social stimuli, which is likely associated with abnormalities of frontostriatal circuitry. The striatum is characterized by a diversity of interneuron subtypes, including inhibitory parvalbumin-positive interneurons (PV+) and excitatory cholinergic interneurons (Ch+). Animal model research has identified an important role for these specialized cells in regulating social approach behavior. Previous research in humans identified a depletion of interneuron subtypes associated with neuropsychiatric disorders. Here, we examined the density of PV+ and Ch+ interneurons in the striatum of 13 WS and neurotypical (NT) subjects. We found a significant reduction in the density of Ch+ interneurons in the medial caudate nucleus and nucleus accumbens, important regions receiving cortical afferents from the orbitofrontal and ventromedial prefrontal cortex, and circuitry involved in language and reward systems. No significant difference in the distribution of PV+ interneurons was found. The pattern of decreased Ch+ interneuron densities in WS differs from patterns of interneuron depletion found in other disorders.


Cholinergic Neurons/pathology , Corpus Striatum/pathology , Interneurons/pathology , Williams Syndrome/pathology , Adolescent , Adult , Aged , Choline O-Acetyltransferase/analysis , Female , Humans , Male , Middle Aged , Parvalbumins/analysis , Young Adult
16.
Mol Autism ; 11(1): 12, 2020 02 05.
Article En | MEDLINE | ID: mdl-32024554

BACKGROUND: Williams syndrome (WS) and autism spectrum disorder (ASD) are neurodevelopmental disorders that demonstrate overlapping genetic associations, dichotomous sociobehavioral phenotypes, and dichotomous pathological differences in neuronal distribution in key social brain areas, including the prefrontal cortex and the amygdala. The serotonergic system is critical to many processes underlying neurodevelopment and is additionally an important neuromodulator associated with behavioral variation. The amygdala is heavily innervated by serotonergic projections, suggesting that the serotonergic system is a significant mediator of neuronal activity. Disruptions to the serotonergic system, and atypical structure and function of the amygdala, are implicated in both WS and ASD. METHODS: We quantified the serotonergic axon density in the four major subdivisions of the amygdala in the postmortem brains of individuals diagnosed with ASD and WS and neurotypical (NT) brains. RESULTS: We found opposing directions of change in serotonergic innervation in the two disorders, with ASD displaying an increase in serotonergic axons compared to NT and WS displaying a decrease. Significant differences (p < 0.05) were observed between WS and ASD data sets across multiple amygdala nuclei. LIMITATIONS: This study is limited by the availability of human postmortem tissue. Small sample size is an unavoidable limitation of most postmortem human brain research and particularly postmortem research in rare disorders. CONCLUSIONS: Differential alterations to serotonergic innervation of the amygdala may contribute to differences in sociobehavioral phenotype in WS and ASD. These findings will inform future work identifying targets for future therapeutics in these and other disorders characterized by atypical social behavior.


Amygdala/pathology , Autism Spectrum Disorder/pathology , Axons/pathology , Serotonin , Williams Syndrome/pathology , Adolescent , Adult , Aged , Female , Humans , Infant , Male , Middle Aged , Young Adult
17.
Mol Neurobiol ; 57(2): 765-777, 2020 Feb.
Article En | MEDLINE | ID: mdl-31471877

Williams-Beuren syndrome (WBS) is a rare neurodevelopmental disorder characterized by moderate intellectual disability and learning difficulties alongside behavioral abnormalities such as hypersociability. Several structural and functional brain alterations are characteristic of this syndrome, as well as disturbed sleep and sleeping patterns. However, the detailed physiological mechanisms underlying WBS are mostly unknown. Here, we characterized the cortical dynamics in a mouse model of WBS previously reported to replicate most of the behavioral alterations described in humans. We recorded the laminar local field potential generated in the frontal cortex during deep anesthesia and characterized the properties of the emergent slow oscillation activity. Moreover, we performed micro-electrocorticogram recordings using multielectrode arrays covering the cortical surface of one hemisphere. We found significant differences between the cortical emergent activity and functional connectivity between wild-type mice and WBS model mice. Slow oscillations displayed Up states with diminished firing rate and lower high-frequency content in the gamma range. Lower firing rates were also recorded in the awake WBS animals while performing a marble burying task and could be associated with the decreased spine density and thus synaptic connectivity in this cortical area. We also found an overall increase in functional connectivity between brain areas, reflected in lower clustering and abnormally high integration, especially in the gamma range. These results expand previous findings in humans, suggesting that the cognitive deficits characterizing WBS might be associated with reduced excitability, plus an imbalance in the capacity to functionally integrate and segregate information.


Neocortex/pathology , Williams Syndrome/pathology , Animals , Dendritic Spines/metabolism , Disease Models, Animal , Male , Mice, Inbred C57BL , Neocortex/physiopathology , Nerve Net/pathology , Nerve Net/physiopathology , Wakefulness , Williams Syndrome/physiopathology
18.
Prog Brain Res ; 250: 109-127, 2019.
Article En | MEDLINE | ID: mdl-31703898

The prefrontal cortex consists of several cytoarchitectonically defined areas that are involved in higher-order cognitive and emotional processing. The areas are highly variable in terms of organization of cortical layers and distribution of specific neuronal classes, and are affected in neurodevelopmental and psychiatric disorders. Here the focus is on microstructural anatomical characteristics of human prefrontal cortex in an evolutionary context with special emphasis on Williams syndrome. We include a pilot analysis of distribution of neurons labeled with an antibody to non-phosphorylated neurofilament protein (SMI-32) in the frontal pole of Williams syndrome to further examine microstructural characteristics of the prefrontal cortex in Williams syndrome and implications of the distribution of SMI-32 immunoreactive neurons for connectivity between the frontal pole and other cortical areas in the disorder.


Biological Evolution , Nerve Net/anatomy & histology , Neurodevelopmental Disorders/pathology , Prefrontal Cortex/anatomy & histology , Williams Syndrome/pathology , Humans , Nerve Net/cytology , Nerve Net/pathology , Prefrontal Cortex/cytology , Prefrontal Cortex/pathology
19.
Hum Mol Genet ; 28(20): 3443-3465, 2019 10 15.
Article En | MEDLINE | ID: mdl-31418010

Williams syndrome (WS) is a neurodevelopmental disorder caused by a 1.5-1.8 Mbp deletion on chromosome 7q11.23, affecting the copy number of 26-28 genes. Phenotypes of WS include cardiovascular problems, craniofacial dysmorphology, deficits in visual-spatial cognition and a characteristic hypersocial personality. There are still no genes in the region that have been consistently linked to the cognitive and behavioral phenotypes, although human studies and mouse models have led to the current hypothesis that the general transcription factor 2 I family of genes, GTF2I and GTF2IRD1, are responsible. Here we test the hypothesis that these two transcription factors are sufficient to reproduce the phenotypes that are caused by deletion of the WS critical region (WSCR). We compare a new mouse model with loss of function mutations in both Gtf2i and Gtf2ird1 to an established mouse model lacking the complete WSCR. We show that the complete deletion (CD) model has deficits across several behavioral domains including social communication, motor functioning and conditioned fear that are not explained by loss of function mutations in Gtf2i and Gtf2ird1. Furthermore, transcriptome profiling of the hippocampus shows changes in synaptic genes in the CD model that are not seen in the double mutants. Thus, we have thoroughly defined a set of molecular and behavioral consequences of complete WSCR deletion and shown that genes or combinations of genes beyond Gtf2i and Gtf2ird1 are necessary to produce these phenotypic effects.


Muscle Proteins/genetics , Mutation/genetics , Trans-Activators/genetics , Transcription Factors, TFII/genetics , Williams Syndrome/genetics , Williams Syndrome/pathology , Animals , Female , Hippocampus/metabolism , Male , Mice , Phenotype , Vocalization, Animal/physiology
20.
J Med Genet ; 56(12): 801-808, 2019 12.
Article En | MEDLINE | ID: mdl-31413120

BACKGROUND: The hallmark of the neurobehavioural phenotype of Williams-Beuren syndrome (WBS) is increased sociability and relatively preserved language skills, often described as opposite to autism spectrum disorders (ASD). However, the prevalence of ASD in WBS is 6-10 times higher than in the general population. We have investigated the genetic factors that could contribute to the ASD phenotype in individuals with WBS. METHODS: We studied four males and four females with WBS and a confirmed diagnosis of ASD by the Autism Diagnostic Interview-Revised. We performed a detailed molecular characterisation of the deletion and searched for genomic variants using exome sequencing. RESULTS: A de novo deletion of 1.55 Mb (6 cases) or 1.83 Mb (2 cases) at 7q11.23 was detected, being in 7/8 patients of paternal origin. No common breakpoint, deletion mechanism or size was found. Two cases were hemizygous for the rare T allele at rs12539160 in MLXIPL, previously associated with ASD. Inherited rare variants in ASD-related or functionally constrained genes and a de novo nonsense mutation in the UBR5 gene were identified in six cases, with higher burden in females compared with males (p=0.016). CONCLUSIONS: The increased susceptibility to ASD in patients with WBS might be due to additive effects of the common WBS deletion, inherited and de novo rare sequence variants in ASD-related genes elsewhere in the genome, with higher burden of deleterious mutations required for females, and possible hypomorphic variants in the hemizygous allele or cis-acting mechanisms on imprinting.


Autism Spectrum Disorder/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics , Williams Syndrome/genetics , Alleles , Autism Spectrum Disorder/pathology , Child , Child, Preschool , Chromosome Deletion , Female , Genetic Predisposition to Disease , Genome, Human/genetics , Hemizygote , Humans , Infant , Male , Phenotype , Exome Sequencing , Williams Syndrome/pathology
...