Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 737
1.
Top Magn Reson Imaging ; 33(3): e0312, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38836588

BACKGROUND: Altered size in the corpus callosum (CC) has been reported in individuals with autism spectrum disorder (ASD), but few studies have investigated younger children. Moreover, knowledge about the age-related changes in CC size in individuals with ASD is limited. OBJECTIVES: Our objective was to investigate the age-related size of the CC and compare them with age-matched healthy controls between the ages of 2 and 18 years. METHODS: Structural-weighted images were acquired in 97 male patients diagnosed with ASD; published data were used for the control group. The CC was segmented into 7 distinct subregions (rostrum, genu, rostral body, anterior midbody, posterior midbody, isthmus, and splenium) as per Witelson's technique using ITK-SNAP software. We calculated both the total length and volume of the CC as well as the length and height of its 7 subregions. The length of the CC measures was studied as both continuous and categorical forms. For the continuous form, Pearson's correlation was used, while categorical forms were based on age ranges reflecting brain expansion during early postnatal years. Differences in CC measures between adjacent age groups in individuals with ASD were assessed using a Student t-test. Mean and standard deviation scores were compared between ASD and control groups using the Welch t-test. RESULTS: Age showed a moderate positive association with the total length of the CC (r = 0.43; Padj = 0.003) among individuals with ASD. Among the subregions, a positive association was observed only in the anterior midbody of the CC (r = 0.41; Padj = 0.01). No association was found between the age and the height of individual subregions or with the total volume of the CC. In comparison with healthy controls, individuals with ASD exhibited shorter lengths and heights of the genu and splenium of the CC across wide age ranges. CONCLUSION: Overall, our results highlight a distinct abnormal developmental trajectory of CC in ASD, particularly in the genu and splenium structures, potentially reflecting underlying pathophysiological mechanisms that warrant further investigation.


Autism Spectrum Disorder , Corpus Callosum , Magnetic Resonance Imaging , Humans , Male , Corpus Callosum/diagnostic imaging , Corpus Callosum/pathology , Autism Spectrum Disorder/diagnostic imaging , Autism Spectrum Disorder/pathology , Child , Adolescent , Child, Preschool , Female , Image Processing, Computer-Assisted
2.
Cereb Cortex ; 34(13): 146-160, 2024 May 02.
Article En | MEDLINE | ID: mdl-38696608

Autism spectrum disorder is a neurodevelopmental disability that includes sensory disturbances. Hearing is frequently affected and ranges from deafness to hypersensitivity. In utero exposure to the antiepileptic valproic acid is associated with increased risk of autism spectrum disorder in humans and timed valproic acid exposure is a biologically relevant and validated animal model of autism spectrum disorder. Valproic acid-exposed rats have fewer neurons in their auditory brainstem and thalamus, fewer calbindin-positive neurons, reduced ascending projections to the midbrain and thalamus, elevated thresholds, and delayed auditory brainstem responses. Additionally, in the auditory cortex, valproic acid exposure results in abnormal responses, decreased phase-locking, elevated thresholds, and abnormal tonotopic maps. We therefore hypothesized that in utero, valproic acid exposure would result in fewer neurons in auditory cortex, neuronal dysmorphology, fewer calbindin-positive neurons, and reduced connectivity. We approached this hypothesis using morphometric analyses, immunohistochemistry, and retrograde tract tracing. We found thinner cortical layers but no changes in the density of neurons, smaller pyramidal and non-pyramidal neurons in several regions, fewer neurons immunoreactive for calbindin-positive, and fewer cortical neurons projecting to the inferior colliculus. These results support the widespread impact of the auditory system in autism spectrum disorder and valproic acid-exposed animals and emphasize the utility of simple, noninvasive auditory screening for autism spectrum disorder.


Auditory Cortex , Autism Spectrum Disorder , Calbindins , Disease Models, Animal , Valproic Acid , Animals , Autism Spectrum Disorder/pathology , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/chemically induced , Valproic Acid/toxicity , Female , Calbindins/metabolism , Auditory Cortex/pathology , Auditory Cortex/drug effects , Auditory Cortex/metabolism , Pregnancy , Neurons/pathology , Neurons/metabolism , Rats , Male , Auditory Pathways/pathology , Auditory Pathways/drug effects , Prenatal Exposure Delayed Effects/pathology , Rats, Sprague-Dawley , Anticonvulsants
3.
Cereb Cortex ; 34(13): 121-128, 2024 May 02.
Article En | MEDLINE | ID: mdl-38696601

Previous studies in autism spectrum disorder demonstrated an increased number of excitatory pyramidal cells and a decreased number of inhibitory parvalbumin+ chandelier interneurons in the prefrontal cortex of postmortem brains. How these changes in cellular composition affect the overall abundance of excitatory and inhibitory synapses in the cortex is not known. Herein, we quantified the number of excitatory and inhibitory synapses in the prefrontal cortex of 10 postmortem autism spectrum disorder brains and 10 control cases. To identify excitatory synapses, we used VGlut1 as a marker of the presynaptic component and postsynaptic density protein-95 as marker of the postsynaptic component. To identify inhibitory synapses, we used the vesicular gamma-aminobutyric acid transporter as a marker of the presynaptic component and gephyrin as a marker of the postsynaptic component. We used Puncta Analyzer to quantify the number of co-localized pre- and postsynaptic synaptic components in each area of interest. We found an increase in the number of excitatory synapses in upper cortical layers and a decrease in inhibitory synapses in all cortical layers in autism spectrum disorder brains compared with control cases. The alteration in the number of excitatory and inhibitory synapses could lead to neuronal dysfunction and disturbed network connectivity in the prefrontal cortex in autism spectrum disorder.


Membrane Proteins , Prefrontal Cortex , Synapses , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Humans , Male , Female , Synapses/pathology , Synapses/metabolism , Adult , Middle Aged , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/pathology , Young Adult , Adolescent , Child , Autistic Disorder/metabolism , Autistic Disorder/pathology , Neural Inhibition/physiology , Vesicular Glutamate Transport Protein 1/metabolism
4.
Cereb Cortex ; 34(13): 94-103, 2024 May 02.
Article En | MEDLINE | ID: mdl-38696597

Autism (or autism spectrum disorder) was initially defined as a psychiatric disorder, with the likely cause maternal behavior (the very destructive "refrigerator mother" theory). It took several decades for research into brain mechanisms to become established. Both neuropathological and imaging studies found differences in the cerebellum in autism spectrum disorder, the most widely documented being a decreased density of Purkinje cells in the cerebellar cortex. The popular interpretation of these results is that cerebellar neuropathology is a critical cause of autism spectrum disorder. We challenge that view by arguing that if fewer Purkinje cells are critical for autism spectrum disorder, then any condition that causes the loss of Purkinje cells should also cause autism spectrum disorder. We will review data on damage to the cerebellum from cerebellar lesions, tumors, and several syndromes (Joubert syndrome, Fragile X, and tuberous sclerosis). Collectively, these studies raise the question of whether the cerebellum really has a role in autism spectrum disorder. Autism spectrum disorder is now recognized as a genetically caused developmental disorder. A better understanding of the genes that underlie the differences in brain development that result in autism spectrum disorder is likely to show that these genes affect the development of the cerebellum in parallel with the development of the structures that do underlie autism spectrum disorder.


Cerebellum , Humans , Cerebellum/pathology , Autism Spectrum Disorder/pathology , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/physiopathology , Autism Spectrum Disorder/diagnostic imaging , Animals , Autistic Disorder/pathology , Autistic Disorder/genetics , Autistic Disorder/physiopathology , Purkinje Cells/pathology
5.
Sci Transl Med ; 16(749): eadh9974, 2024 May 29.
Article En | MEDLINE | ID: mdl-38781321

Many psychiatric disorders exhibit sex differences, but the underlying mechanisms remain poorly understood. We analyzed transcriptomics data from 2160 postmortem adult prefrontal cortex brain samples from the PsychENCODE consortium in a sex-stratified study design. We compared transcriptomics data of postmortem brain samples from patients with schizophrenia (SCZ), bipolar disorder (BD), and autism spectrum disorder (ASD) with transcriptomics data of postmortem control brains from individuals without a known history of psychiatric disease. We found that brain samples from females with SCZ, BD, and ASD showed a higher burden of transcriptomic dysfunction than did brain samples from males with these disorders. This observation was supported by the larger number of differentially expressed genes (DEGs) and a greater magnitude of gene expression changes observed in female versus male brain specimens. In addition, female patient brain samples showed greater overall connectivity dysfunction, defined by a higher proportion of gene coexpression modules with connectivity changes and higher connectivity burden, indicating a greater degree of gene coexpression variability. We identified several gene coexpression modules enriched in sex-biased DEGs and identified genes from a genome-wide association study that were involved in immune and synaptic functions across different brain cell types. We found a number of genes as hubs within these modules, including those encoding SCN2A, FGF14, and C3. Our results suggest that in the context of psychiatric diseases, males and females exhibit different degrees of transcriptomic dysfunction and implicate immune and synaptic-related pathways in these sex differences.


Autopsy , Brain , Mental Disorders , Sex Characteristics , Transcriptome , Humans , Female , Male , Transcriptome/genetics , Brain/metabolism , Brain/pathology , Mental Disorders/genetics , Mental Disorders/pathology , Bipolar Disorder/genetics , Bipolar Disorder/metabolism , Bipolar Disorder/pathology , Schizophrenia/genetics , Schizophrenia/metabolism , Schizophrenia/pathology , Gene Expression Profiling , Genome-Wide Association Study , Adult , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/pathology , Gene Regulatory Networks , Middle Aged
6.
Mol Autism ; 15(1): 22, 2024 May 25.
Article En | MEDLINE | ID: mdl-38790065

BACKGROUND: Social affective and communication symptoms are central to autism spectrum disorder (ASD), yet their severity differs across toddlers: Some toddlers with ASD display improving abilities across early ages and develop good social and language skills, while others with "profound" autism have persistently low social, language and cognitive skills and require lifelong care. The biological origins of these opposite ASD social severity subtypes and developmental trajectories are not known. METHODS: Because ASD involves early brain overgrowth and excess neurons, we measured size and growth in 4910 embryonic-stage brain cortical organoids (BCOs) from a total of 10 toddlers with ASD and 6 controls (averaging 196 individual BCOs measured/subject). In a 2021 batch, we measured BCOs from 10 ASD and 5 controls. In a 2022 batch, we  tested replicability of BCO size and growth effects by generating and measuring an independent batch of BCOs from 6 ASD and 4 control subjects. BCO size was analyzed within the context of our large, one-of-a-kind social symptom, social attention, social brain and social and language psychometric normative datasets ranging from N = 266 to N = 1902 toddlers. BCO growth rates were examined by measuring size changes between 1- and 2-months of organoid development. Neurogenesis markers at 2-months were examined at the cellular level. At the molecular level, we measured activity and expression of Ndel1; Ndel1 is a prime target for cell cycle-activated kinases; known to regulate cell cycle, proliferation, neurogenesis, and growth; and known to be involved in neuropsychiatric conditions. RESULTS: At the BCO level, analyses showed BCO size was significantly enlarged by 39% and 41% in ASD in the 2021 and 2022 batches. The larger the embryonic BCO size, the more severe the ASD social symptoms. Correlations between BCO size and social symptoms were r = 0.719 in the 2021 batch and r = 0. 873 in the replication 2022 batch. ASD BCOs grew at an accelerated rate nearly 3 times faster than controls. At the cell level, the two largest ASD BCOs had accelerated neurogenesis. At the molecular level, Ndel1 activity was highly correlated with the growth rate and size of BCOs. Two BCO subtypes were found in ASD toddlers: Those in one subtype had very enlarged BCO size with accelerated rate of growth and neurogenesis; a profound autism clinical phenotype displaying severe social symptoms, reduced social attention, reduced cognitive, very low language and social IQ; and substantially altered growth in specific cortical social, language and sensory regions. Those in a second subtype had milder BCO enlargement and milder social, attention, cognitive, language and cortical differences. LIMITATIONS: Larger samples of ASD toddler-derived BCO and clinical phenotypes may reveal additional ASD embryonic subtypes. CONCLUSIONS: By embryogenesis, the biological bases of two subtypes of ASD social and brain development-profound autism and mild autism-are already present and measurable and involve dysregulated cell proliferation and accelerated neurogenesis and growth. The larger the embryonic BCO size in ASD, the more severe the toddler's social symptoms and the more reduced the social attention, language ability, and IQ, and the more atypical the growth of social and language brain regions.


Autism Spectrum Disorder , Organoids , Humans , Autism Spectrum Disorder/pathology , Autism Spectrum Disorder/physiopathology , Organoids/pathology , Male , Female , Child, Preschool , Cerebral Cortex/pathology , Social Behavior , Organ Size , Infant , Severity of Illness Index , Brain/pathology
7.
Genes (Basel) ; 15(4)2024 Mar 28.
Article En | MEDLINE | ID: mdl-38674358

Pathogenic ASH1L variants have been reported in probands with broad phenotypic presentations, including intellectual disability, autism spectrum disorder, attention deficit hyperactivity disorder, seizures, congenital anomalies, and other skeletal, muscular, and sleep differences. Here, we review previously published individuals with pathogenic ASH1L variants and report three further probands with novel ASH1L variants and previously unreported phenotypic features, including mixed receptive language disorder and gait disturbances. These novel data from the Brain Gene Registry, an accessible repository of clinically derived genotypic and phenotypic data, have allowed for the expansion of the phenotypic and genotypic spectrum of this condition.


Histone-Lysine N-Methyltransferase , Neurodevelopmental Disorders , Phenotype , Humans , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Male , Histone-Lysine N-Methyltransferase/genetics , Female , Child , Genotype , DNA-Binding Proteins/genetics , Intellectual Disability/genetics , Intellectual Disability/pathology , Transcription Factors/genetics , Child, Preschool , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Mutation , Adolescent
8.
Nat Neurosci ; 27(6): 1075-1086, 2024 Jun.
Article En | MEDLINE | ID: mdl-38649755

Human brain organization involves the coordinated expression of thousands of genes. For example, the first principal component (C1) of cortical transcription identifies a hierarchy from sensorimotor to association regions. In this study, optimized processing of the Allen Human Brain Atlas revealed two new components of cortical gene expression architecture, C2 and C3, which are distinctively enriched for neuronal, metabolic and immune processes, specific cell types and cytoarchitectonics, and genetic variants associated with intelligence. Using additional datasets (PsychENCODE, Allen Cell Atlas and BrainSpan), we found that C1-C3 represent generalizable transcriptional programs that are coordinated within cells and differentially phased during fetal and postnatal development. Autism spectrum disorder and schizophrenia were specifically associated with C1/C2 and C3, respectively, across neuroimaging, differential expression and genome-wide association studies. Evidence converged especially in support of C3 as a normative transcriptional program for adolescent brain development, which can lead to atypical supragranular cortical connectivity in people at high genetic risk for schizophrenia.


Cerebral Cortex , Schizophrenia , Transcriptome , Humans , Schizophrenia/genetics , Schizophrenia/pathology , Cerebral Cortex/growth & development , Cerebral Cortex/pathology , Cerebral Cortex/metabolism , Female , Male , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Adolescent , Autistic Disorder/genetics , Autistic Disorder/pathology , Genome-Wide Association Study , Child , Adult , Neuroimaging/methods
9.
Cell Rep Med ; 5(5): 101534, 2024 May 21.
Article En | MEDLINE | ID: mdl-38670100

Thalamocortical (TC) circuits are essential for sensory information processing. Clinical and preclinical studies of autism spectrum disorders (ASDs) have highlighted abnormal thalamic development and TC circuit dysfunction. However, mechanistic understanding of how TC dysfunction contributes to behavioral abnormalities in ASDs is limited. Here, our study on a Shank3 mouse model of ASD reveals TC neuron hyperexcitability with excessive burst firing and a temporal mismatch relationship with slow cortical rhythms during sleep. These TC electrophysiological alterations and the consequent sensory hypersensitivity and sleep fragmentation in Shank3 mutant mice are causally linked to HCN2 channelopathy. Restoring HCN2 function early in postnatal development via a viral approach or lamotrigine (LTG) ameliorates sensory and sleep problems. A retrospective case series also supports beneficial effects of LTG treatment on sensory behavior in ASD patients. Our study identifies a clinically relevant circuit mechanism and proposes a targeted molecular intervention for ASD-related behavioral impairments.


Autism Spectrum Disorder , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Nerve Tissue Proteins , Thalamus , Animals , Thalamus/metabolism , Thalamus/pathology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Mice , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/physiopathology , Autism Spectrum Disorder/pathology , Lamotrigine/pharmacology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Channelopathies/genetics , Channelopathies/metabolism , Channelopathies/pathology , Humans , Disease Models, Animal , Male , Neurons/metabolism , Female , Mice, Inbred C57BL , Mutation/genetics , Sleep/physiology , Sleep/drug effects , Sleep/genetics , Potassium Channels
10.
Int J Dev Neurosci ; 84(4): 314-327, 2024 Jun.
Article En | MEDLINE | ID: mdl-38584149

Autism spectrum disorder (ASD) is a fast-growing neurodevelopmental disorder throughout the world. Experiencing early life stresses (ELS) like maternal separation (MS) is associated with autistic-like behaviors. It has been proposed that disturbance in the gut-brain axis-mediated psychiatric disorders following MS. The role of disruption in the integrity of gut-brain barrier in ASD remains unclear. Addressing this knowledge gap, in this study we aimed to investigate role of the gut-brain barrier integrity in mediating autistic-like behaviors in mouse models of MS stress. To do this, mice neonates are separated daily from their mothers from postnatal day (PND) 2 to PND 14 for 3 hours. During PND58-60, behavioral tests related to autistic-like behaviors including three-chamber sociability, shuttle box, and resident-intruder tests were performed. Then, prefrontal cortex (PFC), hippocampus, and colon samples were dissected out for histopathological and molecular evaluations. Results showed that MS is associated with impaired sociability and social preference indexes, aggressive behaviors, and impaired passive avoidance memory. The gene expression of CLDN1 decreased in the colon, and the gene expression of CLDN5, CLDN12, and MMP9 increased in the PFC of the MS mice. MS is associated with decrease in the diameter of CA1 and CA3 areas of the hippocampus. In addition, MS led to histopathological changes in the colon. We concluded that, probably, disturbance in the gut-brain barrier integrities mediated the autistic-like behavior in MS stress in mice.


Disease Models, Animal , Maternal Deprivation , Stress, Psychological , Animals , Mice , Stress, Psychological/pathology , Brain-Gut Axis/physiology , Female , Behavior, Animal/physiology , Male , Hippocampus/pathology , Hippocampus/metabolism , Prefrontal Cortex/pathology , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Autism Spectrum Disorder/physiopathology , Social Behavior , Autistic Disorder/pathology , Autistic Disorder/genetics , Autistic Disorder/physiopathology , Blood-Brain Barrier/pathology , Animals, Newborn , Colon/pathology
11.
Neuroimage ; 291: 120595, 2024 May 01.
Article En | MEDLINE | ID: mdl-38554782

Multimodal magnetic resonance imaging (MRI) provides complementary information for investigating brain structure and function; for example, an in vivo microstructure-sensitive proxy can be estimated using the ratio between T1- and T2-weighted structural MRI. However, acquiring multiple imaging modalities is challenging in patients with inattentive disorders. In this study, we proposed a comprehensive framework to provide multiple imaging features related to the brain microstructure using only T1-weighted MRI. Our toolbox consists of (i) synthesizing T2-weighted MRI from T1-weighted MRI using a conditional generative adversarial network; (ii) estimating microstructural features, including intracortical covariance and moment features of cortical layer-wise microstructural profiles; and (iii) generating a microstructural gradient, which is a low-dimensional representation of the intracortical microstructure profile. We trained and tested our toolbox using T1- and T2-weighted MRI scans of 1,104 healthy young adults obtained from the Human Connectome Project database. We found that the synthesized T2-weighted MRI was very similar to the actual image and that the synthesized data successfully reproduced the microstructural features. The toolbox was validated using an independent dataset containing healthy controls and patients with episodic migraine as well as the atypical developmental condition of autism spectrum disorder. Our toolbox may provide a new paradigm for analyzing multimodal structural MRI in the neuroscience community and is openly accessible at https://github.com/CAMIN-neuro/GAN-MAT.


Autism Spectrum Disorder , Connectome , Humans , Autism Spectrum Disorder/diagnostic imaging , Autism Spectrum Disorder/pathology , Magnetic Resonance Imaging/methods , Brain/diagnostic imaging , Brain/pathology , Multimodal Imaging , Image Processing, Computer-Assisted/methods
12.
Neurobiol Dis ; 194: 106470, 2024 May.
Article En | MEDLINE | ID: mdl-38485094

Pathogenic variants in KCNB1 are associated with a neurodevelopmental disorder spectrum that includes global developmental delays, cognitive impairment, abnormal electroencephalogram (EEG) patterns, and epilepsy with variable age of onset and severity. Additionally, there are prominent behavioral disturbances, including hyperactivity, aggression, and features of autism spectrum disorder. The most frequently identified recurrent variant is KCNB1-p.R306C, a missense variant located within the S4 voltage-sensing transmembrane domain. Individuals with the R306C variant exhibit mild to severe developmental delays, behavioral disorders, and a diverse spectrum of seizures. Previous in vitro characterization of R306C described altered sensitivity and cooperativity of the voltage sensor and impaired capacity for repetitive firing of neurons. Existing Kcnb1 mouse models include dominant negative missense variants, as well as knockout and frameshifts alleles. While all models recapitulate key features of KCNB1 encephalopathy, mice with dominant negative alleles were more severely affected. In contrast to existing loss-of-function and dominant-negative variants, KCNB1-p.R306C does not affect channel expression, but rather affects voltage-sensing. Thus, modeling R306C in mice provides a novel opportunity to explore impacts of a voltage-sensing mutation in Kcnb1. Using CRISPR/Cas9 genome editing, we generated the Kcnb1R306C mouse model and characterized the molecular and phenotypic effects. Consistent with the in vitro studies, neurons from Kcnb1R306C mice showed altered excitability. Heterozygous and homozygous R306C mice exhibited hyperactivity, altered susceptibility to chemoconvulsant-induced seizures, and frequent, long runs of slow spike wave discharges on EEG, reminiscent of the slow spike and wave activity characteristic of Lennox Gastaut syndrome. This novel model of channel dysfunction in Kcnb1 provides an additional, valuable tool to study KCNB1 encephalopathies. Furthermore, this allelic series of Kcnb1 mouse models will provide a unique platform to evaluate targeted therapies.


Autism Spectrum Disorder , Brain Diseases , Epilepsy , Animals , Mice , Autism Spectrum Disorder/pathology , Brain Diseases/pathology , Epilepsy/pathology , Mutation , Phenotype , Seizures
13.
Int J Dev Neurosci ; 84(3): 163-176, 2024 May.
Article En | MEDLINE | ID: mdl-38488315

INTRODUCTION: Recent research indicates that some brain structures show alterations in conditions such as Autism Spectrum Disorder (ASD). Among them, are the basal ganglia that are involved in motor, cognitive and behavioral neural circuits. OBJECTIVE: Review the literature that describes possible volumetric alterations in the basal ganglia of individuals with ASD and the impacts that these changes have on the severity of the condition. METHODOLOGY: This systematic review was registered in the design and reported according to the PRISMA Items and registered in PROSPERO (CRD42023394787). The study analyzed data from published clinical, case-contemplate, and cohort trials. The following databases were consulted: PubMed, Embase, Scopus, and Cochrane Central Register of Controlled Trials, using the Medical Subject Titles (MeSH) "Autism Spectrum Disorder" and "Basal Ganglia". The last search was carried out on February 28, 2023. RESULTS: Thirty-five eligible articles were collected, analyzed, and grouped according to the levels of alterations. CONCLUSION: The present study showed important volumetric alterations in the basal ganglia in ASD. However, the examined studies have methodological weaknesses that do not allow generalization and correlation with ASD manifestations.


Autism Spectrum Disorder , Basal Ganglia , Humans , Autism Spectrum Disorder/diagnostic imaging , Autism Spectrum Disorder/pathology , Autism Spectrum Disorder/physiopathology , Basal Ganglia/pathology , Basal Ganglia/diagnostic imaging
14.
Exp Neurol ; 376: 114756, 2024 Jun.
Article En | MEDLINE | ID: mdl-38508482

Overexpression of the Ube3a gene and the resulting increase in Ube3a protein are linked to autism spectrum disorder (ASD). However, the cellular and molecular processes underlying Ube3a-dependent ASD remain unclear. Using both male and female mice, we find that neurons in the somatosensory cortex of the Ube3a 2× Tg ASD mouse model display reduced dendritic spine density and increased immature filopodia density. Importantly, the increased gene dosage of Ube3a in astrocytes alone is sufficient to confer alterations in neurons as immature dendritic protrusions, as observed in primary hippocampal neuron cultures. We show that Ube3a overexpression in astrocytes leads to a loss of astrocyte-derived spinogenic protein, thrombospondin-2 (TSP2), due to a suppression of TSP2 gene transcription. By neonatal intraventricular injection of astrocyte-specific virus, we demonstrate that Ube3a overexpression in astrocytes in vivo results in a reduction in dendritic spine maturation in prelimbic cortical neurons, accompanied with autistic-like behaviors in mice. These findings reveal an astrocytic dominance in initiating ASD pathobiology at the neuronal and behavior levels. SIGNIFICANCE STATEMENT: Increased gene dosage of Ube3a is tied to autism spectrum disorders (ASDs), yet cellular and molecular alterations underlying autistic phenotypes remain unclear. We show that Ube3a overexpression leads to impaired dendritic spine maturation, resulting in reduced spine density and increased filopodia density. We find that dysregulation of spine development is not neuron autonomous, rather, it is mediated by an astrocytic mechanism. Increased gene dosage of Ube3a in astrocytes leads to reduced production of the spinogenic glycoprotein thrombospondin-2 (TSP2), leading to abnormalities in spines. Astrocyte-specific Ube3a overexpression in the brain in vivo confers dysregulated spine maturation concomitant with autistic-like behaviors in mice. These findings indicate the importance of astrocytes in aberrant neurodevelopment and brain function in Ube3a-depdendent ASD.


Autism Spectrum Disorder , Dendritic Spines , Neuroglia , Ubiquitin-Protein Ligases , Animals , Mice , Astrocytes/metabolism , Astrocytes/pathology , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Cells, Cultured , Dendritic Spines/pathology , Dendritic Spines/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis/physiology , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology , Somatosensory Cortex/metabolism , Somatosensory Cortex/pathology , Thrombospondins/metabolism , Thrombospondins/genetics , Thrombospondins/biosynthesis , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
15.
Am J Pathol ; 194(6): 1062-1077, 2024 Jun.
Article En | MEDLINE | ID: mdl-38492733

Autism spectrum disorder (ASD) is a prevalent neurodevelopmental disorder with a complex etiology. Recent evidence suggests that dopamine plays a crucial role in neural development. However, whether and how disrupted dopaminergic signaling during development contributes to ASD remains unknown. In this study, human brain RNA sequencing transcriptome analysis revealed a significant correlation between changes in dopaminergic signaling pathways and neural developmental signaling in ASD patients. In the zebrafish model, disrupted developmental dopaminergic signaling led to neural circuit abnormalities and behavior reminiscent of autism. Dopaminergic signaling may impact neuronal specification by potentially modulating integrins. These findings shed light on the mechanisms underlying the link between disrupted developmental dopamine signaling and ASD, and they point to the possibility of targeting dopaminergic signaling in early development for ASD treatment.


Autism Spectrum Disorder , Dopamine , Phenotype , Signal Transduction , Zebrafish , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Animals , Humans , Dopamine/metabolism , Brain/metabolism , Brain/pathology , Disease Models, Animal , Male , Neural Pathways/metabolism , Female , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology
16.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167097, 2024 Jun.
Article En | MEDLINE | ID: mdl-38408544

Zika virus (ZIKV) infection was first associated with Central Nervous System (CNS) infections in Brazil in 2015, correlated with an increased number of newborns with microcephaly, which ended up characterizing the Congenital Zika Syndrome (CZS). Here, we investigated the impact of ZIKV infection on the functionality of iPSC-derived astrocytes. Besides, we extrapolated our findings to a Brazilian cohort of 136 CZS children and validated our results using a mouse model. Interestingly, ZIKV infection in neuroprogenitor cells compromises cell migration and causes apoptosis but does not interfere in astrocyte generation. Moreover, infected astrocytes lost their ability to uptake glutamate while expressing more glutamate transporters and secreted higher levels of IL-6. Besides, infected astrocytes secreted factors that impaired neuronal synaptogenesis. Since these biological endophenotypes were already related to Autism Spectrum Disorder (ASD), we extrapolated these results to a cohort of children, now 6-7 years old, and found seven children with ASD diagnosis (5.14 %). Additionally, mice infected by ZIKV revealed autistic-like behaviors, with a significant increase of IL-6 mRNA levels in the brain. Considering these evidence, we inferred that ZIKV infection during pregnancy might lead to synaptogenesis impairment and neuroinflammation, which could increase the risk for ASD.


Astrocytes , Autism Spectrum Disorder , Neuroinflammatory Diseases , Synapses , Zika Virus Infection , Zika Virus , Zika Virus Infection/pathology , Zika Virus Infection/metabolism , Zika Virus Infection/virology , Zika Virus Infection/complications , Autism Spectrum Disorder/virology , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/pathology , Humans , Animals , Mice , Zika Virus/physiology , Female , Child , Synapses/metabolism , Synapses/pathology , Neuroinflammatory Diseases/virology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/etiology , Astrocytes/virology , Astrocytes/metabolism , Astrocytes/pathology , Male , Interleukin-6/metabolism , Interleukin-6/genetics , Pregnancy , Risk Factors , Induced Pluripotent Stem Cells/virology , Induced Pluripotent Stem Cells/metabolism , Brazil/epidemiology , Disease Models, Animal , Neurogenesis
17.
Neuroimage ; 288: 120534, 2024 Mar.
Article En | MEDLINE | ID: mdl-38340881

Autism spectrum disorder is a common neurodevelopmental condition that manifests as a disruption in sensory and social skills. Although it has been shown that the brain morphology of individuals with autism is asymmetric, how this differentially affects the structural connectome organization of each hemisphere remains under-investigated. We studied whole-brain structural connectivity-based brain asymmetry in individuals with autism using diffusion magnetic resonance imaging obtained from the Autism Brain Imaging Data Exchange initiative. By leveraging dimensionality reduction techniques, we constructed low-dimensional representations of structural connectivity and calculated their asymmetry index. Comparing the asymmetry index between individuals with autism and neurotypical controls, we found atypical structural connectome asymmetry in the sensory and default-mode regions, particularly showing weaker asymmetry towards the right hemisphere in autism. Network communication provided topological underpinnings by demonstrating that the inferior temporal cortex and limbic and frontoparietal regions showed reduced global network communication efficiency and decreased send-receive network navigation in the inferior temporal and lateral visual cortices in individuals with autism. Finally, supervised machine learning revealed that structural connectome asymmetry could be used as a measure for predicting communication-related autistic symptoms and nonverbal intelligence. Our findings provide insights into macroscale structural connectome alterations in autism and their topological underpinnings.


Autism Spectrum Disorder , Autistic Disorder , Connectome , Humans , Autistic Disorder/diagnostic imaging , Autism Spectrum Disorder/diagnostic imaging , Autism Spectrum Disorder/pathology , Magnetic Resonance Imaging/methods , Brain/diagnostic imaging , Brain/pathology
18.
Cereb Cortex ; 34(2)2024 01 31.
Article En | MEDLINE | ID: mdl-38220572

Autism spectrum disorder is a complex neurodevelopmental condition with diverse genetic and brain involvement. Despite magnetic resonance imaging advances, autism spectrum disorder diagnosis and understanding its neurogenetic factors remain challenging. We propose a dual-branch graph neural network that effectively extracts and fuses features from bimodalities, achieving 73.9% diagnostic accuracy. To explain the mechanism distinguishing autism spectrum disorder from healthy controls, we establish a perturbation model for brain imaging markers and perform a neuro-transcriptomic joint analysis using partial least squares regression and enrichment to identify potential genetic biomarkers. The perturbation model identifies brain imaging markers related to structural magnetic resonance imaging in the frontal, temporal, parietal, and occipital lobes, while functional magnetic resonance imaging markers primarily reside in the frontal, temporal, occipital lobes, and cerebellum. The neuro-transcriptomic joint analysis highlights genes associated with biological processes, such as "presynapse," "behavior," and "modulation of chemical synaptic transmission" in autism spectrum disorder's brain development. Different magnetic resonance imaging modalities offer complementary information for autism spectrum disorder diagnosis. Our dual-branch graph neural network achieves high accuracy and identifies abnormal brain regions and the neuro-transcriptomic analysis uncovers important genetic biomarkers. Overall, our study presents an effective approach for assisting in autism spectrum disorder diagnosis and identifying genetic biomarkers, showing potential for enhancing the diagnosis and treatment of this condition.


Autism Spectrum Disorder , Autistic Disorder , Deep Learning , Humans , Autistic Disorder/pathology , Autism Spectrum Disorder/diagnostic imaging , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Brain , Magnetic Resonance Imaging/methods , Biomarkers , Brain Mapping/methods
19.
Acad Radiol ; 31(5): 2074-2084, 2024 May.
Article En | MEDLINE | ID: mdl-38185571

RATIONALE AND OBJECTIVES: This study employed tract-based spatial statistics (TBSS) to investigate abnormalities in the white matter microstructure among children with autism spectrum disorder (ASD). Additionally, an eXtreme Gradient Boosting (XGBoost) model was developed to effectively classify individuals with ASD and typical developing children (TDC). METHODS AND MATERIALS: Multi-shell diffusion weighted images were acquired from 62 children with ASD and 44 TDC. Using the Pydesigner procedure, diffusion tensor (DT), diffusion kurtosis (DK), and white matter tract integrity (WMTI) metrics were computed. Subsequently, TBSS analysis was applied to discern differences in these diffusion parameters between ASD and TDC groups. The XGBoost model was then trained using metrics showing significant differences, and Shapley Additive explanations (SHAP) values were computed to assess the feature importance in the model's predictions. RESULTS: TBSS analysis revealed a significant reduction in axonal diffusivity (AD) in the left posterior corona radiata and the right superior corona radiata. Among the DK indicators, mean kurtosis, axial kurtosis, and kurtosis fractional anisotropy were notably increased in children with ASD, with no significant difference in radial kurtosis. WMTI metrics such as axonal water fraction, axonal diffusivity of the extra-axonal space (EAS_AD), tortuosity of the extra-axonal space (EAS_TORT), and diffusivity of intra-axonal space (IAS_Da) were significantly increased, primarily in the corpus callosum and fornix. Notably, there was no significant difference in radial diffusivity of the extra-axial space (EAS_RD). The XGBoost model demonstrated excellent classification ability, and the SHAP analysis identified EAS_TORT as the feature with the highest importance in the model's predictions. CONCLUSION: This study utilized TBSS analyses with multi-shell diffusion data to examine white matter abnormalities in pediatric autism. Additionally, the developed XGBoost model showed outstanding performance in classifying ASD and TDC. The ranking of SHAP values based on the XGBoost model underscored the significance of features in influencing model predictions.


Autism Spectrum Disorder , Diffusion Tensor Imaging , Machine Learning , White Matter , Humans , Autism Spectrum Disorder/diagnostic imaging , Autism Spectrum Disorder/pathology , White Matter/diagnostic imaging , White Matter/pathology , Male , Female , Child, Preschool , Diffusion Tensor Imaging/methods , Diffusion Magnetic Resonance Imaging/methods , Child , Image Interpretation, Computer-Assisted/methods
20.
Genet Med ; 26(5): 101087, 2024 May.
Article En | MEDLINE | ID: mdl-38288683

PURPOSE: Interneuronopathies are a group of neurodevelopmental disorders characterized by deficient migration and differentiation of gamma-aminobutyric acidergic interneurons resulting in a broad clinical spectrum, including autism spectrum disorders, early-onset epileptic encephalopathy, intellectual disability, and schizophrenic disorders. SP9 is a transcription factor belonging to the Krüppel-like factor and specificity protein family, the members of which harbor highly conserved DNA-binding domains. SP9 plays a central role in interneuron development and tangential migration, but it has not yet been implicated in a human neurodevelopmental disorder. METHODS: Cases with SP9 variants were collected through international data-sharing networks. To address the specific impact of SP9 variants, in silico and in vitro assays were carried out. RESULTS: De novo heterozygous variants in SP9 cause a novel form of interneuronopathy. SP9 missense variants affecting the glutamate 378 amino acid result in severe epileptic encephalopathy because of hypomorphic and neomorphic DNA-binding effects, whereas SP9 loss-of-function variants result in a milder phenotype with epilepsy, developmental delay, and autism spectrum disorder. CONCLUSION: De novo heterozygous SP9 variants are responsible for a neurodevelopmental disease. Interestingly, variants located in conserved DNA-binding domains of KLF/SP family transcription factors may lead to neomorphic DNA-binding functions resulting in a combination of loss- and gain-of-function effects.


Autism Spectrum Disorder , Epilepsy , Intellectual Disability , Interneurons , Sp Transcription Factors , Transcription Factors , Adolescent , Child , Child, Preschool , Female , Humans , Male , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Epilepsy/genetics , Epilepsy/pathology , Heterozygote , Intellectual Disability/genetics , Intellectual Disability/pathology , Interneurons/metabolism , Interneurons/pathology , Mutation, Missense/genetics , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Phenotype , Transcription Factors/genetics , Transcription Factors/metabolism , Sp Transcription Factors/genetics
...