Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Intellect Disabil Res ; 67(7): 679-689, 2023 07.
Article in English | MEDLINE | ID: mdl-37129092

ABSTRACT

BACKGROUND: Investigating copy number variations (CNVs) such as microdeletions or microduplications can significantly contribute to discover the aetiology of neurodevelopmental disorders. 15q11.2 genomic region, including NIPA1 and NIPA2 genes, contains a recurrent but rare CNV, flanked by the break points BP1 and BP2. Both BP1-BP2 microdeletion and microduplication have been associated with intellectual disability (ID), neuropsychiatric/behavioural disturbances and mild clinical features, even if with incomplete penetrance and variable expressivity. The pathogenic role of this CNV is quite unclear though. Unknown variants in other DNA regions and parent-of-origin effect (POE) are some of the mechanisms that have been proposed as an explanation of the wide phenotypic variability. As NIPA1 and NIPA2 encode for proteins that mediate magnesium (Mg2+ ) metabolism, it has been suggested that urinary Mg2+ levels could potentially represent informative and affordable biomarkers for a rapid screening of 15q11.2 duplications or deletions. Furthermore, magnesium supplementation has been proposed as possible therapeutic strategy. METHODS: Thirty one children with ID and/or other neurodevelopmental disorders carrying either a duplication or a deletion in 15q11.2 BP1-BP2 region have been recruited. When available, blood samples from parents have been analysed to identify the CNV origin. All participants underwent family and medical data collection, physical examination and neuropsychiatric assessment. Electroencephalogram (EEG) and brain magnetic resonance imaging (MRI) scan were performed in 15 children. In addition, 11 families agreed to participate to the assessment of blood and urinary Mg2+ levels. RESULTS: We observed a highly variable phenotypic spectrum of developmental issues encompassing ID in most subjects as well as a variety of behavioural disorders such as autism and attention-deficit disorder/attention-deficit hyperactivity disorder. Dysmorphic traits and malformations were detected only in a minority of the participants, and no clear association with growth anomalies was found. Abnormal brain MRI and/or EEG were reported respectively in 64% and 92% of the subjects. Inheritance assessment highlighted an excess of duplication of maternal origin, while cardiac alterations were detected only in children with 15q11.2 CNV inherited from the father. We found great variability in Mg2+ urinary values, without correlation with 15q11.2 copy numbers. However, the variance of urinary Mg2+ levels largely increases in individuals with 15q11.2 deletion/duplication. CONCLUSIONS: This study provides further evidence that 15q11.2 BP1-BP2 CNV is associated with a broad spectrum of neurodevelopmental disorders and POE might be an explanation for clinical variability. However, some issues may question the real impact of 15q11.2 CNV on the phenotype in the carriers: DNA sequencing could be useful to exclude other pathogenic gene mutations. Our results do not support the possibility that urinary Mg2+ levels can be used as biomarkers to screen children with neurodevelopmental disorders for 15q11.2 duplication/deletion. However, there are evidences of correlations between 15q11.2 BP1-BP2 CNV and Mg2+ metabolism and future studies may pave the way to new therapeutic options.


Subject(s)
Intellectual Disability , Neurodevelopmental Disorders , Humans , Chromosome Aberrations , Magnesium , DNA Copy Number Variations/genetics , Neurodevelopmental Disorders/genetics , Intellectual Disability/genetics , Biomarkers
2.
Dev Med Child Neurol ; 64(9): 1077-1084, 2022 09.
Article in English | MEDLINE | ID: mdl-35661141

ABSTRACT

Precision medicine refers to treatments that are targeted to an individual's unique characteristics. Precision medicine for neurodevelopmental disorders (such as cerebral palsy, attention-deficit/hyperactivity disorder, obsessive-compulsive disorder, Tourette syndrome, and autism spectrum disorder) in children has predominantly focused on advances in genomic sequencing technologies to increase our ability to identify single gene mutations, diagnose a multitude of rare neurodevelopmental disorders, and gain insights into pathogenesis. Although targeting specific gene variants with high penetrance will help some children with rare disease, this approach will not help most children with neurodevelopmental disorders. A 'pathway' driven approach targeting the cumulative influence of psychosocial, epigenetic, or cellular factors is likely to be more effective. To optimize the therapeutic potential of precision medicine, we present a biopsychosocial integrated framework to examine the 'gene-environment neuroscience interaction'. Such an approach would be supported through harnessing the power of big data, transdiagnostic assessment, impact and implementation evaluation, and a bench-to-bedside scientific discovery agenda with ongoing clinician and patient engagement. WHAT THIS PAPER ADDS: Precision medicine has predominantly focused on genetic risk factors. The impact of environmental risk factors, particularly inflammatory, metabolic, and psychosocial risks, is understudied. A holistic biopsychosocial model of neurodevelopmental disorder causal pathways is presented. The model will provide precision medicine across the full spectrum of neurodevelopmental disorders.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Autism Spectrum Disorder , Neurodevelopmental Disorders , Attention Deficit Disorder with Hyperactivity/diagnosis , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/therapy , Child , Genomics , Humans , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/therapy , Precision Medicine
3.
Nutrients ; 13(11)2021 Oct 28.
Article in English | MEDLINE | ID: mdl-34836113

ABSTRACT

Iron deficiency (ID) anemia is the foremost micronutrient deficiency worldwide, affecting around 40% of pregnant women and young children. ID during the prenatal and early postnatal periods has a pronounced effect on neurodevelopment, resulting in long-term effects such as cognitive impairment and increased risk for neuropsychiatric disorders. Treatment of ID has been complicated as it does not always resolve the long-lasting neurodevelopmental deficits. In animal models, developmental ID results in abnormal hippocampal structure and function associated with dysregulation of genes involved in neurotransmission and synaptic plasticity. Dysregulation of these genes is a likely proximate cause of the life-long deficits that follow developmental ID. However, a direct functional link between iron and gene dysregulation has yet to be elucidated. Iron-dependent epigenetic modifications are one mechanism by which ID could alter gene expression across the lifespan. The jumonji and AT-rich interaction domain-containing (JARID) protein and the Ten-Eleven Translocation (TET) proteins are two families of iron-dependent epigenetic modifiers that play critical roles during neural development by establishing proper gene regulation during critical periods of brain development. Therefore, JARIDs and TETs can contribute to the iron-mediated epigenetic mechanisms by which early-life ID directly causes stable changes in gene regulation across the life span.


Subject(s)
Anemia, Iron-Deficiency/genetics , Epigenesis, Genetic/physiology , Hippocampus/metabolism , Infant Nutritional Physiological Phenomena/genetics , Maternal Nutritional Physiological Phenomena/genetics , Anemia, Iron-Deficiency/complications , Animals , Animals, Newborn , Child Development/physiology , Epigenomics , Female , Hippocampus/growth & development , Humans , Infant , Infant, Newborn , Neurodevelopmental Disorders/genetics , Neurogenesis/physiology , Neuronal Plasticity/physiology , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Synaptic Transmission/physiology
4.
Dev Neurosci ; 43(3-4): 253-261, 2021.
Article in English | MEDLINE | ID: mdl-33940579

ABSTRACT

Brain development is a complex process that requires a series of precise and coordinated events to take place. When alterations in some of those events occur, neurodevelopmental disorders (NDDs) may appear, with their characteristic symptoms, including cognitive, social motor deficits, and epilepsy. While pharmacologic treatments have been the only therapeutic options for many years, more recently the research is turning to the direct removal of the underlying genetic cause of each specific NDD. This is possible thanks to the increased knowledge of genetic basis of those diseases and the enormous advances in genome-editing tools. Together with clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9-based strategies, there is a great development also of nuclease defective Cas9 (dCas9) tools that, with an extreme flexibility, allow the recruitment of specific protein functions to the desired genomic sites. In this work, we review dCas9-based tools and discuss all the published applications in the setting of therapeutic approaches for NDDs at the preclinical level. In particular, dCas9-based therapeutic strategies for Dravet syndrome, transcallosal dysconnectivity caused by mutations in C11orf46 gene, and Fragile X syndrome are presented and discussed. A direct comparison with other possible therapeutic strategies, such as classic gene replacement or CRISPR/Cas9-based strategies, is provided. We also highlight not only those aspects that constitute a clear advantage compared to previous strategies but also the main technical hurdles related to their applications that need to be overcome.


Subject(s)
CRISPR-Cas Systems , Neurodevelopmental Disorders , CRISPR-Cas Systems/genetics , Gene Editing , Genetic Therapy , Humans , Mutation , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/therapy
5.
RNA Biol ; 18(7): 1063-1084, 2021 07.
Article in English | MEDLINE | ID: mdl-33499699

ABSTRACT

Single-cell RNA-sequencing (scRNA-seq) has emerged in recent years as a breakthrough technology to understand RNA metabolism at cellular resolution. In addition to allowing new cell types and states to be identified, scRNA-seq can permit cell-type specific differential gene expression changes, pre-mRNA processing events, gene regulatory networks and single-cell developmental trajectories to be uncovered. More recently, a new wave of multi-omic adaptations and complementary spatial transcriptomics workflows have been developed that facilitate the collection of even more holistic information from individual cells. These developments have unprecedented potential to provide penetrating new insights into the basic neural cell dynamics and molecular mechanisms relevant to the nervous system in both health and disease. In this review we discuss this maturation of single-cell RNA-sequencing over the past decade, and review the different adaptations of the technology that can now be applied both at different scales and for different purposes. We conclude by highlighting how these methods have already led to many exciting discoveries across neuroscience that have furthered our cellular understanding of the neurological disease.


Subject(s)
Brain/metabolism , Nerve Tissue Proteins/genetics , Neurodegenerative Diseases/genetics , Neurodevelopmental Disorders/genetics , Neurons/metabolism , RNA, Messenger/genetics , Single-Cell Analysis/methods , Animals , Brain/pathology , Computational Biology/methods , DNA Barcoding, Taxonomic , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , High-Throughput Nucleotide Sequencing , Humans , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurodevelopmental Disorders/metabolism , Neurodevelopmental Disorders/pathology , Neurons/pathology , RNA, Messenger/metabolism , Sequence Analysis, RNA/methods , Transcriptome
6.
Curr Opin Genet Dev ; 68: 18-25, 2021 06.
Article in English | MEDLINE | ID: mdl-33454514

ABSTRACT

Among medical disciplines, diagnosis in psychiatry depends highly upon descriptive signs and symptoms, rather than biomarkers. Clear descriptions of specific genetic etiologies have been lacking; genomic technologies, however, are rapidly changing that landscape. Notably, chromosomal microarrays-which detect gene copy number variants (CNVs)-are a recommended standard of care for neurodevelopmental disorders. As a result, an increasing number of patients now receive a clinical diagnosis based on the Diagnostic and Statistical Manual of Mental Disorders (DSM-5) and an identified genetic etiological variant. However, psychiatric and genetic diagnoses are frequently communicated and managed as two disconnected diagnostic parameters. Here, we advocate for a transition model, allowing the integration of genetic etiological information-starting with diagnostically proven CNVs-within the DSM-5 classification framework.


Subject(s)
DNA Copy Number Variations , Delivery of Health Care, Integrated , Evidence-Based Medicine , Gene Dosage , Genetic Predisposition to Disease , Neurodevelopmental Disorders/diagnosis , Neurodevelopmental Disorders/genetics , Diagnostic Techniques, Neurological , Genetic Testing/methods , Humans
7.
Mol Genet Metab ; 131(3): 316-324, 2020 11.
Article in English | MEDLINE | ID: mdl-33127324

ABSTRACT

Propionic acidemia (PA) is caused by inherited deficiency of mitochondrial propionyl-CoA carboxylase (PCC) and results in significant neurodevelopmental and cardiac morbidity. However, relationships among therapeutic intervention, biochemical markers, and disease progression are poorly understood. Sixteen individuals homozygous for PCCB c.1606A > G (p.Asn536Asp) variant PA participated in a two-week suspension of therapy. Standard metabolic markers (plasma amino acids, blood spot methylcitrate, plasma/urine acylcarnitines, urine organic acids) were obtained before and after stopping treatment. These same markers were obtained in sixteen unaffected siblings. Echocardiography and electrocardiography were obtained from all subjects. We characterized the baseline biochemical phenotype of untreated PCCB c.1606A > G homozygotes and impact of treatment on PCC deficiency biomarkers. Therapeutic regimens varied widely. Suspension of therapy did not significantly alter branched chain amino acid levels, their alpha-ketoacid derivatives, or urine ketones. Carnitine supplementation significantly increased urine propionylcarnitine and its ratio to total carnitine. Methylcitrate blood spot and urine levels did not correlate with other biochemical measures or cardiac outcomes. Treatment of PCCB c.1606A > G homozygotes with protein restriction, prescription formula, and/or various dietary supplements has a limited effect on core biomarkers of PCC deficiency. These patients require further longitudinal study with standardized approaches to better understand the relationship between biomarkers and disease burden.


Subject(s)
Carbon-Carbon Ligases/genetics , Heart/physiopathology , Neurodevelopmental Disorders/genetics , Propionic Acidemia/genetics , Acids/blood , Acids/urine , Adolescent , Adult , Amino Acids/blood , Amino Acids/urine , Biomarkers/blood , Biomarkers/urine , Carbon-Carbon Ligases/blood , Carbon-Carbon Ligases/urine , Carnitine/blood , Carnitine/urine , Child , Child, Preschool , Echocardiography , Female , Heart/diagnostic imaging , Humans , Male , Mitochondria/genetics , Mitochondria/metabolism , Mutation/genetics , Neurodevelopmental Disorders/blood , Neurodevelopmental Disorders/diagnostic imaging , Neurodevelopmental Disorders/urine , Organic Chemicals/blood , Organic Chemicals/urine , Phenotype , Propionic Acidemia/blood , Propionic Acidemia/diagnostic imaging , Propionic Acidemia/urine , Young Adult
8.
Brain Dev ; 42(9): 691-695, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32773162

ABSTRACT

INTRODUCTION: KCNT2 was recently recognized as a gene associated with neurodevelopmental disorder and epilepsy. CASE REPORT: We present an additional observation of a 16-year-old male patient with a novel de novo KCNT2 likely pathogenic variant and review the five previously reported cases of de novo variants in this gene. DISCUSSION: Whole exome sequencing identified the missense variant c.725C > A p.(Thr242Asn), which was confirmed by Sanger sequencing. Our patient has a refractory stereotyped and monomorphic type of hyperkinetic focal motor seizure, similar to what is seen in frontal lobe epilepsy, occurring only during sleep. This type of seizure is not usually seen in epileptic encephalopathies.


Subject(s)
Brain Diseases/genetics , Epilepsy, Frontal Lobe/genetics , Potassium Channels, Sodium-Activated/genetics , Adolescent , Brain Diseases/metabolism , Child , Epilepsy, Frontal Lobe/diagnosis , Epilepsy, Generalized/genetics , Female , Humans , Male , Mutation, Missense/genetics , Neurodevelopmental Disorders/diagnosis , Neurodevelopmental Disorders/genetics , Phenotype , Potassium Channels, Sodium-Activated/metabolism , Exome Sequencing , Young Adult
9.
Am J Med Genet A ; 182(9): 2129-2132, 2020 09.
Article in English | MEDLINE | ID: mdl-32627382

ABSTRACT

YY1 mutations cause Gabriele-de Vries syndrome, a recently described condition involving cognitive impairment, facial dysmorphism and intrauterine growth restriction. Movement disorders were reported in 5/10 cases of the original series, but no detailed description was provided. Here we present a 21-year-old woman with a mild intellectual deficit, facial dysmorphism and a complex movement disorder including an action tremor, cerebellar ataxia, dystonia, and partial ocular apraxia as the presenting and most striking feature. Whole-exome sequencing revealed a novel heterozygous de novo mutation in YY1 [NM: 003403.4 (YY1): c.907 T > C; p.(Cys303Arg)], classified as pathogenic according to the ACMG guidelines.


Subject(s)
Movement Disorders/genetics , Neurodevelopmental Disorders/genetics , YY1 Transcription Factor/genetics , Child , Child, Preschool , Exome/genetics , Female , Genetic Predisposition to Disease , Heterozygote , Humans , Infant , Infant, Newborn , Intellectual Disability/genetics , Intellectual Disability/pathology , Movement Disorders/pathology , Neurodevelopmental Disorders/pathology , Phenotype , Exome Sequencing
10.
Proc Natl Acad Sci U S A ; 117(12): 6836-6843, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32144139

ABSTRACT

Visuomotor impairments characterize numerous neurological disorders and neurogenetic syndromes, such as autism spectrum disorder (ASD) and Dravet, Fragile X, Prader-Willi, Turner, and Williams syndromes. Despite recent advances in systems neuroscience, the biological basis underlying visuomotor functional impairments associated with these clinical conditions is poorly understood. In this study, we used neuroimaging connectomic approaches to map the visuomotor integration (VMI) system in the human brain and investigated the topology approximation of the VMI network to the Allen Human Brain Atlas, a whole-brain transcriptome-wide atlas of cortical genetic expression. We found the genetic expression of four genes-TBR1, SCN1A, MAGEL2, and CACNB4-to be prominently associated with visuomotor integrators in the human cortex. TBR1 gene transcripts, an ASD gene whose expression is related to neural development of the cortex and the hippocampus, showed a central spatial allocation within the VMI system. Our findings delineate gene expression traits underlying the VMI system in the human cortex, where specific genes, such as TBR1, are likely to play a central role in its neuronal organization, as well as on specific phenotypes of neurogenetic syndromes.


Subject(s)
Calcium Channels/genetics , Motor Cortex/physiopathology , NAV1.1 Voltage-Gated Sodium Channel/genetics , Neurodevelopmental Disorders/pathology , Proteins/genetics , T-Box Domain Proteins/genetics , Visual Cortex/physiopathology , Adult , Aged , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Brain Mapping , Cohort Studies , Epilepsies, Myoclonic/genetics , Epilepsies, Myoclonic/pathology , Female , Humans , Male , Middle Aged , Neurodevelopmental Disorders/genetics , Prader-Willi Syndrome/genetics , Prader-Willi Syndrome/pathology , Psychomotor Performance , Visual Perception
11.
Cereb Cortex ; 30(4): 2358-2371, 2020 04 14.
Article in English | MEDLINE | ID: mdl-31812984

ABSTRACT

2p16.3 deletions, involving heterozygous NEUREXIN1 (NRXN1) deletion, dramatically increase the risk of developing neurodevelopmental disorders, including autism and schizophrenia. We have little understanding of how NRXN1 heterozygosity increases the risk of developing these disorders, particularly in terms of the impact on brain and neurotransmitter system function and brain network connectivity. Thus, here we characterize cerebral metabolism and functional brain network connectivity in Nrxn1α heterozygous mice (Nrxn1α+/- mice), and assess the impact of ketamine and dextro-amphetamine on cerebral metabolism in these animals. We show that heterozygous Nrxn1α deletion alters cerebral metabolism in neural systems implicated in autism and schizophrenia including the thalamus, mesolimbic system, and select cortical regions. Nrxn1α heterozygosity also reduces the efficiency of functional brain networks, through lost thalamic "rich club" and prefrontal cortex (PFC) hub connectivity and through reduced thalamic-PFC and thalamic "rich club" regional interconnectivity. Subanesthetic ketamine administration normalizes the thalamic hypermetabolism and partially normalizes thalamic disconnectivity present in Nrxn1α+/- mice, while cerebral metabolic responses to dextro-amphetamine are unaltered. The data provide new insight into the systems-level impact of heterozygous Nrxn1α deletion and how this increases the risk of developing neurodevelopmental disorders. The data also suggest that the thalamic dysfunction induced by heterozygous Nrxn1α deletion may be NMDA receptor-dependent.


Subject(s)
Calcium-Binding Proteins/genetics , Ketamine/administration & dosage , Neural Cell Adhesion Molecules/genetics , Neurodevelopmental Disorders/diagnostic imaging , Neurodevelopmental Disorders/genetics , Prefrontal Cortex/diagnostic imaging , Thalamus/diagnostic imaging , Animals , Disease Models, Animal , Gene Deletion , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Net/diagnostic imaging , Nerve Net/drug effects , Neurodevelopmental Disorders/drug therapy , Prefrontal Cortex/drug effects , Thalamus/drug effects
12.
J Child Neurol ; 34(12): 778-781, 2019 10.
Article in English | MEDLINE | ID: mdl-31282308

ABSTRACT

BACKGROUND: WARS2 encodes a tryptophanyl tRNA synthetase, which is involved in mitochondrial protein synthesis. Biallelic mutations in WARS2 are rare and have been associated with a spectrum of clinical presentations, including neurodevelopmental disorder with abnormal movements, lactic acidosis with or without seizures (NEMMLAS). CASE PRESENTATION: Here we present the case of an 8-year-old girl with ataxia and parkinsonism with periventricular white matter abnormalities on magnetic resonance imaging (MRI) and global developmental delay. The initial investigations revealed an elevated lactate level. Extensive metabolic testing, including a muscle biopsy, was inconclusive. Cerebrospinal fluid (CSF) neurotransmitter levels were low; however, a trial of levodopa was unremarkable. The chromosomal microarray and initial ataxia gene panel was normal. Zinc supplementation for a heterozygous variant of unknown significance in the CP gene on the ataxia exome panel was not effective in treating her symptoms. Reanalysis of the ataxia exome panel highlighted biallelic mutations in WARS2, which lead to the diagnosis of neurodevelopmental disorder, mitochondrial, with abnormal movements and lactic acidosis, with or without seizures (NEMMLAS). This lead to parental genetic testing, redirected therapy, and helped to expand the symptomology of this rare condition. CONCLUSION: Here we emphasize the importance of imminent and repeat expanded genetic testing to ensure early diagnosis and treatment for rare pediatric disorders. The patient is being trialed on a mitochondrial cocktail in an attempt to compensate for defects in mitochondrial protein synthesis associated with this variant. Longitudinal monitoring of disease manifestation will help establish the currently unknown natural history of this condition.


Subject(s)
Acidosis, Lactic/diagnosis , Dyskinesias/diagnosis , Neurodevelopmental Disorders/diagnosis , Seizures/diagnosis , Tryptophan-tRNA Ligase/genetics , Acidosis, Lactic/diagnostic imaging , Acidosis, Lactic/genetics , Brain/diagnostic imaging , Child , Dyskinesias/diagnostic imaging , Dyskinesias/genetics , Female , Humans , Magnetic Resonance Imaging , Mitochondria/genetics , Neurodevelopmental Disorders/diagnostic imaging , Neurodevelopmental Disorders/genetics , Phenotype , Seizures/diagnostic imaging , Seizures/genetics , Syndrome , White Matter/diagnostic imaging
13.
Clin Genet ; 95(2): 241-252, 2019 02.
Article in English | MEDLINE | ID: mdl-30047142

ABSTRACT

There is growing evidence that epigenetic dysregulation plays a role in neurodevelopmental disorders. In humans, folate is one of the main donors of the methyl group required for the synthesis of S-adenosylmethionine, which in turn is needed for DNA and histone methylation as key neurodevelopment processes. Folate deficiency during pregnancy has been correlated with neural tube defects and with a higher incidence of neurocognitive and/or neurobehavioral deficits. A similar outcome may be exerted by gene polymorphisms in folate or folate-related pathways. This has been documented by numerous case/control association studies performed on neurodevelopmental disorders such as autism spectrum disorder and attention deficit hyperactivity disorder. In this regard, the folate cycle represents a "perfect model" of how genetics influences epigenetics. Gene variants in folate and folate-related pathways can be considered risk factors for neurodevelopmental disorders and should therefore be assessed by genetic testing in pregnant women. High-risk women should be considered for folate supplementation during pregnancy. Here, we review all published case/control association studies on gene polymorphisms in folate and folate-related pathways performed on neurodevelopmental disorders, provide an overview of neurodevelopment and DNA methylation changes occurring at this time, and describe the biological basis of neurodevelopmental disorders and recent evidence of their epigenetic dysregulation.


Subject(s)
Epigenesis, Genetic , Folic Acid/metabolism , Genetic Predisposition to Disease , Genetic Variation , Metabolic Networks and Pathways , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/metabolism , Animals , Biomarkers , DNA Methylation , Diet , Gene Expression Regulation , Humans , Metabolic Networks and Pathways/genetics , Neurogenesis/genetics
15.
Neuron ; 98(2): 282-295, 2018 04 18.
Article in English | MEDLINE | ID: mdl-29673480

ABSTRACT

Diagnoses of behavioral disorders such as autism spectrum disorder and schizophrenia are based on symptomatic descriptions that have been difficult to connect to mechanism. Although psychiatric genetics provide insight into the genetic underpinning of such disorders, with a majority of cases explained by polygenic factors, it remains difficult to design rational treatments. In this review, we highlight the value of understanding neural circuit function both as an intermediate level of explanatory description that links gene to behavior and as a pathway for developing rational diagnostics and therapeutics for behavioral disorders. As neural circuits perform hierarchically organized computational functions and give rise to network-level processes (e.g., macroscopic rhythms and goal-directed or homeostatic behaviors), correlated network-level deficits may indicate perturbation of a specific circuit. Therefore, identifying such correlated deficits or a circuit endophenotype would provide a mechanistic point of entry, enhancing both diagnosis and treatment of a given behavioral disorder. We focus on a circuit endophenotype of the thalamic reticular nucleus (TRN) and how its impairment in neurodevelopmental disorders gives rise to a correlated set of readouts across sleep and attention. Because TRN neurons express several disorder-relevant genes identified through genome-wide association studies, exploring the consequences of different TRN disruptions may be of broad translational significance.


Subject(s)
Endophenotypes/metabolism , Midbrain Reticular Formation/metabolism , Nerve Net/metabolism , Neurodevelopmental Disorders/metabolism , Thalamus/metabolism , Animals , Humans , Midbrain Reticular Formation/physiopathology , Nerve Net/physiopathology , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/physiopathology , Thalamus/physiopathology
16.
Nutr Neurosci ; 21(9): 602-613, 2018 Nov.
Article in English | MEDLINE | ID: mdl-28553986

ABSTRACT

Many aspects of human development and disease are influenced by the interaction between genetic and environmental factors. Understanding how our genes respond to the environment is central to managing health and disease, and is one of the major contemporary challenges in human genetics. Various epigenetic processes affect chromosome structure and accessibility of deoxyribonucleic acid (DNA) to the enzymatic machinery that leads to expression of genes. One important epigenetic mechanism that appears to underlie the interaction between environmental factors, including diet, and our genome, is chemical modification of the DNA. The best understood of these modifications is methylation of cytosine residues in DNA. It is now recognized that the pattern of methylated cytosines throughout our genomes (the 'methylome') can change during development and in response to environmental cues, often with profound effects on gene expression. Many dietary constituents may indirectly influence genomic pathways that methylate DNA, and there is evidence for biochemical links between nutritional quality and mental health. Deficiency of both macro- and micronutrients has been associated with increased behavioural problems, and nutritional supplementation has proven efficacious in treatment of certain neuropsychiatric disorders. In this review we examine evidence from the fields of nutrition, developmental biology, and mental health that supports dietary impacts on epigenetic processes, particularly DNA methylation. We then consider whether such processes could underlie the demonstrated efficacy of dietary supplementation in treatment of mental disorders, and whether targeted manipulation of DNA methylation patterns using controlled dietary supplementation may be of wider clinical value.


Subject(s)
Epigenesis, Genetic , Mental Health , Nutritional Status , Animals , DNA Methylation/drug effects , Diet , Dietary Supplements , Disease Models, Animal , Gene Expression Regulation , Humans , Mental Disorders/diagnosis , Mental Disorders/diet therapy , Mental Disorders/genetics , Micronutrients/administration & dosage , Micronutrients/deficiency , Neurodevelopmental Disorders/diagnosis , Neurodevelopmental Disorders/diet therapy , Neurodevelopmental Disorders/genetics
18.
J Neurosci ; 35(46): 15326-38, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26586820

ABSTRACT

Selenium (Se) is essential for both brain development and male fertility. Male mice lacking two key genes involved in Se metabolism (Scly(-/-)Sepp1(-/-) mice), selenoprotein P (Sepp1) and Sec lyase (Scly), develop severe neurological dysfunction, neurodegeneration, and audiogenic seizures that manifest beginning in early adulthood. We demonstrate that prepubescent castration of Scly(-/-)Sepp1(-/-) mice prevents behavioral deficits, attenuates neurodegeneration, rescues maturation of GABAergic inhibition, and increases brain selenoprotein levels. Moreover, castration also yields similar neuroprotective benefits to Sepp1(-/-) and wild-type mice challenged with Se-deficient diets. Our data show that, under Se-compromised conditions, the brain and testes compete for Se utilization, with concomitant effects on neurodevelopment and neurodegeneration. SIGNIFICANCE STATEMENT: Selenium is an essential trace element that promotes male fertility and brain function. Herein, we report that prepubescent castration provides neuroprotection by increasing selenium-dependent antioxidant activity in the brain, revealing a competition between the brain and testes for selenium utilization. These findings provide novel insight into the interaction of sex and oxidative stress upon the developing brain and have potentially significant implications for the prevention of neurodevelopmental disorders characterized by aberrant excitatory/inhibitory balance, such as schizophrenia and epilepsy.


Subject(s)
Brain/metabolism , Lyases/metabolism , Neurodevelopmental Disorders/genetics , Selenium/metabolism , Selenoprotein P/metabolism , Age Factors , Animals , Brain/drug effects , Brain/pathology , Castration , Dizocilpine Maleate/pharmacology , Epilepsy, Reflex/genetics , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glutamate Decarboxylase/metabolism , Lyases/genetics , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/genetics , Nerve Tissue Proteins/metabolism , Neurodevelopmental Disorders/metabolism , Neurodevelopmental Disorders/pathology , Neurodevelopmental Disorders/prevention & control , Selenoprotein P/genetics , Sex Factors , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL