1
|
Slevin H, Kehinde F, Begum-Ali J, Ellis C, Burkitt-Wright E, Green J, Johnson MH, Pasco G, Charman T, Jones EJH, Garg S. Developmental trajectories in infants and pre-school children with Neurofibromatosis 1. Mol Autism 2024; 15:45. [PMID: 39407332 PMCID: PMC11481376 DOI: 10.1186/s13229-024-00621-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 09/11/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Children with Neurofibromatosis 1 (NF1) show cognitive, behavioural and social differences compared to their peers. However, the age and sequence at which these differences begin to emerge is not fully understood. This prospective cohort study examines the cognitive, behavioural, ADHD trait and autism symptom development in infant and pre-school children with NF1 compared with typically developing (TD) children without a family history of neurodevelopmental conditions. METHODS Data from standardised tests was gathered at 5, 10, 14, 24 and 36 months of age (NF1 n = 35, TD n = 29). Developmental trajectories of cognitive (Mullen Scales of Early Learning, MSEL) and adaptive behavioural (Vineland Adaptive Behavior Scales, VABS) development from 5 to 36 months were analysed using linear mixed modelling. Measures of ADHD (Child Behavior Checklist) and autism traits (ADOS-2, BOSA-MV and ADI-R) were assessed at 24 and 36 months. RESULTS The developmental trajectory of cognitive skills (all domains of the MSEL) and behavioural skills (four domains of the VABS) differed significantly between NF1 and TD groups. Post-hoc tests demonstrated that the NF1 participants scored significantly lower than TD participants at 24 months on all MSEL and VABS domains. The NF1 cohort demonstrated higher mean autism and ADHD traits at 24 months and 14% of the NF1 cohort met a research diagnostic classification for autism at 36 months. LIMITATIONS The study has a relatively small sample size due to variable retention and rolling recruitment. Due to limitations imposed by the COVID-19 pandemic, we utilised the Brief Observation of Symptoms of Autism for Minimally Verbal children (BOSA-MV) for some participants, which was administered online and may not gather as accurate a picture of traits as ADOS-2. The BOSA-MV was utilised for 41% of participants with NF1 at 36 months compared to 11% at 24 months. This may explain the reduction in the percentage of children with NF1 that met autism criteria at 36 months. CONCLUSIONS By 24 months of age, the NF1 cohort show lower cognitive skills and adaptive behaviour and higher levels of autism and ADHD traits as compared to TD children. This has implications for developmental monitoring and referral for early interventions. TRIAL REGISTRATION Not applicable.
Collapse
Affiliation(s)
- Hannah Slevin
- Division of Psychology and Mental Health, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Fiona Kehinde
- Division of Psychology and Mental Health, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jannath Begum-Ali
- Centre for Brain and Cognitive Development and Department of Psychological Sciences, Birkbeck, University of London, London, UK
| | - Ceri Ellis
- Division of Psychology, Communication and Human Neuroscience, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Emma Burkitt-Wright
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, UK
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jonathan Green
- Division of Psychology and Mental Health, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Harrington Building, Royal Manchester Children's Hospital, Oxford Rd, Manchester, M13 9WL, UK
| | - Mark H Johnson
- Centre for Brain and Cognitive Development and Department of Psychological Sciences, Birkbeck, University of London, London, UK
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - Greg Pasco
- Psychology Department, Institute of Psychiatry, King's College London, Psychology & Neuroscience, London, UK
| | - Tony Charman
- Psychology Department, Institute of Psychiatry, King's College London, Psychology & Neuroscience, London, UK
| | - Emily J H Jones
- Centre for Brain and Cognitive Development and Department of Psychological Sciences, Birkbeck, University of London, London, UK
| | - Shruti Garg
- Division of Psychology and Mental Health, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
- Harrington Building, Royal Manchester Children's Hospital, Oxford Rd, Manchester, M13 9WL, UK.
| |
Collapse
|
2
|
McGhee CA, Honari H, Siqueiros-Sanchez M, Serur Y, van Staalduinen EK, Stevenson D, Bruno JL, Raman MM, Green T. Influences of RASopathies on Neuroanatomical Variation in Children. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2024; 9:858-870. [PMID: 38621478 PMCID: PMC11381177 DOI: 10.1016/j.bpsc.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 03/09/2024] [Accepted: 04/04/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND RASopathies are a group of disorders characterized by pathogenic mutations in the Ras/mitogen-activated protein kinase (Ras/MAPK) signaling pathway. Distinct pathogenic variants in genes encoding proteins in the Ras/MAPK pathway cause Noonan syndrome (NS) and neurofibromatosis type 1 (NF1), which are associated with increased risk for autism spectrum disorder and attention-deficit/hyperactivity disorder. METHODS This study examined the effect of RASopathies (NS and NF1) on human neuroanatomy, specifically on surface area (SA), cortical thickness (CT), and subcortical volumes. Using vertex-based analysis for cortical measures and Desikan region of interest parcellation for subcortical volumes, we compared structural T1-weighted images of children with RASopathies (n = 91, mean age = 8.81 years, SD = 2.12) to those of sex- and age-matched typically developing children (n = 74, mean age = 9.07 years, SD = 1.77). RESULTS Compared with typically developing children, RASopathies had convergent effects on SA and CT, exhibiting increased SA in the precentral gyrus, decreased SA in occipital regions, and thinner CT in the precentral gyrus. RASopathies exhibited divergent effects on subcortical volumes, with syndrome-specific influences from NS and NF1. Overall, children with NS showed decreased volumes in striatal and thalamic structures, and children with NF1 displayed increased volumes in the hippocampus, amygdala, and thalamus. CONCLUSIONS Our study reveals the converging and diverging neuroanatomical effects of RASopathies on human neurodevelopment. The convergence of cortical effects on SA and CT indicates a shared influence of Ras/MAPK hyperactivation on the human brain. Therefore, considering these measures as objective outcome indicators for targeted treatments is imperative.
Collapse
Affiliation(s)
- Chloe Alexa McGhee
- Department of Psychiatry & Behavioral Sciences, Stanford University, Stanford, California.
| | - Hamed Honari
- Department of Psychiatry & Behavioral Sciences, Stanford University, Stanford, California
| | | | - Yaffa Serur
- Department of Psychiatry & Behavioral Sciences, Stanford University, Stanford, California
| | - Eric K van Staalduinen
- Department of Psychiatry & Behavioral Sciences, Stanford University, Stanford, California
| | - David Stevenson
- Division of Medical Genetics, Stanford University, Stanford, California
| | - Jennifer L Bruno
- Department of Psychiatry & Behavioral Sciences, Stanford University, Stanford, California
| | - Mira Michelle Raman
- Department of Psychiatry & Behavioral Sciences, Stanford University, Stanford, California
| | - Tamar Green
- Department of Psychiatry & Behavioral Sciences, Stanford University, Stanford, California
| |
Collapse
|
3
|
Suarez GO, Kumar DS, Brunner H, Emel J, Teel J, Knauss A, Botero V, Broyles CN, Stahl A, Bidaye SS, Tomchik SM. Neurofibromin deficiency alters the patterning and prioritization of motor behaviors in a state-dependent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.08.607070. [PMID: 39149363 PMCID: PMC11326213 DOI: 10.1101/2024.08.08.607070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Genetic disorders such as neurofibromatosis type 1 increase vulnerability to cognitive and behavioral disorders, such as autism spectrum disorder and attention-deficit/hyperactivity disorder. Neurofibromatosis type 1 results from loss-of-function mutations in the neurofibromin gene and subsequent reduction in the neurofibromin protein (Nf1). While the mechanisms have yet to be fully elucidated, loss of Nf1 may alter neuronal circuit activity leading to changes in behavior and susceptibility to cognitive and behavioral comorbidities. Here we show that mutations decreasing Nf1 expression alter motor behaviors, impacting the patterning, prioritization, and behavioral state dependence in a Drosophila model of neurofibromatosis type 1. Loss of Nf1 increases spontaneous grooming in a nonlinear spatial and temporal pattern, differentially increasing grooming of certain body parts, including the abdomen, head, and wings. This increase in grooming could be overridden by hunger in food-deprived foraging animals, demonstrating that the Nf1 effect is plastic and internal state-dependent. Stimulus-evoked grooming patterns were altered as well, with nf1 mutants exhibiting reductions in wing grooming when coated with dust, suggesting that hierarchical recruitment of grooming command circuits was altered. Yet loss of Nf1 in sensory neurons and/or grooming command neurons did not alter grooming frequency, suggesting that Nf1 affects grooming via higher-order circuit alterations. Changes in grooming coincided with alterations in walking. Flies lacking Nf1 walked with increased forward velocity on a spherical treadmill, yet there was no detectable change in leg kinematics or gait. Thus, loss of Nf1 alters motor function without affecting overall motor coordination, in contrast to other genetic disorders that impair coordination. Overall, these results demonstrate that loss of Nf1 alters the patterning and prioritization of repetitive behaviors, in a state-dependent manner, without affecting motor coordination.
Collapse
Affiliation(s)
- Genesis Omana Suarez
- Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
- H.L. Wilkes Honors College, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Divya S. Kumar
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Hannah Brunner
- Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Jalen Emel
- Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Jensen Teel
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Anneke Knauss
- Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Valentina Botero
- Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Connor N. Broyles
- Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Aaron Stahl
- Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Salil S. Bidaye
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Seth M. Tomchik
- Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
- Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
- H.L. Wilkes Honors College, Florida Atlantic University, Jupiter, FL 33458, USA
| |
Collapse
|
4
|
Hocking DR, Sun X, Haebich K, Darke H, North KN, Vivanti G, Payne JM. Delineating Visual Habituation Profiles in Preschoolers with Neurofibromatosis Type 1 and Autism Spectrum Disorder: A Cross-Syndrome Study. J Autism Dev Disord 2024; 54:1998-2011. [PMID: 36877426 DOI: 10.1007/s10803-023-05913-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2023] [Indexed: 03/07/2023]
Abstract
Atypical habituation to repetitive information has been commonly reported in Autism Spectrum Disorder (ASD) but it is not yet clear whether similar abnormalities are present in Neurofibromatosis Type 1 (NF1). We employed a cross-syndrome design using a novel eye tracking paradigm to measure habituation in preschoolers with NF1, children with idiopathic ASD and typically developing (TD) children. Eye movements were recorded to examine fixation duration to simultaneously presented repeating and novel stimuli. Children with NF1 showed a bias for longer look durations to repeating stimuli at the expense of novel stimuli, and slower habituation in NF1 was associated with elevated ASD traits. These findings could indicate aberrant modulation of bottom-up attentional networks that interact with the emergence of ASD phenotypes.
Collapse
Affiliation(s)
- Darren R Hocking
- School of Psychology and Public Health, La Trobe University, Melbourne, Australia.
| | - Xiaoyun Sun
- School of Psychology and Public Health, La Trobe University, Melbourne, Australia
| | - Kristina Haebich
- Murdoch Children's Research Institute, Parkville, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Hayley Darke
- Murdoch Children's Research Institute, Parkville, Australia
| | - Kathryn N North
- Murdoch Children's Research Institute, Parkville, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Giacomo Vivanti
- A.J. Drexel Autism Institute, Drexel University, 3020 Market Street, Suite 560, 19104-3734, Philadelphia, PA, USA
| | - Jonathan M Payne
- Murdoch Children's Research Institute, Parkville, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
5
|
Kereszturi É. Diversity and Classification of Genetic Variations in Autism Spectrum Disorder. Int J Mol Sci 2023; 24:16768. [PMID: 38069091 PMCID: PMC10706722 DOI: 10.3390/ijms242316768] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/19/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition with symptoms that affect the whole personality and all aspects of life. Although there is a high degree of heterogeneity in both its etiology and its characteristic behavioral patterns, the disorder is well-captured along the autistic triad. Currently, ASD status can be confirmed following an assessment of behavioral features, but there is a growing emphasis on conceptualizing autism as a spectrum, which allows for establishing a diagnosis based on the level of support need, free of discrete categories. Since ASD has a high genetic predominance, the number of genetic variations identified in the background of the condition is increasing exponentially as genetic testing methods are rapidly evolving. However, due to the huge amount of data to be analyzed, grouping the different DNA variations is still challenging. Therefore, in the present review, a multidimensional classification scheme was developed to accommodate most of the currently known genetic variants associated with autism. Genetic variations have been grouped according to six criteria (extent, time of onset, information content, frequency, number of genes involved, inheritance pattern), which are themselves not discrete categories, but form a coherent continuum in line with the autism spectrum approach.
Collapse
Affiliation(s)
- Éva Kereszturi
- Department of Molecular Biology, Semmelweis University, H-1085 Budapest, Hungary
| |
Collapse
|
6
|
Santos S, Martins B, Sereno J, Martins J, Castelo-Branco M, Gonçalves J. Neurobehavioral sex-related differences in Nf1 +/- mice: female show a "camouflaging"-type behavior. Biol Sex Differ 2023; 14:24. [PMID: 37101298 PMCID: PMC10131355 DOI: 10.1186/s13293-023-00509-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/17/2023] [Indexed: 04/28/2023] Open
Abstract
BACKGROUND Neurofibromatosis type 1 (NF1) is an inherited neurocutaneous disorder associated with neurodevelopmental disorders including autism spectrum disorder (ASD). This condition has been associated with an increase of gamma-aminobutyric acid (GABA) neurotransmission and, consequently, an excitation/inhibition imbalance associated with autistic-like behavior in both human and animal models. Here, we explored the influence of biological sex in the GABAergic system and behavioral alterations induced by the Nf1+/- mutation in a murine model. METHODS Juvenile male and female Nf1+/- mice and their wild-type (WT) littermates were used. Hippocampus size was assessed by conventional toluidine blue staining and structural magnetic resonance imaging (MRI). Hippocampal GABA and glutamate levels were determined by magnetic resonance spectroscopy (MRS), which was complemented by western blot for the GABA(A) receptor. Behavioral evaluation of on anxiety, memory, social communication, and repetitive behavior was performed. RESULTS We found that juvenile female Nf1+/- mice exhibited increased hippocampal GABA levels. Moreover, mutant female displays a more prominent anxious-like behavior together with better memory performance and social behavior. On the other hand, juvenile Nf1+/- male mice showed increased hippocampal volume and thickness, with a decrease in GABA(A) receptor levels. We observed that mutant males had higher tendency for repetitive behavior. CONCLUSIONS Our results suggested a sexually dimorphic impact of Nf1+/- mutation in hippocampal neurochemistry, and autistic-like behaviors. For the first time, we identified a "camouflaging"-type behavior in females of an animal model of ASD, which masked their autistic traits. Accordingly, like observed in human disorder, in this animal model of ASD, females show larger anxiety levels but better executive functions and production of normative social patterns, together with an imbalance of inhibition/excitation ratio. Contrary, males have more externalizing disorders, such as hyperactivity and repetitive behaviors, with memory deficits. The ability of females to camouflage their autistic traits creates a phenotypic evaluation challenge that mimics the diagnosis difficulty observed in humans. Thus, we propose the study of the Nf1+/- mouse model to better understand the sexual dimorphisms of ASD phenotypes and to create better diagnostic tools.
Collapse
Affiliation(s)
- Sofia Santos
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra, Portugal
- Institute of Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| | - Beatriz Martins
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra, Portugal
- Institute of Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| | - José Sereno
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra, Portugal
- Institute of Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| | - João Martins
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra, Portugal
- Institute of Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| | - Miguel Castelo-Branco
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra, Portugal.
- Institute of Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal.
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| | - Joana Gonçalves
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, Coimbra, Portugal.
- Institute of Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
7
|
Murari K, Abushaibah A, Rho JM, Turner RW, Cheng N. A clinically relevant selective ERK-pathway inhibitor reverses core deficits in a mouse model of autism. EBioMedicine 2023; 91:104565. [PMID: 37088035 PMCID: PMC10149189 DOI: 10.1016/j.ebiom.2023.104565] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 03/07/2023] [Accepted: 03/29/2023] [Indexed: 04/25/2023] Open
Abstract
BACKGROUND Extracellular signal-regulated kinase (ERK/MAPK) pathway in the brain is hypothesized to be a critical convergent node in the development of autism spectrum disorder. We reasoned that selectively targeting this pathway could reverse core autism-like phenotype in animal models. METHODS Here we tested a clinically relevant, selective inhibitor of ERK pathway, PD325901 (Mirdametinib), in a mouse model of idiopathic autism, the BTBR mice. FINDINGS We report that treating juvenile mice with PD325901 reduced ERK pathway activation, dose and duration-dependently reduced core disease-modeling deficits in sociability, vocalization and repetitive behavior, and reversed abnormal EEG signals. Further analysis revealed that subchronic treatment did not affect weight gain, locomotion, or neuronal density in the brain. Parallel treatment in the C57BL/6J mice did not alter their phenotype. INTERPRETATION Our data indicate that selectively inhibiting ERK pathway using PD325901 is beneficial in the BTBR model, thus further support the notion that ERK pathway is critically involved in the pathophysiology of autism. These results suggest that a similar approach could be applied to animal models of syndromic autism with dysregulated ERK signaling, to further test selectively targeting ERK pathway as a new approach for treating autism. FUNDING This has beenwork was supported by Alberta Children's Hospital Research Foundation (JMR & NC), University of Calgary Faculty of Veterinary Medicine (NC), Kids Brain Health Network (NC), and Natural Sciences and Engineering Research Council of Canada (NC).
Collapse
Affiliation(s)
- Kartikeya Murari
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Canada; Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Canada; Department of Electrical and Software Engineering, Schulich School of Engineering, University of Calgary, Canada
| | - Abdulrahman Abushaibah
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Canada; Bachelor of Health Sciences, Cumming School of Medicine, University of Calgary, Canada
| | - Jong M Rho
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Canada; Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Canada
| | - Ray W Turner
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Canada; Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Canada; Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Canada
| | - Ning Cheng
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Canada; Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Canada; Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Canada.
| |
Collapse
|
8
|
Fournier H, Calcagni N, Morice-Picard F, Quintard B. Psychosocial implications of rare genetic skin diseases affecting appearance on daily life experiences, emotional state, self-perception and quality of life in adults: a systematic review. Orphanet J Rare Dis 2023; 18:39. [PMID: 36823650 PMCID: PMC9951542 DOI: 10.1186/s13023-023-02629-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
BACKGROUND Since the beginning of human genetic research, there are very few publications sharing insights of the negative impact of rare genetic skin diseases (RGSD) on patients' experiences. This systematic review assessed the psychosocial implications of these conditions in terms of daily life experiences, emotional state, self-perception, and Quality of Life (QoL). METHODOLOGY A systematic review was carried out on albinism, neurofibromatosis type 1 (NF1), birthmarks and inherited ichthyosis. The PubMed, Scopus, PsycArticle, PsychInfo, Psychology and Behavioral Sciences Collection, and SOCindex databases were queried. Inclusion criteria were adult patients with one of these RGSDs. Simple descriptive statistics and qualitative content analysis were conducted to summarize the main results reported by the authors. RESULTS Of the 9987 articles retrieved, 48 articles were included: albinism (16), NF1 (16), inherited ichthyosis (10), birthmarks (6). The majority of the studies on albinism were conducted in Africa. Twenty-seven studies quantitatively assessed diverse psychological parameters: 13 showed a significant impact of the disease on QoL, five on emotional state, two on self-representation and two others on psychiatric comorbidities. Disease severity and visibility were good predictors of QoL (except for albinism). Body image and appearance concerns were also associated with QoL and emotional state. The 19 qualitative studies highlighted recurring themes across each of these diseases: discrimination and stigma during childhood and adolescence, discomfort in social interactions, guilt of transmission, the importance of social support from family and friends, altered daily life functioning, altered romantic and sex life, limited academic and professional aspirations, lack of interest and support from the medical field, and the unpredictability of the evolution of the disease. The only two mixed-method studies in this review were unable to contribute to any inferential analyses but could corroborate some of the qualitative findings. CONCLUSION These results showed that RGSDs have a significant impact on different aspects of patients' lives. This review has demonstrated that there is a real need for support systems for patients with these diseases. Such systems should be developed to provide them with necessary information and to guide them through an appropriate care pathway.
Collapse
Affiliation(s)
- Hugo Fournier
- Laboratoire de Psychologie (LabPsy) EA4139, Univ. Bordeaux, 3 ter Place de la Victoire, Bâtiment A - 1er étage, 33000 Bordeaux, France
| | | | | | - Bruno Quintard
- Laboratoire de Psychologie (LabPsy) EA4139, Univ. Bordeaux, 3 ter Place de la Victoire, Bâtiment A - 1er étage, 33000 Bordeaux, France
| |
Collapse
|
9
|
Singla R, Mishra A, Cao R. The trilateral interactions between mammalian target of rapamycin (mTOR) signaling, the circadian clock, and psychiatric disorders: an emerging model. Transl Psychiatry 2022; 12:355. [PMID: 36045116 PMCID: PMC9433414 DOI: 10.1038/s41398-022-02120-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 08/08/2022] [Accepted: 08/12/2022] [Indexed: 02/07/2023] Open
Abstract
Circadian (~24 h) rhythms in physiology and behavior are evolutionarily conserved and found in almost all living organisms. The rhythms are endogenously driven by daily oscillatory activities of so-called "clock genes/proteins", which are widely distributed throughout the mammalian brain. Mammalian (mechanistic) target of rapamycin (mTOR) signaling is a fundamental intracellular signal transduction cascade that controls important neuronal processes including neurodevelopment, synaptic plasticity, metabolism, and aging. Dysregulation of the mTOR pathway is associated with psychiatric disorders including autism spectrum disorders (ASD) and mood disorders (MD), in which patients often exhibit disrupted daily physiological rhythms and abnormal circadian gene expression in the brain. Recent work has found that the activities of mTOR signaling are temporally controlled by the circadian clock and exhibit robust circadian oscillations in multiple systems. In the meantime, mTOR signaling regulates fundamental properties of the central and peripheral circadian clocks, including period length, entrainment, and synchronization. Whereas the underlying mechanisms remain to be fully elucidated, increasing clinical and preclinical evidence support significant crosstalk between mTOR signaling, the circadian clock, and psychiatric disorders. Here, we review recent progress in understanding the trilateral interactions and propose an "interaction triangle" model between mTOR signaling, the circadian clock, and psychiatric disorders (focusing on ASD and MD).
Collapse
Affiliation(s)
- Rubal Singla
- grid.17635.360000000419368657Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812 USA
| | - Abhishek Mishra
- grid.17635.360000000419368657Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812 USA
| | - Ruifeng Cao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, 55812, USA. .,Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.
| |
Collapse
|
10
|
Sex- and age-related differences in autistic behaviours in children with neurofibromatosis type 1. J Autism Dev Disord 2022:10.1007/s10803-022-05571-6. [PMID: 35445370 DOI: 10.1007/s10803-022-05571-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2022] [Indexed: 10/18/2022]
Abstract
This study investigated sex and age differences in autistic behaviours in children with neurofibromatosis type 1 (NF1) who scored within the clinical range on the Social Responsiveness Scale - Second Edition (T score ≥ 60). Thirty-four males and 28 females (3-16 years) were assessed with the Autism Diagnostic Observation Schedule - Second Edition and Autism Diagnostic Interview - Revised. Across both measures, males exhibited greater social communication deficits relative to females. Age-related abatement of social communication difficulties was observed for males but not females. Conversely, no sex differences were found for restricted/repetitive behaviours, which were stable over time for both males and females. The findings are discussed within the context of broader neurodevelopmental considerations that are common in NF1.
Collapse
|
11
|
Foy AMH, Hudock RL, Shanley R, Pierpont EI. Social behavior in RASopathies and idiopathic autism. J Neurodev Disord 2022; 14:5. [PMID: 35021989 PMCID: PMC8753327 DOI: 10.1186/s11689-021-09414-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 12/24/2021] [Indexed: 11/22/2022] Open
Abstract
Background RASopathies are genetic syndromes that result from pathogenic variants in the RAS-MAPK cellular signaling pathway. These syndromes, which include neurofibromatosis type 1, Noonan syndrome, cardiofaciocutaneous syndrome, and Costello syndrome, are associated with a complex array of medical and behavioral health complications. Despite a heightened risk for social challenges and autism spectrum disorder (ASD), few studies have compared different aspects of social behavior across these conditions. It is also unknown whether the underlying neuropsychological characteristics that contribute to social competence and socially empathetic (“prosocial”) behaviors differ in children with RASopathies as compared to children with nonsyndromic (i.e., idiopathic) ASD. Methods In this cross-sectional, survey-based investigation, caregivers of preschool and school-aged children with RASopathies (n = 202) or with idiopathic ASD (n = 109) provided demographic, medical, and developmental information about their child, including psychiatric comorbidities. For children who were able to communicate verbally, caregivers also completed standardized rating scales to assess social competence and empathetic behavior as well as symptoms of hyperactivity/inattention and emotional problems. Results As compared to children with idiopathic ASD, children with RASopathies were rated as demonstrating more resilience in the domain of empathy relative to their overall social competence. Similarities and differences emerged in the psychological factors that predicted social behavior in these two groups. Stronger communication skills and fewer hyperactive-impulsive behaviors were associated with increased empathy and social competence for both groups. Greater emotional challenges were associated with lower social competence for children with RASopathies and stronger empathy for children with idiopathic ASD. Among children with RASopathy and a co-occurring ASD diagnosis, socially empathetic behaviors were observed more often as compared to children with idiopathic ASD. Conclusions Findings suggest that the development of social behavior among children with RASopathies involves a distinct pattern of strengths and weaknesses as compared to a behaviorally defined disorder (idiopathic ASD). Identification of areas of resilience as well as behavioral and social challenges will support more targeted intervention. Supplementary Information The online version contains supplementary material available at 10.1186/s11689-021-09414-w.
Collapse
Affiliation(s)
- Allison M H Foy
- Department of Pediatrics, Division of Clinical Behavioral Neuroscience, University of Minnesota Medical School, 2025 East River Parkway, Minneapolis, MN, 55414, USA.,Department of Educational Psychology, University of Wisconsin-Madison, Madison, USA
| | - Rebekah L Hudock
- Department of Pediatrics, Division of Clinical Behavioral Neuroscience, University of Minnesota Medical School, 2025 East River Parkway, Minneapolis, MN, 55414, USA
| | - Ryan Shanley
- Biostatistics Core, Masonic Cancer Center, University of Minnesota, Minneapolis, USA
| | - Elizabeth I Pierpont
- Department of Pediatrics, Division of Clinical Behavioral Neuroscience, University of Minnesota Medical School, 2025 East River Parkway, Minneapolis, MN, 55414, USA.
| |
Collapse
|
12
|
Chisholm AK, Haebich KM, Pride NA, Walsh KS, Lami F, Ure A, Maloof T, Brignell A, Rouel M, Granader Y, Maier A, Barton B, Darke H, Dabscheck G, Anderson VA, Williams K, North KN, Payne JM. Delineating the autistic phenotype in children with neurofibromatosis type 1. Mol Autism 2022; 13:3. [PMID: 34983638 PMCID: PMC8729013 DOI: 10.1186/s13229-021-00481-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 12/14/2021] [Indexed: 12/15/2022] Open
Abstract
Background Existing research has demonstrated elevated autistic behaviours in children with neurofibromatosis type 1 (NF1), but the autistic phenotype and its relationship to other neurodevelopmental manifestations of NF1 remains unclear. To address this gap, we performed detailed characterisation of autistic behaviours in children with NF1 and investigated their association with other common NF1 child characteristics. Methods Participants were drawn from a larger cross-sectional study examining autism in children with NF1. The population analysed in this study scored above threshold on the Social Responsiveness Scale-Second Edition (T-score ≥ 60; 51% larger cohort) and completed the Autism Diagnostic Interview-Revised (ADI-R) and/or the Autism Diagnostic Observation Schedule-Second Edition (ADOS-2). All participants underwent evaluation of their intellectual function, and behavioural data were collected via parent questionnaires. Results The study cohort comprised 68 children (3–15 years). Sixty-three per cent met the ADOS-2 ‘autism spectrum’ cut-off, and 34% exceeded the more stringent threshold for ‘autistic disorder’ on the ADI-R. Social communication symptoms were common and wide-ranging, while restricted and repetitive behaviours (RRBs) were most commonly characterised by ‘insistence on sameness’ (IS) behaviours such as circumscribed interests and difficulties with minor changes. Autistic behaviours were weakly correlated with hyperactive/impulsive attention deficit hyperactivity disorder (ADHD) symptoms but not with inattentive ADHD or other behavioural characteristics. Language and verbal IQ were weakly related to social communication behaviours but not to RRBs. Limitations Lack of genetic validation of NF1, no clinical diagnosis of autism, and a retrospective assessment of autistic behaviours in early childhood. Conclusions Findings provide strong support for elevated autistic behaviours in children with NF1. While these behaviours were relatively independent of other NF1 comorbidities, the importance of taking broader child characteristics into consideration when interpreting data from autism-specific measures in this population is highlighted. Social communication deficits appear similar to those observed in idiopathic autism and are coupled with a unique RRB profile comprising prominent IS behaviours. This autistic phenotype and its relationship to common NF1 comorbidities such as anxiety and executive dysfunction will be important to examine in future research. Current findings have important implications for the early identification of autism in NF1 and clinical management. Supplementary Information The online version contains supplementary material available at 10.1186/s13229-021-00481-3.
Collapse
Affiliation(s)
- Anita K Chisholm
- Murdoch Children's Research Institute, 50 Flemington Road, Parkville, VIC, 3052, Australia.,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, 3010, Australia.,The Royal Children's Hospital, 50 Flemington Road, Parkville, VIC, 3052, Australia
| | - Kristina M Haebich
- Murdoch Children's Research Institute, 50 Flemington Road, Parkville, VIC, 3052, Australia.,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Natalie A Pride
- Kids Neuroscience Centre, The Children's Hospital at Westmead, 178A Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Karin S Walsh
- Center for Neuroscience and Behavioral Medicine, Children's National Hospital, Michigan Avenue NW, Washington, DC, 20310, USA
| | - Francesca Lami
- Murdoch Children's Research Institute, 50 Flemington Road, Parkville, VIC, 3052, Australia.,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Alex Ure
- Murdoch Children's Research Institute, 50 Flemington Road, Parkville, VIC, 3052, Australia.,Department of Paediatrics, School of Clinical Sciences, Monash University, 246 Clayton Road, Clayton, VIC, 3168, Australia.,Developmental Paediatrics, Monash Children's Hospital, 246 Clayton Road, Clayton, VIC, 3168, Australia
| | - Tiba Maloof
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, 3010, Australia.,The Royal Children's Hospital, 50 Flemington Road, Parkville, VIC, 3052, Australia
| | - Amanda Brignell
- Murdoch Children's Research Institute, 50 Flemington Road, Parkville, VIC, 3052, Australia.,Department of Paediatrics, School of Clinical Sciences, Monash University, 246 Clayton Road, Clayton, VIC, 3168, Australia
| | - Melissa Rouel
- Kids Neuroscience Centre, The Children's Hospital at Westmead, 178A Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Yael Granader
- Center for Neuroscience and Behavioral Medicine, Children's National Hospital, Michigan Avenue NW, Washington, DC, 20310, USA
| | - Alice Maier
- Murdoch Children's Research Institute, 50 Flemington Road, Parkville, VIC, 3052, Australia.,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Belinda Barton
- Kids Neuroscience Centre, The Children's Hospital at Westmead, 178A Hawkesbury Road, Westmead, NSW, 2145, Australia.,Sydney Medical School, University of Sydney, Camperdown, NSW, 2050, Australia.,Children's Hospital Education Research Institute, The Children's Hospital at Westmead, 178A Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Hayley Darke
- Murdoch Children's Research Institute, 50 Flemington Road, Parkville, VIC, 3052, Australia
| | - Gabriel Dabscheck
- Murdoch Children's Research Institute, 50 Flemington Road, Parkville, VIC, 3052, Australia.,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, 3010, Australia.,The Royal Children's Hospital, 50 Flemington Road, Parkville, VIC, 3052, Australia
| | - Vicki A Anderson
- Murdoch Children's Research Institute, 50 Flemington Road, Parkville, VIC, 3052, Australia.,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, 3010, Australia.,The Royal Children's Hospital, 50 Flemington Road, Parkville, VIC, 3052, Australia
| | - Katrina Williams
- Murdoch Children's Research Institute, 50 Flemington Road, Parkville, VIC, 3052, Australia.,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, 3010, Australia.,Department of Paediatrics, School of Clinical Sciences, Monash University, 246 Clayton Road, Clayton, VIC, 3168, Australia.,Developmental Paediatrics, Monash Children's Hospital, 246 Clayton Road, Clayton, VIC, 3168, Australia
| | - Kathryn N North
- Murdoch Children's Research Institute, 50 Flemington Road, Parkville, VIC, 3052, Australia.,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jonathan M Payne
- Murdoch Children's Research Institute, 50 Flemington Road, Parkville, VIC, 3052, Australia. .,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, 3010, Australia. .,The Royal Children's Hospital, 50 Flemington Road, Parkville, VIC, 3052, Australia.
| |
Collapse
|
13
|
Lubbers K, Stijl EM, Dierckx B, Hagenaar DA, Ten Hoopen LW, Legerstee JS, de Nijs PFA, Rietman AB, Greaves-Lord K, Hillegers MHJ, Dieleman GC, Mous SE. Autism Symptoms in Children and Young Adults With Fragile X Syndrome, Angelman Syndrome, Tuberous Sclerosis Complex, and Neurofibromatosis Type 1: A Cross-Syndrome Comparison. Front Psychiatry 2022; 13:852208. [PMID: 35651825 PMCID: PMC9149157 DOI: 10.3389/fpsyt.2022.852208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 04/26/2022] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE The etiology of autism spectrum disorder (ASD) remains unclear, due to genetic heterogeneity and heterogeneity in symptoms across individuals. This study compares ASD symptomatology between monogenetic syndromes with a high ASD prevalence, in order to reveal syndrome specific vulnerabilities and to clarify how genetic variations affect ASD symptom presentation. METHODS We assessed ASD symptom severity in children and young adults (aged 0-28 years) with Fragile X Syndrome (FXS, n = 60), Angelman Syndrome (AS, n = 91), Neurofibromatosis Type 1 (NF1, n = 279) and Tuberous Sclerosis Complex (TSC, n = 110), using the Autism Diagnostic Observation Schedule and Social Responsiveness Scale. Assessments were part of routine clinical care at the ENCORE expertise center in Rotterdam, the Netherlands. First, we compared the syndrome groups on the ASD classification prevalence and ASD severity scores. Then, we compared individuals in our syndrome groups with an ASD classification to a non-syndromic ASD group (nsASD, n = 335), on both ASD severity scores and ASD symptom profiles. Severity scores were compared using MANCOVAs with IQ and gender as covariates. RESULTS Overall, ASD severity scores were highest for the FXS group and lowest for the NF1 group. Compared to nsASD, individuals with an ASD classification in our syndrome groups showed less problems on the instruments' social domains. We found a relative strength in the AS group on the social cognition, communication and motivation domains and a relative challenge in creativity; a relative strength of the NF1 group on the restricted interests and repetitive behavior scale; and a relative challenge in the FXS and TSC groups on the restricted interests and repetitive behavior domain. CONCLUSION The syndrome-specific strengths and challenges we found provide a frame of reference to evaluate an individual's symptoms relative to the larger syndromic population and to guide treatment decisions. Our findings support the need for personalized care and a dimensional, symptom-based diagnostic approach, in contrast to a dichotomous ASD diagnosis used as a prerequisite for access to healthcare services. Similarities in ASD symptom profiles between AS and FXS, and between NF1 and TSC may reflect similarities in their neurobiology. Deep phenotyping studies are required to link neurobiological markers to ASD symptomatology.
Collapse
Affiliation(s)
- Kyra Lubbers
- ENCORE Expertise Centre for Neurodevelopmental Disorders, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Child- and Adolescent Psychiatry and Psychology, Erasmus University Medical Center, Rotterdam, Netherlands.,Child Brain Center, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Eefje M Stijl
- ENCORE Expertise Centre for Neurodevelopmental Disorders, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Child- and Adolescent Psychiatry and Psychology, Erasmus University Medical Center, Rotterdam, Netherlands.,Child Brain Center, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Bram Dierckx
- ENCORE Expertise Centre for Neurodevelopmental Disorders, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Child- and Adolescent Psychiatry and Psychology, Erasmus University Medical Center, Rotterdam, Netherlands.,Child Brain Center, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Doesjka A Hagenaar
- ENCORE Expertise Centre for Neurodevelopmental Disorders, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Child- and Adolescent Psychiatry and Psychology, Erasmus University Medical Center, Rotterdam, Netherlands.,Child Brain Center, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of General Paediatrics, Erasmus MC, Rotterdam, Netherlands
| | - Leontine W Ten Hoopen
- ENCORE Expertise Centre for Neurodevelopmental Disorders, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Child- and Adolescent Psychiatry and Psychology, Erasmus University Medical Center, Rotterdam, Netherlands.,Child Brain Center, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jeroen S Legerstee
- ENCORE Expertise Centre for Neurodevelopmental Disorders, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Child- and Adolescent Psychiatry and Psychology, Erasmus University Medical Center, Rotterdam, Netherlands.,Child Brain Center, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Pieter F A de Nijs
- ENCORE Expertise Centre for Neurodevelopmental Disorders, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Child- and Adolescent Psychiatry and Psychology, Erasmus University Medical Center, Rotterdam, Netherlands.,Child Brain Center, Erasmus University Medical Center, Rotterdam, Netherlands
| | - André B Rietman
- ENCORE Expertise Centre for Neurodevelopmental Disorders, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Child- and Adolescent Psychiatry and Psychology, Erasmus University Medical Center, Rotterdam, Netherlands.,Child Brain Center, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Kirstin Greaves-Lord
- Department of Child- and Adolescent Psychiatry and Psychology, Erasmus University Medical Center, Rotterdam, Netherlands.,Clinical Psychology and Experimental Psychopathology Unit, Department of Psychology, Rijksuniversiteit Groningen, Groningen, Netherlands.,Yulius Mental Health, Dordrecht, Netherlands.,Jonx Autism Team Northern-Netherlands, Lentis Mental Health, Groningen, Netherlands
| | - Manon H J Hillegers
- ENCORE Expertise Centre for Neurodevelopmental Disorders, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Child- and Adolescent Psychiatry and Psychology, Erasmus University Medical Center, Rotterdam, Netherlands.,Child Brain Center, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Gwendolyn C Dieleman
- ENCORE Expertise Centre for Neurodevelopmental Disorders, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Child- and Adolescent Psychiatry and Psychology, Erasmus University Medical Center, Rotterdam, Netherlands.,Child Brain Center, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Sabine E Mous
- ENCORE Expertise Centre for Neurodevelopmental Disorders, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Child- and Adolescent Psychiatry and Psychology, Erasmus University Medical Center, Rotterdam, Netherlands.,Child Brain Center, Erasmus University Medical Center, Rotterdam, Netherlands
| | | |
Collapse
|
14
|
Vasic V, Jones MSO, Haslinger D, Knaus LS, Schmeisser MJ, Novarino G, Chiocchetti AG. Translating the Role of mTOR- and RAS-Associated Signalopathies in Autism Spectrum Disorder: Models, Mechanisms and Treatment. Genes (Basel) 2021; 12:genes12111746. [PMID: 34828352 PMCID: PMC8624393 DOI: 10.3390/genes12111746] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 12/23/2022] Open
Abstract
Mutations affecting mTOR or RAS signaling underlie defined syndromes (the so-called mTORopathies and RASopathies) with high risk for Autism Spectrum Disorder (ASD). These syndromes show a broad variety of somatic phenotypes including cancers, skin abnormalities, heart disease and facial dysmorphisms. Less well studied are the neuropsychiatric symptoms such as ASD. Here, we assess the relevance of these signalopathies in ASD reviewing genetic, human cell model, rodent studies and clinical trials. We conclude that signalopathies have an increased liability for ASD and that, in particular, ASD individuals with dysmorphic features and intellectual disability (ID) have a higher chance for disruptive mutations in RAS- and mTOR-related genes. Studies on rodent and human cell models confirm aberrant neuronal development as the underlying pathology. Human studies further suggest that multiple hits are necessary to induce the respective phenotypes. Recent clinical trials do only report improvements for comorbid conditions such as epilepsy or cancer but not for behavioral aspects. Animal models show that treatment during early development can rescue behavioral phenotypes. Taken together, we suggest investigating the differential roles of mTOR and RAS signaling in both human and rodent models, and to test drug treatment both during and after neuronal development in the available model systems.
Collapse
Affiliation(s)
- Verica Vasic
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany; (V.V.); (M.J.S.)
| | - Mattson S. O. Jones
- Autism Therapy and Research Center of Excellence, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, 60528 Frankfurt am Main, Germany; (M.S.O.J.); (D.H.)
- Center for Personalized Translational Epilepsy Research (CePTER), Goethe University Frankfurt, 60528 Frankfurt am Main, Germany
| | - Denise Haslinger
- Autism Therapy and Research Center of Excellence, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, 60528 Frankfurt am Main, Germany; (M.S.O.J.); (D.H.)
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria; (L.S.K.); (G.N.)
| | - Lisa S. Knaus
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria; (L.S.K.); (G.N.)
| | - Michael J. Schmeisser
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany; (V.V.); (M.J.S.)
- Focus Program Translational Neurosciences (FTN), University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Gaia Novarino
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria; (L.S.K.); (G.N.)
| | - Andreas G. Chiocchetti
- Autism Therapy and Research Center of Excellence, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, 60528 Frankfurt am Main, Germany; (M.S.O.J.); (D.H.)
- Center for Personalized Translational Epilepsy Research (CePTER), Goethe University Frankfurt, 60528 Frankfurt am Main, Germany
- Correspondence: ; Tel.: +49-69-6301-80658
| |
Collapse
|
15
|
Janusz JA, Klein-Tasman BP, Payne JM, Wolters PL, Thompson HL, Martin S, de Blank P, Ullrich N, Del Castillo A, Hussey M, Hardy KK, Haebich K, Rosser T, Toledo-Tamula MA, Walsh KS. Recommendations for Social Skills End Points for Clinical Trials in Neurofibromatosis Type 1. Neurology 2021; 97:S73-S80. [PMID: 34230205 PMCID: PMC8594002 DOI: 10.1212/wnl.0000000000012422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 06/07/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To review parent-report social skills measures to identify and recommend consensus outcomes for use in clinical trials of social deficit in children and adolescents (ages 6-18 years) with neurofibromatosis type 1 (NF1). METHODS Searches were conducted via PubMed and ClinicalTrials.gov to identity social skills outcome measures with English language versions used in clinical trials in the past 5 years with populations with known social skills deficits, including attention-deficit/hyperactivity disorder and autism spectrum disorder (ASD). Measures were rated by the Response Evaluation in Neurofibromatosis and Schwannomatosis (REiNS) Neurocognitive Committee on patient characteristics, use in published studies, domains assessed, availability of standard scores, psychometric properties, and feasibility to determine their appropriateness for use in NF1 clinical trials. RESULTS Two measures were ultimately recommended by the committee: the Social Responsiveness Scale-2 (SRS-2) and the Social Skills Improvement System-Rating Scale (SSIS-RS). CONCLUSIONS Each of the 2 measures assesses different aspects of social functioning. The SSIS-RS is appropriate for studies focused on broader social functioning; the SRS-2 is best for studies targeting problematic social behaviors associated with ASD. Researchers will need to consider the goals of their study when choosing a measure, and specific recommendations for their use are provided.
Collapse
Affiliation(s)
- Jennifer A Janusz
- From the Children's Hospital Colorado and University of Colorado School of Medicine (J.A.J.), Aurora; Department of Psychology (B.P.K.-T.), University of Wisconsin-Milwaukee; Murdoch Children's Research Institute and Department of Pediatrics (J.M.P., K.H.), University of Melbourne, Australia; Pediatric Oncology Branch (P.L.W., S.M., M.A.T.-T.), National Cancer Institute, Bethesda, MD; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; University of Cincinnati Medical Center (P.d.B.), OH; Boston Children's Hospital (N.U.), MA; Children's National Hospital, Gilbert NF Institute (A.d.C., K.K.H., K.S.W.), Washington, DC; Children's Tumor Foundation (M.H.), New York, NY; The George Washington School of Medicine (K.K.H., K.S.W.), Washington, DC; Children's Hospital Los Angeles (T.R.), CA; and Leidos Biomedical Research, Inc. (M.A.T.-T.), Frederick, MD.
| | - Bonita P Klein-Tasman
- From the Children's Hospital Colorado and University of Colorado School of Medicine (J.A.J.), Aurora; Department of Psychology (B.P.K.-T.), University of Wisconsin-Milwaukee; Murdoch Children's Research Institute and Department of Pediatrics (J.M.P., K.H.), University of Melbourne, Australia; Pediatric Oncology Branch (P.L.W., S.M., M.A.T.-T.), National Cancer Institute, Bethesda, MD; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; University of Cincinnati Medical Center (P.d.B.), OH; Boston Children's Hospital (N.U.), MA; Children's National Hospital, Gilbert NF Institute (A.d.C., K.K.H., K.S.W.), Washington, DC; Children's Tumor Foundation (M.H.), New York, NY; The George Washington School of Medicine (K.K.H., K.S.W.), Washington, DC; Children's Hospital Los Angeles (T.R.), CA; and Leidos Biomedical Research, Inc. (M.A.T.-T.), Frederick, MD
| | - Jonathan M Payne
- From the Children's Hospital Colorado and University of Colorado School of Medicine (J.A.J.), Aurora; Department of Psychology (B.P.K.-T.), University of Wisconsin-Milwaukee; Murdoch Children's Research Institute and Department of Pediatrics (J.M.P., K.H.), University of Melbourne, Australia; Pediatric Oncology Branch (P.L.W., S.M., M.A.T.-T.), National Cancer Institute, Bethesda, MD; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; University of Cincinnati Medical Center (P.d.B.), OH; Boston Children's Hospital (N.U.), MA; Children's National Hospital, Gilbert NF Institute (A.d.C., K.K.H., K.S.W.), Washington, DC; Children's Tumor Foundation (M.H.), New York, NY; The George Washington School of Medicine (K.K.H., K.S.W.), Washington, DC; Children's Hospital Los Angeles (T.R.), CA; and Leidos Biomedical Research, Inc. (M.A.T.-T.), Frederick, MD
| | - Pamela L Wolters
- From the Children's Hospital Colorado and University of Colorado School of Medicine (J.A.J.), Aurora; Department of Psychology (B.P.K.-T.), University of Wisconsin-Milwaukee; Murdoch Children's Research Institute and Department of Pediatrics (J.M.P., K.H.), University of Melbourne, Australia; Pediatric Oncology Branch (P.L.W., S.M., M.A.T.-T.), National Cancer Institute, Bethesda, MD; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; University of Cincinnati Medical Center (P.d.B.), OH; Boston Children's Hospital (N.U.), MA; Children's National Hospital, Gilbert NF Institute (A.d.C., K.K.H., K.S.W.), Washington, DC; Children's Tumor Foundation (M.H.), New York, NY; The George Washington School of Medicine (K.K.H., K.S.W.), Washington, DC; Children's Hospital Los Angeles (T.R.), CA; and Leidos Biomedical Research, Inc. (M.A.T.-T.), Frederick, MD
| | - Heather L Thompson
- From the Children's Hospital Colorado and University of Colorado School of Medicine (J.A.J.), Aurora; Department of Psychology (B.P.K.-T.), University of Wisconsin-Milwaukee; Murdoch Children's Research Institute and Department of Pediatrics (J.M.P., K.H.), University of Melbourne, Australia; Pediatric Oncology Branch (P.L.W., S.M., M.A.T.-T.), National Cancer Institute, Bethesda, MD; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; University of Cincinnati Medical Center (P.d.B.), OH; Boston Children's Hospital (N.U.), MA; Children's National Hospital, Gilbert NF Institute (A.d.C., K.K.H., K.S.W.), Washington, DC; Children's Tumor Foundation (M.H.), New York, NY; The George Washington School of Medicine (K.K.H., K.S.W.), Washington, DC; Children's Hospital Los Angeles (T.R.), CA; and Leidos Biomedical Research, Inc. (M.A.T.-T.), Frederick, MD
| | - Staci Martin
- From the Children's Hospital Colorado and University of Colorado School of Medicine (J.A.J.), Aurora; Department of Psychology (B.P.K.-T.), University of Wisconsin-Milwaukee; Murdoch Children's Research Institute and Department of Pediatrics (J.M.P., K.H.), University of Melbourne, Australia; Pediatric Oncology Branch (P.L.W., S.M., M.A.T.-T.), National Cancer Institute, Bethesda, MD; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; University of Cincinnati Medical Center (P.d.B.), OH; Boston Children's Hospital (N.U.), MA; Children's National Hospital, Gilbert NF Institute (A.d.C., K.K.H., K.S.W.), Washington, DC; Children's Tumor Foundation (M.H.), New York, NY; The George Washington School of Medicine (K.K.H., K.S.W.), Washington, DC; Children's Hospital Los Angeles (T.R.), CA; and Leidos Biomedical Research, Inc. (M.A.T.-T.), Frederick, MD
| | - Peter de Blank
- From the Children's Hospital Colorado and University of Colorado School of Medicine (J.A.J.), Aurora; Department of Psychology (B.P.K.-T.), University of Wisconsin-Milwaukee; Murdoch Children's Research Institute and Department of Pediatrics (J.M.P., K.H.), University of Melbourne, Australia; Pediatric Oncology Branch (P.L.W., S.M., M.A.T.-T.), National Cancer Institute, Bethesda, MD; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; University of Cincinnati Medical Center (P.d.B.), OH; Boston Children's Hospital (N.U.), MA; Children's National Hospital, Gilbert NF Institute (A.d.C., K.K.H., K.S.W.), Washington, DC; Children's Tumor Foundation (M.H.), New York, NY; The George Washington School of Medicine (K.K.H., K.S.W.), Washington, DC; Children's Hospital Los Angeles (T.R.), CA; and Leidos Biomedical Research, Inc. (M.A.T.-T.), Frederick, MD
| | - Nicole Ullrich
- From the Children's Hospital Colorado and University of Colorado School of Medicine (J.A.J.), Aurora; Department of Psychology (B.P.K.-T.), University of Wisconsin-Milwaukee; Murdoch Children's Research Institute and Department of Pediatrics (J.M.P., K.H.), University of Melbourne, Australia; Pediatric Oncology Branch (P.L.W., S.M., M.A.T.-T.), National Cancer Institute, Bethesda, MD; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; University of Cincinnati Medical Center (P.d.B.), OH; Boston Children's Hospital (N.U.), MA; Children's National Hospital, Gilbert NF Institute (A.d.C., K.K.H., K.S.W.), Washington, DC; Children's Tumor Foundation (M.H.), New York, NY; The George Washington School of Medicine (K.K.H., K.S.W.), Washington, DC; Children's Hospital Los Angeles (T.R.), CA; and Leidos Biomedical Research, Inc. (M.A.T.-T.), Frederick, MD
| | - Allison Del Castillo
- From the Children's Hospital Colorado and University of Colorado School of Medicine (J.A.J.), Aurora; Department of Psychology (B.P.K.-T.), University of Wisconsin-Milwaukee; Murdoch Children's Research Institute and Department of Pediatrics (J.M.P., K.H.), University of Melbourne, Australia; Pediatric Oncology Branch (P.L.W., S.M., M.A.T.-T.), National Cancer Institute, Bethesda, MD; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; University of Cincinnati Medical Center (P.d.B.), OH; Boston Children's Hospital (N.U.), MA; Children's National Hospital, Gilbert NF Institute (A.d.C., K.K.H., K.S.W.), Washington, DC; Children's Tumor Foundation (M.H.), New York, NY; The George Washington School of Medicine (K.K.H., K.S.W.), Washington, DC; Children's Hospital Los Angeles (T.R.), CA; and Leidos Biomedical Research, Inc. (M.A.T.-T.), Frederick, MD
| | - Maureen Hussey
- From the Children's Hospital Colorado and University of Colorado School of Medicine (J.A.J.), Aurora; Department of Psychology (B.P.K.-T.), University of Wisconsin-Milwaukee; Murdoch Children's Research Institute and Department of Pediatrics (J.M.P., K.H.), University of Melbourne, Australia; Pediatric Oncology Branch (P.L.W., S.M., M.A.T.-T.), National Cancer Institute, Bethesda, MD; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; University of Cincinnati Medical Center (P.d.B.), OH; Boston Children's Hospital (N.U.), MA; Children's National Hospital, Gilbert NF Institute (A.d.C., K.K.H., K.S.W.), Washington, DC; Children's Tumor Foundation (M.H.), New York, NY; The George Washington School of Medicine (K.K.H., K.S.W.), Washington, DC; Children's Hospital Los Angeles (T.R.), CA; and Leidos Biomedical Research, Inc. (M.A.T.-T.), Frederick, MD
| | - Kristina K Hardy
- From the Children's Hospital Colorado and University of Colorado School of Medicine (J.A.J.), Aurora; Department of Psychology (B.P.K.-T.), University of Wisconsin-Milwaukee; Murdoch Children's Research Institute and Department of Pediatrics (J.M.P., K.H.), University of Melbourne, Australia; Pediatric Oncology Branch (P.L.W., S.M., M.A.T.-T.), National Cancer Institute, Bethesda, MD; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; University of Cincinnati Medical Center (P.d.B.), OH; Boston Children's Hospital (N.U.), MA; Children's National Hospital, Gilbert NF Institute (A.d.C., K.K.H., K.S.W.), Washington, DC; Children's Tumor Foundation (M.H.), New York, NY; The George Washington School of Medicine (K.K.H., K.S.W.), Washington, DC; Children's Hospital Los Angeles (T.R.), CA; and Leidos Biomedical Research, Inc. (M.A.T.-T.), Frederick, MD
| | - Kristina Haebich
- From the Children's Hospital Colorado and University of Colorado School of Medicine (J.A.J.), Aurora; Department of Psychology (B.P.K.-T.), University of Wisconsin-Milwaukee; Murdoch Children's Research Institute and Department of Pediatrics (J.M.P., K.H.), University of Melbourne, Australia; Pediatric Oncology Branch (P.L.W., S.M., M.A.T.-T.), National Cancer Institute, Bethesda, MD; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; University of Cincinnati Medical Center (P.d.B.), OH; Boston Children's Hospital (N.U.), MA; Children's National Hospital, Gilbert NF Institute (A.d.C., K.K.H., K.S.W.), Washington, DC; Children's Tumor Foundation (M.H.), New York, NY; The George Washington School of Medicine (K.K.H., K.S.W.), Washington, DC; Children's Hospital Los Angeles (T.R.), CA; and Leidos Biomedical Research, Inc. (M.A.T.-T.), Frederick, MD
| | - Tena Rosser
- From the Children's Hospital Colorado and University of Colorado School of Medicine (J.A.J.), Aurora; Department of Psychology (B.P.K.-T.), University of Wisconsin-Milwaukee; Murdoch Children's Research Institute and Department of Pediatrics (J.M.P., K.H.), University of Melbourne, Australia; Pediatric Oncology Branch (P.L.W., S.M., M.A.T.-T.), National Cancer Institute, Bethesda, MD; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; University of Cincinnati Medical Center (P.d.B.), OH; Boston Children's Hospital (N.U.), MA; Children's National Hospital, Gilbert NF Institute (A.d.C., K.K.H., K.S.W.), Washington, DC; Children's Tumor Foundation (M.H.), New York, NY; The George Washington School of Medicine (K.K.H., K.S.W.), Washington, DC; Children's Hospital Los Angeles (T.R.), CA; and Leidos Biomedical Research, Inc. (M.A.T.-T.), Frederick, MD
| | - Mary Anne Toledo-Tamula
- From the Children's Hospital Colorado and University of Colorado School of Medicine (J.A.J.), Aurora; Department of Psychology (B.P.K.-T.), University of Wisconsin-Milwaukee; Murdoch Children's Research Institute and Department of Pediatrics (J.M.P., K.H.), University of Melbourne, Australia; Pediatric Oncology Branch (P.L.W., S.M., M.A.T.-T.), National Cancer Institute, Bethesda, MD; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; University of Cincinnati Medical Center (P.d.B.), OH; Boston Children's Hospital (N.U.), MA; Children's National Hospital, Gilbert NF Institute (A.d.C., K.K.H., K.S.W.), Washington, DC; Children's Tumor Foundation (M.H.), New York, NY; The George Washington School of Medicine (K.K.H., K.S.W.), Washington, DC; Children's Hospital Los Angeles (T.R.), CA; and Leidos Biomedical Research, Inc. (M.A.T.-T.), Frederick, MD
| | - Karin S Walsh
- From the Children's Hospital Colorado and University of Colorado School of Medicine (J.A.J.), Aurora; Department of Psychology (B.P.K.-T.), University of Wisconsin-Milwaukee; Murdoch Children's Research Institute and Department of Pediatrics (J.M.P., K.H.), University of Melbourne, Australia; Pediatric Oncology Branch (P.L.W., S.M., M.A.T.-T.), National Cancer Institute, Bethesda, MD; Department of Communication Sciences and Disorders (H.L.T.), California State University, Sacramento; University of Cincinnati Medical Center (P.d.B.), OH; Boston Children's Hospital (N.U.), MA; Children's National Hospital, Gilbert NF Institute (A.d.C., K.K.H., K.S.W.), Washington, DC; Children's Tumor Foundation (M.H.), New York, NY; The George Washington School of Medicine (K.K.H., K.S.W.), Washington, DC; Children's Hospital Los Angeles (T.R.), CA; and Leidos Biomedical Research, Inc. (M.A.T.-T.), Frederick, MD
| |
Collapse
|
16
|
Borrie SC, Plasschaert E, Callaerts-Vegh Z, Yoshimura A, D'Hooge R, Elgersma Y, Kushner SA, Legius E, Brems H. MEK inhibition ameliorates social behavior phenotypes in a Spred1 knockout mouse model for RASopathy disorders. Mol Autism 2021; 12:53. [PMID: 34311771 PMCID: PMC8314535 DOI: 10.1186/s13229-021-00458-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 07/12/2021] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND RASopathies are a group of disorders that result from mutations in genes coding for proteins involved in regulating the Ras-MAPK signaling pathway, and have an increased incidence of autism spectrum disorder (ASD). Legius syndrome is a rare RASopathy caused by loss-of-function mutations in the SPRED1 gene. The patient phenotype is similar to, but milder than, Neurofibromatosis type 1-another RASopathy caused by loss-of-function mutations in the NF1 gene. RASopathies exhibit increased activation of Ras-MAPK signaling and commonly manifest with cognitive impairments and ASD. Here, we investigated if a Spred1-/- mouse model for Legius syndrome recapitulates ASD-like symptoms, and whether targeting the Ras-MAPK pathway has therapeutic potential in this RASopathy mouse model. METHODS We investigated social and communicative behaviors in Spred1-/- mice and probed therapeutic mechanisms underlying the observed behavioral phenotypes by pharmacological targeting of the Ras-MAPK pathway with the MEK inhibitor PD325901. RESULTS Spred1-/- mice have robust increases in social dominance in the automated tube test and reduced adult ultrasonic vocalizations during social communication. Neonatal ultrasonic vocalization was also altered, with significant differences in spectral properties. Spred1-/- mice also exhibit impaired nesting behavior. Acute MEK inhibitor treatment in adulthood with PD325901 reversed the enhanced social dominance in Spred1-/- mice to normal levels, and improved nesting behavior in adult Spred1-/- mice. LIMITATIONS This study used an acute treatment protocol to administer the drug. It is not known what the effects of longer-term treatment would be on behavior. Further studies titrating the lowest dose of this drug that is required to alter Spred1-/- social behavior are still required. Finally, our findings are in a homozygous mouse model, whereas patients carry heterozygous mutations. These factors should be considered before any translational conclusions are drawn. CONCLUSIONS These results demonstrate for the first time that social behavior phenotypes in a mouse model for RASopathies (Spred1-/-) can be acutely reversed. This highlights a key role for Ras-MAPK dysregulation in mediating social behavior phenotypes in mouse models for ASD, suggesting that proper regulation of Ras-MAPK signaling is important for social behavior.
Collapse
Affiliation(s)
- Sarah C Borrie
- Department of Human Genetics, KU Leuven, O&N1 Herestraat 49, Box 607, 3000, Leuven, Belgium
| | - Ellen Plasschaert
- Department of Human Genetics, KU Leuven, O&N1 Herestraat 49, Box 607, 3000, Leuven, Belgium
| | | | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Rudi D'Hooge
- Laboratory for Biological Psychology, KU Leuven, Leuven, Belgium
| | - Ype Elgersma
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Department of Neuroscience, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Steven A Kushner
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Department of Psychiatry, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Eric Legius
- Department of Human Genetics, KU Leuven, O&N1 Herestraat 49, Box 607, 3000, Leuven, Belgium
| | - Hilde Brems
- Department of Human Genetics, KU Leuven, O&N1 Herestraat 49, Box 607, 3000, Leuven, Belgium.
| |
Collapse
|
17
|
Begum-Ali J, Kolesnik-Taylor A, Quiroz I, Mason L, Garg S, Green J, Johnson MH, Jones EJH. Early differences in auditory processing relate to Autism Spectrum Disorder traits in infants with Neurofibromatosis Type I. J Neurodev Disord 2021; 13:22. [PMID: 34049498 PMCID: PMC8161667 DOI: 10.1186/s11689-021-09364-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 04/03/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Sensory modulation difficulties are common in children with conditions such as Autism Spectrum Disorder (ASD) and could contribute to other social and non-social symptoms. Positing a causal role for sensory processing differences requires observing atypical sensory reactivity prior to the emergence of other symptoms, which can be achieved through prospective studies. METHODS In this longitudinal study, we examined auditory repetition suppression and change detection at 5 and 10 months in infants with and without Neurofibromatosis Type 1 (NF1), a condition associated with higher likelihood of developing ASD. RESULTS In typically developing infants, suppression to vowel repetition and enhanced responses to vowel/pitch change decreased with age over posterior regions, becoming more frontally specific; age-related change was diminished in the NF1 group. Whilst both groups detected changes in vowel and pitch, the NF1 group were largely slower to show a differentiated neural response. Auditory responses did not relate to later language, but were related to later ASD traits. CONCLUSIONS These findings represent the first demonstration of atypical brain responses to sounds in infants with NF1 and suggest they may relate to the likelihood of later ASD.
Collapse
Affiliation(s)
- Jannath Begum-Ali
- Centre for Brain and Cognitive Development, Birkbeck, University of London, Henry Wellcome Building, Malet Street, London, WC1E 7HX, UK.
| | - Anna Kolesnik-Taylor
- Centre for Brain and Cognitive Development, Birkbeck, University of London, Henry Wellcome Building, Malet Street, London, WC1E 7HX, UK
- Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Isabel Quiroz
- Centre for Brain and Cognitive Development, Birkbeck, University of London, Henry Wellcome Building, Malet Street, London, WC1E 7HX, UK
| | - Luke Mason
- Centre for Brain and Cognitive Development, Birkbeck, University of London, Henry Wellcome Building, Malet Street, London, WC1E 7HX, UK
| | - Shruti Garg
- Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
| | - Jonathan Green
- Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
| | - Mark H Johnson
- Centre for Brain and Cognitive Development, Birkbeck, University of London, Henry Wellcome Building, Malet Street, London, WC1E 7HX, UK
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - Emily J H Jones
- Centre for Brain and Cognitive Development, Birkbeck, University of London, Henry Wellcome Building, Malet Street, London, WC1E 7HX, UK.
| |
Collapse
|
18
|
Santoro C, Giugliano T, Bernardo P, Palladino F, Torella A, Del Vecchio Blanco F, Onore ME, Carotenuto M, Nigro V, Piluso G. A novel RAB39B mutation and concurrent de novo NF1 mutation in a boy with neurofibromatosis type 1, intellectual disability, and autism: a case report. BMC Neurol 2020; 20:327. [PMID: 32873259 PMCID: PMC7460788 DOI: 10.1186/s12883-020-01911-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Mutations in RAB39B at Xq28 causes a rare form of X-linked intellectual disability (ID) and Parkinson's disease. Neurofibromatosis type 1 (NF1) is caused by heterozygous mutations in NF1 occurring de novo in about 50% of cases, usually due to paternal gonadal mutations. This case report describes clinical and genetic findings in a boy with the occurrence of two distinct causative mutations in NF1 and RAB39B explaining the observed phenotype. CASE PRESENTATION Here we report a 7-year-old boy with multiple café-au-lait macules (CALMs) and freckling, severe macrocephaly, peculiar facial gestalt, severe ID with absent speech, epilepsy, autistic traits, self-harming, and aggressiveness. Proband is an only child born to a father aged 47. Parents did not present signs of NF1, while a maternal uncle showed severe ID, epilepsy, and tremors.By RNA analysis of NF1, we identified a de novo splicing variant (NM_000267.3:c.6579+2T>C) in proband, which explained NF1 clinical features but not the severe ID, behavioral problems, and aggressiveness. Family history suggested an X-linked condition and massively parallel sequencing of X-exome identified a novel RAB39B mutation (NM_171998.2:c.436_447del) in proband, his mother, and affected maternal uncle, subsequently validated by Sanger sequencing in these and other family members. CONCLUSIONS The case presented here highlights how concurrent genetic defects should be considered in NF1 patients when NF1 mutations cannot reasonably explain all the observed clinical features.
Collapse
Affiliation(s)
- Claudia Santoro
- Department of Physical and Mental Health, and Preventive Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy.,Department of Women, Children, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Teresa Giugliano
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via Luigi De Crecchio,7 -, 80138, Naples, Italy
| | - Pia Bernardo
- Department of Neurosciences, Pediatric Hospital Santobono-Pausilipon, Naples, Italy
| | - Federica Palladino
- Department of Women, Children, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Annalaura Torella
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via Luigi De Crecchio,7 -, 80138, Naples, Italy
| | - Francesca Del Vecchio Blanco
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via Luigi De Crecchio,7 -, 80138, Naples, Italy
| | - Maria Elena Onore
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via Luigi De Crecchio,7 -, 80138, Naples, Italy
| | - Marco Carotenuto
- Department of Physical and Mental Health, and Preventive Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Vincenzo Nigro
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via Luigi De Crecchio,7 -, 80138, Naples, Italy.,Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Giulio Piluso
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via Luigi De Crecchio,7 -, 80138, Naples, Italy.
| |
Collapse
|
19
|
Brain-wide structural and functional disruption in mice with oligodendrocyte-specific Nf1 deletion is rescued by inhibition of nitric oxide synthase. Proc Natl Acad Sci U S A 2020; 117:22506-22513. [PMID: 32839340 PMCID: PMC7486714 DOI: 10.1073/pnas.2008391117] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
This study assessed the effects of myelin decompaction on motor behavior and brain-wide structural and functional connectivity, and the effect of nitric oxide synthase inhibition by N-nitro-l-arginine methyl ester (L-NAME) on these imaging measures. We report that inducible oligodendrocyte-specific inactivation of the Nf1 gene, which causes myelin decompaction, results in reduced initial motor coordination. Using diffusion-based magnetic resonance imaging (MRI), we show reduced myelin integrity, and using functional MRI, we show reduced functional connectivity in awake passive mice. L-NAME administration results in rescue of the pathology at the mesoscopic level, as measured using imaging procedures that can be directly applied to humans to study treatment efficacy in clinical trials. Neurofibromin gene (NF1) mutation causes neurofibromatosis type 1 (NF1), a disorder in which brain white matter deficits identified by neuroimaging are common, yet of unknown cellular etiology. In mice, Nf1 loss in adult oligodendrocytes causes myelin decompaction and increases oligodendrocyte nitric oxide (NO) levels. Nitric oxide synthase (NOS) inhibitors rescue this pathology. Whether oligodendrocyte pathology is sufficient to affect brain-wide structure and account for NF1 imaging findings is unknown. Here we show that Nf1 gene inactivation in adult oligodendrocytes (Plp-Nf1fl/+ mice) results in a motor coordination deficit. Magnetic resonance imaging in awake mice showed that fractional anisotropy is reduced in Plp-Nf1fl/+ corpus callosum and that interhemispheric functional connectivity in the motor cortex is also reduced, consistent with disrupted myelin integrity. Furthermore, NOS-specific inhibition rescued both measures. These results suggest that oligodendrocyte defects account for aspects of brain dysfunction in NF1 that can be identified by neuroimaging and ameliorated by NOS inhibition.
Collapse
|
20
|
Walsh KS, del Castillo A, Kennedy T, Karim AI, Semerjian C. A Review of Psychological, Social, and Behavioral Functions in the RASopathies. JOURNAL OF PEDIATRIC NEUROPSYCHOLOGY 2020. [DOI: 10.1007/s40817-020-00088-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
21
|
Cohen R, Halevi A, Aharoni S, Aronson B, Diamond G. Impairments in communication and social interaction in children with neurofibromatosis type 1: Characteristics and role of ADHD and language delay. APPLIED NEUROPSYCHOLOGY. CHILD 2020; 11:220-225. [PMID: 32569512 DOI: 10.1080/21622965.2020.1780924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Background: Neurofibromatosis type 1 (NF1) is a multisystem neurocutaneous disorder with increased risk of tumor formation and higher incidence of autism spectrum disorder (ASD) than the general population. The aim of the study was to assess the presence of ASD symptoms in young children with NF1 and to examine their potential association with attention deficit hyperactivity disorder (ADHD) and speech delay.Methods: The cohort included 30 patients with NF1 attending the multidisciplinary NF1 clinic of a tertiary pediatric medical center from September 2015 through September 2016. The parents/caregivers completed the Social Communication Questionnaire (SCQ) and the Vineland Adaptive Behavior Scales (VABS II).Results: Sixteen patients (53%) had a previous diagnosis of ADHD. There was a positive association between the presence of ADHD and a low score on the VABS II interpersonal relationships subscale of the Socialization domain. Language delay, documented in 12 children (40%), also correlated with a low interpersonal relationships score.Conclusions: ADHD appears to be more a marker than an actual independent risk factor of ASD in NF1. The early evaluation of children with NF1 for interpersonal communication problems and ASD, especially those with a speech delay or ADHD, will alert clinicians to initiate appropriate and timely treatment.
Collapse
Affiliation(s)
- Rony Cohen
- Department of Pediatric Neurology and Epilepsy Center, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ayelet Halevi
- Department of Pediatric Neurology and Epilepsy Center, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sharon Aharoni
- Department of Pediatric Neurology and Epilepsy Center, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Bosmat Aronson
- The Child Development and Rehabilitation Institute, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.,Department of Developmental and Educational Psychology, Clalit Health Services, Tel Aviv, Israel
| | - Gary Diamond
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,The Child Development and Rehabilitation Institute, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| |
Collapse
|
22
|
Lukkes JL, Drozd HP, Fitz SD, Molosh AI, Clapp DW, Shekhar A. Guanfacine treatment improves ADHD phenotypes of impulsivity and hyperactivity in a neurofibromatosis type 1 mouse model. J Neurodev Disord 2020; 12:2. [PMID: 31941438 PMCID: PMC6961243 DOI: 10.1186/s11689-019-9304-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 12/16/2019] [Indexed: 02/07/2023] Open
Abstract
Background Neurofibromatosis type 1 (NF1) is an autosomal dominant disorder with a mutation in one copy of the neurofibromin gene (NF1+/−). Even though approximately 40–60% of children with NF1 meet the criteria for attention deficit hyperactivity disorder (ADHD), very few preclinical studies, if any, have investigated alterations in impulsivity and risk-taking behavior. Mice with deletion of a single NF1 gene (Nf1+/−) recapitulate many of the phenotypes of NF1 patients. Methods We compared wild-type (WT) and Nf1+/− mouse strains to investigate differences in impulsivity and hyperactivity using the delay discounting task (DDT), cliff avoidance reaction (CAR) test, and open field. We also investigated whether treatment with the clinically effective alpha-2A adrenergic receptor agonist, guanfacine (0.3 mg/kg, i.p.), would reverse deficits observed in behavioral inhibition. Results Nf1+/− mice chose a higher percentage of smaller rewards when both 10- and 20-s delays were administered compared to WT mice, suggesting Nf1+/− mice are more impulsive. When treated with guanfacine (0.3 mg/kg, i.p.), Nf1+/− mice exhibited decreased impulsive choice by waiting for the larger, delayed reward. Nf1+/− mice also exhibited deficits in behavioral inhibition compared to WT mice in the CAR test by repetitively entering the outer edge of the platform where they risk falling. Treatment with guanfacine ameliorated these deficits. In addition, Nf1+/− mice exhibited hyperactivity as increased distance was traveled compared to WT controls in the open field. This hyperactivity in Nf1+/− mice was reduced with guanfacine pre-treatment. Conclusions Overall, our study confirms that Nf1+/− mice exhibit deficits in behavioral inhibition in multiple contexts, a key feature of ADHD, and can be used as a model system to identify alterations in neural circuitry associated with symptoms of ADHD in children with NF1.
Collapse
Affiliation(s)
- J L Lukkes
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, Indianapolis, IN, 46202, USA.
| | - H P Drozd
- Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, Indianapolis, IN, 46202, USA.,Program in Medical Neurosciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - S D Fitz
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, Indianapolis, IN, 46202, USA
| | - A I Molosh
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, Indianapolis, IN, 46202, USA
| | - D W Clapp
- Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, Indianapolis, IN, 46202, USA.,Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A Shekhar
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, Indianapolis, IN, 46202, USA.,Program in Medical Neurosciences, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana Clinical and Translation Sciences Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
23
|
Deutsch SI, Burket JA. An Evolving Therapeutic Rationale for Targeting the α 7 Nicotinic Acetylcholine Receptor in Autism Spectrum Disorder. Curr Top Behav Neurosci 2020; 45:167-208. [PMID: 32468495 DOI: 10.1007/7854_2020_136] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Abnormalities of cholinergic nuclei, cholinergic projections, and cholinergic receptors, as well as abnormalities of growth factors involved in the maturation and maintenance of cholinergic neurons, have been described in postmortem brains of persons with autism spectrum disorder (ASD). Further, microdeletions of the 15q13.3 locus that encompasses CHRNA7, the gene coding the α7 nicotinic acetylcholine receptor (α7 nAChR), are associated with a spectrum of neurodevelopmental disorders, including ASD. The heterozygous 15q13.3 microdeletion syndrome suggests that diminished or impaired transduction of the acetylcholine (ACh) signal by the α7 nAChR can be a pathogenic mechanism of ASD. The α7 nAChR has a role in regulating the firing and function of parvalbumin (PV)-expressing GABAergic projections, which synchronize the oscillatory output of assemblies of pyramidal neurons onto which they project. Synchronous oscillatory output is an electrophysiological substrate for higher executive functions, such as working memory, and functional connectivity between discrete anatomic areas of the brain. The α7 nAChR regulates PV expression and works cooperatively with the co-expressed NMDA receptor in subpopulations of GABAergic interneurons in mouse models of ASD. An evolving literature supports therapeutic exploration of selectively targeted cholinergic interventions for the treatment of ASD, especially compounds that target the α7 nAChR subtype. Importantly, development and availability of high-affinity, brain-penetrable, α7 nAChR-selective agonists, partial agonists, allosteric agonists, and positive allosteric modulators (PAMs) should facilitate "proof-of-principle/concept" clinical trials. nAChRs are pentameric allosteric proteins that function as ligand-gated ion channel receptors constructed from five constituent polypeptide subunits, all of which share a common structural motif. Importantly, in addition to α7 nAChR-gated Ca2+ conductance causing membrane depolarization, there are emerging data consistent with possible metabotropic functions of this ionotropic receptor. The ability of α7-selective type II PAMs to "destabilize" the desensitized state and promote ion channel opening may afford them therapeutic advantages over orthosteric agonists. The current chapter reviews historic and recent literature supporting selective therapeutic targeting of the α7 nAChR in persons affected with ASD.
Collapse
Affiliation(s)
- Stephen I Deutsch
- Department of Psychiatry and Behavioral Sciences, Eastern Virginia Medical School, Norfolk, VA, USA.
| | - Jessica A Burket
- Department of Molecular Biology and Chemistry, Christopher Newport University, Newport News, VA, USA
| |
Collapse
|
24
|
Kang E, Kim YM, Seo GH, Oh A, Yoon HM, Ra YS, Kim EK, Kim H, Heo SH, Kim GH, Osborn MJ, Tolar J, Yoo HW, Lee BH. Phenotype categorization of neurofibromatosis type I and correlation to NF1 mutation types. J Hum Genet 2019; 65:79-89. [PMID: 31776437 DOI: 10.1038/s10038-019-0695-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 11/04/2019] [Accepted: 11/12/2019] [Indexed: 01/17/2023]
Abstract
Neurofibromatosis type 1 (NF1) is caused by heterozygous mutation in the NF1 gene. NF1 is one of the most common human genetic diseases. However, the overall genotype-phenotype correlation has not been known, due to a wide spectrum of genotypic and phenotypic heterogeneity. Here we describe the detailed clinical and genetic features of 427 Korean NF1 patients from 389 unrelated families. Long range PCR and sequencing of genomic DNA with multiplex ligation-dependent probe amplification analysis identified 250 different NF1 mutations in 363 families (93%), including 94 novel mutations. With an emphasis on phenotypes requiring medical attention (classified and termed: NF1+), we investigated the correlation of NF1+ and mutation types. NF1+ was more prevalent in patients with truncating/splicing mutations and large deletions than in those with missense mutations (59.6%, 64.3% vs. 36.6%, p = 0.001). This difference was especially significant in the patients younger than age 19 years. The number of items in NF1+ was a higher in the former groups (0.95 ± 0.06, 1.18 ± 0.20 vs. 0.56 ± 0.10, p = 0.002). These results suggest that mutation types are associated not only with higher prevalence of severe phenotypes in NF1 but also with their earlier onset.
Collapse
Affiliation(s)
- Eungu Kang
- Department of Pediatrics, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea
| | - Yoon-Myung Kim
- Department of Pediatrics, Gangneung Asan Hospital, Gangneung, Republic of Korea
| | - Go Hun Seo
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88, Olympic-ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Arum Oh
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88, Olympic-ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Hee Mang Yoon
- Department of Radiology and Research Institute of Radiology, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Young-Shin Ra
- Departments of Neurosurgery, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eun Key Kim
- Department of Plastic Surgery, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Heyry Kim
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88, Olympic-ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Sun-Hee Heo
- Asan Institute for Life Sciences, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Gu-Hwan Kim
- Medical Genetics Center, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Mark J Osborn
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Jakub Tolar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Han-Wook Yoo
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88, Olympic-ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea.,Medical Genetics Center, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Beom Hee Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88, Olympic-ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea. .,Medical Genetics Center, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
25
|
Wester Oxelgren U, Åberg M, Myrelid Å, Annerén G, Westerlund J, Gustafsson J, Fernell E. Autism needs to be considered in children with Down Syndrome. Acta Paediatr 2019; 108:2019-2026. [PMID: 31090964 DOI: 10.1111/apa.14850] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 05/08/2019] [Accepted: 05/13/2019] [Indexed: 02/01/2023]
Abstract
AIM To analyse levels and profiles of autism symptoms in children with Down Syndrome (DS) with and without diagnosed autism spectrum disorder (ASD) and to specifically study the groups with severe Intellectual Disability (ID). METHODS From a population-based cohort of 60 children with DS (age 5-17 years) with 41 participating children, scores obtained from the Autism Diagnostic Observation Schedule (ADOS) Module-1 algorithm were compared between those with and without diagnosed ASD. Children with DS and ASD were also compared to a cohort of children with idiopathic ASD, presented in the ADOS manual. RESULTS Children with DS and ASD had significantly higher ADOS scores in all domains compared to those without ASD. When the groups with DS, with and without ASD, were restricted to those with severe ID, the difference remained. When the children with DS and ASD and the idiopathic autism group were compared, the ADOS profiles were similar. CONCLUSION A considerable proportion of children with DS has ASD, but there is also a group of children with DS and severe ID without autism. There is a need to increase awareness of the high prevalence of autism in children with DS to ensure that appropriate measures and care are provided.
Collapse
Affiliation(s)
| | - Marie Åberg
- Department of Health and Habilitation Kungsgärdet Center Uppsala Sweden
| | - Åsa Myrelid
- Department of Women’s and Children´s Health Uppsala University Uppsala Sweden
| | - Göran Annerén
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory Uppsala University Uppsala Sweden
| | - Joakim Westerlund
- Department of Psychology Stockholm University Stockholm Sweden
- Gillberg Neuropsychiatry Centre, Department of Neuroscience and Physiology Gothenburg University Gothenburg Sweden
| | - Jan Gustafsson
- Department of Women’s and Children´s Health Uppsala University Uppsala Sweden
| | - Elisabeth Fernell
- Gillberg Neuropsychiatry Centre, Department of Neuroscience and Physiology Gothenburg University Gothenburg Sweden
| |
Collapse
|
26
|
Haebich KM, Pride NA, Walsh KS, Chisholm A, Rouel M, Maier A, Anderson V, Barton B, Silk T, Korgaonkar M, Seal M, Lami F, Lorenzo J, Williams K, Dabscheck G, Rae CD, Kean M, North KN, Payne JM. Understanding autism spectrum disorder and social functioning in children with neurofibromatosis type 1: protocol for a cross-sectional multimodal study. BMJ Open 2019; 9:e030601. [PMID: 31558455 PMCID: PMC6773330 DOI: 10.1136/bmjopen-2019-030601] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION Children with the single-gene disorder neurofibromatosis type 1 (NF1) appear to be at an increased risk for autism spectrum disorder (ASD) and exhibit a unique social-cognitive phenotype compared with children with idiopathic ASD. A complete framework is required to better understand autism in NF1, from neurobiological levels through to behavioural and functional outcomes. The primary aims of this study are to establish the frequency of ASD in children with NF1, examine the social cognitive phenotype, investigate the neuropsychological processes contributing to ASD symptoms and poor social functioning in children with NF1, and to investigate novel structural and functional neurobiological markers of ASD and social dysfunction in NF1. The secondary aim of this study is to compare the neuropsychological and neurobiological features of ASD in children with NF1 to a matched group of patients with idiopathic ASD. METHODS AND ANALYSIS This is an international, multisite, prospective, cross-sectional cohort study of children with NF1, idiopathic ASD and typically developing (TD) controls. Participants will be 200 children with NF1 (3-15 years of age), 70 TD participants (3-15 years) and 35 children with idiopathic ASD (7-15 years). Idiopathic ASD and NF1 cases will be matched on age, sex and intelligence. All participants will complete cognitive testing and parents will rate their child's behaviour on standardised questionnaires. Neuroimaging will be completed by a subset of participants aged 7 years and older. Children with NF1 that screen at risk for ASD on the parent-rated Social Responsiveness Scale 2nd Edition will be invited back to complete the Autism Diagnostic Observation Scale 2nd Edition and Autism Diagnostic Interview-Revised to determine whether they fulfil ASD diagnostic criteria. ETHICS AND DISSEMINATION This study has hospital ethics approval and the results will be disseminated through peer-reviewed publications and international conferences.
Collapse
Affiliation(s)
- Kristina M Haebich
- Brain and Mind, Murdoch Children's Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Science, University of Melbourne, Parkville, VIC, Australia
| | - Natalie A Pride
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Westmead, NSW, Australia
- Discipline of Child and Adolescent Health, University of Sydney Medical School, Westmead, NSW, Australia
| | - Karin S Walsh
- Center for Neuroscience and Behavioral Medicine, Children's National Health System, Washington, DC, United States
- Departments of Pediatrics and Psychiatry, The George Washington University School of Medicine, Washington, DC, United States
| | - Anita Chisholm
- Brain and Mind, Murdoch Children's Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Science, University of Melbourne, Parkville, VIC, Australia
| | - Melissa Rouel
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - Alice Maier
- Brain and Mind, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Vicki Anderson
- Brain and Mind, Murdoch Children's Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Science, University of Melbourne, Parkville, VIC, Australia
| | - Belinda Barton
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Westmead, NSW, Australia
- Discipline of Child and Adolescent Health, University of Sydney Medical School, Westmead, NSW, Australia
- Children's Hospital Education Research Institute, The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - Tim Silk
- School of Psychology, Deakin University, Burwood, VIC, Australia
| | - Mayuresh Korgaonkar
- Brain Dynamics Centre, Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
| | - Marc Seal
- Developmental Imaging, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Francesca Lami
- Brain and Mind, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Jennifer Lorenzo
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - Katrina Williams
- Department of Paediatrics, Monash University, Clayton, VIC, Australia
| | - Gabriel Dabscheck
- Department of Neurology, Royal Children's Hospital Melbourne, Parkville, VIC, Australia
| | - Caroline D Rae
- Neuroscience Research Australia, University of New South Wales, Randwick, NSW, Australia
| | - Michael Kean
- Imaging Department, Royal Children's Hospital Melbourne, Parkville, VIC, Australia
| | - Kathryn N North
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Science, University of Melbourne, Parkville, VIC, Australia
- Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Jonathan M Payne
- Brain and Mind, Murdoch Children's Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Science, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
27
|
Lewis AK, Porter MA, Williams TA, Bzishvili S, North KN, Payne JM. Attention to faces in social context in children with neurofibromatosis type 1. Dev Med Child Neurol 2019; 61:174-180. [PMID: 29873078 DOI: 10.1111/dmcn.13928] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/18/2018] [Indexed: 02/06/2023]
Abstract
AIM To examine visual attention to faces within social scenes in children with neurofibromatosis type 1 (NF1) and typically developing peers. METHOD Using eye-tracking technology we investigated the time taken to fixate on a face and the percentage of time spent attending to faces relative to the rest of the screen within social scenes in 24 children with NF1 (17 females, seven males; mean age 10y 4mo [SD 1y 9mo]). Results were compared with those of 24 age-matched typically developing controls (11 females, 13 males; mean age 10y 3mo [SD 2y]). RESULTS There was no significant between-group differences in time taken to initially fixate on a face (p=0.617); however, children with NF1 spent less time attending to faces within scenes than controls (p=0.048). Decreased attention to faces was associated with elevated autism traits in children with NF1. INTERPRETATION Children with NF1 spend less time attending to faces than typically developing children when presented in social scenes. Our findings contribute to a growing body of literature suggesting that abnormal face processing is a key aspect of the social-cognitive phenotype of NF1 and appears to be related to autism spectrum disorder traits. Clinicians should consider the impact of reduced attention to faces when designing and implementing treatment programmes for social dysfunction in this population. WHAT THIS PAPER ADDS Children with neurofibromatosis type 1 (NF1) demonstrated atypical gaze behaviour when attending to faces. NF1 gaze behaviour was characterized by normal initial fixation on faces but shorter face dwell time. Decreased attention to faces was associated with elevated autism traits in the sample with NF1.
Collapse
Affiliation(s)
- Amelia K Lewis
- Department of Psychology, Macquarie University, Sydney, NSW, Australia
| | - Melanie A Porter
- Department of Psychology, Macquarie University, Sydney, NSW, Australia.,ARC Centre of Excellence in Cognition and its Disorders, Macquarie University, Sydney, NSW, Australia
| | - Tracey A Williams
- Kids Rehab, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | | | - Kathryn N North
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Vic., Australia.,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Vic., Australia
| | - Jonathan M Payne
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Vic., Australia.,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Vic., Australia
| |
Collapse
|
28
|
Drozd HP, Karathanasis SF, Molosh AI, Lukkes JL, Clapp DW, Shekhar A. From bedside to bench and back: Translating ASD models. PROGRESS IN BRAIN RESEARCH 2018; 241:113-158. [PMID: 30447753 DOI: 10.1016/bs.pbr.2018.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Autism spectrum disorders (ASD) represent a heterogeneous group of disorders defined by deficits in social interaction/communication and restricted interests, behaviors, or activities. Models of ASD, developed based on clinical data and observations, are used in basic science, the "bench," to better understand the pathophysiology of ASD and provide therapeutic options for patients in the clinic, the "bedside." Translational medicine creates a bridge between the bench and bedside that allows for clinical and basic science discoveries to challenge one another to improve the opportunities to bring novel therapies to patients. From the clinical side, biomarker work is expanding our understanding of possible mechanisms of ASD through measures of behavior, genetics, imaging modalities, and serum markers. These biomarkers could help to subclassify patients with ASD in order to better target treatments to a more homogeneous groups of patients most likely to respond to a candidate therapy. In turn, basic science has been responding to developments in clinical evaluation by improving bench models to mechanistically and phenotypically recapitulate the ASD phenotypes observed in clinic. While genetic models are identifying novel therapeutics targets at the bench, the clinical efforts are making progress by defining better outcome measures that are most representative of meaningful patient responses. In this review, we discuss some of these challenges in translational research in ASD and strategies for the bench and bedside to bridge the gap to achieve better benefits to patients.
Collapse
Affiliation(s)
- Hayley P Drozd
- Program in Medical Neurobiology, Stark Neurosciences Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sotirios F Karathanasis
- Program in Medical Neurobiology, Stark Neurosciences Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Andrei I Molosh
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jodi L Lukkes
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
| | - D Wade Clapp
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Anantha Shekhar
- Program in Medical Neurobiology, Stark Neurosciences Institute, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States; Indiana Clinical and Translation Sciences Institute, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
29
|
Molosh AI, Shekhar A. Neurofibromatosis type 1 as a model system to study molecular mechanisms of autism spectrum disorder symptoms. PROGRESS IN BRAIN RESEARCH 2018; 241:37-62. [PMID: 30447756 DOI: 10.1016/bs.pbr.2018.09.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Neurofibromatosis type 1 (NF1) is monogenic neurodevelopmental disorder caused by mutation of NF1 gene, which leads to increased susceptibility to various tumors formations. Additionally, majority of patients with NF1 are experience high incidence of cognitive deficits. Particularly, we review the growing number of reports demonstrated a higher incidence of autism spectrum disorder (ASD) in individuals with NF1. In this review we also discuss face validity of preclinical Nf1 mouse models. Then we describe discoveries from these animal models that have uncovered the deficiencies in the regulation of Ras and other intracellular pathways as critical mechanisms underlying the Nf1 cognitive problems. We also summarize and interpret recent preclinical and clinical studies that point toward potential pharmacological therapies for NF1 patients.
Collapse
Affiliation(s)
- Andrei I Molosh
- Department of Psychiatry, Institute of Psychiatric Research, IU School of Medicine, Indianapolis, IN, United States; Stark Neurosciences Research Institute, IU School of Medicine, Indianapolis, IN, United States.
| | - Anantha Shekhar
- Department of Psychiatry, Institute of Psychiatric Research, IU School of Medicine, Indianapolis, IN, United States; Stark Neurosciences Research Institute, IU School of Medicine, Indianapolis, IN, United States; Department of Pharmacology & Toxicology, IU School of Medicine, Indianapolis, IN, United States; Indiana Clinical and Translational Institute, IU School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
30
|
Social Function and Autism Spectrum Disorder in Children and Adults with Neurofibromatosis Type 1: a Systematic Review and Meta-Analysis. Neuropsychol Rev 2018; 28:317-340. [DOI: 10.1007/s11065-018-9380-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/10/2018] [Indexed: 12/27/2022]
|
31
|
Abstract
The mechanistic target of rapamycin (mTOR) is an important signaling hub that integrates environmental information regarding energy availability and stimulates anabolic molecular processes and cell growth. Abnormalities in this pathway have been identified in several syndromes in which autism spectrum disorder (ASD) is highly prevalent. Several studies have investigated mTOR signaling in developmental and neuronal processes that, when dysregulated, could contribute to the development of ASD. Although many potential mechanisms still remain to be fully understood, these associations are of great interest because of the clinical availability of mTOR inhibitors. Clinical trials evaluating the efficacy of mTOR inhibitors to improve neurodevelopmental outcomes have been initiated.
Collapse
Affiliation(s)
- Kellen D. Winden
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Darius Ebrahimi-Fakhari
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Mustafa Sahin
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
32
|
Bizaoui V, Gage J, Brar R, Rauen KA, Weiss LA. RASopathies are associated with a distinct personality profile. Am J Med Genet B Neuropsychiatr Genet 2018; 177:434-446. [PMID: 29659143 PMCID: PMC6039190 DOI: 10.1002/ajmg.b.32632] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 02/02/2018] [Accepted: 03/01/2018] [Indexed: 11/08/2022]
Abstract
Personality is a complex, yet partially heritable, trait. Although some Mendelian diseases like Williams-Beuren syndrome are associated with a particular personality profile, studies have failed to assign the personality features to a single gene or pathway. As a family of monogenic disorders caused by mutations in the Ras/MAPK pathway known to influence social behavior, RASopathies are likely to provide insight into the genetic basis of personality. Eighty subjects diagnosed with cardiofaciocutaneous syndrome, Costello syndrome, neurofibromatosis type 1, and Noonan syndrome were assessed using a parent-report BFQ-C (Big Five Questionnaire for Children) evaluating agreeableness, extraversion, conscientiousness, intellect/openness, and neuroticism, along with 55 unaffected sibling controls. A short questionnaire was added to assess sense of humor. RASopathy subjects and sibling controls were compared for individual components of personality, multidimensional personality profiles, and individual questions using Student tests, analysis of variance, and principal component analysis. RASopathy subjects were given lower scores on average compared to sibling controls in agreeableness, extraversion, conscientiousness, openness, and sense of humor, and similar scores in neuroticism. When comparing the multidimensional personality profile between groups, RASopathies showed a distinct profile from unaffected siblings, but no difference in this global profile was found within RASopathies, revealing a common profile for the Ras/MAPK-related disorders. In addition, several syndrome-specific strengths or weaknesses were observed in individual domains. We describe for the first time an association between a single pathway and a specific personality profile, providing a better understanding of the genetics underlying personality, and new tools for tailoring educational and behavioral approaches for individuals with RASopathies.
Collapse
Affiliation(s)
- Varoona Bizaoui
- Department of Psychiatry and Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA,Institute for Human Genetics, University of California San Francisco, San Francisco, California, USA,Laboratoire de Génétique moléculaire et Histocompatibilité, CHRU de Brest, Brest, France,Inserm UMR1078, Etablissement français du sang – Bretagne, Brest, France, Association Gaëtan Saleun
| | - Jessica Gage
- Department of Psychiatry and Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA,Institute for Human Genetics, University of California San Francisco, San Francisco, California, USA,Department of Biological Sciences, California State University, Stanislaus, Turlock, California, USA
| | - Rita Brar
- Department of Psychiatry and Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA,Institute for Human Genetics, University of California San Francisco, San Francisco, California, USA
| | - Katherine A Rauen
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, USA,Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Lauren A Weiss
- Department of Psychiatry and Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA,Institute for Human Genetics, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
33
|
Green J, Garg S. Annual Research Review: The state of autism intervention science: progress, target psychological and biological mechanisms and future prospects. J Child Psychol Psychiatry 2018; 59:424-443. [PMID: 29574740 DOI: 10.1111/jcpp.12892] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/18/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND There has been recent systematic review of key evidence in psychosocial intervention in autism but little review of biological treatments. METHODS We analyse the current literature from the perspective of intervention and mechanism targets across social and biological development. RESULTS The overall quality of trials evidence in autism intervention remains relatively low, despite some recent progress. Many treatments in common use have little or no evidence base. This is very concerning in such an important disorder. A variety of psychosocial interventions can show effect to improve some short-term effects on children's immediate dyadic social interactions, for instance with caregivers. But showing true effectiveness in this developmental disorder requires generalisation of such effects into wider social contexts, on autism symptoms and in long-term progress in development. Only a few interventions so far have begun to show this. A number of early phase interventions on biological targets have shown real promise, but none has yet progressed to larger scale effectiveness trials on behavioural or symptom outcomes. CONCLUSIONS There has been enough progress in psychosocial intervention research now to be able to begin to identify some evidence-based practice in autism treatment. To consolidate and improve outcomes, the next phase of intervention research needs improved trial design, and an iterative approach building on success. It may also include the testing of potential synergies between promising biological and psychosocial interventions.
Collapse
Affiliation(s)
- Jonathan Green
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK.,Manchester University NHS Foundation Trust, Manchester, UK.,Greater Manchester Mental Health NHS Foundation Trust, Manchester, UK
| | - Shruti Garg
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK.,Manchester University NHS Foundation Trust, Manchester, UK.,Greater Manchester Mental Health NHS Foundation Trust, Manchester, UK
| |
Collapse
|
34
|
Garg S, Green J. Studying child development in genetic models of ASD. PROGRESS IN BRAIN RESEARCH 2018; 241:159-192. [DOI: 10.1016/bs.pbr.2018.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
35
|
Kolesnik AM, Jones EJH, Garg S, Green J, Charman T, Johnson MH. Early development of infants with neurofibromatosis type 1: a case series. Mol Autism 2017; 8:62. [PMID: 29204259 PMCID: PMC5701449 DOI: 10.1186/s13229-017-0178-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 10/31/2017] [Indexed: 12/26/2022] Open
Abstract
Background Prospective studies of infants at familial risk for autism spectrum disorder (ASD) have yielded insights into the earliest signs of the disorder but represent heterogeneous samples of unclear aetiology. Complementing this approach by studying cohorts of infants with monogenic syndromes associated with high rates of ASD offers the opportunity to elucidate the factors that lead to ASD. Methods We present the first report from a prospective study of ten 10-month-old infants with neurofibromatosis type 1 (NF1), a monogenic disorder with high prevalence of ASD or ASD symptomatology. We compared data from infants with NF1 to a large cohort of infants at familial risk for ASD, separated by outcome at age 3 of ASD (n = 34), atypical development (n = 44), or typical development (n = 89), and low-risk controls (n = 75). Domains assessed at 10 months by parent report and examiner observation include cognitive and adaptive function, sensory processing, social engagement, and temperament. Results Infants with NF1 showed striking impairments in motor functioning relative to low-risk infants; this pattern was seen in infants with later ASD from the familial cohort (HR-ASD). Both infants with NF1 and the HR-ASD group showed communication delays relative to low-risk infants. Conclusions Ten-month-old infants with NF1 show a range of developmental difficulties that were particularly striking in motor and communication domains. As with HR-ASD infants, social skills at this age were not notably impaired. This is some of the first information on early neurodevelopment in NF1. Strong inferences are limited by the sample size, but the findings suggest implications for early comparative developmental science and highlight motor functioning as an important domain to inform the development of relevant animal models. The findings have clinical implications in indicating an important focus for early surveillance and remediation in this early diagnosed genetic disorder.
Collapse
Affiliation(s)
- Anna May Kolesnik
- Centre for Brain and Cognitive Development and Department of Psychology, Birkbeck, University of London, Malet Street, London, WC1E 7HX UK
| | - Emily Jane Harrison Jones
- Centre for Brain and Cognitive Development and Department of Psychology, Birkbeck, University of London, Malet Street, London, WC1E 7HX UK
| | - Shruti Garg
- Neuroscience & Experimental Psychology, Manchester Academic Health Science Centre, University of Manchester and Royal Manchester Children’s Hospital, Central Manchester University Hospitals NHS Foundation, Manchester, UK
| | - Jonathan Green
- Neuroscience & Experimental Psychology, Manchester Academic Health Science Centre, University of Manchester and Royal Manchester Children’s Hospital, Central Manchester University Hospitals NHS Foundation, Manchester, UK
| | - Tony Charman
- Department of Psychology, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Mark Henry Johnson
- Centre for Brain and Cognitive Development and Department of Psychology, Birkbeck, University of London, Malet Street, London, WC1E 7HX UK
| |
Collapse
|
36
|
Gonçalves J, Violante IR, Sereno J, Leitão RA, Cai Y, Abrunhosa A, Silva AP, Silva AJ, Castelo-Branco M. Testing the excitation/inhibition imbalance hypothesis in a mouse model of the autism spectrum disorder: in vivo neurospectroscopy and molecular evidence for regional phenotypes. Mol Autism 2017; 8:47. [PMID: 28932379 PMCID: PMC5605987 DOI: 10.1186/s13229-017-0166-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 09/12/2017] [Indexed: 12/26/2022] Open
Abstract
Background Excitation/inhibition (E/I) imbalance remains a widely discussed hypothesis in autism spectrum disorders (ASD). The presence of such an imbalance may potentially define a therapeutic target for the treatment of cognitive disabilities related to this pathology. Consequently, the study of monogenic disorders related to autism, such as neurofibromatosis type 1 (NF1), represents a promising approach to isolate mechanisms underlying ASD-related cognitive disabilities. However, the NF1 mouse model showed increased γ-aminobutyric acid (GABA) neurotransmission, whereas the human disease showed reduced cortical GABA levels. It is therefore important to clarify whether the E/I imbalance hypothesis holds true. We hypothesize that E/I may depend on distinct pre- and postsynaptic push-pull mechanisms that might be are region-dependent. Methods In current study, we assessed two critical components of E/I regulation: the concentration of neurotransmitters and levels of GABA(A) receptors. Measurements were performed across the hippocampi, striatum, and prefrontal cortices by combined in vivo magnetic resonance spectroscopy (MRS) and molecular approaches in this ASD-related animal model, the Nf1+/− mouse. Results Cortical and striatal GABA/glutamate ratios were increased. At the postsynaptic level, very high receptor GABA(A) receptor expression was found in hippocampus, disproportionately to the small reduction in GABA levels. Gabaergic tone (either by receptor levels change or GABA/glutamate ratios) seemed therefore to be enhanced in all regions, although by a different mechanism. Conclusions Our data provides support for the hypothesis of E/I imbalance in NF1 while showing that pre- and postsynaptic changes are region-specific. All these findings are consistent with our previous physiological evidence of increased inhibitory tone. Such heterogeneity suggests that therapeutic approaches to address neurochemical imbalance in ASD may need to focus on targets where convergent physiological mechanisms can be found.
Collapse
Affiliation(s)
- Joana Gonçalves
- CiBIT, Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal.,Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute for Biomedical Imaging and Life Sciences (CNC.IBILI) Research Unit, University of Coimbra, Coimbra, Portugal
| | - Inês R Violante
- School of Psychology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - José Sereno
- CiBIT, Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal.,Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute for Biomedical Imaging and Life Sciences (CNC.IBILI) Research Unit, University of Coimbra, Coimbra, Portugal
| | - Ricardo A Leitão
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute for Biomedical Imaging and Life Sciences (CNC.IBILI) Research Unit, University of Coimbra, Coimbra, Portugal.,Laboratory of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ying Cai
- Department of Neurobiology, Integrative Center for Learning and Memory, Brain Research Institute, University of California Los Angeles, Los Angeles, CA USA
| | - Antero Abrunhosa
- CiBIT, Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal.,Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute for Biomedical Imaging and Life Sciences (CNC.IBILI) Research Unit, University of Coimbra, Coimbra, Portugal
| | - Ana Paula Silva
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute for Biomedical Imaging and Life Sciences (CNC.IBILI) Research Unit, University of Coimbra, Coimbra, Portugal.,Laboratory of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Alcino J Silva
- Department of Neurobiology, Integrative Center for Learning and Memory, Brain Research Institute, University of California Los Angeles, Los Angeles, CA USA
| | - Miguel Castelo-Branco
- CiBIT, Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal.,Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute for Biomedical Imaging and Life Sciences (CNC.IBILI) Research Unit, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
37
|
Monroe CL, Dahiya S, Gutmann DH. Dissecting Clinical Heterogeneity in Neurofibromatosis Type 1. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 12:53-74. [PMID: 28135565 DOI: 10.1146/annurev-pathol-052016-100228] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neurofibromatosis type 1 (NF1) is a common neurogenetic disorder in which affected children and adults are predisposed to the development of benign and malignant nervous system tumors. Caused by a germline mutation in the NF1 tumor suppressor gene, individuals with NF1 are prone to optic gliomas, malignant gliomas, neurofibromas, and malignant peripheral nerve sheath tumors, as well as behavioral, cognitive, motor, bone, cardiac, and pigmentary abnormalities. Although NF1 is a classic monogenic syndrome, the clinical features of the disorder and their impact on patient morbidity are variable, even within individuals who bear the same germline NF1 gene mutation. As such, NF1 affords unique opportunities to define the factors that contribute to disease heterogeneity and to develop therapies personalized to a given individual (precision medicine). This review highlights the clinical features of NF1 and the use of genetically engineered mouse models to define the molecular and cellular pathogenesis of NF1-associated nervous system tumors.
Collapse
Affiliation(s)
- Courtney L Monroe
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110;
| | - Sonika Dahiya
- Division of Neuropathology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110;
| |
Collapse
|
38
|
Rietman AB, Oostenbrink R, Bongers S, Gaukema E, van Abeelen S, Hendriksen JG, Looman CWN, de Nijs PFA, de Wit MC. Motor problems in children with neurofibromatosis type 1. J Neurodev Disord 2017; 9:19. [PMID: 28529667 PMCID: PMC5437487 DOI: 10.1186/s11689-017-9198-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 05/02/2017] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Children with the neurogenetic disorder neurofibromatosis type 1 (NF1) often have problems with learning and behaviour. In both parent reports and neuropsychological assessment, motor problems are reported in approximately one third to one half of the children with NF1. Studies using broad motor performance test batteries with relatively large groups of children with NF1 are limited. The aim of this cross-sectional observational study was to describe the severity of motor problems in children with NF1 and to explore the predictive value of demographics, intelligence, and behavioural problems. METHODS From 2002 to 2014, 69 children with NF1, aged 4 to 16 years (age = 9.5 ± 2.8 years; 29 girls) had a motor, psychological, and neurological evaluation in an NF1 expertise centre. Data were collected about (1) motor performance (M-ABC: Movement Assessment Battery for Children), (2) intelligence, and (3) emotional and behavioural problems as rated by parents. RESULTS Sixty-one percent of these children scored within the clinical range of the M-ABC. In ordinal logistic regression analyses, motor problems were associated with symptoms of attention-deficit/hyperactivity disorder (ADHD), symptoms of autism spectrum disorder (ASD), and externalising behavioural problems. Motor outcome was not predicted by age, intelligence, scoliosis, hypotonia, nor hypermobility. CONCLUSIONS Motor problems are among the most common comorbid developmental problems in children with NF1, and these problems do not diminish with age. Because of their impact on daily functioning, motor problems need to be specifically addressed in diagnosis, follow-up, and treatment of NF1.
Collapse
Affiliation(s)
- André B. Rietman
- Department of Child and Adolescent Psychiatry/Psychology, ENCORE NF1 Expertise Centre for Neurodevelopmental Disorders, Erasmus Medical Centre–Sophia Children’s Hospital, Rotterdam, The Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Sophia Children’s Hospital, Room Sp 2478, P.O. Box 2060, 3000 CB Rotterdam, The Netherlands
| | - Rianne Oostenbrink
- Department of General Paediatrics, ENCORE NF1, Erasmus Medical Centre–Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Sanne Bongers
- Department of Child and Adolescent Psychiatry/Psychology, ENCORE NF1 Expertise Centre for Neurodevelopmental Disorders, Erasmus Medical Centre–Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Eddy Gaukema
- Kempenhaeghe Centre for neurological learning disabilities, Heeze, The Netherlands
| | - Sandra van Abeelen
- Kempenhaeghe Centre for neurological learning disabilities, Heeze, The Netherlands
| | - Jos G. Hendriksen
- Kempenhaeghe Centre for neurological learning disabilities, Heeze, The Netherlands
| | - Caspar W. N. Looman
- Department of Public Health, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Pieter F. A. de Nijs
- Department of Child and Adolescent Psychiatry/Psychology, ENCORE NF1 Expertise Centre for Neurodevelopmental Disorders, Erasmus Medical Centre–Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Marie-Claire de Wit
- Department of Paediatric Neurology, ENCORE NF1, Erasmus Medical Centre–Sophia Children’s Hospital, Rotterdam, The Netherlands
| |
Collapse
|
39
|
López-Juárez A, Titus HE, Silbak SH, Pressler JW, Rizvi TA, Bogard M, Bennett MR, Ciraolo G, Williams MT, Vorhees CV, Ratner N. Oligodendrocyte Nf1 Controls Aberrant Notch Activation and Regulates Myelin Structure and Behavior. Cell Rep 2017; 19:545-557. [PMID: 28423318 PMCID: PMC5828008 DOI: 10.1016/j.celrep.2017.03.073] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 02/23/2017] [Accepted: 03/27/2017] [Indexed: 11/29/2022] Open
Abstract
The RASopathy neurofibromatosis type 1 (NF1) is one of the most common autosomal dominant genetic disorders. In NF1 patients, neurological issues may result from damaged myelin, and mice with a neurofibromin gene (Nf1) mutation show white matter (WM) defects including myelin decompaction. Using mouse genetics, we find that altered Nf1 gene-dose in mature oligodendrocytes results in progressive myelin defects and behavioral abnormalities mediated by aberrant Notch activation. Blocking Notch, upstream mitogen-activated protein kinase (MAPK), or nitric oxide signaling rescues myelin defects in hemizygous Nf1 mutants, and pharmacological gamma secretase inhibition rescues aberrant behavior with no effects in wild-type (WT) mice. Concomitant pathway inhibition rescues myelin abnormalities in homozygous mutants. Notch activation is also observed in Nf1+/− mouse brains, and cells containing active Notch are increased in NF1 patient WM. We thus identify Notch as an Nf1 effector regulating myelin structure and behavior in a RASopathy and suggest that inhibition of Notch signaling may be a therapeutic strategy for NF1.
Collapse
Affiliation(s)
- Alejandro López-Juárez
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Haley E Titus
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Sadiq H Silbak
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Joshua W Pressler
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Tilat A Rizvi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Madeleine Bogard
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Michael R Bennett
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Georgianne Ciraolo
- Division of Pathology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Michael T Williams
- Division of Neurology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Charles V Vorhees
- Division of Neurology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Nancy Ratner
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA.
| |
Collapse
|
40
|
Wang P, Mokhtari R, Pedrosa E, Kirschenbaum M, Bayrak C, Zheng D, Lachman HM. CRISPR/Cas9-mediated heterozygous knockout of the autism gene CHD8 and characterization of its transcriptional networks in cerebral organoids derived from iPS cells. Mol Autism 2017; 8:11. [PMID: 28321286 PMCID: PMC5357816 DOI: 10.1186/s13229-017-0124-1] [Citation(s) in RCA: 183] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 02/15/2017] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND CHD8 (chromodomain helicase DNA-binding protein 8), which codes for a member of the CHD family of ATP-dependent chromatin-remodeling factors, is one of the most commonly mutated genes in autism spectrum disorders (ASD) identified in exome-sequencing studies. Loss of function mutations in the gene have also been found in schizophrenia (SZ) and intellectual disabilities and influence cancer cell proliferation. We previously reported an RNA-seq analysis carried out on neural progenitor cells (NPCs) and monolayer neurons derived from induced pluripotent stem (iPS) cells that were heterozygous for CHD8 knockout (KO) alleles generated using CRISPR-Cas9 gene editing. A significant number of ASD and SZ candidate genes were among those that were differentially expressed in a comparison of heterozygous KO lines (CHD8+/-) vs isogenic controls (CHD8+/-), including the SZ and bipolar disorder (BD) candidate gene TCF4, which was markedly upregulated in CHD8+/- neuronal cells. METHODS In the current study, RNA-seq was carried out on CHD8+/- and isogenic control (CHD8+/+) cerebral organoids, which are 3-dimensional structures derived from iPS cells that model the developing human telencephalon. RESULTS TCF4 expression was, again, significantly upregulated. Pathway analysis carried out on differentially expressed genes (DEGs) revealed an enrichment of genes involved in neurogenesis, neuronal differentiation, forebrain development, Wnt/β-catenin signaling, and axonal guidance, similar to our previous study on NPCs and monolayer neurons. There was also significant overlap in our CHD8+/- DEGs with those found in a transcriptome analysis carried out by another group using cerebral organoids derived from a family with idiopathic ASD. Remarkably, the top DEG in our respective studies was the non-coding RNA DLX6-AS1, which was markedly upregulated in both studies; DLX6-AS1 regulates the expression of members of the DLX (distal-less homeobox) gene family. DLX1 was also upregulated in both studies. DLX genes code for transcription factors that play a key role in GABAergic interneuron differentiation. Significant overlap was also found in a transcriptome study carried out by another group using iPS cell-derived neurons from patients with BD, a condition characterized by dysregulated WNT/β-catenin signaling in a subgroup of affected individuals. CONCLUSIONS Overall, the findings show that distinct ASD, SZ, and BD candidate genes converge on common molecular targets-an important consideration for developing novel therapeutics in genetically heterogeneous complex traits.
Collapse
Affiliation(s)
- Ping Wang
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY USA
| | - Ryan Mokhtari
- Department of Psychiatry and Behavioral Sciences, Erciyes University School of Medicine, Kayseri, Turkey
| | - Erika Pedrosa
- Department of Psychiatry and Behavioral Sciences, Erciyes University School of Medicine, Kayseri, Turkey
| | - Michael Kirschenbaum
- Department of Psychiatry and Behavioral Sciences, Erciyes University School of Medicine, Kayseri, Turkey
| | - Can Bayrak
- Erciyes University School of Medicine, Kayseri, Turkey
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY USA
- Department of Neurology, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY USA
- Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY USA
| | - Herbert M. Lachman
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY USA
- Department of Psychiatry and Behavioral Sciences, Erciyes University School of Medicine, Kayseri, Turkey
- Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY USA
- Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY USA
| |
Collapse
|
41
|
Robert C, Pasquier L, Cohen D, Fradin M, Canitano R, Damaj L, Odent S, Tordjman S. Role of Genetics in the Etiology of Autistic Spectrum Disorder: Towards a Hierarchical Diagnostic Strategy. Int J Mol Sci 2017; 18:E618. [PMID: 28287497 PMCID: PMC5372633 DOI: 10.3390/ijms18030618] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/17/2017] [Accepted: 02/20/2017] [Indexed: 12/27/2022] Open
Abstract
Progress in epidemiological, molecular and clinical genetics with the development of new techniques has improved knowledge on genetic syndromes associated with autism spectrum disorder (ASD). The objective of this article is to show the diversity of genetic disorders associated with ASD (based on an extensive review of single-gene disorders, copy number variants, and other chromosomal disorders), and consequently to propose a hierarchical diagnostic strategy with a stepwise evaluation, helping general practitioners/pediatricians and child psychiatrists to collaborate with geneticists and neuropediatricians, in order to search for genetic disorders associated with ASD. The first step is a clinical investigation involving: (i) a child psychiatric and psychological evaluation confirming autism diagnosis from different observational sources and assessing autism severity; (ii) a neuropediatric evaluation examining neurological symptoms and developmental milestones; and (iii) a genetic evaluation searching for dysmorphic features and malformations. The second step involves laboratory and if necessary neuroimaging and EEG studies oriented by clinical results based on clinical genetic and neuropediatric examinations. The identification of genetic disorders associated with ASD has practical implications for diagnostic strategies, early detection or prevention of co-morbidity, specific treatment and follow up, and genetic counseling.
Collapse
Affiliation(s)
- Cyrille Robert
- Pôle Hospitalo-Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (PHUPEA), University of Rennes 1 and Centre Hospitalier Guillaume Régnier, 35200 Rennes, France.
- Service de Génétique Clinique, Centre de Référence Maladies Rares Anomalies du Développement (Centre Labellisé pour les Anomalies du Développement de l'Ouest: CLAD Ouest), Hôpital Sud, Centre Hospitalier Universitaire de Rennes, 35200 Rennes, France.
| | - Laurent Pasquier
- Service de Génétique Clinique, Centre de Référence Maladies Rares Anomalies du Développement (Centre Labellisé pour les Anomalies du Développement de l'Ouest: CLAD Ouest), Hôpital Sud, Centre Hospitalier Universitaire de Rennes, 35200 Rennes, France.
| | - David Cohen
- Hospital-University Department of Child and Adolescent Psychiatry, Pitié-Salpétrière Hospital, Paris 6 University, 75013 Paris, France.
| | - Mélanie Fradin
- Service de Génétique Clinique, Centre de Référence Maladies Rares Anomalies du Développement (Centre Labellisé pour les Anomalies du Développement de l'Ouest: CLAD Ouest), Hôpital Sud, Centre Hospitalier Universitaire de Rennes, 35200 Rennes, France.
| | - Roberto Canitano
- Division of Child and Adolescent Neuropsychiatry, University Hospital of Siena, 53100 Siena, Italy.
| | - Léna Damaj
- Service de Génétique Clinique, Centre de Référence Maladies Rares Anomalies du Développement (Centre Labellisé pour les Anomalies du Développement de l'Ouest: CLAD Ouest), Hôpital Sud, Centre Hospitalier Universitaire de Rennes, 35200 Rennes, France.
| | - Sylvie Odent
- Service de Génétique Clinique, Centre de Référence Maladies Rares Anomalies du Développement (Centre Labellisé pour les Anomalies du Développement de l'Ouest: CLAD Ouest), Hôpital Sud, Centre Hospitalier Universitaire de Rennes, 35200 Rennes, France.
| | - Sylvie Tordjman
- Pôle Hospitalo-Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (PHUPEA), University of Rennes 1 and Centre Hospitalier Guillaume Régnier, 35200 Rennes, France.
- Laboratory of Psychology of Perception, University Paris Descartes, 75270 Paris, France.
| |
Collapse
|
42
|
Mitra I, Lavillaureix A, Yeh E, Traglia M, Tsang K, Bearden CE, Rauen KA, Weiss LA. Reverse Pathway Genetic Approach Identifies Epistasis in Autism Spectrum Disorders. PLoS Genet 2017; 13:e1006516. [PMID: 28076348 PMCID: PMC5226683 DOI: 10.1371/journal.pgen.1006516] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 12/01/2016] [Indexed: 02/08/2023] Open
Abstract
Although gene-gene interaction, or epistasis, plays a large role in complex traits in model organisms, genome-wide by genome-wide searches for two-way interaction have limited power in human studies. We thus used knowledge of a biological pathway in order to identify a contribution of epistasis to autism spectrum disorders (ASDs) in humans, a reverse-pathway genetic approach. Based on previous observation of increased ASD symptoms in Mendelian disorders of the Ras/MAPK pathway (RASopathies), we showed that common SNPs in RASopathy genes show enrichment for association signal in GWAS (P = 0.02). We then screened genome-wide for interactors with RASopathy gene SNPs and showed strong enrichment in ASD-affected individuals (P < 2.2 x 10-16), with a number of pairwise interactions meeting genome-wide criteria for significance. Finally, we utilized quantitative measures of ASD symptoms in RASopathy-affected individuals to perform modifier mapping via GWAS. One top region overlapped between these independent approaches, and we showed dysregulation of a gene in this region, GPR141, in a RASopathy neural cell line. We thus used orthogonal approaches to provide strong evidence for a contribution of epistasis to ASDs, confirm a role for the Ras/MAPK pathway in idiopathic ASDs, and to identify a convergent candidate gene that may interact with the Ras/MAPK pathway.
Collapse
Affiliation(s)
- Ileena Mitra
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Alinoë Lavillaureix
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
- Université Paris Descartes, Sorbonne Paris Cité, Faculty of Medicine, Paris, France
| | - Erika Yeh
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Michela Traglia
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Kathryn Tsang
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Carrie E. Bearden
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Psychology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Katherine A. Rauen
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
- Department of Pediatrics, School of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Lauren A. Weiss
- Department of Psychiatry, University of California San Francisco, San Francisco, California, United States of America
- Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
43
|
Torres Nupan MM, Velez Van Meerbeke A, López Cabra CA, Herrera Gomez PM. Cognitive and Behavioral Disorders in Children with Neurofibromatosis Type 1. Front Pediatr 2017; 5:227. [PMID: 29164079 PMCID: PMC5670111 DOI: 10.3389/fped.2017.00227] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 10/05/2017] [Indexed: 11/13/2022] Open
Abstract
AIM The last systematic review of research on the behavior of children with neurofibromatosis type 1 (NF1) was in 2012. Since then, several important findings have been published. Therefore, the study aim was to synthesize recent relevant work related to this issue. METHOD We conducted a systematic review of the literature. Relevant articles were identified using the electronic databases PubMed, PsycINFO, and Scopus and a manual search of references lists. Thirty of 156 articles identified met the inclusion criteria. A quality evaluation of the articles was performed and the information was synthesized using a narrative approach. RESULTS Compared with controls, children and adolescents with NF1 present significant alterations in language, reading, visuospatial skills, motor function, executive function, attention, behavior, emotion, and social skills. The prevalence of attention-deficit/hyperactivity disorder (ADHD) is important and can affect cognition and executive function variables. A high prevalence of autistic traits and autistic spectrum disorder were reported. The benefits of using statins to treat cognitive deficits are unclear. However, children with NF1 and ADHD seem to benefit from methylphenidate treatment. The presence of hyperintensities in brain magnetic resonance imaging data seem to be related to poor cognitive performance. Analysis of these lesions could help to predict cognitive alterations in children with NF1. INTERPRETATION There has been important progress to evaluate cognitive characteristics of children with NF1 and to determine the physiological mechanisms of the concomitant disorders. However, discrepancies in relation to intelligence, learning disabilities, attention deficits, and treatment remain. Further investigations on this topic are recommended.
Collapse
Affiliation(s)
- Martha Milade Torres Nupan
- Neurosciences Research Group, Medicine and Health Sciences School, Universidad del Rosario, Bogota, Colombia
| | - Alberto Velez Van Meerbeke
- Neurosciences Research Group, Medicine and Health Sciences School, Universidad del Rosario, Bogota, Colombia
| | | | - Paula Marcela Herrera Gomez
- Neurosciences Research Group, Medicine and Health Sciences School, Universidad del Rosario, Bogota, Colombia
| |
Collapse
|
44
|
Morris SM, Acosta MT, Garg S, Green J, Huson S, Legius E, North KN, Payne JM, Plasschaert E, Frazier TW, Weiss LA, Zhang Y, Gutmann DH, Constantino JN. Disease Burden and Symptom Structure of Autism in Neurofibromatosis Type 1: A Study of the International NF1-ASD Consortium Team (INFACT). JAMA Psychiatry 2016; 73:1276-1284. [PMID: 27760236 PMCID: PMC5298203 DOI: 10.1001/jamapsychiatry.2016.2600] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
IMPORTANCE Recent reports have demonstrated a higher incidence of autism spectrum disorder (ASD) and substantially elevated autistic trait burden in individuals with neurofibromatosis type 1 (NF1). However, important discrepancies regarding the distribution of autistic traits, sex predominance, and association between ASD symptoms and attentional problems have emerged, and critical features of the ASD phenotype within NF1 have never been adequately explored. Establishing NF1 as a monogenic cause for ASD has important implications for affected patients and for future research focused on establishing convergent pathogenic mechanisms relevant to the potential treatment targets for ASD. OBJECTIVE To characterize the quantitative autistic trait (QAT) burden in a pooled NF1 data set. DESIGN, SETTING, AND PARTICIPANTS Anonymized, individual-level primary data were accumulated from 6 tertiary referral centers in the United States, Belgium, United Kingdom, and Australia. A total of 531 individuals recruited from NF1 clinical centers were included in the study. MAIN OUTCOMES AND MEASURES Distribution of ASD traits (Social Responsiveness Scale, second edition [SRS-2], with T scores of ≥75 associated with a categorical ASD diagnosis); attention-deficit/hyperactivity disorder (ADHD) traits (4 versions of Conners Rating Scale, with T scores of ≥65 indicating clinically significant ADHD symptoms); ASD symptom structure, latent structure, base rate derived from mixture modeling; and familiality. RESULTS Of the 531 patients included in the analysis, 247 were male (46.5%); median age was 11 years (range, 2.5-83.9 years). QAT scores were continuously distributed and pathologically shifted; 13.2% (95% CI, 10.3%-16.1%) of individuals scored within the most severe range (ie, above the first percentile of the general population distribution) in which the male to female ratio was markedly attenuated (1.6:1) relative to idiopathic ASD. Autistic symptoms in this NF1 cohort demonstrated a robust unitary factor structure, with the first principal component explaining 30.9% of the variance in SRS-2 scores, and a strong association with ADHD symptoms (r = 0.61). Within-family correlation for QAT burden (intraclass correlation coefficient, 0.73 in NF1-affected first-degree relatives) exceeded that observed in the general population and ASD family samples. CONCLUSIONS AND RELEVANCE This study provides confirmation that the diversity of mutations that give rise to NF1 function as quantitative trait loci for ASD. Moreover, the within-family correlation implicates a high degree of mutational specificity for this associated phenotype. Clinicians should be alerted to the increased frequency of this disabling comorbidity, and the scientific community should be aware of the potential for this monogenic disorder to help elucidate the biological features of idiopathic autism.
Collapse
Affiliation(s)
- Stephanie M. Morris
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - Maria T. Acosta
- Center for Neuroscience and Behavioral Medicine at Children’s National Health System, Washington, DC
| | - Shruti Garg
- Institute of Brain Behavior and Mental Health, The University of Manchester, Manchester, England4Manchester Academic Health Sciences Centre, Manchester, England5Central Manchester University NHS Foundation Trust, Manchester, England
| | - Jonathan Green
- Institute of Brain Behavior and Mental Health, The University of Manchester, Manchester, England4Manchester Academic Health Sciences Centre, Manchester, England5Central Manchester University NHS Foundation Trust, Manchester, England
| | - Susan Huson
- Central Manchester University NHS Foundation Trust, Manchester, England
| | - Eric Legius
- Department of Human Genetics, Laboratory for Neurofibromatosis Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Kathryn N. North
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Australia8Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Jonathan M. Payne
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Australia8Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Ellen Plasschaert
- Department of Human Genetics, Laboratory for Neurofibromatosis Research, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Thomas W. Frazier
- Center for Pediatric Behavioral Health, Pediatric Institute, Cleveland Clinic, Cleveland, Ohio
| | - Lauren A. Weiss
- Department of Psychiatry and Institute for Human Genetics, University of California, San Francisco
| | - Yi Zhang
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri
| | - David H. Gutmann
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - John N. Constantino
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri12Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
45
|
Chromosomal microarray testing in adults with intellectual disability presenting with comorbid psychiatric disorders. Eur J Hum Genet 2016; 25:66-72. [PMID: 27650969 PMCID: PMC5159755 DOI: 10.1038/ejhg.2016.107] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 06/23/2016] [Accepted: 06/28/2016] [Indexed: 12/12/2022] Open
Abstract
Chromosomal copy-number variations (CNVs) are a class of genetic variants highly implicated in the aetiology of neurodevelopmental disorders, including intellectual disabilities (ID), schizophrenia and autism spectrum disorders (ASD). Yet the majority of adults with idiopathic ID presenting to psychiatric services have not been tested for CNVs. We undertook genome-wide chromosomal microarray analysis (CMA) of 202 adults with idiopathic ID recruited from community and in-patient ID psychiatry services across England. CNV pathogenicity was assessed using standard clinical diagnostic methods and participants underwent comprehensive medical and psychiatric phenotyping. We found an 11% yield of likely pathogenic CNVs (22/202). CNVs at recurrent loci, including the 15q11-q13 and 16p11.2-p13.11 regions were most frequently observed. We observed an increased frequency of 16p11.2 duplications compared with those reported in single-disorder cohorts. CNVs were also identified in genes known to effect neurodevelopment, namely NRXN1 and GRIN2B. Furthermore deletions at 2q13, 12q21.2-21.31 and 19q13.32, and duplications at 4p16.3, 13q32.3-33.3 and Xq24-25 were observed. Routine CMA in ID psychiatry could uncover ~11% new genetic diagnoses with potential implications for patient management. We advocate greater consideration of CMA in the assessment of adults with idiopathic ID presenting to psychiatry services.
Collapse
|
46
|
Garg S, Heuvelman H, Huson S, Tobin H, Green J. Sex bias in autism spectrum disorder in neurofibromatosis type 1. J Neurodev Disord 2016; 8:26. [PMID: 27516813 PMCID: PMC4980803 DOI: 10.1186/s11689-016-9159-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/05/2016] [Indexed: 01/29/2023] Open
Abstract
Background Despite extensive literature, little is known about the mechanisms underlying sex bias in autism spectrum disorder (ASD). This study investigates the sex differences in ASD associated with neurofibromatosis type 1, a single-gene model of syndromic autism. Methods We analysed data from n = 194 children aged 4–16 years with neurofibromatosis type 1. Sex differences were evaluated across the Autism Diagnostic Interview-Revised (ADI-R), Autism Diagnostic Observation Schedule (ADOS), verbal IQ, Social Responsiveness Scale (SRS) and Conners questionnaires. Results There was 2.68:1 male:female ratio in children meeting ASD criteria on the deep phenotyping measures. On symptom profile, males with neurofibromatosis type 1 (NF1) + ASD were more impaired on reciprocal social interaction and communication domains of the ADI-R but we found no differences on the restricted, repetitive behaviours (RRBs) domain of the ADI-R and no differences on the social on the ADOS. NF1 ASD males and females were comparable on verbal IQ, and the inattention/hyperactivity domains of the Conners questionnaire. Conclusions There is a significant male bias in the prevalence of ASD in NF1. The phenotypic profile of NF1 + ASD cases includes greater social communication impairment in males. We discuss the implications of our findings and the rationale for using NF1 as a model for investigating sex bias in idiopathic ASD.
Collapse
Affiliation(s)
- Shruti Garg
- Institute of Brain Behaviour and Mental Health, University of Manchester, Oxford Road, Manchester, M13 9PL UK ; Child and Adolescent Mental Health Service, Royal Manchester Children's Hospital, Oxford Road, Manchester, M13 9WL UK
| | - Hein Heuvelman
- Centre for Biostatistics, Institute for Population Health, University of Manchester, Manchester, UK
| | - Susan Huson
- Genomic Medicine, Manchester Academic Health Science Centre, Institute of Human Development, The University of Manchester, Central Manchester University Hospitals NHS Trust, St Mary's Hospital, Oxford Road, Manchester, M13 9WL UK
| | - Hannah Tobin
- Institute of Brain Behaviour and Mental Health, University of Manchester, Oxford Road, Manchester, M13 9PL UK
| | - Jonathan Green
- Institute of Brain Behaviour and Mental Health, University of Manchester, Oxford Road, Manchester, M13 9PL UK
| | | |
Collapse
|
47
|
van der Vaart T, Rietman AB, Plasschaert E, Legius E, Elgersma Y, Moll HA. Behavioral and cognitive outcomes for clinical trials in children with neurofibromatosis type 1. Neurology 2015; 86:154-60. [PMID: 26519538 DOI: 10.1212/wnl.0000000000002118] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 09/09/2015] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To evaluate the appropriateness of cognitive and behavioral outcome measures in clinical trials in neurofibromatosis type 1 (NF1) by analyzing the degree of deficits compared to reference groups, test-retest reliability, and how scores correlate between outcome measures. METHODS Data were analyzed from the Simvastatin for cognitive deficits and behavioral problems in patients with neurofibromatosis type 1 (NF1-SIMCODA) trial, a randomized placebo-controlled trial of simvastatin for cognitive deficits and behavioral problems in children with NF1. Outcome measures were compared with age-specific reference groups to identify domains of dysfunction. Pearson r was computed for before and after measurements within the placebo group to assess test-retest reliability. Principal component analysis was used to identify the internal structure in the outcome data. RESULTS Strongest mean score deviations from the reference groups were observed for full-scale intelligence (-1.1 SD), Rey Complex Figure Test delayed recall (-2.0 SD), attention problems (-1.2 SD), and social problems (-1.1 SD). Long-term test-retest reliability were excellent for Wechsler scales (r > 0.88), but poor to moderate for other neuropsychological tests (r range 0.52-0.81) and Child Behavioral Checklist subscales (r range 0.40-0.79). The correlation structure revealed 2 strong components in the outcome measures behavior and cognition, with no correlation between these components. Scores on psychosocial quality of life correlate strongly with behavioral problems and less with cognitive deficits. CONCLUSIONS Children with NF1 show distinct deficits in multiple domains. Many outcome measures showed weak test-retest correlations over the 1-year trial period. Cognitive and behavioral outcomes are complementary. This analysis demonstrates the need to include reliable outcome measures on a variety of cognitive and behavioral domains in clinical trials for NF1.
Collapse
Affiliation(s)
- Thijs van der Vaart
- From the Departments of Neuroscience (T.v.d.V., Y.E.), Pediatrics (T.v.d.V., H.A.M.), and Neurology (A.B.R.), and ENCORE-Expertise Center for Neurodevelopmental Disorders (T.v.d.V., A.B.R., Y.E., H.A.M.), Erasmus MC: University Medical Centre, Rotterdam, the Netherlands; the Department of Human Genetics (E.P., E.L.), Catholic University Leuven; and the Centre for Human Genetics at University Hospital Leuven (E.P., E.L.), Belgium
| | - André B Rietman
- From the Departments of Neuroscience (T.v.d.V., Y.E.), Pediatrics (T.v.d.V., H.A.M.), and Neurology (A.B.R.), and ENCORE-Expertise Center for Neurodevelopmental Disorders (T.v.d.V., A.B.R., Y.E., H.A.M.), Erasmus MC: University Medical Centre, Rotterdam, the Netherlands; the Department of Human Genetics (E.P., E.L.), Catholic University Leuven; and the Centre for Human Genetics at University Hospital Leuven (E.P., E.L.), Belgium
| | - Ellen Plasschaert
- From the Departments of Neuroscience (T.v.d.V., Y.E.), Pediatrics (T.v.d.V., H.A.M.), and Neurology (A.B.R.), and ENCORE-Expertise Center for Neurodevelopmental Disorders (T.v.d.V., A.B.R., Y.E., H.A.M.), Erasmus MC: University Medical Centre, Rotterdam, the Netherlands; the Department of Human Genetics (E.P., E.L.), Catholic University Leuven; and the Centre for Human Genetics at University Hospital Leuven (E.P., E.L.), Belgium
| | - Eric Legius
- From the Departments of Neuroscience (T.v.d.V., Y.E.), Pediatrics (T.v.d.V., H.A.M.), and Neurology (A.B.R.), and ENCORE-Expertise Center for Neurodevelopmental Disorders (T.v.d.V., A.B.R., Y.E., H.A.M.), Erasmus MC: University Medical Centre, Rotterdam, the Netherlands; the Department of Human Genetics (E.P., E.L.), Catholic University Leuven; and the Centre for Human Genetics at University Hospital Leuven (E.P., E.L.), Belgium
| | - Ype Elgersma
- From the Departments of Neuroscience (T.v.d.V., Y.E.), Pediatrics (T.v.d.V., H.A.M.), and Neurology (A.B.R.), and ENCORE-Expertise Center for Neurodevelopmental Disorders (T.v.d.V., A.B.R., Y.E., H.A.M.), Erasmus MC: University Medical Centre, Rotterdam, the Netherlands; the Department of Human Genetics (E.P., E.L.), Catholic University Leuven; and the Centre for Human Genetics at University Hospital Leuven (E.P., E.L.), Belgium
| | - Henriëtte A Moll
- From the Departments of Neuroscience (T.v.d.V., Y.E.), Pediatrics (T.v.d.V., H.A.M.), and Neurology (A.B.R.), and ENCORE-Expertise Center for Neurodevelopmental Disorders (T.v.d.V., A.B.R., Y.E., H.A.M.), Erasmus MC: University Medical Centre, Rotterdam, the Netherlands; the Department of Human Genetics (E.P., E.L.), Catholic University Leuven; and the Centre for Human Genetics at University Hospital Leuven (E.P., E.L.), Belgium.
| | | |
Collapse
|
48
|
Abadin SS, Zoellner NL, Schaeffer M, Porcelli B, Gutmann DH, Johnson KJ. Racial/Ethnic Differences in Pediatric Brain Tumor Diagnoses in Patients with Neurofibromatosis Type 1. J Pediatr 2015; 167:613-20.e1-2. [PMID: 26028287 PMCID: PMC4784699 DOI: 10.1016/j.jpeds.2015.04.076] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 04/06/2015] [Accepted: 04/30/2015] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To evaluate evidence for differences in pediatric brain tumor diagnoses by race and ethnicity using a cross-sectional study design in individuals with neurofibromatosis type 1 (NF1). STUDY DESIGN Subjects with NF1 were ascertained from the NF1 Patient Registry Initiative and through a clinical record database of patients at a large academic medical center. Logistic regression was employed to calculate ORs and 95% CIs to analyze differences in the odds of brain tumor diagnosis by race (White, Black, Asian, other/unknown) and ethnic (Hispanic vs non-Hispanic) groups. RESULTS Data from a total of 1546, 629, and 2038 individuals who were ascertained from the NF1 Patient Registry Initiative, clinical records, and pooled datasets were analyzed, respectively. After adjusting for birth year, we observed a significantly reduced odds of brain tumor diagnoses in individuals self-identified or clinically reported as Black (OR = 0.13, 95% CI 0.05-0.31), Asian (OR = 0.15, 95% CI 0.04-0.64), and other/unknown (OR = 0.61, 95% CI 0.41-0.93) race compared with those with reported as White race. There was no significant difference in the odds of pediatric brain tumor diagnosis by Hispanic ethnicity. CONCLUSIONS Consistent with prior smaller studies, these data suggest that pediatric brain tumor diagnoses vary by race in individuals with NF1. Reasons underlying observed differences by race warrant further investigation.
Collapse
Affiliation(s)
| | | | | | - Bree Porcelli
- School of Medicine, Washington University, St. Louis, MO
| | - David H. Gutmann
- Department of Neurology, School of Medicine, Washington University, St. Louis, MO
| | - Kimberly J. Johnson
- Brown School, Washington University, St. Louis, MO,Department of Pediatrics, School of Medicine, Washington University in St. Louis, MO
| |
Collapse
|
49
|
Burket JA, Benson AD, Tang AH, Deutsch SI. NMDA receptor activation regulates sociability by its effect on mTOR signaling activity. Prog Neuropsychopharmacol Biol Psychiatry 2015; 60:60-5. [PMID: 25703582 PMCID: PMC5549784 DOI: 10.1016/j.pnpbp.2015.02.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 01/22/2015] [Accepted: 02/15/2015] [Indexed: 11/20/2022]
Abstract
Tuberous Sclerosis Complex is one example of a syndromic form of autism spectrum disorder associated with disinhibited activity of mTORC1 in neurons (e.g., cerebellar Purkinje cells). mTORC1 is a complex protein possessing serine/threonine kinase activity and a key downstream molecule in a signaling cascade beginning at the cell surface with the transduction of neurotransmitters (e.g., glutamate and acetylcholine) and nerve growth factors (e.g., Brain-Derived Neurotrophic Factor). Interestingly, the severity of the intellectual disability in Tuberous Sclerosis Complex may relate more to this metabolic disturbance (i.e., overactivity of mTOR signaling) than the density of cortical tubers. Several recent reports showed that rapamycin, an inhibitor of mTORC1, improved sociability and other symptoms in mouse models of Tuberous Sclerosis Complex and autism spectrum disorder, consistent with mTORC1 overactivity playing an important pathogenic role. NMDA receptor activation may also dampen mTORC1 activity by at least two possible mechanisms: regulating intraneuronal accumulation of arginine and the phosphorylation status of a specific extracellular signal regulating kinase (i.e., ERK1/2), both of which are "drivers" of mTORC1 activity. Conceivably, the prosocial effects of targeting the NMDA receptor with agonists in mouse models of autism spectrum disorders result from their ability to dampen mTORC1 activity in neurons. Strategies for dampening mTORC1 overactivity by NMDA receptor activation may be preferred to its direct inhibition in chronic neurodevelopmental disorders, such as autism spectrum disorders.
Collapse
Affiliation(s)
- Jessica A Burket
- Department of Psychiatry and Behavioral Sciences, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Andrew D Benson
- Department of Psychiatry and Behavioral Sciences, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Amy H Tang
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Stephen I Deutsch
- Department of Psychiatry and Behavioral Sciences, Eastern Virginia Medical School, Norfolk, VA, United States.
| |
Collapse
|