1
|
Rava A, Buzzelli V, Feo A, Ascone F, Di Trapano M, Schiavi S, Carbone E, Pasquadibisceglie A, Polticelli F, Manduca A, Trezza V. Role of peroxisome proliferator-activated receptors α and γ in mediating the beneficial effects of β-caryophyllene in a rat model of fragile X syndrome. Prog Neuropsychopharmacol Biol Psychiatry 2024; 136:111234. [PMID: 39725014 DOI: 10.1016/j.pnpbp.2024.111234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/17/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024]
Abstract
β-Caryophyllene (BCP) is a naturally occurring sesquiterpene found in numerous plant species, including Cannabis sativa. BCP has shown a high safety profile and a wide range of biological functions, including beneficial effects in neurodegenerative and inflammatory diseases. Here, we used behavioral, pharmacological, and in-silico docking analyses to investigate the effects and mechanism of action of BCP in Fragile X Syndrome (FXS), the most common inherited cause of Autism Spectrum Disorder (ASD) and intellectual disability. To this aim, we used the recently validated Fmr1-Δexon 8 rat model of FXS, that is also a genetic rat model of ASD. Acute and repeated oral administration of BCP rescued the cognitive deficits displayed by Fmr1-Δexon 8 rats, without inducing tolerance after repeated administration. These beneficial effects were mediated by activation of hippocampal peroxisome proliferator-activated receptors (PPARs) α and γ, and were mimicked by the PPARα agonist Fenofibrate and the PPARγ agonist Pioglitazone. Conversely, CB2 cannabinoid receptors were not involved. Docking analyses further confirmed the ability of BCP to bind rat PPARs. Together, our findings demonstrate that hippocampal PPARs α and γ play a role in the cognitive deficits observed in a rat model of FXS, and provide first preclinical evidence about the efficacy and mechanism of action of BCP in neurodevelopmental disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Antonia Manduca
- Dept. Science, Roma Tre University, Rome, Italy; Dept. Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Neuroendocrinology, Metabolism and Neuropharmacology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Viviana Trezza
- Dept. Science, Roma Tre University, Rome, Italy; Neuroendocrinology, Metabolism and Neuropharmacology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy.
| |
Collapse
|
2
|
Thurman AJ, Nunnally AD, Nguyen V, Berry-Kravis E, Sterling A, Edgin J, Hamilton D, Aschkenasy J, Abbeduto L. Short-term and Long-term Stability of the Autism Diagnostic Observation Schedule (ADOS-2) Calibrated Comparison Scores (CCS) and Classification Scores in Youth with Down Syndrome or Fragile X Syndrome with Intellectual Disability. J Autism Dev Disord 2024:10.1007/s10803-024-06535-8. [PMID: 39251531 DOI: 10.1007/s10803-024-06535-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2024] [Indexed: 09/11/2024]
Abstract
Autism diagnosis in individuals with fragile X syndrome (FXS) or Down syndrome (DS) with co-occurring intellectual disability is complex since clinicians often must consider other co-occurring behavioral features. Understanding how best to assess the features of autism in individuals with these conditions is crucial. In this study, we consider the short-term and long-term psychometric consistency of the Autism Diagnostic Observation Schedule-2 (ADOS-2) calibrated comparison scores (CCSs) and ASD classifications in individuals with FXS or DS. 76 individuals with DS (39 males; Mage = 15.27) and 90 individuals with FXS (71 males; Mage = 14.52 years) completed an assessment battery (ADOS-2, abbreviated IQ assessment and semi-structured language sample) at three timepoints (initial visit, short-term stability visit, long-term stability visit). All CCSs were found to have short-and long-term consistency for both groups, with lowest reliability scores for the repetitive behaviors (RRB) CCSs. Decreased reliability of RRB CCSs was found in the DS group than the FXS group. Variable short- and long-term ASD classifications were observed in both groups, with significantly higher variability in the DS group. Across groups, participants with variable classifications had lower ADOS-2 CCSs and higher language scores than those with stable ASD classifications. In the FXS group, those with variable classifications earned higher cognitive scores than did participants with stable ASD classifications. These findings highlight the high incidence of autism symptomatology in individuals with DS or FXS and co-occurring intellectual disability, while elucidating the short- and long-term variability of symptom expression in the context of structured observational tasks such as the ADOS-2.
Collapse
Affiliation(s)
- Angela John Thurman
- MIND Institute, University of California, Davis, 2825 50th Street, Room 2335, Sacramento, CA, 95817, USA.
- Department of Psychiatry and Behavioral Sciences, University of California Davis Medical Center, Sacramento, USA.
| | - Amanda Dimachkie Nunnally
- MIND Institute, University of California, Davis, 2825 50th Street, Room 2335, Sacramento, CA, 95817, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis Medical Center, Sacramento, USA
| | - Vivian Nguyen
- MIND Institute, University of California, Davis, 2825 50th Street, Room 2335, Sacramento, CA, 95817, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis Medical Center, Sacramento, USA
| | - Elizabeth Berry-Kravis
- Departments of Pediatrics, Neurological Sciences and Anatomy and Cell Biology, Rush University Medical Center, Chicago, USA
| | - Audra Sterling
- Waisman Center and Department of Communication Sciences and Disorders, University of Wisconsin-Madison, Madison, USA
| | - Jamie Edgin
- Department of Psychology, Virginia Tech, Blacksburg, USA
| | - Debra Hamilton
- Department of Human Genetics, Emory University, Atlanta, USA
| | | | - Leonard Abbeduto
- MIND Institute, University of California, Davis, 2825 50th Street, Room 2335, Sacramento, CA, 95817, USA
- Department of Psychiatry and Behavioral Sciences, University of California Davis Medical Center, Sacramento, USA
| |
Collapse
|
3
|
Proteau-Lemieux M, Knoth IS, Davoudi S, Martin CO, Bélanger AM, Fontaine V, Côté V, Agbogba K, Vachon K, Whitlock K, Biag HMB, Thurman AJ, Rosenfelt C, Tassone F, Frei J, Capano L, Abbeduto L, Jacquemont S, Hessl D, Hagerman RJ, Schneider A, Bolduc F, Anagnostou E, Lippe S. Specific EEG resting state biomarkers in FXS and ASD. J Neurodev Disord 2024; 16:53. [PMID: 39251926 PMCID: PMC11382468 DOI: 10.1186/s11689-024-09570-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 08/23/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) and autism spectrum disorder (ASD) are neurodevelopmental conditions that often have a substantial impact on daily functioning and quality of life. FXS is the most common cause of inherited intellectual disability (ID) and the most common monogenetic cause of ASD. Previous literature has shown that electrophysiological activity measured by electroencephalogram (EEG) during resting state is perturbated in FXS and ASD. However, whether electrophysiological profiles of participants with FXS and ASD are similar remains unclear. The aim of this study was to compare EEG alterations found in these two clinical populations presenting varying degrees of cognitive and behavioral impairments. METHODS Resting state EEG signal complexity, alpha peak frequency (APF) and power spectral density (PSD) were compared between 47 participants with FXS (aged between 5-20), 49 participants with ASD (aged between 6-17), and 52 neurotypical (NT) controls with a similar age distribution using MANCOVAs with age as covariate when appropriate. MANCOVAs controlling for age, when appropriate, and nonverbal intelligence quotient (NVIQ) score were subsequently performed to determine the impact of cognitive functioning on EEG alterations. RESULTS Our results showed that FXS participants manifested decreased signal complexity and APF compared to ASD participants and NT controls, as well as altered power in the theta, alpha and low gamma frequency bands. ASD participants showed exaggerated beta power compared to FXS participants and NT controls, as well as enhanced low and high gamma power compared to NT controls. However, ASD participants did not manifest altered signal complexity or APF. Furthermore, when controlling for NVIQ, results of decreased complexity in higher scales and lower APF in FXS participants compared to NT controls and ASD participants were not replicated. CONCLUSIONS These findings suggest that signal complexity and APF might reflect cognitive functioning, while altered power in the low gamma frequency band might be associated with neurodevelopmental conditions, particularly FXS and ASD.
Collapse
Affiliation(s)
- Mélodie Proteau-Lemieux
- Department of Psychology, University of Montreal, Montreal, QC, Canada
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada
| | - Inga Sophia Knoth
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada
| | - Saeideh Davoudi
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada
- Department of Neuroscience, University of Montreal, Montreal, QC, Canada
| | | | - Anne-Marie Bélanger
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada
| | - Valérie Fontaine
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada
| | - Valérie Côté
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada
| | - Kristian Agbogba
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada
| | | | | | - Hazel Maridith Barlahan Biag
- Department of Pediatrics and MIND Institute, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Angela John Thurman
- Department of Psychiatry and Behavioral Sciences and MIND Institute, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Cory Rosenfelt
- Department of Pediatric Neurology, University of Alberta, Edmonton, AB, Canada
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Julia Frei
- McMaster University of Ottawa, Ottawa, ON, Canada
| | - Lucia Capano
- Queen's University of Kingston, Kingston, ON, Canada
| | - Leonard Abbeduto
- Department of Psychiatry and Behavioral Sciences and MIND Institute, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Sébastien Jacquemont
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada
- Department of Pediatrics, University of Montreal, Montreal, QC, Canada
| | - David Hessl
- Department of Psychiatry and Behavioral Sciences and MIND Institute, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Randi Jenssen Hagerman
- Department of Pediatrics and MIND Institute, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Andrea Schneider
- Department of Pediatrics and MIND Institute, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Francois Bolduc
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Evdokia Anagnostou
- Department of Paediatrics, University of Toronto, Toronto, ON, Canada
- Holland Bloorview Research Center, Toronto, ON, Canada
| | - Sarah Lippe
- Department of Psychology, University of Montreal, Montreal, QC, Canada.
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada.
| |
Collapse
|
4
|
Thacker JS, Bettio L, Liang S, Shkolnikov I, Collingridge GL, Christie BR. Adiponectin rescues synaptic plasticity in the dentate gyrus of a mouse model of Fragile X Syndrome. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230221. [PMID: 38853554 PMCID: PMC11343265 DOI: 10.1098/rstb.2023.0221] [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] [Received: 10/13/2023] [Revised: 01/15/2024] [Accepted: 01/23/2024] [Indexed: 06/11/2024] Open
Abstract
Fragile X syndrome (FXS) is the most common inherited cause of intellectual disability and is the leading known single-gene cause of autism spectrum disorder. Patients with FXS display varied behavioural deficits that include mild to severe cognitive impairments in addition to mood disorders. Currently, there is no cure for this condition; however, there is an emerging focus on therapies that inhibit mechanistic target of rapamycin (mTOR)-dependent protein synthesis owing to the clinical effectiveness of metformin for alleviating some behavioural symptoms in FXS. Adiponectin (APN) is a neurohormone that is released by adipocytes and provides an alternative means to inhibit mTOR activation in the brain. In these studies, we show that Fmr1 knockout mice, like patients with FXS, show reduced levels of circulating APN and that both long-term potentiation (LTP) and long-term depression (LTD) in the dentate gyrus (DG) are impaired. Brief (20 min) incubation of hippocampal slices in APN (50 nM) was able to rescue both LTP and LTD in the DG and increased both the surface expression and phosphorylation of GluA1 receptors. These results provide evidence for reduced APN levels in FXS playing a role in decreasing bidirectional synaptic plasticity and show that therapies which enhance APN levels may have therapeutic potential for this and related conditions.This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.
Collapse
Affiliation(s)
- Jonathan S. Thacker
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Health System, Toronto, OntarioM5G 1X5, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, OntarioM5S 1A8, Canada
- Division of Medical Sciences, University of Victoria, Victoria, British ColumbiaV8P 5C2, Canada
| | - Luis Bettio
- Division of Medical Sciences, University of Victoria, Victoria, British ColumbiaV8P 5C2, Canada
| | - Stanley Liang
- Division of Medical Sciences, University of Victoria, Victoria, British ColumbiaV8P 5C2, Canada
| | - Irene Shkolnikov
- Division of Medical Sciences, University of Victoria, Victoria, British ColumbiaV8P 5C2, Canada
| | - Graham L. Collingridge
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Health System, Toronto, OntarioM5G 1X5, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, OntarioM5S 1A8, Canada
| | - Brian R. Christie
- Division of Medical Sciences, University of Victoria, Victoria, British ColumbiaV8P 5C2, Canada
- Island Medical Program, University of British Columbia, Victoria, British ColumbiaV8P 5C2, Canada
- Center for Behavioral Teratology, San Diego State University, San Diego, CA92120, USA
| |
Collapse
|
5
|
Metkar SK, Yan Y, Lu Y, Lu J, Zhu X, Du F, Xu Y. Phosphodiesterase 2 and Its Isoform A as Therapeutic Targets in the Central Nervous System Disorders. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:941-955. [PMID: 37855295 DOI: 10.2174/1871527323666230811093126] [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: 01/06/2023] [Revised: 06/15/2023] [Accepted: 07/07/2023] [Indexed: 10/20/2023]
Abstract
Cyclic adenosine monophosphates (cAMP) and cyclic guanosine monophosphate (cGMP) are two essential second messengers, which are hydrolyzed by phosphodiesterase's (PDEs), such as PDE-2. Pharmacological inhibition of PDE-2 (PDE2A) in the central nervous system improves cAMP and cGMP signaling, which controls downstream proteins related to neuropsychiatric, neurodegenerative, and neurodevelopmental disorders. Considering that there are no specific treatments for these disorders, PDE-2 inhibitors' development has gained more attention in the recent decade. There is high demand for developing new-generation drugs targeting PDE2 for treating diseases in the central nervous and peripheral systems. This review summarizes the relationship between PDE-2 with neuropsychiatric, neurodegenerative, and neurodevelopmental disorders as well as its possible treatment, mainly involving inhibitors of PDE2.
Collapse
Affiliation(s)
- Sanjay K Metkar
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Yuqing Yan
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Yue Lu
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Jianming Lu
- Codex BioSolutions Inc. 12358 Parklawn Drive, Suite 250A, Rockville, MD 20852, Maryland
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio 44106; USA
| | - Fu Du
- FD NeuroTechnologies Consulting & Services, Inc., Columbia, MD 21046, Maryland
| | - Ying Xu
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| |
Collapse
|
6
|
Sharma T, Kundu N, Kaur S, Shankaraswamy J, Saxena S. Why to target G-quadruplexes using peptides: Next-generation G4-interacting ligands. J Pept Sci 2023; 29:e3491. [PMID: 37009771 DOI: 10.1002/psc.3491] [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] [Received: 11/18/2022] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/04/2023]
Abstract
Guanine-rich oligonucleotides existing in both DNA and RNA are able to fold into four-stranded DNA secondary structures via Hoogsteen type hydrogen-bonding, where four guanines self-assemble into a square planar arrangement, which, when stacked upon each other, results in the formation of higher-order structures called G-quadruplexes. Their distribution is not random; they are more frequently present at telomeres, proto-oncogenic promoters, introns, 5'- and 3'-untranslated regions, stem cell markers, ribosome binding sites and so forth and are associated with various biological functions, all of which play a pivotal role in various incurable diseases like cancer and cellular ageing. Several studies have suggested that G-quadruplexes could not regulate biological processes by themselves; instead, various proteins take part in this regulation and can be important therapeutic targets. There are certain limitations in using whole G4-protein for therapeutics purpose because of its high manufacturing cost, laborious structure prediction, dynamic nature, unavailability for oral administration due to its degradation in the gut and inefficient penetration to reach the target site because of the large size. Hence, biologically active peptides can be the potential candidates for therapeutic intervention instead of the whole G4-protein complex. In this review, we aimed to clarify the biological roles of G4s, how we can identify them throughout the genome via bioinformatics, the proteins interacting with G4s and how G4-interacting peptide molecules may be the potential next-generation ligands for targeting the G4 motifs located in biologically important regions.
Collapse
Affiliation(s)
- Taniya Sharma
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Nikita Kundu
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Sarvpreet Kaur
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Jadala Shankaraswamy
- Department of Fruit Science, College of Horticulture, Mojerla, Sri Konda Laxman Telangana State Horticultural University, Budwel, Telangana, India
| | - Sarika Saxena
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| |
Collapse
|
7
|
Martin GE, Lee M, Bicknell K, Goodkind A, Maltman N, Losh M. A longitudinal investigation of pragmatic language across contexts in autism and related neurodevelopmental conditions. Front Neurol 2023; 14:1155691. [PMID: 37545730 PMCID: PMC10402743 DOI: 10.3389/fneur.2023.1155691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 07/10/2023] [Indexed: 08/08/2023] Open
Abstract
Background Pragmatic language, or the use of language in social contexts, is a critical skill in daily life, supporting social interactions and the development of meaningful social relationships. Pragmatic language is universally impacted in autism spectrum disorder (ASD) and pragmatic deficits are also common in other neurodevelopmental conditions, particularly those related to ASD, such as fragile X syndrome (FXS). This study used a multi-method, longitudinal approach to characterize potentially unique pragmatic profiles across different neurodevelopmental disabilities, and across contexts that varied in degree of social demand. The utility of computational linguistic analyses, as an efficient tool for capturing pragmatic abilities, was also explored. Methods Pragmatic skills of boys with idiopathic ASD (ASD-O, n = 43), FXS with and without ASD (FXS-ASD, n = 57; FXS-O, n = 14), Down syndrome (DS, n = 22), and typical development (TD, n = 24) were compared using variables obtained from a standardized measure, narrative, and semi-naturalistic conversation at up to three time points. Results Pragmatic language was most significantly impacted among males with ASD-O and FXS-ASD across all three contexts, with more difficulties in the least structured context (conversation), and also some differences based on FXS comorbidity. Patterns of group differences were more nuanced for boys with FXS-O and DS, with context having less of an impact. Clinical groups demonstrated minimal changes in pragmatic skills with age, with some exceptions. Computational language measurement tools showed some utility for measuring pragmatic skills, but were not as successful as traditional methods at capturing differences between clinical groups. Conclusion Overlap and differences between ASD and other forms of neurodevelopmental disability in general, and between idiopathic and syndromic ASD in particular, have important implications for developing precisely tailored assessment and intervention approaches, consistent with a personalized medicine approach to clinical study and care in ASD.
Collapse
Affiliation(s)
- Gary E. Martin
- Department of Communication Sciences and Disorders, St. John’s University, Staten Island, NY, United States
| | - Michelle Lee
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, United States
- Department of Child and Adolescent Psychiatry, Child Study Center, Hassenfeld Children’s Hospital at NYU Langone, New York, NY, United States
| | - Klinton Bicknell
- Department of Linguistics, Northwestern University, Evanston, IL, United States
- Duolingo, Pittsburgh, PA, United States
| | - Adam Goodkind
- Department of Linguistics, Northwestern University, Evanston, IL, United States
- Department of Communication Studies, Northwestern University, Evanston, IL, United States
| | - Nell Maltman
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, United States
- Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Molly Losh
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, United States
| |
Collapse
|
8
|
Schiavi S, Manduca A, Carbone E, Buzzelli V, Rava A, Feo A, Ascone F, Morena M, Campolongo P, Hill MN, Trezza V. Anandamide and 2-arachidonoylglycerol differentially modulate autistic-like traits in a genetic model of autism based on FMR1 deletion in rats. Neuropsychopharmacology 2023; 48:897-907. [PMID: 36114286 PMCID: PMC10156791 DOI: 10.1038/s41386-022-01454-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/20/2022] [Accepted: 09/01/2022] [Indexed: 11/09/2022]
Abstract
Autism spectrum disorder (ASD) has a multifactorial etiology. Major efforts are underway to understand the neurobiological bases of ASD and to develop efficacious treatment strategies. Recently, the use of cannabinoid compounds in children with neurodevelopmental disorders including ASD has received increasing attention. Beyond anecdotal reports of efficacy, however, there is limited current evidence supporting such an intervention and the clinical studies currently available have intrinsic limitations that make the interpretation of the findings challenging. Furthermore, as the mechanisms underlying the beneficial effects of cannabinoid compounds in neurodevelopmental disorders are still largely unknown, the use of drugs targeting the endocannabinoid system remains controversial. Here, we studied the role of endocannabinoid neurotransmission in the autistic-like traits displayed by the recently validated Fmr1-Δexon 8 rat model of autism. Fmr1-Δexon 8 rats showed reduced anandamide levels in the hippocampus and increased 2-arachidonoylglycerol (2-AG) content in the amygdala. Systemic and intra-hippocampal potentiation of anandamide tone through administration of the anandamide hydrolysis inhibitor URB597 ameliorated the cognitive deficits displayed by Fmr1-Δexon 8 rats along development, as assessed through the novel object and social discrimination tasks. Moreover, blockade of amygdalar 2-AG signaling through intra-amygdala administration of the CB1 receptor antagonist SR141716A prevented the altered sociability displayed by Fmr1-Δexon 8 rats. These findings demonstrate that anandamide and 2-AG differentially modulate specific autistic-like traits in Fmr1-Δexon 8 rats in a brain region-specific manner, suggesting that fine changes in endocannabinoid mechanisms contribute to ASD-related behavioral phenotypes.
Collapse
Affiliation(s)
- Sara Schiavi
- Department of Science, Roma Tre University, Rome, Italy
| | - Antonia Manduca
- Department of Science, Roma Tre University, Rome, Italy
- Neuroendocrinology, Metabolism and Neuropharmacology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| | | | | | | | | | | | - Maria Morena
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- Neuropsychopharmacology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
- Departments of Cell Biology and Anatomy & Psychiatry, Hotchkiss Brain Institute and Mathison Center for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Patrizia Campolongo
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- Neuropsychopharmacology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Matthew N Hill
- Departments of Cell Biology and Anatomy & Psychiatry, Hotchkiss Brain Institute and Mathison Center for Mental Health Research and Education, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Viviana Trezza
- Department of Science, Roma Tre University, Rome, Italy.
| |
Collapse
|
9
|
Reyes ZMD, Lynch E, Henry J, De Simone LM, Sobotka SA. Diagnosis of autism in a rare case of tyrosine hydroxylase deficiency: a case report. BMC Med Genomics 2023; 16:78. [PMID: 37041529 PMCID: PMC10088295 DOI: 10.1186/s12920-023-01510-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 04/04/2023] [Indexed: 04/13/2023] Open
Abstract
BACKGROUND Tyrosine hydroxylase deficiency (THD) is a rare movement disorder with broad phenotypic expression caused by bi-allelic mutations in the TH gene, which encode for tyrosine hydroxylase (TH) protein. Some patients with THD have improvement in dystonia with carbidopa-levodopa, a synthetic form of dopamine typically used in Parkinson's disease, and are considered to have dopa-responsive THD. THD has been found in 0.5-1 per million persons, although due to overlapping symptoms with other disorders and the need for genetic testing, prevalence is likely underestimated. Existing literature describes some patients with THD having intellectual disability, but comorbid autism spectrum disorder (ASD) has not been reported. CASE PRESENTATION A nearly 3-year-old boy was referred to pediatric neurology due to hypotonia, delayed motor milestones, and expressive speech delay. Whole exome sequencing confirmed tyrosine hydroxylase deficiency, detecting a novel variant p.S307C first reported here. The child was treated with carbidopa-levodopa with an excellent response, resulting in improved balance, fewer falls, and improved ability to jump, run and climb stairs. He was determined to have dopa-responsive THD. Due to his delays in expressive speech, the boy also had an assessment with a developmental and behavioral pediatrician, who identified a pattern of social pragmatic speech delay, sensory sensitivities, and restricted interests, and determined that he met criteria for a diagnosis of ASD. CONCLUSIONS While ASD can stand alone as a clinical diagnosis, it is also a cardinal feature of other genetically-based neurological disorders. To our knowledge, this is the first case that describes a patient with both disorders. Perhaps THD may be among the genetic disorders linked with ASD.
Collapse
Affiliation(s)
| | - Emma Lynch
- Section of Developmental and Behavioral Pediatrics, Department of Pediatrics, The University of Chicago, 950 East 61St Street, Suite 207, Chicago, IL, 60637, USA
| | - Julia Henry
- Section of Pediatric Neurology, Department of Pediatrics, The University of Chicago, Chicago, USA
| | | | - Sarah A Sobotka
- Section of Developmental and Behavioral Pediatrics, Department of Pediatrics, The University of Chicago, 950 East 61St Street, Suite 207, Chicago, IL, 60637, USA.
| |
Collapse
|
10
|
Sex-Related Changes in the Clinical, Genetic, Electrophysiological, Connectivity, and Molecular Presentations of ASD: A Comparison between Human and Animal Models of ASD with Reference to Our Data. Int J Mol Sci 2023; 24:ijms24043287. [PMID: 36834699 PMCID: PMC9965966 DOI: 10.3390/ijms24043287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/28/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
The etiology of autism spectrum disorder (ASD) is genetic, environmental, and epigenetic. In addition to sex differences in the prevalence of ASD, which is 3-4 times more common in males, there are also distinct clinical, molecular, electrophysiological, and pathophysiological differences between sexes. In human, males with ASD have more externalizing problems (i.e., attention-deficit hyperactivity disorder), more severe communication and social problems, as well as repetitive movements. Females with ASD generally exhibit fewer severe communication problems, less repetitive and stereotyped behavior, but more internalizing problems, such as depression and anxiety. Females need a higher load of genetic changes related to ASD compared to males. There are also sex differences in brain structure, connectivity, and electrophysiology. Genetic or non-genetic experimental animal models of ASD-like behavior, when studied for sex differences, showed some neurobehavioral and electrophysiological differences between male and female animals depending on the specific model. We previously carried out studies on behavioral and molecular differences between male and female mice treated with valproic acid, either prenatally or early postnatally, that exhibited ASD-like behavior and found distinct differences between the sexes, the female mice performing better on tests measuring social interaction and undergoing changes in the expression of more genes in the brain compared to males. Interestingly, co-administration of S-adenosylmethionine alleviated the ASD-like behavioral symptoms and the gene-expression changes to the same extent in both sexes. The mechanisms underlying the sex differences are not yet fully understood.
Collapse
|
11
|
Maltman N, Hilvert E, Friedman L, Sterling A. Comparison of Linguistic Error Production in Conversational Language Among Boys With Fragile X Syndrome + Autism Spectrum Disorder and Autistic Boys. JOURNAL OF SPEECH, LANGUAGE, AND HEARING RESEARCH : JSLHR 2023; 66:296-313. [PMID: 36599155 PMCID: PMC10023176 DOI: 10.1044/2022_jslhr-22-00078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 07/18/2022] [Accepted: 10/07/2022] [Indexed: 06/17/2023]
Abstract
PURPOSE Expressive language impairments are common among school-age boys with fragile X syndrome (FXS) and autistic boys. Given the high co-occurrence of autism spectrum disorder (ASD) among individuals with FXS, cross-condition comparisons can elucidate the specificity of such impairments as they relate to ASD. Language samples can provide fruitful information regarding individuals' grammatical skills in less structured formats relative to standardized measures. This study examined grammatical errors produced during a conversational language sample among 20 boys with FXS and co-occurring ASD (FXS + ASD) and 19 autistic boys matched on ASD severity. METHOD Language samples were coded for omissions and errors at the word and utterance levels. Participants' grammatical errors were also compared to separate mental age-matched and mean length of utterance-matched boys from a reference database. RESULTS Boys with FXS + ASD and autistic boys produced similar rates of errors across all categories. Relative to their matched comparison groups, boys with FXS + ASD and autistic boys produced significantly more omissions during conversation. CONCLUSIONS These findings suggest that omissions may be a unique grammatical marker associated with the ASD phenotype. Further examination of omissions across diagnostic groups would aid in clarifying the specificity of omissions in the language phenotype of ASD.
Collapse
Affiliation(s)
| | | | - Laura Friedman
- Waisman Center, University of Wisconsin–Madison
- Department of Communication Sciences and Disorders, University of South Carolina, Columbia
| | - Audra Sterling
- Waisman Center, University of Wisconsin–Madison
- Department of Communication Sciences and Disorders, University of Wisconsin–Madison
| |
Collapse
|
12
|
Buzzelli V, Carbone E, Manduca A, Schiavi S, Feo A, Perederiy JV, Ambert KH, Hausman M, Trezza V. Psilocybin mitigates the cognitive deficits observed in a rat model of Fragile X syndrome. Psychopharmacology (Berl) 2023; 240:137-147. [PMID: 36469097 DOI: 10.1007/s00213-022-06286-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/20/2022] [Indexed: 12/12/2022]
Abstract
RATIONALE Fragile X syndrome (FXS) is the most common form of inherited intellectual disability (ID) and the leading monogenic cause of autism spectrum disorder (ASD). Serotonergic neurotransmission has a key role in the modulation of neuronal activity during development, and therefore, it has been hypothesized to be involved in ASD and co-occurring conditions including FXS. As serotonin is involved in synaptic remodeling and maturation, serotonergic insufficiency during childhood may have a compounding effect on brain patterning in neurodevelopmental disorders, manifesting as behavioral and emotional symptoms. Thus, compounds that stimulate serotonergic signaling such as psilocybin may offer promise as effective early interventions for developmental disorders such as ASD and FXS. OBJECTIVES The aim of the present study was to test whether different protocols of psilocybin administration mitigate cognitive deficits displayed by the recently validated Fmr1-Δexon 8 rat model of ASD, which is also a model of FXS. RESULTS Our results revealed that systemic and oral administration of psilocybin microdoses normalizes the aberrant cognitive performance displayed by adolescent Fmr1-Δexon 8 rats in the short-term version of the novel object recognition test-a measure of exploratory behavior, perception, and recognition. CONCLUSIONS These data support the hypothesis that serotonin-modulating drugs such as psilocybin may be useful to ameliorate ASD-related cognitive deficits. Overall, this study provides evidence of the beneficial effects of different schedules of psilocybin treatment in mitigating the short-term cognitive deficit observed in a rat model of FXS.
Collapse
Affiliation(s)
- Valeria Buzzelli
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Viale G. Marconi 446, 00146, Rome, Italy
| | - Emilia Carbone
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Viale G. Marconi 446, 00146, Rome, Italy
| | - Antonia Manduca
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Viale G. Marconi 446, 00146, Rome, Italy.,Neuroendocrinology, Metabolism and Neuropharmacology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Sara Schiavi
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Viale G. Marconi 446, 00146, Rome, Italy
| | - Alessandro Feo
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Viale G. Marconi 446, 00146, Rome, Italy
| | | | - Kyle H Ambert
- Nova Mentis Life Science Corp., Vancouver, BC, Canada
| | | | - Viviana Trezza
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Viale G. Marconi 446, 00146, Rome, Italy.
| |
Collapse
|
13
|
D’Elia A, Schiavi S, Manduca A, Rava A, Buzzelli V, Ascone F, Orsini T, Putti S, Soluri A, Galli F, Soluri A, Mattei M, Cicconi R, Massari R, Trezza V. FMR1 deletion in rats induces hyperactivity with no changes in striatal dopamine transporter availability. Sci Rep 2022; 12:22535. [PMID: 36581671 PMCID: PMC9800572 DOI: 10.1038/s41598-022-26986-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 12/22/2022] [Indexed: 12/30/2022] Open
Abstract
Autism Spectrum Disorder (ASD) is a pervasive neurodevelopmental disorder emerging in early life characterized by impairments in social interaction, poor verbal and non-verbal communication, and repetitive patterns of behaviors. Among the best-known genetic risk factors for ASD, there are mutations causing the loss of the Fragile X Messenger Ribonucleoprotein 1 (FMRP) leading to Fragile X syndrome (FXS), a common form of inherited intellectual disability and the leading monogenic cause of ASD. Being a pivotal regulator of motor activity, motivation, attention, and reward processing, dopaminergic neurotransmission has a key role in several neuropsychiatric disorders, including ASD. Fmr1 Δexon 8 rats have been validated as a genetic model of ASD based on FMR1 deletion, and they are also a rat model of FXS. Here, we performed behavioral, biochemical and in vivo SPECT neuroimaging experiments to investigate whether Fmr1 Δexon 8 rats display ASD-like repetitive behaviors associated with changes in striatal dopamine transporter (DAT) availability assessed through in vivo SPECT neuroimaging. At the behavioral level, Fmr1 Δexon 8 rats displayed hyperactivity in the open field test in the absence of repetitive behaviors in the hole board test. However, these behavioral alterations were not associated with changes in striatal DAT availability as assessed by non-invasive in vivo SPECT and Western blot analyses.
Collapse
Affiliation(s)
- Annunziata D’Elia
- grid.5326.20000 0001 1940 4177Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), c/o International Campus “A. Buzzati-Traverso”, Via E. Ramarini, 32, 00015 Monterotondo Scalo (Rome), Italy ,grid.8509.40000000121622106Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Viale G. Marconi 446, 00146 Rome, Italy
| | - Sara Schiavi
- grid.8509.40000000121622106Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Viale G. Marconi 446, 00146 Rome, Italy
| | - Antonia Manduca
- grid.8509.40000000121622106Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Viale G. Marconi 446, 00146 Rome, Italy ,grid.417778.a0000 0001 0692 3437Neuroendocrinology, Metabolism and Neuropharmacology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Alessandro Rava
- grid.8509.40000000121622106Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Viale G. Marconi 446, 00146 Rome, Italy
| | - Valeria Buzzelli
- grid.8509.40000000121622106Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Viale G. Marconi 446, 00146 Rome, Italy
| | - Fabrizio Ascone
- grid.8509.40000000121622106Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Viale G. Marconi 446, 00146 Rome, Italy
| | - Tiziana Orsini
- grid.5326.20000 0001 1940 4177Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), c/o International Campus “A. Buzzati-Traverso”, Via E. Ramarini, 32, 00015 Monterotondo Scalo (Rome), Italy
| | - Sabrina Putti
- grid.5326.20000 0001 1940 4177Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), c/o International Campus “A. Buzzati-Traverso”, Via E. Ramarini, 32, 00015 Monterotondo Scalo (Rome), Italy
| | - Andrea Soluri
- grid.5326.20000 0001 1940 4177Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), c/o International Campus “A. Buzzati-Traverso”, Via E. Ramarini, 32, 00015 Monterotondo Scalo (Rome), Italy ,grid.9657.d0000 0004 1757 5329Unit of Molecular Neurosciences, University Campus Bio-Medico, Rome, Rome, Italy
| | - Filippo Galli
- grid.7841.aNuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, Faculty of Medicine and Psychology, “Sapienza” University of Rome, Rome, Italy
| | - Alessandro Soluri
- grid.5326.20000 0001 1940 4177Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), c/o International Campus “A. Buzzati-Traverso”, Via E. Ramarini, 32, 00015 Monterotondo Scalo (Rome), Italy
| | - Maurizio Mattei
- grid.6530.00000 0001 2300 0941Department of Biology and Centro di Servizi Interdipartimentale-Stazione per la Tecnologia Animale, “Tor Vergata” University, Rome, Italy
| | - Rosella Cicconi
- grid.6530.00000 0001 2300 0941Department of Biology and Centro di Servizi Interdipartimentale-Stazione per la Tecnologia Animale, “Tor Vergata” University, Rome, Italy
| | - Roberto Massari
- grid.5326.20000 0001 1940 4177Institute of Biochemistry and Cell Biology (IBBC), National Research Council of Italy (CNR), c/o International Campus “A. Buzzati-Traverso”, Via E. Ramarini, 32, 00015 Monterotondo Scalo (Rome), Italy
| | - Viviana Trezza
- grid.8509.40000000121622106Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Viale G. Marconi 446, 00146 Rome, Italy
| |
Collapse
|
14
|
Wilkinson EH, Britton TC, Hall SS. Examining Phenotypic Differences in Gaze Avoidance Between Autism Spectrum Disorder and Fragile X Syndrome. AMERICAN JOURNAL ON INTELLECTUAL AND DEVELOPMENTAL DISABILITIES 2022; 127:435-454. [PMID: 36306410 PMCID: PMC9667749 DOI: 10.1352/1944-7558-127.6.435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 08/31/2021] [Indexed: 05/22/2023]
Abstract
We examined potential phenotypic differences in eye gaze avoidance exhibited by boys with autism spectrum disorder (ASD) and boys with fragile X syndrome (FXS). In Study 1, the Eye Contact Avoidance Scale (ECAS) was administered to caregivers of boys aged 7-18 years with FXS (n = 148), ASD (n = 168), and mixed developmental disabilities (MDD; n = 128). In Study 2, subsets of boys with FXS (n = 31) and boys with ASD (n = 25) received a brief behavioral treatment probe to improve eye contact. Results showed that boys with FXS obtained significantly higher scores on the ECAS compared to boys with ASD and MDD. Exposure to the brief behavioral treatment probe resulted in significant decreases in scores for boys with FXS, but not for boys with ASD.
Collapse
Affiliation(s)
- Ellen H Wilkinson
- Ellen H. Wilkinson, Tobias C. Britton, and Scott S. Hall, Stanford University School of Medicine
| | - Tobias C Britton
- Ellen H. Wilkinson, Tobias C. Britton, and Scott S. Hall, Stanford University School of Medicine
| | - Scott S Hall
- Ellen H. Wilkinson, Tobias C. Britton, and Scott S. Hall, Stanford University School of Medicine
| |
Collapse
|
15
|
Abolghasemi A, Carullo MP, Aguilera EC, Laroui A, Plantefeve R, Rojas D, Benachenhou S, Ramírez MV, Proteau-Lemieux M, Lepage JF, Corbin F, Plourde M, Farez M, Cogram P, Çaku A. Alteration of Fatty Acid Profile in Fragile X Syndrome. Int J Mol Sci 2022; 23:ijms231810815. [PMID: 36142726 PMCID: PMC9502195 DOI: 10.3390/ijms231810815] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/28/2022] [Accepted: 09/12/2022] [Indexed: 11/16/2022] Open
Abstract
Fragile X Syndrome (FXS) is the most prevalent monogenic cause of Autism Spectrum Disorders (ASDs). Despite a common genetic etiology, the affected individuals display heterogenous metabolic abnormalities including hypocholesterolemia. Although changes in the metabolism of fatty acids (FAs) have been reported in various neuropsychiatric disorders, it has not been explored in humans with FXS. In this study, we investigated the FA profiles of two different groups: (1) an Argentinian group, including FXS individuals and age- and sex-matched controls, and (2) a French-Canadian group, including FXS individuals and their age- and sex-matched controls. Since phospholipid FAs are an indicator of medium-term diet and endogenous metabolism, we quantified the FA profile in plasma phospholipids using gas chromatography. Our results showed significantly lower levels in various plasma FAs including saturated, monosaturated, ω-6 polyunsaturated, and ω-3 polyunsaturated FAs in FXS individuals compared to the controls. A decrease in the EPA/ALA (eicosapentaenoic acid/alpha linoleic acid) ratio and an increase in the DPA/EPA (docosapentaenoic acid/eicosapentaenoic acid) ratio suggest an alteration associated with desaturase and elongase activity, respectively. We conclude that FXS individuals present an abnormal profile of FAs, specifically FAs belonging to the ω-3 family, that might open new avenues of treatment to improve core symptoms of the disorder.
Collapse
Affiliation(s)
- Armita Abolghasemi
- Centre de Recherche du CHUS, Department of Biochemistry, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Maria Paulina Carullo
- Department of Child Neurology, Raúl Carrea Institute for Neurological Research (FLENI), Buenos Aires C1428AQK, Argentina
| | - Ester Cisneros Aguilera
- Centre de Recherche sur le Vieillissement, Departments of Medicine, University of Sherbrooke, Sherbrooke, QC J1H 4C4, Canada
| | - Asma Laroui
- Centre de Recherche du CHUS, Department of Biochemistry, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Rosalie Plantefeve
- Centre de Recherche du CHUS, Department of Biochemistry, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Daniela Rojas
- Centre de Recherche du CHUS, Department of Biochemistry, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Serine Benachenhou
- Centre de Recherche du CHUS, Department of Biochemistry, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - María Victoria Ramírez
- Department of Child Neurology, Raúl Carrea Institute for Neurological Research (FLENI), Buenos Aires C1428AQK, Argentina
| | - Mélodie Proteau-Lemieux
- Department of Pediatrics and Centre de Recherche du CHUS, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Jean-François Lepage
- Department of Pediatrics and Centre de Recherche du CHUS, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - François Corbin
- Centre de Recherche du CHUS, Department of Biochemistry, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Mélanie Plourde
- Centre de Recherche sur le Vieillissement, Departments of Medicine, University of Sherbrooke, Sherbrooke, QC J1H 4C4, Canada
| | - Mauricio Farez
- Department of Child Neurology, Raúl Carrea Institute for Neurological Research (FLENI), Buenos Aires C1428AQK, Argentina
| | - Patricia Cogram
- Biomedicine Division, Centre for Systems Biotechnology, Fraunhofer Chile Research Foundation, Santiago 7500588, Chile
| | - Artuela Çaku
- Centre de Recherche du CHUS, Department of Biochemistry, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Correspondence:
| |
Collapse
|
16
|
Klusek J, O'Connor SL, Hickey A, Hills KJ, Abbeduto L, Roberts JE. Attention/Deficit Hyperactivity Disorder in Adolescent and Young Adult Males With Fragile X Syndrome. AMERICAN JOURNAL ON INTELLECTUAL AND DEVELOPMENTAL DISABILITIES 2022; 127:213-230. [PMID: 35443049 PMCID: PMC9414675 DOI: 10.1352/1944-7558-127.3.213] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/26/2021] [Indexed: 05/03/2023]
Abstract
This study characterized the rates of attention-deficit/hyperactivity disorder (ADHD) in adolescent and young adult males with fragile X syndrome (FXS) using a multi-method approach integrating a DSM-based parent interview (Children's Interview for Psychiatric Syndromes; P-ChIPS, Fristad et al., 1998) and a parent rating scale (Child Behavior Checklist; CBCL, Achenbach, 2001). Thirty-one males with FXS, aged 16-24 years, participated. Forty-two percent met DSM-5 criteria for ADHD and 35% exceeded the CBCL cut-offs. Agreement between the two classification methods was fair (κ = 0.38). Autism symptom severity and nonverbal cognitive ability did not predict ADHD diagnoses/symptoms. Results show high rates of ADHD in males with FXS during late adolescence and young adulthood, which are not accounted for by impaired nonverbal cognitive skills or autism symptom severity. DSM-based ADHD-specific scales are recommended over broadband symptom scales to improve accurate identification.
Collapse
Affiliation(s)
- Jessica Klusek
- Jessica Klusek, Shannon L. O'Connor, Alexandra Hickey and Kimberly J. Hills, University of South Carolina
| | - Shannon L O'Connor
- Jessica Klusek, Shannon L. O'Connor, Alexandra Hickey and Kimberly J. Hills, University of South Carolina
| | - Alexandra Hickey
- Jessica Klusek, Shannon L. O'Connor, Alexandra Hickey and Kimberly J. Hills, University of South Carolina
| | - Kimberly J Hills
- Jessica Klusek, Shannon L. O'Connor, Alexandra Hickey and Kimberly J. Hills, University of South Carolina
| | | | | |
Collapse
|
17
|
Budimirovic DB, Protic DD, Delahunty CM, Andrews HF, Choo TH, Xu Q, Berry-Kravis E, Kaufmann WE. Sleep problems in fragile X syndrome: Cross-sectional analysis of a large clinic-based cohort. Am J Med Genet A 2022; 188:1029-1039. [PMID: 34889523 PMCID: PMC11057226 DOI: 10.1002/ajmg.a.62601] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 11/20/2021] [Accepted: 11/24/2021] [Indexed: 11/11/2022]
Abstract
Fragile X syndrome (FXS), the leading cause of inherited intellectual disability and autism spectrum disorder, is associated with multiple neurobehavioral abnormalities including sleep difficulties. Nonetheless, frequency, severity, and consequences of sleep problems are still unclear. The Fragile X Online Registry with Accessible Research Database (FORWARD-version-3), including Clinician Report and Parent Report forms, was analyzed for frequency, severity, relationship with behavioral problems, and impact of sleep difficulties in a mainly pediatric cohort. A focused evaluation of sleep apnea was also conducted. Six surveyed sleep difficulties were moderately frequent (~23%-46%), relatively mild, affected predominantly younger males, and considered a problem for 7%-20% of families. Snoring was more prevalent in older individuals. All sleep difficulties were associated with irritability/aggression and most also to hyperactivity. Only severe snoring was correlated with sleep apnea (loud snoring: 30%; sleep apnea: 2%-3%). Sleep difficulties are prevalent in children with FXS and, although they tend to be mild, they are associated with behavioral problems and negative impact to families. Because of its cross-sectional nature, clinic-origin, use of ad hoc data collection forms, and lack of treatment data, the present study should be considered foundational for future research aiming at better recognition and management of sleep problems in FXS.
Collapse
Affiliation(s)
- Dejan B. Budimirovic
- Department of Psychiatry, Fragile X Clinic, Kennedy Krieger Institute, JHMI, Baltimore, MD, USA
- Department of Psychiatry & Behavioral Sciences-Child Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | - Howard F. Andrews
- Departments of Psychiatry and Biostatistics, Columbia University, Irving Medical Center, New York, NY, USA
- Department of Psychiatry, Mental Health Data Science, Columbia University Medical Center and NY State Psychiatric Institute, New York, NY, USA
| | - Tse-Hwei Choo
- Department of Psychiatry, Mental Health Data Science, Columbia University Medical Center and NY State Psychiatric Institute, New York, NY, USA
| | - Qing Xu
- Department of Psychiatry, Mental Health Data Science, Columbia University Medical Center and NY State Psychiatric Institute, New York, NY, USA
| | - Elizabeth Berry-Kravis
- Departments of Pediatrics, Neurological Sciences, and Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - Walter E. Kaufmann
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
- Boston Children’s Hospital, Boston, MA, USA
| |
Collapse
|
18
|
Eye-Tracking Studies in Adults with Autism Spectrum Disorder: A Systematic Review and Meta-analysis. J Autism Dev Disord 2022; 53:2430-2443. [PMID: 35355174 DOI: 10.1007/s10803-022-05524-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2022] [Indexed: 10/18/2022]
Abstract
Eye-tracking studies have shown potential in effectively discriminating between autism spectrum disorder (ASD) and non-ASD groups. The main objective of the present study was to conduct a systematic review and meta-analysis of eye-tracking studies in adults with ASD. A total of 22 studies were included for meta-analysis. Eyes and Non-Social regions proved better for discriminating between ASD and non-ASD adults, while fixation duration seems to be the outcome to choose. Active engaged tasks seem to reduce differences between ASD and non-ASD adults, regardless of the emotional content of the stimuli/task. Proportional fixation duration on eyes and non-social areas in non-active tasks (e.g. free viewing) seems to be the best eye-tracking design for increasing the sensitivity and specificity in ASD adults.
Collapse
|
19
|
Schiavi S, Carbone E, Melancia F, di Masi A, Jarjat M, Brau F, Cardarelli S, Giorgi M, Bardoni B, Trezza V. Phosphodiesterase 2A inhibition corrects the aberrant behavioral traits observed in genetic and environmental preclinical models of Autism Spectrum Disorder. Transl Psychiatry 2022; 12:119. [PMID: 35338117 PMCID: PMC8956682 DOI: 10.1038/s41398-022-01885-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/01/2022] [Accepted: 03/07/2022] [Indexed: 11/20/2022] Open
Abstract
Pharmacological inhibition of phosphodiesterase 2A (PDE2A), which catalyzes the hydrolysis of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP), has recently been proposed as a novel therapeutic tool for Fragile X Syndrome (FXS), the leading monogenic cause of Autism Spectrum Disorder (ASD). Here, we investigated the role of PDE2A in ASD pathogenesis using two rat models that reflect one of either the genetic or environmental factors involved in the human disease: the genetic Fmr1-Δexon 8 rat model and the environmental rat model based on prenatal exposure to valproic acid (VPA, 500 mg/kg). Prior to behavioral testing, the offspring was treated with the PDE2A inhibitor BAY607550 (0.05 mg/kg at infancy, 0.1 mg/kg at adolescence and adulthood). Socio-communicative symptoms were assessed in both models through the ultrasonic vocalization test at infancy and three-chamber test at adolescence and adulthood, while cognitive impairments were assessed by the novel object recognition test in Fmr1-Δexon 8 rats (adolescence and adulthood) and by the inhibitory avoidance test in VPA-exposed rats (adulthood). PDE2A enzymatic activity in VPA-exposed infant rats was also assessed. In line with the increased PDE2A enzymatic activity previously observed in the brain of Fmr1-KO animals, we found an altered upstream regulation of PDE2A activity in the brain of VPA-exposed rats at an early developmental age (p < 0.05). Pharmacological inhibition of PDE2A normalized the communicative (p < 0.01, p < 0.05), social (p < 0.001, p < 0.05), and cognitive impairment (p < 0.001) displayed by both Fmr1-Δexon 8 and VPA-exposed rats. Altogether, these data highlight a key role of PDE2A in brain development and point to PDE2A inhibition as a promising pharmacological approach for the deficits common to both FXS and ASD.
Collapse
Affiliation(s)
- Sara Schiavi
- Deptartment of Science, University "Roma Tre", Rome, Italy
| | - Emilia Carbone
- Deptartment of Science, University "Roma Tre", Rome, Italy
| | | | | | | | - Fréderic Brau
- Université Côte d'Azur, CNRS, IPMC, 06560, Valbonne, France
| | - Silvia Cardarelli
- Deptartment of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185, Rome, Italy
| | - Mauro Giorgi
- Deptartment of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185, Rome, Italy
| | - Barbara Bardoni
- Université Côte d'Azur, Inserm, CNRS, IPMC, 06560, Valbonne, France.
| | - Viviana Trezza
- Deptartment of Science, University "Roma Tre", Rome, Italy.
| |
Collapse
|
20
|
Bangert K, Scott KS, Adams C, Kisenwether JS, Giuffre L, Reed J, Thurman AJ, Abbeduto L, Klusek J. Cluttering in the Speech of Young Men With Fragile X Syndrome. JOURNAL OF SPEECH, LANGUAGE, AND HEARING RESEARCH : JSLHR 2022; 65:954-969. [PMID: 35196138 PMCID: PMC9150725 DOI: 10.1044/2021_jslhr-21-00446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/29/2021] [Accepted: 11/22/2021] [Indexed: 06/14/2023]
Abstract
PURPOSE Cluttering is a fluency disorder that has been noted clinically in individuals with fragile X syndrome (FXS). Yet, cluttering has not been systematically characterized in this population, hindering identification and intervention efforts. This study examined the rates of cluttering in male young adults with FXS using expert clinical opinion, the alignment between expert clinical opinion and objectively quantified features of cluttering from language transcripts, and the association between cluttering and aspects of the FXS phenotype. METHOD Thirty-six men with FXS (aged 18-26 years; M = 22, SD = 2.35) contributed language samples and completed measures of nonverbal cognition, autism symptoms, anxiety, and symptoms of attention-deficit/hyperactivity disorder (ADHD). The presence of cluttering was determined by the consensus of two clinical experts in fluency disorders based on characteristics exhibited in the language sample. Cluttering features (speech rate, disfluencies, etc.) were also objectively quantified from the language transcripts. RESULTS Clinical experts determined that 50% of participants met the criteria for a cluttering diagnosis. Phrase repetitions were the most salient feature that distinguished individuals who cluttered. Although the presence of cluttering was not associated with autism symptoms or mean length of utterance, cluttering was more likely to occur when nonverbal cognitive ability was higher, ADHD symptoms were elevated, and anxiety symptoms were low. CONCLUSIONS Half of the male young adults with FXS exhibited cluttering, which supports FXS as a genetic diagnosis that is highly enriched for risk of cluttering. Cluttering was associated with increased ADHD symptoms and cognitive ability and reduced anxiety symptoms. This study contributes a new description of the clinical presentation of cluttering in men with FXS and may lead to improved understanding of the potential underlying mechanisms of cluttering and eventual refinements to treatment and diagnosis.
Collapse
Affiliation(s)
- Katherine Bangert
- Department of Communication Sciences and Disorders, University of South Carolina, Columbia
- Department of Psychology, University of South Carolina, Columbia
| | | | - Charley Adams
- Department of Communication Sciences and Disorders, University of South Carolina, Columbia
| | | | - Lisa Giuffre
- Department of Speech-Language Pathology, Misericordia University, Dallas, PA
| | - Jenna Reed
- Department of Speech-Language Pathology, Misericordia University, Dallas, PA
| | - Angela John Thurman
- Department of Psychiatry and Behavioral Sciences, University of California Davis Health, Sacramento
- MIND Institute, University of California Davis Health, Sacramento
| | - Leonard Abbeduto
- Department of Psychiatry and Behavioral Sciences, University of California Davis Health, Sacramento
- MIND Institute, University of California Davis Health, Sacramento
| | - Jessica Klusek
- Department of Communication Sciences and Disorders, University of South Carolina, Columbia
| |
Collapse
|
21
|
Behavior Problems and Social Competence in Fragile X Syndrome: A Systematic Review. Genes (Basel) 2022; 13:genes13020280. [PMID: 35205326 PMCID: PMC8871871 DOI: 10.3390/genes13020280] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 02/06/2023] Open
Abstract
Fragile X syndrome (FXS) causes intellectual disability and is the known leading cause of autism. Common problems in FXS include behavior and social problems. Along with syndromic characteristics and autism comorbidity, environmental factors might influence these difficulties. This systematic review focuses on the last 20 years of studies concerning behavior and social problems in FXS, considering environmental and personal variables that might influence both problems. Three databases were reviewed, leading to fifty-one studies meeting the inclusion criteria. Attention deficit hyperactivity disorder (ADHD) problems remain the greatest behavior problems, with behavioral problems and social competence being stable during the 20 years. Some developmental trajectories might have changed due to higher methodological control, such as aggressive behavior and attention problems. The socialization trajectory from childhood to adolescence remains unclear. Comorbidity with autism in individuals with FXS increased behavior problems and worsened social competence profiles. At the same time, comparisons between individuals with comorbid FXS and autism and individuals with autism might help define the comorbid phenotype. Environmental factors and parental characteristics influenced behavior problems and social competence. Higher methodological control is needed in studies including autism symptomatology and parental characteristics. More studies comparing autism in FXS with idiopathic autism are needed to discern differences between conditions.
Collapse
|
22
|
Change in Behavior Problems from Childhood Through Adolescence for Children with Fragile X Syndrome. J Autism Dev Disord 2021; 52:4056-4066. [PMID: 34536164 PMCID: PMC8449523 DOI: 10.1007/s10803-021-05270-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2021] [Indexed: 11/01/2022]
Abstract
In this study, we examined trajectories of specific domains of behavior problems (i.e., attention problems, depression/anxiety, and aggressive behavior) from age 6 to 18 in a sample of 55 children with fragile X syndrome. We also examined autism status and early parenting as predictors of subsequent behavioral trajectories. We found that attention problems and aggressive behavior declined steadily from childhood through adolescence whereas anxious/depressed behavior demonstrated relative stability over the same period. Youth with highly flexible mothers displayed more optional trajectories of improvement in attention problems.
Collapse
|
23
|
Sechi S, Karimpour-Ghahnavieh A, Frappaolo A, Di Francesco L, Piergentili R, Schininà E, D’Avino PP, Giansanti MG. Identification of GOLPH3 Partners in Drosophila Unveils Potential Novel Roles in Tumorigenesis and Neural Disorders. Cells 2021; 10:cells10092336. [PMID: 34571985 PMCID: PMC8468827 DOI: 10.3390/cells10092336] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 12/28/2022] Open
Abstract
Golgi phosphoprotein 3 (GOLPH3) is a highly conserved peripheral membrane protein localized to the Golgi apparatus and the cytosol. GOLPH3 binding to Golgi membranes depends on phosphatidylinositol 4-phosphate [PI(4)P] and regulates Golgi architecture and vesicle trafficking. GOLPH3 overexpression has been correlated with poor prognosis in several cancers, but the molecular mechanisms that link GOLPH3 to malignant transformation are poorly understood. We recently showed that PI(4)P-GOLPH3 couples membrane trafficking with contractile ring assembly during cytokinesis in dividing Drosophila spermatocytes. Here, we use affinity purification coupled with mass spectrometry (AP-MS) to identify the protein-protein interaction network (interactome) of Drosophila GOLPH3 in testes. Analysis of the GOLPH3 interactome revealed enrichment for proteins involved in vesicle-mediated trafficking, cell proliferation and cytoskeleton dynamics. In particular, we found that dGOLPH3 interacts with the Drosophila orthologs of Fragile X mental retardation protein and Ataxin-2, suggesting a potential role in the pathophysiology of disorders of the nervous system. Our findings suggest novel molecular targets associated with GOLPH3 that might be relevant for therapeutic intervention in cancers and other human diseases.
Collapse
Affiliation(s)
- Stefano Sechi
- Istituto di Biologia e Patologia Molecolari del CNR, c/o Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, Piazzale A. Moro 5, 00185 Roma, Italy; (S.S.); (A.K.-G.); (A.F.); (R.P.)
| | - Angela Karimpour-Ghahnavieh
- Istituto di Biologia e Patologia Molecolari del CNR, c/o Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, Piazzale A. Moro 5, 00185 Roma, Italy; (S.S.); (A.K.-G.); (A.F.); (R.P.)
| | - Anna Frappaolo
- Istituto di Biologia e Patologia Molecolari del CNR, c/o Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, Piazzale A. Moro 5, 00185 Roma, Italy; (S.S.); (A.K.-G.); (A.F.); (R.P.)
| | - Laura Di Francesco
- Dipartimento di Scienze Biochimiche A. Rossi Fanelli, Sapienza Università di Roma, Piazzale A. Moro 5, 00185 Roma, Italy; (L.D.F.); (E.S.)
| | - Roberto Piergentili
- Istituto di Biologia e Patologia Molecolari del CNR, c/o Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, Piazzale A. Moro 5, 00185 Roma, Italy; (S.S.); (A.K.-G.); (A.F.); (R.P.)
| | - Eugenia Schininà
- Dipartimento di Scienze Biochimiche A. Rossi Fanelli, Sapienza Università di Roma, Piazzale A. Moro 5, 00185 Roma, Italy; (L.D.F.); (E.S.)
| | - Pier Paolo D’Avino
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK;
| | - Maria Grazia Giansanti
- Istituto di Biologia e Patologia Molecolari del CNR, c/o Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, Piazzale A. Moro 5, 00185 Roma, Italy; (S.S.); (A.K.-G.); (A.F.); (R.P.)
- Correspondence: ; Tel.: +39-064-991-2555
| |
Collapse
|
24
|
Marlborough M, Welham A, Jones C, Reckless S, Moss J. Autism spectrum disorder in females with fragile X syndrome: a systematic review and meta-analysis of prevalence. J Neurodev Disord 2021; 13:28. [PMID: 34294028 PMCID: PMC8299695 DOI: 10.1186/s11689-021-09362-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 04/03/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Whilst up to 60% of males with fragile X syndrome (FXS) meet criteria for autism spectrum disorder (ASD), the prevalence and nature of ASD in females with FXS remains unclear. METHOD A systematic literature search identified papers reporting ASD prevalence and/or symptomatology in females with FXS. RESULTS AND CONCLUSION Meta-analysis suggested that rates of ASD for females with FXS are reliably higher than for females in the general population (a random effects model estimated weighted average prevalence at 14%, 95% CI 13-18%). Whilst papers highlighted a number of social and repetitive difficulties for females with FXS, characteristic profiles of impairment are not clear. Possible associations between ASD traits and IQ, and between ASD and levels of fragile X mental retardation protein, are suggested, but data are equivocal.
Collapse
Affiliation(s)
- M Marlborough
- School of Psychology, George Davis Centre, University of Leicester, Leicester, UK
| | - A Welham
- School of Psychology, George Davis Centre, University of Leicester, Leicester, UK.
| | - C Jones
- School of Psychology, George Davis Centre, University of Leicester, Leicester, UK
| | - S Reckless
- School of Psychology, George Davis Centre, University of Leicester, Leicester, UK
| | - J Moss
- School of Psychology, George Davis Centre, University of Leicester, Leicester, UK
| |
Collapse
|
25
|
Banco MT, Ferré-D'Amaré AR. The emerging structural complexity of G-quadruplex RNAs. RNA (NEW YORK, N.Y.) 2021; 27:390-402. [PMID: 33483368 PMCID: PMC7962482 DOI: 10.1261/rna.078238.120] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
G-quadruplexes (G4s) are four-stranded nucleic acid structures that arise from the stacking of G-quartets, cyclic arrangements of four guanines engaged in Hoogsteen base-pairing. Until recently, most RNA G4 structures were thought to conform to a sequence pattern in which guanines stacking within the G4 would also be contiguous in sequence (e.g., four successive guanine trinucleotide tracts separated by loop nucleotides). Such a sequence restriction, and the stereochemical constraints inherent to RNA (arising, in particular, from the presence of the 2'-OH), dictate relatively simple RNA G4 structures. Recent crystallographic and solution NMR structure determinations of a number of in vitro selected RNA aptamers have revealed RNA G4 structures of unprecedented complexity. Structures of the Sc1 aptamer that binds an RGG peptide from the Fragile-X mental retardation protein, various fluorescence turn-on aptamers (Corn, Mango, and Spinach), and the spiegelmer that binds the complement protein C5a, in particular, reveal complexity hitherto unsuspected in RNA G4s, including nucleotides in syn conformation, locally inverted strand polarity, and nucleotide quartets that are not all-G. Common to these new structures, the sequences folding into G4s do not conform to the requirement that guanine stacks arise from consecutive (contiguous in sequence) nucleotides. This review highlights how emancipation from this constraint drastically expands the structural possibilities of RNA G-quadruplexes.
Collapse
Affiliation(s)
- Michael T Banco
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland 20892-8012, USA
| | - Adrian R Ferré-D'Amaré
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, Maryland 20892-8012, USA
| |
Collapse
|
26
|
Wilkinson CL, Nelson CA. Increased aperiodic gamma power in young boys with Fragile X Syndrome is associated with better language ability. Mol Autism 2021; 12:17. [PMID: 33632320 PMCID: PMC7908768 DOI: 10.1186/s13229-021-00425-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/16/2021] [Indexed: 02/17/2023] Open
Abstract
Background The lack of robust and reliable clinical biomarkers in Fragile X Syndrome (FXS), the most common inherited form of intellectual disability, has limited the successful translation of bench-to-bedside therapeutics. While numerous drugs have shown promise in reversing synaptic and behavioral phenotypes in mouse models of FXS, none have demonstrated clinical efficacy in humans. Electroencephalographic (EEG) measures have been identified as candidate biomarkers as EEG recordings of both adults with FXS and mouse models of FXS consistently exhibit alterations in resting state and task-related activity. However, the developmental timing of these EEG differences is not known as thus far EEG studies have not focused on young children with FXS. Further, understanding how EEG differences are associated with core symptoms of FXS is crucial to successful use of EEG as a biomarker, and may improve our understanding of the disorder. Methods Resting-state EEG was collected from FXS boys with full mutation of Fmr1 (2.5–7 years old, n = 11) and compared with both age-matched (n = 12) and cognitive-matched (n = 12) typically developing boys. Power spectra (including aperiodic and periodic components) were compared using non-parametric cluster-based permutation testing. Associations between 30 and 50 Hz gamma power and cognitive, language, and behavioral measures were evaluated using Pearson correlation and linear regression with age as a covariate. Results FXS participants showed increased power in the beta/gamma range (~ 25–50 Hz) across multiple brain regions. Both a reduction in the aperiodic (1/f) slope and increase in beta/gamma periodic activity contributed to the significant increase in high-frequency power. Increased gamma power, driven by the aperiodic component, was associated with better language ability in the FXS group. No association was observed between gamma power and parent report measures of behavioral challenges, sensory hypersensitivities, or adaptive behaviors. Limitations The study sample size was small, although comparable to other human studies in rare-genetic disorders. Findings are also limited to males in the age range studied. Conclusions Resting-state EEG measures from this study in young boys with FXS identified similar increases in gamma power previously reported in adults and mouse models. The observed positive association between resting state aperiodic gamma power and language development supports hypotheses that alterations in some EEG measures may reflect ongoing compensatory mechanisms. Supplementary Information The online version contains supplementary material available at 10.1186/s13229-021-00425-x.
Collapse
Affiliation(s)
- Carol L Wilkinson
- Division of Developmental Medicine, Boston Children's Hospital, 1 Autumn Street, 6th Floor, Boston, MA, 02115, USA.
| | - Charles A Nelson
- Division of Developmental Medicine, Boston Children's Hospital, 1 Autumn Street, 6th Floor, Boston, MA, 02115, USA
| |
Collapse
|
27
|
Winston M, Nayar K, Landau E, Maltman N, Sideris J, Zhou L, Sharp K, Berry-Kravis E, Losh M. A Unique Visual Attention Profile Associated With the FMR1 Premutation. Front Genet 2021; 12:591211. [PMID: 33633778 PMCID: PMC7901883 DOI: 10.3389/fgene.2021.591211] [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: 08/04/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
Atypical visual attention patterns have been observed among carriers of the fragile X mental retardation gene (FMR1) premutation (PM), with some similarities to visual attention patterns observed in autism spectrum disorder (ASD) and among clinically unaffected relatives of individuals with ASD. Patterns of visual attention could constitute biomarkers that can help to inform the neurocognitive profile of the PM, and that potentially span diagnostic boundaries. This study examined patterns of eye movement across an array of fixation measurements from three distinct eye-tracking tasks in order to investigate potentially overlapping profiles of visual attention among PM carriers, ASD parents, and parent controls. Logistic regression analyses were conducted to examine whether variables constituting a PM-specific looking profile were able to effectively predict group membership. Participants included 65PM female carriers, 188 ASD parents, and 84 parent controls. Analyses of fixations across the eye-tracking tasks, and their corresponding areas of interest, revealed a distinct visual attention pattern in carriers of the FMR1 PM, characterized by increased fixations on the mouth when viewing faces, more intense focus on bodies in socially complex scenes, and decreased fixations on salient characters and faces while narrating a wordless picture book. This set of variables was able to successfully differentiate individuals with the PM from controls (Sensitivity = 0.76, Specificity = 0.85, Accuracy = 0.77) as well as from ASD parents (Sensitivity = 0.70, Specificity = 0.80, Accuracy = 0.72), but did not show a strong distinction between ASD parents and controls (Accuracy = 0.62), indicating that this set of variables comprises a profile that is unique to PM carriers. Regarding predictive power, fixations toward the mouth when viewing faces was able to differentiate PM carriers from both ASD parents and controls, whereas fixations toward other social stimuli did not differentiate PM carriers from ASD parents, highlighting some overlap in visual attention patterns that could point toward shared neurobiological mechanisms. Results demonstrate a profile of visual attention that appears strongly associated with the FMR1 PM in women, and may constitute a meaningful biomarker.
Collapse
Affiliation(s)
- Molly Winston
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, United States
| | - Kritika Nayar
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, United States
| | - Emily Landau
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, United States
| | - Nell Maltman
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, United States
| | - John Sideris
- Chan Division of Occupational Science and Occupational Therapy, University of Southern California, Los Angeles, CA, United States
| | - Lili Zhou
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, United States
| | - Kevin Sharp
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, United States
| | | | - Molly Losh
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, United States
| |
Collapse
|
28
|
Shukla T, de la Peña JB, Perish JM, Ploski JE, Stumpf CR, Webster KR, Thorn CA, Campbell ZT. A Highly Selective MNK Inhibitor Rescues Deficits Associated with Fragile X Syndrome in Mice. Neurotherapeutics 2021; 18:624-639. [PMID: 33006091 PMCID: PMC8116363 DOI: 10.1007/s13311-020-00932-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2020] [Indexed: 12/22/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common inherited source of intellectual disability in humans. FXS is caused by mutations that trigger epigenetic silencing of the Fmr1 gene. Loss of Fmr1 results in increased activity of the mitogen-activated protein kinase (MAPK) pathway. An important downstream consequence is activation of the mitogen-activated protein kinase interacting protein kinase (MNK). MNK phosphorylates the mRNA cap-binding protein, eukaryotic initiation factor 4E (eIF4E). Excessive phosphorylation of eIF4E has been directly implicated in the cognitive and behavioral deficits associated with FXS. Pharmacological reduction of eIF4E phosphorylation is one potential strategy for FXS treatment. We demonstrate that systemic dosing of a highly specific, orally available MNK inhibitor, eFT508, attenuates numerous deficits associated with loss of Fmr1 in mice. eFT508 resolves a range of phenotypic abnormalities associated with FXS including macroorchidism, aberrant spinogenesis, and alterations in synaptic plasticity. Key behavioral deficits related to anxiety, social interaction, obsessive and repetitive activities, and object recognition are ameliorated by eFT508. Collectively, this work establishes eFT508 as a potential means to reverse deficits associated with FXS.
Collapse
Affiliation(s)
- Tarjani Shukla
- Department of Biological Sciences, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - June Bryan de la Peña
- Department of Biological Sciences, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - John M Perish
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Jonathan E Ploski
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, 75080, USA
| | | | | | - Catherine A Thorn
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Zachary T Campbell
- Department of Biological Sciences, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA.
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA.
| |
Collapse
|
29
|
Ezell J, Hogan A, Will EA, Smith K, Roberts J. Cardiac Startle Response and Clinical Outcomes in Preschool Children With Fragile X Syndrome and Autism Spectrum Disorder. Front Psychiatry 2021; 12:729127. [PMID: 35046845 PMCID: PMC8761627 DOI: 10.3389/fpsyt.2021.729127] [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: 06/22/2021] [Accepted: 11/09/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: Poor physiological regulation in response to threat is linked to multiple negative developmental outcomes including anxiety, which is highly prevalent and impairing in young children with neurodevelopmental disabilities like fragile X syndrome (FXS) and autism spectrum disorder (ASD). The present study contrasted cardiac startle response in pre-school-aged children with FXS, with and without ASD, to children with non-syndromic ASD (nsASD) and neurotypical controls (NT). The relationship of cardiac startle to non-verbal mental age (NVMA), ASD severity, and parent-reported anxiety was also examined. Method: Four age-matched groups of pre-school children participated including those with FXS without ASD (FXS-Only, n = 21), FXS with ASD (FXS+ASD, n = 17), nsASD (n = 42), and NT children (n = 27). Participants viewed a silent movie during which a single 200 ms 98-decibel white noise burst occurred. Cardiac activity was analyzed for pre-stimulus respiratory sinus arrhythmia (RSA) and the inter-beat intervals (IBI) at the auditory stimulus and 10 s post-stimulus. The Spence Pre-school Anxiety Scale, Autism Diagnostic Observation Schedule-2nd Edition, and Mullen Scales of Early Learning were examined in relation to startle response. Results: The nsASD group demonstrated heightened cardiac activity at the auditory stimulus and 10 s post-stimulus compared to the NT controls. Neither of the FXS groups showed differences from any other group. Higher pre-stimulus RSA was associated with reduced cardiac response across groups, while the relationship between cognitive ability and ASD severity to cardiac response varied between groups. Parent-reported anxiety was not associated with cardiac response for any group. Conclusion: These findings demonstrate group distinctions in cardiac responses to auditory startle. Although FXS and ASD share behavioral characteristics, the nsASD group showed a heightened cardiac startle response compared to the NT group that was not present in the FXS groups with or without ASD. Non-verbal mental age was associated with greater stimulus or post-stimulus reactivity for all groups except the FXS+ASD group, which showed no association between startle response and any clinical outcomes. Increased understanding of the relationship between physiological regulation and clinical outcomes will assist in identifying the timing and targets for effective interventions for individuals with neurodevelopmental disabilities.
Collapse
Affiliation(s)
- Jordan Ezell
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Abigail Hogan
- Department of Psychology, University of South Carolina, Columbia, SC, United States
| | - Elizabeth A Will
- Department of Psychology, University of South Carolina, Columbia, SC, United States
| | - Kayla Smith
- Department of Psychology, University of South Carolina, Columbia, SC, United States
| | - Jane Roberts
- Department of Psychology, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
30
|
Nguyen U, Tinsley B, Sen Y, Stein J, Palacios Y, Ceballos A, Welch C, Nzenkue K, Penn A, Murphy L, Leodones K, Casiquin J, Ivory I, Ghenta K, Danziger K, Widman E, Newman J, Triplehorn M, Hindi Z, Mulligan K. Exposure to bisphenol A differentially impacts neurodevelopment and behavior in Drosophila melanogaster from distinct genetic backgrounds. Neurotoxicology 2020; 82:146-157. [PMID: 33309840 DOI: 10.1016/j.neuro.2020.12.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/30/2020] [Accepted: 12/06/2020] [Indexed: 12/13/2022]
Abstract
Bisphenol A (BPA) is a ubiquitous environmental chemical that has been linked to behavioral differences in children and shown to impact critical neurodevelopmental processes in animal models. Though data is emerging, we still have an incomplete picture of how BPA disrupts neurodevelopment; in particular, how its impacts may vary across different genetic backgrounds. Given the genetic tractability of Drosophila melanogaster, they present a valuable model to address this question. Fruit flies are increasingly being used for assessment of neurotoxicants because of their relatively simple brain structure and variety of measurable behaviors. Here we investigated the neurodevelopmental impacts of BPA across two genetic strains of Drosophila-w1118 (control) and the Fragile X Syndrome (FXS) model-by examining both behavioral and neuronal phenotypes. We show that BPA induces hyperactivity in larvae, increases repetitive grooming behavior in adults, reduces courtship behavior, impairs axon guidance in the mushroom body, and disrupts neural stem cell development in the w1118 genetic strain. Remarkably, for every behavioral and neuronal phenotype examined, the impact of BPA in FXS flies was either insignificant or contrasted with the phenotypes observed in the w1118 strain. This data indicates that the neurodevelopmental impacts of BPA can vary widely depending on genetic background and suggests BPA may elicit a gene-environment interaction with Drosophila fragile X mental retardation 1 (dFmr1)-the ortholog of human FMR1, which causes Fragile X Syndrome and is associated with autism spectrum disorder.
Collapse
Affiliation(s)
- U Nguyen
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - B Tinsley
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - Y Sen
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - J Stein
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - Y Palacios
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - A Ceballos
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - C Welch
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - K Nzenkue
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - A Penn
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - L Murphy
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - K Leodones
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - J Casiquin
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - I Ivory
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - K Ghenta
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - K Danziger
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - E Widman
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - J Newman
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - M Triplehorn
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - Z Hindi
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States
| | - K Mulligan
- Department of Biological Sciences, California State University, Sacramento, 6000 J Street, Sacramento, CA, 95819, United States.
| |
Collapse
|
31
|
Banke TG, Barria A. Transient Enhanced GluA2 Expression in Young Hippocampal Neurons of a Fragile X Mouse Model. Front Synaptic Neurosci 2020; 12:588295. [PMID: 33343326 PMCID: PMC7745073 DOI: 10.3389/fnsyn.2020.588295] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 11/10/2020] [Indexed: 12/29/2022] Open
Abstract
AMPA-type glutamate receptors (AMPARs) are tetrameric ligand-gated channels made up of combinations of GluA1-4 subunits and play important roles in synaptic transmission and plasticity. Here, we have investigated the development of AMPAR-mediated synaptic transmission in the hippocampus of the Fmr1 knock-out (KO) mouse, a widely used model of Fragile X syndrome (FXS). FXS is the leading monogenic cause of intellectual disability and autism spectrum disorders (ASD) and it is considered a neurodevelopmental disorder. For that reason, we investigated synaptic properties and dendritic development in animals from an early stage when synapses are starting to form up to adulthood. We found that hippocampal CA1 pyramidal neurons in the Fmr1-KO mouse exhibit a higher AMPAR-NMDAR ratio early in development but reverses to normal values after P13. This increase was accompanied by a larger presence of the GluA2-subunit in synaptic AMPARs that will lead to altered Ca2+ permeability of AMPARs that could have a profound impact upon neural circuits, learning, and diseases. Following this, we found that young KO animals lack Long-term potentiation (LTP), a well-understood model of synaptic plasticity necessary for proper development of circuits, and exhibit an increased frequency of spontaneous miniature excitatory postsynaptic currents, a measure of synaptic density. Furthermore, post hoc morphological analysis of recorded neurons revealed altered dendritic branching in the KO group. Interestingly, all these anomalies are transitory and revert to normal values in older animals. Our data suggest that loss of FMRP during early development leads to temporary upregulation of the GluA2 subunit and this impacts synaptic plasticity and altering morphological dendritic branching.
Collapse
Affiliation(s)
- Tue G Banke
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States
| | - Andres Barria
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States
| |
Collapse
|
32
|
Improving the Diagnosis of Autism Spectrum Disorder in Fragile X Syndrome by Adapting the Social Communication Questionnaire and the Social Responsiveness Scale-2. J Autism Dev Disord 2020; 50:3276-3295. [PMID: 31342442 DOI: 10.1007/s10803-019-04148-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
We carried out a psychometric assessment of the Social Communication Questionnaire (SCQ) and the Social Responsiveness Scale (SRS-2) in fragile X syndrome (FXS), relative to clinician DSM5-based diagnosis of autism spectrum disorder (ASD) in FXS. This was followed by instrument revisions that included: removal of non-discriminating and/or low face validity items for FXS; use of receiver operating characteristic (ROC) curves to determine optimal cut points for the original and revised measures; an exploratory factor analysis to outline subscales better representing ASD in FXS; and creation of a "triple criteria" diagnosis to better delineate ASD subgroups in FXS. These methods improved the sensitivity and/or specificity of the SCQ and SRS-2, but diagnostic accuracy of ASD remains problematic in FXS.
Collapse
|
33
|
Abstract
Prospective longitudinal studies of idiopathic autism spectrum disorder (ASD) have provided insights into early symptoms and predictors of ASD during infancy, well before ASD can be diagnosed at age 2-3 years. However, research on the emergence of ASD in disorders with a known genetic etiology, contextualized in a developmental framework, is currently lacking. Using a biobehavioral multimethod approach, we (a) determined the rate of ASD in N = 51 preschoolers with fragile X syndrome (FXS) using a clinical best estimate (CBE) procedure with differential diagnoses of comorbid psychiatric disorders and (b) investigated trajectories of ASD symptoms and physiological arousal across infancy as predictors of ASD in preschoolers with FXS. ASD was not diagnosed if intellectual ability or psychiatric disorders better accounted for the symptoms. Our results determined that 60.7% of preschoolers with FXS met the Diagnostic and Statistical Manual of Mental Disorders (fifth edition) (DSM-5) criteria for ASD using the CBE procedure. In addition, 92% of these preschoolers presented with developmental delay and 45.4% also met criteria for psychiatric disorders, either anxiety, ADHD, or both. ASD diagnoses in preschoolers with FXS were predicted by elevated scores on traditional ASD screeners in addition to elevated autonomic arousal and avoidant eye contact from infancy.
Collapse
|
34
|
Begeman A, Son A, Litberg TJ, Wroblewski TH, Gehring T, Huizar Cabral V, Bourne J, Xuan Z, Horowitz S. G-Quadruplexes act as sequence-dependent protein chaperones. EMBO Rep 2020; 21:e49735. [PMID: 32945124 PMCID: PMC7534610 DOI: 10.15252/embr.201949735] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 07/29/2020] [Accepted: 08/05/2020] [Indexed: 01/06/2023] Open
Abstract
Maintaining proteome health is important for cell survival. Nucleic acids possess the ability to prevent protein aggregation more efficiently than traditional chaperone proteins. In this study, we explore the sequence specificity of the chaperone activity of nucleic acids. Evaluating over 500 nucleic acid sequences' effects on protein aggregation, we show that the holdase chaperone effect of nucleic acids is sequence-dependent. G-Quadruplexes prevent protein aggregation via quadruplex:protein oligomerization. They also increase the folded protein level of a biosensor in E. coli. These observations contextualize recent reports of quadruplexes playing important roles in aggregation-related diseases, such as fragile X and amyotrophic lateral sclerosis (ALS), and provide evidence that nucleic acids have the ability to modulate the folding environment of E. coli.
Collapse
Affiliation(s)
- Adam Begeman
- Department of Chemistry & Biochemistry, Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| | - Ahyun Son
- Department of Chemistry & Biochemistry, Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| | - Theodore J Litberg
- Department of Chemistry & Biochemistry, Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| | - Tadeusz H Wroblewski
- Department of Chemistry & Biochemistry, Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| | - Thane Gehring
- Department of Chemistry & Biochemistry, Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| | - Veronica Huizar Cabral
- Department of Chemistry & Biochemistry, Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| | - Jennifer Bourne
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Zhenyu Xuan
- Department of Biological Sciences, Center for Systems Biology, University of Texas at Dallas, Richardson, TX, USA
| | - Scott Horowitz
- Department of Chemistry & Biochemistry, Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| |
Collapse
|
35
|
Doherty BR, Longhi E, Cole V, Karmiloff-Smith A, Cornish K, Scerif G. Disentangling autism spectrum and attention-deficit/hyperactivity symptoms over development in fragile X syndrome. RESEARCH IN DEVELOPMENTAL DISABILITIES 2020; 104:103692. [PMID: 32505083 DOI: 10.1016/j.ridd.2020.103692] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 05/08/2020] [Accepted: 05/08/2020] [Indexed: 06/11/2023]
Abstract
Even genetic disorders associated with monogenic aetiologies are characterized by complex and variable risk for poor outcomes, highlighting the need to follow trajectories longitudinally. Here, we investigated the longitudinal relationships between attention deficit hyperactivity disorder (ADHD) and autism spectrum disorder (ASD) symptoms in a population at high risk for both: boys with fragile X syndrome. 59 boys with fragile X syndrome aged 3-10 years old at entry participated in this study, and were followed up one and two years after their first visit. As expected, we found strong relationships over three timepoints for ADHD symptoms (as measured by the parent-rated Conners scale) and ASD symptoms (as measured by the Social Communication Questionnaire [SCQ]). In addition, using structural equation modeling (SEM) we found that ADHD symptoms at time 2 predicted ASD symptoms at time 3, suggestive of a causal relationship. Importantly, these relationships hold when including chronological age at entry to the study, as well as when including severity of impairment as measured by IQ, and their effects on both ASD and ADHD symptoms do not reach significance. This result highlights the need to study outcomes longitudinally and it informs the comorbidity of the two symptom domains in FXS as well as their potential directionality, both of which have been little researched. In addition, our findings may suggest a future need to study how ADHD symptoms and their treatment impact individuals with ASD.
Collapse
Affiliation(s)
| | - Elena Longhi
- Department of Experimental Psychology, University of Oxford, United Kingdom
| | - Victoria Cole
- Department of Experimental Psychology, University of Oxford, United Kingdom
| | - Annette Karmiloff-Smith
- Department of Experimental Psychology, University of Oxford, United Kingdom; Turner Institute for Brain and Mental Health, Monash University, Melbourne, Australia
| | - Kim Cornish
- Turner Institute for Brain and Mental Health, Monash University, Melbourne, Australia
| | - Gaia Scerif
- Department of Experimental Psychology, University of Oxford, United Kingdom.
| |
Collapse
|
36
|
Gauthier-Boudreault C, Gallagher F, Trépanier J, Corbin F, Couture M. Factors impacting the transition to adulthood of youth with fragile X syndrome and their families: Facilitators, obstacles and needs. RESEARCH IN DEVELOPMENTAL DISABILITIES 2020; 103:103674. [PMID: 32535492 DOI: 10.1016/j.ridd.2020.103674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 04/24/2020] [Accepted: 04/24/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND The transition from school to adulthood is a critical time for families of youth with disabilities. Few studies have focused on the needs of families of youths with fragile X syndrome. This syndrome is often associated with intellectual disability and autism spectrum disorder, which creates specific needs that must be documented to improve transition planning. The aim of the current study was to document factors impacting transition planning and describe parents' experiences during this period. METHOD Individual interviews were conducted with thirteen parents of young people with fragile X syndrome. Two research team members analysed the interviews separately. RESULTS Factors related to the youth, the youth's family and the steps taken by the various institutions involved during this period seem to impact this transition and contributed to families' anxiety. A clear, uniform transition planning process, initiated early enough to have time for exploratory work placements, and gradual integration emerged as crucial facilitators for the parents in this study. CONCLUSIONS Understanding the reality of people with fragile X syndrome and their families will help to adapt services and develop concrete plans for their future.
Collapse
Affiliation(s)
- Camille Gauthier-Boudreault
- Health Sciences Research Programme, Université de Sherbrooke, 3001, 12(e) Avenue Nord, Sherbrooke, Québec, J1H 5N4, Canada.
| | | | | | - Francois Corbin
- Department of Biochemistry and Functional Genomic, Université de Sherbrooke, Canada
| | - Mélanie Couture
- Occupational Therapy Programme, School of Rehabilitation, Université de Sherbrooke, Canada
| |
Collapse
|
37
|
Usher LV, DaWalt LS, Hong J, Greenberg JS, Mailick MR. Trajectories of Change in the Behavioral and Health Phenotype of Adolescents and Adults with Fragile X Syndrome and Intellectual Disability: Longitudinal Trends Over a Decade. J Autism Dev Disord 2020; 50:2779-2792. [PMID: 32040800 PMCID: PMC7377950 DOI: 10.1007/s10803-020-04367-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This study examined trajectories of daily living skills, behavior problems, body mass index (BMI), and health conditions spanning nearly a decade in adolescents and adults with fragile X syndrome (N = 134; age range at study end = 19-49 years), examining influences of sex and autism spectrum disorder (ASD) symptoms. Hierarchical linear modeling revealed early increases in daily living skills, with decreases at older ages. Behavior problems became less severe over time, with some increases at older ages. Individuals gained weight and had increasing health problems over time. Fewer ASD symptoms were associated with greater daily living skills and fewer behavior problems at study start. This study offers some of the first prospective quantitative analyses of behavioral and health life course trajectories in FXS.
Collapse
Affiliation(s)
- Lauren V Usher
- University of Wisconsin-Madison Waisman Center, 1500 Highland Avenue, Room 531A, Madison, WI, 53705, USA.
| | - Leann S DaWalt
- University of Wisconsin-Madison Waisman Center, 1500 Highland Avenue, Room 531A, Madison, WI, 53705, USA
| | - Jinkuk Hong
- University of Wisconsin-Madison Waisman Center, 1500 Highland Avenue, Room 531A, Madison, WI, 53705, USA
| | - Jan S Greenberg
- University of Wisconsin-Madison Waisman Center, 1500 Highland Avenue, Room 531A, Madison, WI, 53705, USA
| | - Marsha R Mailick
- University of Wisconsin-Madison Waisman Center, 1500 Highland Avenue, Room 531A, Madison, WI, 53705, USA
| |
Collapse
|
38
|
Cordeiro L, Braden M, Coan E, Welnick N, Tanda T, Tartaglia N. Evaluating Social Interactions Using the Autism Screening Instrument for Education Planning-3rd Edition (ASIEP-3): Interaction Assessment in Children and Adults with Fragile X Syndrome. Brain Sci 2020; 10:E248. [PMID: 32331269 PMCID: PMC7226214 DOI: 10.3390/brainsci10040248] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/09/2020] [Accepted: 04/17/2020] [Indexed: 11/16/2022] Open
Abstract
An efficient and direct measure of social interactions and autism symptoms is needed for fragile X syndrome (FXS) research and clinical care. The Autism Screening Instrument for Educational Planning-Third Edition (ASIEP-3) Interaction assessment is a brief standardized measure that quantifies social responses under different conditions. The feasibility and validity of the ASIEP-3 was evaluated in 26 males and 13 females with FXS, along with cognitive testing and behavior questionnaires. The videos were scored at 10-second intervals, and the observed behaviors were scored as an interaction, independent play, no response, or aggression. In total, 39/41 participants successfully completed the ASIEP-3 (age M = 14.4 ± 10.2), with a range of cognitive abilities (abbreviated IQ (ABIQ) M = 58.9 ± 17.3, median = 50), behaviors (Aberrant Behavior Checklist (ABC) Total M = 37.00 ± 27.3), and autism diagnoses (N = 22/39). Reliable administration was demonstrated by all team members. The mean coded behaviors included interaction (40.6%), independent play (36.8%), no response (21.1%), and aggressive behavior (<10%). The interaction score was negatively correlated with the Social Communication Questionnaire (SCQ) score (p = 0.037), and the profiles differed by autism spectrum disorder (ASD) diagnosis. The intraclass correlation coefficients (ICCs) ranged from 0.79 to 0.93 for master's level and above. Administration of the ASIEP-3 was feasible for FXS across sex, age, ability, and behavior ratings by a trained research team. Reliable scoring required advanced training in the assessment of social development and FXS experience. The scores correlated to ratings and diagnoses of ASD. The ASIEP-3 shows promise to reliably index social interactions in FXS.
Collapse
Affiliation(s)
- Lisa Cordeiro
- Department of Pediatrics, School of Medicine, University of Colorado, Aurora, CO 80045, USA; (E.C.); (T.T.)
| | - Marcia Braden
- Licensed Psychologist, Private Practice, Colorado Springs, CO 80903, USA;
| | - Elizabeth Coan
- Department of Pediatrics, School of Medicine, University of Colorado, Aurora, CO 80045, USA; (E.C.); (T.T.)
- Developmental Pediatrics, Children’s Hospital Colorado, Aurora, CO 80045, USA;
| | - Nanastasia Welnick
- Developmental Pediatrics, Children’s Hospital Colorado, Aurora, CO 80045, USA;
| | - Tanea Tanda
- Department of Pediatrics, School of Medicine, University of Colorado, Aurora, CO 80045, USA; (E.C.); (T.T.)
| | - Nicole Tartaglia
- Department of Pediatrics, School of Medicine, University of Colorado, Aurora, CO 80045, USA; (E.C.); (T.T.)
- Developmental Pediatrics, Children’s Hospital Colorado, Aurora, CO 80045, USA;
| |
Collapse
|
39
|
Moskowitz LJ, Will EA, Black CJ, Roberts JE. Restricted and Repetitive Behaviors in Males and Females with Fragile X Syndrome: Developmental Trajectories in Toddlers Through Young Adults. J Autism Dev Disord 2020; 50:3957-3966. [PMID: 32221748 DOI: 10.1007/s10803-020-04459-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
There is limited research on the trajectory of restricted and repetitive behaviors (RRBs) in fragile X syndrome (FXS), with previous studies only examining males and/or examining RRBs as a unitary construct rather than delineating subtypes of RRBs. Thus, we described the trajectory of five subtypes of RRBs in 153 males and females with FXS (aged 1-18 years) with repeated measurement over time (445 total assessments). Multilevel modeling was used to test age-related differences in RRB subtypes between males and females with FXS, controlling for nonverbal IQ. Results showed that lower-order Sensory-Motor behaviors decreased over time for both males and females, while there was no significant change in the higher-order RRBs. The trajectory between males and females differed for Self-Injury.
Collapse
Affiliation(s)
- Lauren J Moskowitz
- Department of Psychology, St. John's University, 8000 Utopia Parkway, Queens, NY, 11439, USA.
| | - Elizabeth A Will
- Department of Psychology, University of South Carolina, Columbia, SC, USA
| | - Conner J Black
- Department of Psychology, University of South Carolina, Columbia, SC, USA
| | - Jane E Roberts
- Department of Psychology, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
40
|
Pharmacologic Interventions for Irritability, Aggression, Agitation and Self-Injurious Behavior in Fragile X Syndrome: An Initial Cross-Sectional Analysis. J Autism Dev Disord 2020; 49:4595-4602. [PMID: 31468273 DOI: 10.1007/s10803-019-04173-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Using a dataset involving 415 individuals with irritability, aggression, agitation and self-injury (IAAS) behaviors from the fragile X syndrome (FXS) FORWARD database, we describe the psychopharmacologic management of IAAS and features of the population of persons with FXS treated with drug therapy for IAAS. Among those with FXS exhibiting IAAS, individuals with FXS receiving drug treatment of IAAS were older, more predominantly male, have more significant intellectual disability, more like to have comorbid autism, hyperarousal, and social impairments. The most commonly utilized medications for IAAS in FXS are antipsychotic medications, specifically aripiprazole and risperidone (37% and 27%, respectively). The majority of subjects (63%) experienced no side effects noted from the use of their psychopharmacologic medications.
Collapse
|
41
|
Hilvert E. Expressive language abilities of boys with idiopathic autism spectrum disorder and boys with fragile X syndrome + autism spectrum disorder: Cross-context comparisons. AUTISM & DEVELOPMENTAL LANGUAGE IMPAIRMENTS 2020; 5:2396941520912118. [PMID: 36381548 PMCID: PMC9620452 DOI: 10.1177/2396941520912118] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
BACKGROUND AND AIMS Understanding the unique expressive language profiles of children with phenotypically similar, but distinct neurodevelopmental disorders, such as idiopathic autism spectrum disorder and fragile X syndrome with a co-diagnosis of autism spectrum disorder (fragile X syndrome + autism spectrum disorder), has both clinical and theoretical implications. However, comparative studies of these two clinical groups have been limited, and results have been inconsistent, partially as a result of different assessment methods being utilized. Thus, the current study compared the expressive language profiles of boys with idiopathic autism spectrum disorder and boys with fragile X syndrome + autism spectrum disorder and examined whether a similar linguistic profile emerged across different language sampling contexts: a semi-structured conversation and the Autism Diagnostic Observation Schedule. METHODS Eighteen boys with autism spectrum disorder (Mage = 13.25 years) and 19 boys with fragile X syndrome + autism spectrum disorder (Mage = 12.19 years), matched on autism spectrum disorder symptom severity and similar in terms of chronological age and mean length of utterance, participated in this study. Boys produced two language samples: one semi-structured conversation and one taken from the Autism Diagnostic Observation Schedule. Language samples were coded for talkativeness, lexical diversity, mean length of utterance, intelligibility, and repetitive or perseverative language. RESULTS Analyses revealed that boys with autism spectrum disorder produced language samples that were more lexically diverse and intelligible, and that included less topic perseveration compared to boys with fragile X syndrome + autism spectrum disorder. With regards to sampling context, boys in both groups were more talkative and produced longer and more intelligible utterances in their conversation sample compared to their Autism Diagnostic Observation Schedule sample. However, boys with autism spectrum disorder and fragile X syndrome + autism spectrum disorder used a higher proportion of topic perseveration during the conversation sample. CONCLUSIONS Overall, we found similarities as well as distinctions in the expressive language profiles of boys with fragile X syndrome + autism spectrum disorder and boys with idiopathic autism spectrum disorder. Moreover, the Autism Diagnostic Observation Schedule sample elicited a relatively different language profile than the conversation sample for boys in both groups. IMPLICATIONS These findings help to further elucidate the unique language phenotypes of boys with idiopathic autism spectrum disorder and boys with fragile X syndrome + autism spectrum disorder. Moreover, our findings indicate that multiple language samples may be needed to obtain a comprehensive account of a child's expressive language ability.
Collapse
Affiliation(s)
- Elizabeth Hilvert
- Elizabeth Hilvert, University of Wisconsin
Madison, 1500 Highland Ave, Madison, WI 53705, USA.
| |
Collapse
|
42
|
Haebig E, Sterling A, Barton-Hulsey A, Friedman L. Rates and Predictors of Co-occurring Autism Spectrum Disorder in Boys with Fragile X Syndrome. AUTISM & DEVELOPMENTAL LANGUAGE IMPAIRMENTS 2020; 5:2396941520905328. [PMID: 35847766 PMCID: PMC9281610 DOI: 10.1177/2396941520905328] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Background and aims Males with fragile X syndrome display many behavioral features of autism spectrum disorder. Despite this overlap, our understanding of autism spectrum disorder symptoms and severity in fragile X syndrome is limited due to variation in assessment methods in the literature. Furthermore, the relationship between autism spectrum disorder symptoms and child characteristics, like age, language, and cognitive abilities, are not well understood in individuals with fragile X syndrome. Therefore, the first research aim was to compare the rates of autism spectrum disorder classifications from three commonly reported autism spectrum disorder assessments in the literature. Our second research aim was to examine the relationship between autism spectrum disorder characteristics and other child characteristics. Methods The present study compared autism spectrum disorder classifications and symptoms using the Autism Diagnostic Observation Schedule, Autism Diagnostic Interview, Revised, and Childhood Autism Rating Scale, second edition in a sample of 33 school-age and adolescent boys with fragile X syndrome. In addition, the participants completed nonverbal IQ testing, expressive vocabulary and grammar tests, and a conversation language sample. Results The majority of the participants met criteria for autism spectrum disorder on the Autism Diagnostic Observation Schedule (96.97%) and Autism Diagnostic Interview, Revised (90.91%), while only half met criteria for autism spectrum disorder on the Childhood Autism Rating Scale, second edition. Sixteen boys (48.48%) met criteria for autism spectrum disorder on all three measures, and all participants met criteria for autism spectrum disorder on at least one measure. Expressive vocabulary accounted for a unique amount of variance in Childhood Autism Rating Scale, second edition and Autism Diagnostic Observation Schedule scores. Additionally, grammatical complexity accounted for a unique amount of variance in Childhood Autism Rating Scale, second edition scores. None of the child variables accounted for the variance found in Autism Diagnostic Interview, Revised scores. Although nonverbal IQ scores did not account for a significant amount of variance on the Autism Diagnostic Observation Schedule, Autism Diagnostic Interview, Revised, and Childhood Autism Rating Scale, Second Edition, boys who met criteria for autism spectrum disorder on all three measures had lower nonverbal IQ compared to the boys who did not. Additionally, mean length of utterance and expressive vocabulary scores were lower in the boys who met criteria for autism spectrum disorder on all three measures than those who did not. Conclusions Our findings identify areas of overlap and difference in the Autism Diagnostic Observation Schedule, Autism Diagnostic Interview, Revised, and Childhood Autism Rating Scale, second edition when used with males with fragile X syndrome. Variation in assessments may differentially identify the phenotypic behaviors of boys with fragile X syndrome that lead to a co-diagnosis of autism spectrum disorder, which contributes to the variation in reported co-morbidity of fragile X syndrome and autism spectrum disorder. Also, expressive language abilities, especially expressive vocabulary, are associated with autism spectrum disorder symptomatology. Implications: When interpreting comorbid fragile X syndrome and autism spectrum disorder rates in the literature, it is important to consider the assessment tool that was used. Although the assessments that we used in the present study yielded scores that were highly correlated (i.e. Autism Diagnostic Observation Schedule and Childhood Autism Rating Scale, second edition), their categorical classifications did not align perfectly. Our findings also highlight the importance of considering language skills when assessing autism spectrum disorder severity in fragile X syndrome.
Collapse
Affiliation(s)
- Eileen Haebig
- Department of Communication Sciences and Disorders, Louisiana State
University, USA
| | - Audra Sterling
- Department of Communication Sciences and Disorders, University of
Wisconsin-Madison, USA; Waisman Center, University of Wisconsin-Madison,
USA
| | - Andrea Barton-Hulsey
- Waisman Center, University of Wisconsin-Madison, USA; School of
Communication Sciences and Disorders, Florida State University, USA
| | - Laura Friedman
- Department of Communication Sciences and Disorders, University of
Wisconsin-Madison, USA; Waisman Center, University of Wisconsin-Madison,
USA
| |
Collapse
|
43
|
Danilina K, Gorbachevskaya N. Adaptive Status, Autistic Symptoms and Cognitive Profile in Patients with Monogenic Form of Autism Spectrum Disorders – Fragile X Syndrome. КЛИНИЧЕСКАЯ И СПЕЦИАЛЬНАЯ ПСИХОЛОГИЯ 2020. [DOI: 10.17759/cpse.2020090204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The article analyzes psychological data of a large group (55 males and 6 females) of subjects with monogenic form of hereditary cognitive impairment with autistic symptoms – Martin-Bell syndrome (FXS) at different age ranges (from 2 to 34 years old). As a result of the analysis, significant cognitive impairments were identified, which persisted throughout the studied age interval (IQ 50 ± 2.1 in males and 60 ± 5.6 in females). Autistic disorders were observed on average in 60% of subjects (less in females) and were most pronounced at 8-12 years. Use of Autism Diagnostic Observation Scale-2 (ADOS-2) allowed us to show that “Social Affect” scale makes the main contribution to overall score of autistic manifestations. Almost all subjects showed a significantly reduced level of adaptive skills. The lowest scores on “Communication”, “Socialization” and “Everyday life skills” scales were observed at the age of 8-12 years. With increase in age, subjects improved only on “Daily Life Skills” scale. It was also shown that a higher degree of adaptation and better nonverbal intelligence was observed in children with less severe autistic symptoms.
Collapse
Affiliation(s)
- K.K. Danilina
- Research and Clinical Center of Pediatric psychoneurology of Moscow Department of Public Health
| | | |
Collapse
|
44
|
Klusek J, Moser C, Schmidt J, Abbeduto L, Roberts JE. A novel eye-tracking paradigm for indexing social avoidance-related behavior in fragile X syndrome. Am J Med Genet B Neuropsychiatr Genet 2020; 183:5-16. [PMID: 31418535 PMCID: PMC6898737 DOI: 10.1002/ajmg.b.32757] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 07/22/2019] [Accepted: 07/26/2019] [Indexed: 11/07/2022]
Abstract
Fragile X syndrome (FXS) is characterized by hallmark features of gaze avoidance, reduced social approach, and social anxiety. The development of therapeutics to manage these symptoms has been hindered, in part, by the lack of sensitive outcome measures. This study investigated the utility of a novel eye-tracking paradigm for indexing social avoidance-related phenotypes. Adolescent/young adult-aged males with FXS (n = 24) and typical development (n = 23) participated in the study. Participants viewed faces displaying direct or averted gaze and the first fixation duration on the eyes was recorded as an index of initial stimulus registration. Fixation durations did not differ across the direction of gaze conditions in either group, although the control group showed longer initial fixations on the eyes relative to the FXS group. Shorter initial fixation on averted gaze in males with FXS was a robust predictor of the severity of their social avoidance behavior exhibited during a social greeting context, whereas parent-reported social avoidance symptoms were not related to performance in the semi-naturalistic context. This eye-tracking paradigm may represent a promising outcome measure for FXS clinical trials because it provides a quantitative index that closely maps onto core social avoidance phenotypes of FXS, can be completed in less than 20 min, and is suitable for use with individuals with low IQ.
Collapse
Affiliation(s)
- Jessica Klusek
- Department of Communication Sciences and Disorders, University of South Carolina, Columbia, South Carolina
| | - Carly Moser
- Department of Communication Sciences and Disorders, University of South Carolina, Columbia, South Carolina
| | - Joseph Schmidt
- Department of Psychology, University of Central Florida, Orlando, Florida
| | - Leonard Abbeduto
- Department of Psychiatry and Behavioral Sciences and MIND Institute, University of California, Davis, California
| | - Jane E Roberts
- Department of Psychology, University of South Carolina, Columbia, South Carolina
| |
Collapse
|
45
|
Baker EK, Arpone M, Vera SA, Bretherton L, Ure A, Kraan CM, Bui M, Ling L, Francis D, Hunter MF, Elliott J, Rogers C, Field MJ, Cohen J, Maria LS, Faundes V, Curotto B, Morales P, Trigo C, Salas I, Alliende AM, Amor DJ, Godler DE. Intellectual functioning and behavioural features associated with mosaicism in fragile X syndrome. J Neurodev Disord 2019; 11:41. [PMID: 31878865 PMCID: PMC6933737 DOI: 10.1186/s11689-019-9288-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023] Open
Abstract
Background Fragile X syndrome (FXS) is a common cause of intellectual disability and autism spectrum disorder (ASD) usually associated with a CGG expansion, termed full mutation (FM: CGG ≥ 200), increased DNA methylation of the FMR1 promoter and silencing of the gene. Mosaicism for presence of cells with either methylated FM or smaller unmethylated pre-mutation (PM: CGG 55–199) alleles in the same individual have been associated with better cognitive functioning. This study compares age- and sex-matched FM-only and PM/FM mosaic individuals on intellectual functioning, ASD features and maladaptive behaviours. Methods This study comprised a large international cohort of 126 male and female participants with FXS (aged 1.15 to 43.17 years) separated into FM-only and PM/FM mosaic groups (90 males, 77.8% FM-only; 36 females, 77.8% FM-only). Intellectual functioning was assessed with age appropriate developmental or intelligence tests. The Autism Diagnostic Observation Schedule-2nd Edition was used to examine ASD features while the Aberrant Behavior Checklist-Community assessed maladaptive behaviours. Results Comparing males and females (FM-only + PM/FM mosaic), males had poorer intellectual functioning on all domains (p < 0.0001). Although females had less ASD features and less parent-reported maladaptive behaviours, these differences were no longer significant after controlling for intellectual functioning. Participants with PM/FM mosaicism, regardless of sex, presented with better intellectual functioning and less maladaptive behaviours compared with their age- and sex-matched FM-only counterparts (p < 0.05). ASD features were similar between FM-only and PM/FM mosaics within each sex, after controlling for overall intellectual functioning. Conclusions Males with FXS had significantly lower intellectual functioning than females with FXS. However, there were no significant differences in ASD features and maladaptive behaviours, after controlling for intellectual functioning, independent of the presence or absence of mosaicism. This suggests that interventions that primarily target cognitive abilities may in turn reduce the severity of maladaptive behaviours including ASD features in FXS.
Collapse
Affiliation(s)
- Emma K Baker
- Diagnosis and Development, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia. .,Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia. .,School of Psychology and Public Health, La Trobe University, Bundoora, VIC, Australia.
| | - Marta Arpone
- Diagnosis and Development, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia.,Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia.,Brain and Mind, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Solange Aliaga Vera
- Diagnosis and Development, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Lesley Bretherton
- Brain and Mind, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Alexandra Ure
- Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia.,Neurodisability and Rehabilitation, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia.,Royal Children's Hospital, Melbourne, VIC, Australia.,Department of Pediatrics, Monash University, Clayton, VIC, Australia
| | - Claudine M Kraan
- Diagnosis and Development, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia.,Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Minh Bui
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Carlton, VIC, Australia
| | - Ling Ling
- Diagnosis and Development, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
| | - David Francis
- Victorian Clinical Genetics Services and Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Matthew F Hunter
- Department of Pediatrics, Monash University, Clayton, VIC, Australia.,Monash Genetics, Monash Health, Melbourne, VIC, Australia
| | - Justine Elliott
- Victorian Clinical Genetics Services and Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Carolyn Rogers
- Genetics of Learning Disability Service, Hunter Genetics, Waratah, NSW, Australia
| | - Michael J Field
- Genetics of Learning Disability Service, Hunter Genetics, Waratah, NSW, Australia
| | - Jonathan Cohen
- Fragile X Alliance Inc, Centre for Developmental Disability Health Victoria, Monash University, North Caulfield, Clayton, VIC, Australia
| | - Lorena Santa Maria
- Laboratory of Molecular Cytogenetics, Department of Genetics and Metabolic Diseases, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Victor Faundes
- Laboratory of Molecular Cytogenetics, Department of Genetics and Metabolic Diseases, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Bianca Curotto
- Laboratory of Molecular Cytogenetics, Department of Genetics and Metabolic Diseases, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Paulina Morales
- Laboratory of Molecular Cytogenetics, Department of Genetics and Metabolic Diseases, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Cesar Trigo
- Laboratory of Molecular Cytogenetics, Department of Genetics and Metabolic Diseases, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Isabel Salas
- Laboratory of Molecular Cytogenetics, Department of Genetics and Metabolic Diseases, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Angelica M Alliende
- Laboratory of Molecular Cytogenetics, Department of Genetics and Metabolic Diseases, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - David J Amor
- Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia.,Neurodisability and Rehabilitation, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia
| | - David E Godler
- Diagnosis and Development, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia.,Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
46
|
Russo FB, Brito A, de Freitas AM, Castanha A, de Freitas BC, Beltrão-Braga PCB. The use of iPSC technology for modeling Autism Spectrum Disorders. Neurobiol Dis 2019; 130:104483. [PMID: 31129084 DOI: 10.1016/j.nbd.2019.104483] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/31/2019] [Accepted: 05/22/2019] [Indexed: 12/28/2022] Open
Abstract
Autism Spectrum Disorders (ASDs) are a group of neurodevelopmental disorders that influence social skills, involving communication, interaction, and behavior, usually with repetitive and restrictive manners. Due to the variety of genes involved in ASDs and several possible environmental factors influence, there is still no answer to what really causes syndromic and non-syndromic types of ASDs, usually affecting each individual in a unique way. However, we know that the mechanism underlying ASDs involves brain functioning. The human brain is a complex structure composed of close to 100 billion cells, which is a big challenge to study counting just with post mortem tissue investigation or genetic approaches. Therefore, human induced pluripotent stem cells (iPSC) technology has been used as a tool to produce viable cells for understanding a working brain. Taking advantage of patient-derived stem cells, researchers are now able to generate neurons, glial cells and brain organoids in vitro to model ASDs. In this review we report data from different studies showing how iPSCs have been a critical tool to study the different phenotypes of ASDs.
Collapse
Affiliation(s)
- Fabiele Baldino Russo
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil
| | - Anita Brito
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil
| | | | - Andrelissa Castanha
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil
| | - Beatriz C de Freitas
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil
| | - Patricia Cristina Baleeiro Beltrão-Braga
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil; Department of Obstetrics, School of Arts Sciences and Humanities, São Paulo, SP 03828-000, Brazil.
| |
Collapse
|
47
|
Zhang F, Kang Y, Wang M, Li Y, Xu T, Yang W, Song H, Wu H, Shu Q, Jin P. Fragile X mental retardation protein modulates the stability of its m6A-marked messenger RNA targets. Hum Mol Genet 2019; 27:3936-3950. [PMID: 30107516 DOI: 10.1093/hmg/ddy292] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 08/07/2018] [Indexed: 12/12/2022] Open
Abstract
N6-methyladenosine (m6A) is the most prevalent internal modification of mammalian messenger RNAs (mRNAs) and long non-coding RNAs. The biological functions of this reversible RNA modification can be interpreted by cytoplasmic and nuclear 'm6A reader' proteins to fine-tune gene expression, such as mRNA degradation and translation initiation. Here we profiled transcriptome-wide m6A sites in adult mouse cerebral cortex, underscoring that m6A is a widespread epitranscriptomic modification in brain. Interestingly, the mRNA targets of fragile X mental retardation protein (FMRP), a selective RNA-binding protein, are enriched for m6A marks. Loss of functional FMRP leads to Fragile X syndrome (FXS), the most common inherited form of intellectual disability. Transcriptome-wide gene expression profiling identified 2035 genes differentially expressed in the absence of FMRP in cortex, and 92.5% of 174 downregulated FMRP targets are marked by m6A. Biochemical analyses indicate that FMRP binds to the m6A sites of its mRNA targets and interacts with m6A reader YTHDF2 in an RNA-independent manner. FMRP maintains the stability of its mRNA targets while YTHDF2 promotes the degradation of these mRNAs. These data together suggest that FMRP regulates the stability of its m6A-marked mRNA targets through YTHDF2, which could potentially contribute to the molecular pathogenesis of FXS.
Collapse
Affiliation(s)
- Feiran Zhang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Yunhee Kang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Mengli Wang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Yujing Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Tianlei Xu
- Department of Biostatistics and Bioinformatics, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Wei Yang
- Department of Neurology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Hongjun Song
- Department of Neuroscience, Mahoney Institute for Neurosciences, Institute for Regenerative Medicine and The Epigenetics Institute, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hao Wu
- Department of Biostatistics and Bioinformatics, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Qiang Shu
- The Children's Hospital and Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Peng Jin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
48
|
Stallworth JL, Dy ME, Buchanan CB, Chen CF, Scott AE, Glaze DG, Lane JB, Lieberman DN, Oberman LM, Skinner SA, Tierney AE, Cutter GR, Percy AK, Neul JL, Kaufmann WE. Hand stereotypies: Lessons from the Rett Syndrome Natural History Study. Neurology 2019; 92:e2594-e2603. [PMID: 31053667 DOI: 10.1212/wnl.0000000000007560] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 01/25/2019] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To characterize hand stereotypies (HS) in a large cohort of participants with Rett syndrome (RTT). METHODS Data from 1,123 girls and women enrolled in the RTT Natural History Study were gathered. Standard tests for continuous and categorical variables were used at baseline. For longitudinal data, we used repeated-measures linear and logistic regression models and nonparametric tests. RESULTS HS were reported in 922 participants with classic RTT (100%), 73 with atypical severe RTT (97.3%), 74 with atypical mild RTT (96.1%), and 17 females with MECP2 mutations without RTT (34.7%). Individuals with RTT who had classic presentation or severe MECP2 mutations had higher frequency and earlier onset of HS. Heterogeneity of HS types was confirmed, but variety decreased over time. At baseline, almost half of the participants with RTT had hand mouthing, which like clapping/tapping, decreased over time. These 2 HS types were more frequently reported than wringing/washing. Increased HS severity (prevalence and frequency) was associated with worsened measures of hand function. Number and type of HS were not related to hand function. Overall clinical severity was worse with decreased hand function but only weakly related to any HS characteristic. While hand function decreased over time, prevalence and frequency of HS remained relatively unchanged and high. CONCLUSIONS Nearly all individuals with RTT have severe and multiple types of HS, with mouthing and clapping/tapping decreasing over time. Interaction between HS frequency and hand function is complex. Understanding the natural history of HS in RTT could assist in clinical care and evaluation of new interventions.
Collapse
Affiliation(s)
- Jennifer L Stallworth
- From the Greenwood Genetic Center (J.L.S., C.B.B., C.-F.C., A.E.S., S.A.S., A.E.T., W.E.K.), Center for Translational Research, SC; Department of Neurology (M.E.D., D.N.L.), Boston Children's Hospital, MA; Department of Pediatrics and Neurology (D.G.G.), Baylor College of Medicine, Houston, TX; Civitan International Research Center (J.B.L.), School of Public Health (G.R.C.), University of Alabama at Birmingham; Department of Psychiatry and Human Behavior (L.M.O.), E.P. Bradley Hospital, Warren Alpert Medical School of Brown University, Providence, RI; Department of Pediatrics, Division of Neurology (A.K.P.), Civitan International Research Center, University of Alabama at Birmingham; Vanderbilt Kennedy Center (J.L.N.), Vanderbilt University Medical Center, Nashville, TN; Department of Pediatrics (W.E.K.), University of South Carolina School of Medicine, Columbia; and Department of Human Genetics (W.E.K.), Emory University School of Medicine, Atlanta, GA
| | - Marisela E Dy
- From the Greenwood Genetic Center (J.L.S., C.B.B., C.-F.C., A.E.S., S.A.S., A.E.T., W.E.K.), Center for Translational Research, SC; Department of Neurology (M.E.D., D.N.L.), Boston Children's Hospital, MA; Department of Pediatrics and Neurology (D.G.G.), Baylor College of Medicine, Houston, TX; Civitan International Research Center (J.B.L.), School of Public Health (G.R.C.), University of Alabama at Birmingham; Department of Psychiatry and Human Behavior (L.M.O.), E.P. Bradley Hospital, Warren Alpert Medical School of Brown University, Providence, RI; Department of Pediatrics, Division of Neurology (A.K.P.), Civitan International Research Center, University of Alabama at Birmingham; Vanderbilt Kennedy Center (J.L.N.), Vanderbilt University Medical Center, Nashville, TN; Department of Pediatrics (W.E.K.), University of South Carolina School of Medicine, Columbia; and Department of Human Genetics (W.E.K.), Emory University School of Medicine, Atlanta, GA
| | - Caroline B Buchanan
- From the Greenwood Genetic Center (J.L.S., C.B.B., C.-F.C., A.E.S., S.A.S., A.E.T., W.E.K.), Center for Translational Research, SC; Department of Neurology (M.E.D., D.N.L.), Boston Children's Hospital, MA; Department of Pediatrics and Neurology (D.G.G.), Baylor College of Medicine, Houston, TX; Civitan International Research Center (J.B.L.), School of Public Health (G.R.C.), University of Alabama at Birmingham; Department of Psychiatry and Human Behavior (L.M.O.), E.P. Bradley Hospital, Warren Alpert Medical School of Brown University, Providence, RI; Department of Pediatrics, Division of Neurology (A.K.P.), Civitan International Research Center, University of Alabama at Birmingham; Vanderbilt Kennedy Center (J.L.N.), Vanderbilt University Medical Center, Nashville, TN; Department of Pediatrics (W.E.K.), University of South Carolina School of Medicine, Columbia; and Department of Human Genetics (W.E.K.), Emory University School of Medicine, Atlanta, GA
| | - Chin-Fu Chen
- From the Greenwood Genetic Center (J.L.S., C.B.B., C.-F.C., A.E.S., S.A.S., A.E.T., W.E.K.), Center for Translational Research, SC; Department of Neurology (M.E.D., D.N.L.), Boston Children's Hospital, MA; Department of Pediatrics and Neurology (D.G.G.), Baylor College of Medicine, Houston, TX; Civitan International Research Center (J.B.L.), School of Public Health (G.R.C.), University of Alabama at Birmingham; Department of Psychiatry and Human Behavior (L.M.O.), E.P. Bradley Hospital, Warren Alpert Medical School of Brown University, Providence, RI; Department of Pediatrics, Division of Neurology (A.K.P.), Civitan International Research Center, University of Alabama at Birmingham; Vanderbilt Kennedy Center (J.L.N.), Vanderbilt University Medical Center, Nashville, TN; Department of Pediatrics (W.E.K.), University of South Carolina School of Medicine, Columbia; and Department of Human Genetics (W.E.K.), Emory University School of Medicine, Atlanta, GA
| | - Alexandra E Scott
- From the Greenwood Genetic Center (J.L.S., C.B.B., C.-F.C., A.E.S., S.A.S., A.E.T., W.E.K.), Center for Translational Research, SC; Department of Neurology (M.E.D., D.N.L.), Boston Children's Hospital, MA; Department of Pediatrics and Neurology (D.G.G.), Baylor College of Medicine, Houston, TX; Civitan International Research Center (J.B.L.), School of Public Health (G.R.C.), University of Alabama at Birmingham; Department of Psychiatry and Human Behavior (L.M.O.), E.P. Bradley Hospital, Warren Alpert Medical School of Brown University, Providence, RI; Department of Pediatrics, Division of Neurology (A.K.P.), Civitan International Research Center, University of Alabama at Birmingham; Vanderbilt Kennedy Center (J.L.N.), Vanderbilt University Medical Center, Nashville, TN; Department of Pediatrics (W.E.K.), University of South Carolina School of Medicine, Columbia; and Department of Human Genetics (W.E.K.), Emory University School of Medicine, Atlanta, GA
| | - Daniel G Glaze
- From the Greenwood Genetic Center (J.L.S., C.B.B., C.-F.C., A.E.S., S.A.S., A.E.T., W.E.K.), Center for Translational Research, SC; Department of Neurology (M.E.D., D.N.L.), Boston Children's Hospital, MA; Department of Pediatrics and Neurology (D.G.G.), Baylor College of Medicine, Houston, TX; Civitan International Research Center (J.B.L.), School of Public Health (G.R.C.), University of Alabama at Birmingham; Department of Psychiatry and Human Behavior (L.M.O.), E.P. Bradley Hospital, Warren Alpert Medical School of Brown University, Providence, RI; Department of Pediatrics, Division of Neurology (A.K.P.), Civitan International Research Center, University of Alabama at Birmingham; Vanderbilt Kennedy Center (J.L.N.), Vanderbilt University Medical Center, Nashville, TN; Department of Pediatrics (W.E.K.), University of South Carolina School of Medicine, Columbia; and Department of Human Genetics (W.E.K.), Emory University School of Medicine, Atlanta, GA
| | - Jane B Lane
- From the Greenwood Genetic Center (J.L.S., C.B.B., C.-F.C., A.E.S., S.A.S., A.E.T., W.E.K.), Center for Translational Research, SC; Department of Neurology (M.E.D., D.N.L.), Boston Children's Hospital, MA; Department of Pediatrics and Neurology (D.G.G.), Baylor College of Medicine, Houston, TX; Civitan International Research Center (J.B.L.), School of Public Health (G.R.C.), University of Alabama at Birmingham; Department of Psychiatry and Human Behavior (L.M.O.), E.P. Bradley Hospital, Warren Alpert Medical School of Brown University, Providence, RI; Department of Pediatrics, Division of Neurology (A.K.P.), Civitan International Research Center, University of Alabama at Birmingham; Vanderbilt Kennedy Center (J.L.N.), Vanderbilt University Medical Center, Nashville, TN; Department of Pediatrics (W.E.K.), University of South Carolina School of Medicine, Columbia; and Department of Human Genetics (W.E.K.), Emory University School of Medicine, Atlanta, GA
| | - David N Lieberman
- From the Greenwood Genetic Center (J.L.S., C.B.B., C.-F.C., A.E.S., S.A.S., A.E.T., W.E.K.), Center for Translational Research, SC; Department of Neurology (M.E.D., D.N.L.), Boston Children's Hospital, MA; Department of Pediatrics and Neurology (D.G.G.), Baylor College of Medicine, Houston, TX; Civitan International Research Center (J.B.L.), School of Public Health (G.R.C.), University of Alabama at Birmingham; Department of Psychiatry and Human Behavior (L.M.O.), E.P. Bradley Hospital, Warren Alpert Medical School of Brown University, Providence, RI; Department of Pediatrics, Division of Neurology (A.K.P.), Civitan International Research Center, University of Alabama at Birmingham; Vanderbilt Kennedy Center (J.L.N.), Vanderbilt University Medical Center, Nashville, TN; Department of Pediatrics (W.E.K.), University of South Carolina School of Medicine, Columbia; and Department of Human Genetics (W.E.K.), Emory University School of Medicine, Atlanta, GA
| | - Lindsay M Oberman
- From the Greenwood Genetic Center (J.L.S., C.B.B., C.-F.C., A.E.S., S.A.S., A.E.T., W.E.K.), Center for Translational Research, SC; Department of Neurology (M.E.D., D.N.L.), Boston Children's Hospital, MA; Department of Pediatrics and Neurology (D.G.G.), Baylor College of Medicine, Houston, TX; Civitan International Research Center (J.B.L.), School of Public Health (G.R.C.), University of Alabama at Birmingham; Department of Psychiatry and Human Behavior (L.M.O.), E.P. Bradley Hospital, Warren Alpert Medical School of Brown University, Providence, RI; Department of Pediatrics, Division of Neurology (A.K.P.), Civitan International Research Center, University of Alabama at Birmingham; Vanderbilt Kennedy Center (J.L.N.), Vanderbilt University Medical Center, Nashville, TN; Department of Pediatrics (W.E.K.), University of South Carolina School of Medicine, Columbia; and Department of Human Genetics (W.E.K.), Emory University School of Medicine, Atlanta, GA
| | - Steven A Skinner
- From the Greenwood Genetic Center (J.L.S., C.B.B., C.-F.C., A.E.S., S.A.S., A.E.T., W.E.K.), Center for Translational Research, SC; Department of Neurology (M.E.D., D.N.L.), Boston Children's Hospital, MA; Department of Pediatrics and Neurology (D.G.G.), Baylor College of Medicine, Houston, TX; Civitan International Research Center (J.B.L.), School of Public Health (G.R.C.), University of Alabama at Birmingham; Department of Psychiatry and Human Behavior (L.M.O.), E.P. Bradley Hospital, Warren Alpert Medical School of Brown University, Providence, RI; Department of Pediatrics, Division of Neurology (A.K.P.), Civitan International Research Center, University of Alabama at Birmingham; Vanderbilt Kennedy Center (J.L.N.), Vanderbilt University Medical Center, Nashville, TN; Department of Pediatrics (W.E.K.), University of South Carolina School of Medicine, Columbia; and Department of Human Genetics (W.E.K.), Emory University School of Medicine, Atlanta, GA
| | - Aubin E Tierney
- From the Greenwood Genetic Center (J.L.S., C.B.B., C.-F.C., A.E.S., S.A.S., A.E.T., W.E.K.), Center for Translational Research, SC; Department of Neurology (M.E.D., D.N.L.), Boston Children's Hospital, MA; Department of Pediatrics and Neurology (D.G.G.), Baylor College of Medicine, Houston, TX; Civitan International Research Center (J.B.L.), School of Public Health (G.R.C.), University of Alabama at Birmingham; Department of Psychiatry and Human Behavior (L.M.O.), E.P. Bradley Hospital, Warren Alpert Medical School of Brown University, Providence, RI; Department of Pediatrics, Division of Neurology (A.K.P.), Civitan International Research Center, University of Alabama at Birmingham; Vanderbilt Kennedy Center (J.L.N.), Vanderbilt University Medical Center, Nashville, TN; Department of Pediatrics (W.E.K.), University of South Carolina School of Medicine, Columbia; and Department of Human Genetics (W.E.K.), Emory University School of Medicine, Atlanta, GA
| | - Gary R Cutter
- From the Greenwood Genetic Center (J.L.S., C.B.B., C.-F.C., A.E.S., S.A.S., A.E.T., W.E.K.), Center for Translational Research, SC; Department of Neurology (M.E.D., D.N.L.), Boston Children's Hospital, MA; Department of Pediatrics and Neurology (D.G.G.), Baylor College of Medicine, Houston, TX; Civitan International Research Center (J.B.L.), School of Public Health (G.R.C.), University of Alabama at Birmingham; Department of Psychiatry and Human Behavior (L.M.O.), E.P. Bradley Hospital, Warren Alpert Medical School of Brown University, Providence, RI; Department of Pediatrics, Division of Neurology (A.K.P.), Civitan International Research Center, University of Alabama at Birmingham; Vanderbilt Kennedy Center (J.L.N.), Vanderbilt University Medical Center, Nashville, TN; Department of Pediatrics (W.E.K.), University of South Carolina School of Medicine, Columbia; and Department of Human Genetics (W.E.K.), Emory University School of Medicine, Atlanta, GA
| | - Alan K Percy
- From the Greenwood Genetic Center (J.L.S., C.B.B., C.-F.C., A.E.S., S.A.S., A.E.T., W.E.K.), Center for Translational Research, SC; Department of Neurology (M.E.D., D.N.L.), Boston Children's Hospital, MA; Department of Pediatrics and Neurology (D.G.G.), Baylor College of Medicine, Houston, TX; Civitan International Research Center (J.B.L.), School of Public Health (G.R.C.), University of Alabama at Birmingham; Department of Psychiatry and Human Behavior (L.M.O.), E.P. Bradley Hospital, Warren Alpert Medical School of Brown University, Providence, RI; Department of Pediatrics, Division of Neurology (A.K.P.), Civitan International Research Center, University of Alabama at Birmingham; Vanderbilt Kennedy Center (J.L.N.), Vanderbilt University Medical Center, Nashville, TN; Department of Pediatrics (W.E.K.), University of South Carolina School of Medicine, Columbia; and Department of Human Genetics (W.E.K.), Emory University School of Medicine, Atlanta, GA
| | - Jeffrey L Neul
- From the Greenwood Genetic Center (J.L.S., C.B.B., C.-F.C., A.E.S., S.A.S., A.E.T., W.E.K.), Center for Translational Research, SC; Department of Neurology (M.E.D., D.N.L.), Boston Children's Hospital, MA; Department of Pediatrics and Neurology (D.G.G.), Baylor College of Medicine, Houston, TX; Civitan International Research Center (J.B.L.), School of Public Health (G.R.C.), University of Alabama at Birmingham; Department of Psychiatry and Human Behavior (L.M.O.), E.P. Bradley Hospital, Warren Alpert Medical School of Brown University, Providence, RI; Department of Pediatrics, Division of Neurology (A.K.P.), Civitan International Research Center, University of Alabama at Birmingham; Vanderbilt Kennedy Center (J.L.N.), Vanderbilt University Medical Center, Nashville, TN; Department of Pediatrics (W.E.K.), University of South Carolina School of Medicine, Columbia; and Department of Human Genetics (W.E.K.), Emory University School of Medicine, Atlanta, GA
| | - Walter E Kaufmann
- From the Greenwood Genetic Center (J.L.S., C.B.B., C.-F.C., A.E.S., S.A.S., A.E.T., W.E.K.), Center for Translational Research, SC; Department of Neurology (M.E.D., D.N.L.), Boston Children's Hospital, MA; Department of Pediatrics and Neurology (D.G.G.), Baylor College of Medicine, Houston, TX; Civitan International Research Center (J.B.L.), School of Public Health (G.R.C.), University of Alabama at Birmingham; Department of Psychiatry and Human Behavior (L.M.O.), E.P. Bradley Hospital, Warren Alpert Medical School of Brown University, Providence, RI; Department of Pediatrics, Division of Neurology (A.K.P.), Civitan International Research Center, University of Alabama at Birmingham; Vanderbilt Kennedy Center (J.L.N.), Vanderbilt University Medical Center, Nashville, TN; Department of Pediatrics (W.E.K.), University of South Carolina School of Medicine, Columbia; and Department of Human Genetics (W.E.K.), Emory University School of Medicine, Atlanta, GA.
| |
Collapse
|
49
|
Baker EK, Arpone M, Aliaga SM, Bretherton L, Kraan CM, Bui M, Slater HR, Ling L, Francis D, Hunter MF, Elliott J, Rogers C, Field M, Cohen J, Cornish K, Santa Maria L, Faundes V, Curotto B, Morales P, Trigo C, Salas I, Alliende AM, Amor DJ, Godler DE. Incomplete silencing of full mutation alleles in males with fragile X syndrome is associated with autistic features. Mol Autism 2019; 10:21. [PMID: 31073396 PMCID: PMC6499941 DOI: 10.1186/s13229-019-0271-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/03/2019] [Indexed: 11/10/2022] Open
Abstract
Background Fragile X syndrome (FXS) is a common monogenic cause of intellectual disability with autism features. While it is caused by loss of the FMR1 product (FMRP), mosaicism for active and inactive FMR1 alleles, including alleles termed premutation (PM: 55-199 CGGs), is not uncommon. Importantly, both PM and active full mutation (FM: ≥ 200 CGGs) alleles often express elevated levels of mRNA that are thought to be toxic. This study determined if complete FMR1 mRNA silencing from FM alleles and/or levels of FMR1 mRNA (if present) in blood are associated with intellectual functioning and autism features in FXS. Methods The study cohort included 98 participants (70.4% male) with FXS (FM-only and PM/FM mosaic) aged 1-43 years. A control group of 14 females were used to establish control FMR1 mRNA reference range. Intellectual functioning and autism features were assessed using the Mullen Scales of Early Learning or an age-appropriate Wechsler Scale and the Autism Diagnostic Observation Schedule-2nd Edition (ADOS-2), respectively. FMR1 mRNA was analysed in venous blood collected at the time of assessments, using the real-time PCR relative standard curve method. Results Females with FXS had significantly higher levels of FMR1 mRNA (p < 0.001) than males. FMR1 mRNA levels were positively associated with age (p < 0.001), but not with intellectual functioning and autistic features in females. FM-only males (aged < 19 years) expressing FM FMR1 mRNA had significantly higher ADOS calibrated severity scores compared to FM-only males with completely silenced FMR1 (p = 0.011). However, there were no significant differences between these subgroups on intellectual functioning. In contrast, decreased levels of FMR1 mRNA were associated with decreased intellectual functioning in FXS males (p = 0.029), but not autism features, when combined with the PM/FM mosaic group. Conclusion Incomplete silencing of toxic FM RNA may be associated with autistic features, but not intellectual functioning in FXS males. While decreased levels of mRNA may be more predictive of intellectual functioning than autism features. If confirmed in future studies, these findings may have implications for patient stratification, outcome measure development, and design of clinical and pre-clinical trials in FXS.
Collapse
Affiliation(s)
- Emma K. Baker
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, 50 Flemington Rd, Parkville, VIC 3052 Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
| | - Marta Arpone
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, 50 Flemington Rd, Parkville, VIC 3052 Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
- Brain and Mind, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Australia
| | - Solange M. Aliaga
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, 50 Flemington Rd, Parkville, VIC 3052 Australia
| | - Lesley Bretherton
- Brain and Mind, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Australia
| | - Claudine M. Kraan
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, 50 Flemington Rd, Parkville, VIC 3052 Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
| | - Minh Bui
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Carlton, Australia
| | - Howard R. Slater
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, 50 Flemington Rd, Parkville, VIC 3052 Australia
| | - Ling Ling
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, 50 Flemington Rd, Parkville, VIC 3052 Australia
| | - David Francis
- Victorian Clinical Genetics Services and Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, VIC Australia
| | - Matthew F. Hunter
- Monash Genetics, Monash Health, Melbourne, VIC Australia
- Department of Paediatrics, Monash University, Clayton, VIC Australia
| | - Justine Elliott
- Victorian Clinical Genetics Services and Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, VIC Australia
| | - Carolyn Rogers
- Genetics of Learning Disability Service, Hunter Genetics, Waratah, NSW Australia
| | - Michael Field
- Genetics of Learning Disability Service, Hunter Genetics, Waratah, NSW Australia
| | - Jonathan Cohen
- Fragile X Alliance Inc, North Caulfield, VIC and Center for Developmental Disability Health Victoria, Monash University, Clayton, Australia
| | - Kim Cornish
- Monash Institute of Cognitive and Clinical Neurosciences, Monash University, Clayton, VIC Australia
| | - Lorena Santa Maria
- Molecular and Cytogenetics Laboratory, INTA, University of Chile, Santiago, Chile
| | - Victor Faundes
- Molecular and Cytogenetics Laboratory, INTA, University of Chile, Santiago, Chile
| | - Bianca Curotto
- Molecular and Cytogenetics Laboratory, INTA, University of Chile, Santiago, Chile
| | - Paulina Morales
- Molecular and Cytogenetics Laboratory, INTA, University of Chile, Santiago, Chile
| | - Cesar Trigo
- Molecular and Cytogenetics Laboratory, INTA, University of Chile, Santiago, Chile
| | - Isabel Salas
- Molecular and Cytogenetics Laboratory, INTA, University of Chile, Santiago, Chile
| | - Angelica M. Alliende
- Molecular and Cytogenetics Laboratory, INTA, University of Chile, Santiago, Chile
| | - David J. Amor
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
- Neurodisability and Rehabilitation, Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, Australia
| | - David E. Godler
- Diagnosis and Development, Murdoch Children’s Research Institute, Royal Children’s Hospital, 50 Flemington Rd, Parkville, VIC 3052 Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
| |
Collapse
|
50
|
Abbeduto L, Thurman AJ, McDuffie A, Klusek J, Feigles RT, Ted Brown W, Harvey DJ, Adayev T, LaFauci G, Dobkins C, Roberts JE. ASD Comorbidity in Fragile X Syndrome: Symptom Profile and Predictors of Symptom Severity in Adolescent and Young Adult Males. J Autism Dev Disord 2019; 49:960-977. [PMID: 30382442 DOI: 10.1007/s10803-018-3796-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Many males with FXS meet criteria for ASD. This study was designed to (1) describe ASD symptoms in adolescent and young adult males with FXS (n = 44) and (2) evaluate the contributions to ASD severity of cognitive, language, and psychiatric factors, as well as FMRP (the protein deficient in FXS). A few ASD symptoms on the ADOS-2 were universal in the sample. There was less impairment in restricted and repetitive behaviors (RRB) than in the social affective (SA) domain. The best predictor of overall ASD severity and SA severity was expressive syntactic ability. RRB severity was best predicted by the psychiatric factors. Implications for clinical practice and for understanding the ASD comorbidity in FXS are discussed.
Collapse
Affiliation(s)
- Leonard Abbeduto
- University of California, Davis, USA. .,UC Davis MIND Institute, 2825 50th St, Sacramento, CA, 95817, USA.
| | | | | | | | | | - W Ted Brown
- New York State Institute for Basic Research in Developmental Disabilities, New York, USA
| | | | - Tatyana Adayev
- New York State Institute for Basic Research in Developmental Disabilities, New York, USA
| | - Giuseppe LaFauci
- New York State Institute for Basic Research in Developmental Disabilities, New York, USA
| | - Carl Dobkins
- New York State Institute for Basic Research in Developmental Disabilities, New York, USA
| | | |
Collapse
|