1
|
Jouira G, Iulian Alexe D, Rekik G, Ioana Alexe C, Čaušević D, Setiawan E, Sahli S. Effects of visual and auditory cognitive tasks on postural balance in adolescents with intellectual disability: A comparative analysis of trained versus non-trained individuals. Neurosci Lett 2024; 842:137968. [PMID: 39236799 DOI: 10.1016/j.neulet.2024.137968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 08/13/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024]
Abstract
This study aimed to investigate the influence of visual and auditory cognitive tasks on postural balance in adolescents with intellectual disabilities (ID). Participants included two groups: a trained group (n = 11) with experience in athletic activities and a sedentary group (n = 14). The experiment used a comparative cross-sectional design, with data collection involving single-task (ST) and Visual dual-task (VDT), and auditory dual-task (ADT) conditions in both firm and foam surface conditions. Cognitive tests included the Working Memory Test (WMT) and the Selective Attention Test (SAT). Results revealed that the trained group demonstrated significantly superior balance performance (p < 0.05). During the SAT, VDT conditions had lower center of pressure (CoP) values than ADT conditions in the sedentary group (p < 0.01), this result was observed in the training group only in the WMT, suggesting greater postural instability during ADT. These findings highlight the complex relationship between cognitive function and motor control in adolescents with ID, highlighting the potential benefits of regular physical activity interventions to improve postural balance abilities in this population.
Collapse
Affiliation(s)
- Ghada Jouira
- Research Laboratory Education, Motricité, Sport et Santé (EM2S) LR19JS01, High Institute of Sport and Physical Education of Sfax, University of Sfax, Tunisia
| | - Dan Iulian Alexe
- Department of Physical and Occupational Therapy, "Vasile Alecsandri" University of Bacău, Bacău, Romania.
| | - Ghazi Rekik
- Research Laboratory Education, Motricité, Sport et Santé (EM2S) LR19JS01, High Institute of Sport and Physical Education of Sfax, University of Sfax, Tunisia; High Institute of Sport and Physical Education, University of Sfax, Sfax, Tunisia; Tanyu Research Laboratory, Taipei 112, Taiwan
| | - Cristina Ioana Alexe
- Department of Physical Education and Sports Performance, "Vasile Alecsandri" University of Bacău, Bacău, Romania
| | - Denis Čaušević
- Faculty of Sport and Physical Education, University of Sarajevo, Bosnia and Herzegovina
| | - Edi Setiawan
- Faculty of Teacher Training and Education, Suryakancana University, Cianjur Kidul, West Java, Indonesia
| | - Sonia Sahli
- Research Laboratory Education, Motricité, Sport et Santé (EM2S) LR19JS01, High Institute of Sport and Physical Education of Sfax, University of Sfax, Tunisia
| |
Collapse
|
2
|
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
|
3
|
Herrera ML, Paraíso-Luna J, Bustos-Martínez I, Barco Á. Targeting epigenetic dysregulation in autism spectrum disorders. Trends Mol Med 2024:S1471-4914(24)00162-X. [PMID: 38971705 DOI: 10.1016/j.molmed.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 07/08/2024]
Abstract
Autism spectrum disorders (ASD) comprise a range of neurodevelopmental pathologies characterized by deficits in social interaction and repetitive behaviors, collectively affecting almost 1% of the worldwide population. Deciphering the etiology of ASD has proven challenging due to the intricate interplay of genetic and environmental factors and the variety of molecular pathways affected. Epigenomic alterations have emerged as key players in ASD etiology. Their research has led to the identification of biomarkers for diagnosis and pinpointed specific gene targets for therapeutic interventions. This review examines the role of epigenetic alterations, resulting from both genetic and environmental influences, as a central causative factor in ASD, delving into its contribution to pathogenesis and treatment strategies.
Collapse
Affiliation(s)
- Macarena L Herrera
- Instituto de Neurociencias (Universidad Miguel Hernández - Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant, 03550 Alicante, Spain
| | - Juan Paraíso-Luna
- Instituto de Neurociencias (Universidad Miguel Hernández - Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant, 03550 Alicante, Spain
| | - Isabel Bustos-Martínez
- Instituto de Neurociencias (Universidad Miguel Hernández - Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant, 03550 Alicante, Spain
| | - Ángel Barco
- Instituto de Neurociencias (Universidad Miguel Hernández - Consejo Superior de Investigaciones Científicas), Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant, 03550 Alicante, Spain.
| |
Collapse
|
4
|
Zhang J, Xu Y, Liu Y, Yue L, Jin H, Chen Y, Wang D, Wang M, Chen G, Yang L, Zhang G, Zhang X, Li S, Zhao H, Zhao Y, Niu G, Gao Y, Cai Z, Yang F, Zhu C, Zhu D. Genetic Testing for Global Developmental Delay in Early Childhood. JAMA Netw Open 2024; 7:e2415084. [PMID: 38837156 PMCID: PMC11154162 DOI: 10.1001/jamanetworkopen.2024.15084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 04/03/2024] [Indexed: 06/06/2024] Open
Abstract
Importance Global developmental delay (GDD) is characterized by a complex etiology, diverse phenotypes, and high individual heterogeneity, presenting challenges for early clinical etiologic diagnosis. Cognitive impairment is the core symptom, and despite the pivotal role of genetic factors in GDD development, the understanding of them remains limited. Objectives To assess the utility of genetic detection in patients with GDD and to examine the potential molecular pathogenesis of GDD to identify targets for early intervention. Design, Setting, and Participants This multicenter, prospective cohort study enrolled patients aged 12 to 60 months with GDD from 6 centers in China from July 4, 2020, to August 31, 2023. Participants underwent trio whole exome sequencing (trio-WES) coupled with copy number variation sequencing (CNV-seq). Bioinformatics analysis was used to unravel pathogenesis and identify therapeutic targets. Main Outcomes and Measures The main outcomes of this study involved enhancing the rate of positive genetic diagnosis for GDD, broadening the scope of genetic testing indications, and investigating the underlying pathogenesis. The classification of children into levels of cognitive impairment was based on the developmental quotient assessed using the Gesell scale. Results The study encompassed 434 patients with GDD (262 [60%] male; mean [SD] age, 25.75 [13.24] months) with diverse degrees of cognitive impairment: mild (98 [23%]), moderate (141 [32%]), severe (122 [28%]), and profound (73 [17%]). The combined use of trio-WES and CNV-seq resulted in a 61% positive detection rate. Craniofacial abnormalities (odds ratio [OR], 2.27; 95% CI, 1.45-3.56), moderate or severe cognitive impairment (OR, 1.69; 95% CI, 1.05-2.70), and age between 12 and 24 months (OR, 1.57; 95% CI, 1.05-2.35) were associated with a higher risk of carrying genetic variants. Additionally, bioinformatics analysis suggested that genetic variants may induce alterations in brain development and function, which may give rise to cognitive impairment. Moreover, an association was found between the dopaminergic pathway and cognitive impairment. Conclusions and Relevance In this cohort study of patients with GDD, combining trio-WES with CNV-seq was a demonstrable, instrumental strategy for advancing the diagnosis of GDD. The close association among genetic variations, brain development, and clinical phenotypes contributed valuable insights into the pathogenesis of GDD. Notably, the dopaminergic pathway emerged as a promising focal point for potential targets in future precision medical interventions for GDD.
Collapse
Affiliation(s)
- Jiamei Zhang
- Department of Rehabilitation Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiran Xu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yun Liu
- Kunming Children’s Hospital, Kunming, China
| | - Ling Yue
- Department of Neurological Rehabilitation, Children’s Hospital of Hebei Province, Shijiazhuang, China
| | - Hongfang Jin
- Qinghai Provincial Women and Children’s Hospital, Xining, China
| | | | - Dong Wang
- Department of Pediatric Neurology, Xi’an Children’s Hospital, Xi’an, China
| | - Mingmei Wang
- Department of Rehabilitation Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Gongxun Chen
- Department of Rehabilitation Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lei Yang
- Department of Rehabilitation Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guangyu Zhang
- Department of Rehabilitation Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoli Zhang
- Department of Pediatric Neurology, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Sansong Li
- Department of Rehabilitation Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huiling Zhao
- Department of Rehabilitation Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yunxia Zhao
- Department of Rehabilitation Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guohui Niu
- Department of Rehabilitation Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yongqiang Gao
- Department of Rehabilitation Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhijun Cai
- Department of Rehabilitation Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | | | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Dengna Zhu
- Department of Rehabilitation Medicine, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
5
|
Bhandari K, Kanodia H, Donato F, Caroni P. Selective vulnerability of the ventral hippocampus-prelimbic cortex axis parvalbumin interneuron network underlies learning deficits of fragile X mice. Cell Rep 2024; 43:114124. [PMID: 38630591 DOI: 10.1016/j.celrep.2024.114124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 03/07/2024] [Accepted: 04/02/2024] [Indexed: 04/19/2024] Open
Abstract
High-penetrance mutations affecting mental health can involve genes ubiquitously expressed in the brain. Whether the specific patterns of dysfunctions result from ubiquitous circuit deficits or might reflect selective vulnerabilities of targetable subnetworks has remained unclear. Here, we determine how loss of ubiquitously expressed fragile X mental retardation protein (FMRP), the cause of fragile X syndrome, affects brain networks in Fmr1y/- mice. We find that in wild-type mice, area-specific knockout of FMRP in the adult mimics behavioral consequences of area-specific silencing. By contrast, the functional axis linking the ventral hippocampus (vH) to the prelimbic cortex (PreL) is selectively affected in constitutive Fmr1y/- mice. A chronic alteration in late-born parvalbumin interneuron networks across the vH-PreL axis rescued by VIP signaling specifically accounts for deficits in vH-PreL theta-band network coherence, ensemble assembly, and learning functions of Fmr1y/- mice. Therefore, vH-PreL axis function exhibits a selective vulnerability to loss of FMRP in the vH or PreL, leading to learning and memory dysfunctions in fragile X mice.
Collapse
Affiliation(s)
- Komal Bhandari
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Harsh Kanodia
- Biozentrum, University of Basel, 4058 Basel, Switzerland
| | - Flavio Donato
- Biozentrum, University of Basel, 4058 Basel, Switzerland
| | - Pico Caroni
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland.
| |
Collapse
|
6
|
Avrahami M, Ben-Dor DH, Ratzon R, Weizman A, Perlman Danieli P. Characterizing the clinical and sociodemographic profiles of hospitalized adolescents with autism spectrum disorder. Glob Ment Health (Camb) 2024; 11:e63. [PMID: 38827333 PMCID: PMC11140491 DOI: 10.1017/gmh.2024.63] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 04/17/2024] [Accepted: 05/02/2024] [Indexed: 06/04/2024] Open
Abstract
The prevalence of autism spectrum disorder (ASD) is increasing worldwide. Youngsters with ASD demonstrate higher rates of intellectual disabilities (IDs), comorbid psychopathology and psychiatric hospitalizations, compared to children in the general population. This study characterizes the demographics and clinical parameters of adolescent psychiatric inpatients with ASD compared to inpatients without ASD, all hospitalized during the study period. Additionally, within the ASD group, those with ID were compared to those without. The rate of males among participants with ASD was significantly higher than among those without ASD, and the duration of hospitalization was longer. In contrast, the rate of cigarette smoking, major depressive disorder and suicidal thoughts among those with ASD was lower. One-third of those with ASD had moderate to severe ID, about 10% had comorbid epilepsy, and about half of them demonstrated aggressive behavior. Most ASD patients showed significant improvement upon discharge, although the extent of improvement was more prominent among ASD patients with no ID. Our findings, consistent with previous research, indicate that hospitalization is beneficial to youths with ASD, both those with and those without ID. Further studies that include long-term follow-up are needed.
Collapse
Affiliation(s)
- Matan Avrahami
- Child and Adolescent Division, Geha Mental Health Center, Petah Tikva, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - David Haim Ben-Dor
- Child and Adolescent Division, Geha Mental Health Center, Petah Tikva, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Roy Ratzon
- Child and Adolescent Division, Geha Mental Health Center, Petah Tikva, Israel
| | - Abraham Weizman
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Laboratory of Molecular and Biological Psychiatry, Felsenstein Medical Research Center, Petah Tikva, Israel
- Research Unit, Geha Mental Health Center, Petah Tikva, Israel
| | - Polina Perlman Danieli
- Child and Adolescent Division, Geha Mental Health Center, Petah Tikva, Israel
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Centre for Addiction and Mental Health, Toronto, ON, Canada
| |
Collapse
|
7
|
Tian Y, Qiao H, Zhu LQ, Man HY. Sexually dimorphic phenotypes and the role of androgen receptors in UBE3A-dependent autism spectrum disorder. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.02.592248. [PMID: 38746146 PMCID: PMC11092617 DOI: 10.1101/2024.05.02.592248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Autism spectrum disorders (ASDs) are characterized by social, communication, and behavioral challenges. UBE3A is one of the most common ASD genes. ASDs display a remarkable sex difference with a 4:1 male to female prevalence ratio; however, the underlying mechanism remains largely unknown. Using the UBE3A-overexpressing mouse model for ASD, we studied sex differences at behavioral, genetic, and molecular levels. We found that male mice with extra copies of Ube3A exhibited greater impairments in social interaction, repetitive self-grooming behavior, memory, and pain sensitivity, whereas female mice with UBE3A overexpression displayed greater olfactory defects. Social communication was impaired in both sexes, with males making more calls and females preferring complex syllables. At the molecular level, androgen receptor (AR) levels were reduced in both sexes due to enhanced degradation mediated by UBE3A. However, AR reduction significantly dysregulated AR target genes only in male, not female, UBE3A-overexpressing mice. Importantly, restoring AR levels in the brain effectively normalized the expression of AR target genes, and rescued the deficits in social preference, grooming behavior, and memory in male UBE3A-overexpressing mice, without affecting females. These findings suggest that AR and its signaling cascade play an essential role in mediating the sexually dimorphic changes in UBE3A-dependent ASD.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Hui Qiao
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Heng-Ye Man
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
- Department of Pharmacology, Physiology & Biophysics, Boston University School of Medicine, 72 East Concord St., Boston, MA 02118, USA
- Center for Systems Neuroscience, Boston University, 610 Commonwealth Ave, Boston, MA 02215, USA
| |
Collapse
|
8
|
Parent HH, Niswender CM. Therapeutic Potential for Metabotropic Glutamate Receptor 7 Modulators in Cognitive Disorders. Mol Pharmacol 2024; 105:348-358. [PMID: 38423750 PMCID: PMC11026152 DOI: 10.1124/molpharm.124.000874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/05/2024] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
Metabotropic glutamate receptor 7 (mGlu7) is the most highly conserved and abundantly expressed mGlu receptor in the human brain. The presynaptic localization of mGlu7, coupled with its low affinity for its endogenous agonist, glutamate, are features that contribute to the receptor's role in modulating neuronal excitation and inhibition patterns, including long-term potentiation, in various brain regions. These characteristics suggest that mGlu7 modulation may serve as a novel therapeutic strategy in disorders of cognitive dysfunction, including neurodevelopmental disorders that cause impairments in learning, memory, and attention. Primary mutations in the GRM7 gene have recently been identified as novel causes of neurodevelopmental disorders, and these patients exhibit profound intellectual and cognitive disability. Pharmacological tools, such as agonists, antagonists, and allosteric modulators, have been the mainstay for targeting mGlu7 in its endogenous homodimeric form to probe effects of its function and modulation in disease models. However, recent research has identified diversity in dimerization, as well as trans-synaptic interacting proteins, that also play a role in mGlu7 signaling and pharmacological properties. These novel findings represent exciting opportunities in the field of mGlu receptor drug discovery and highlight the importance of further understanding the functions of mGlu7 in complex neurologic conditions at both the molecular and physiologic levels. SIGNIFICANCE STATEMENT: Proper expression and function of mGlu7 is essential for learning, attention, and memory formation at the molecular level within neural circuits. The pharmacological targeting of mGlu7 is undergoing a paradigm shift by incorporating an understanding of receptor interaction with other cis- and trans- acting synaptic proteins, as well as various intracellular signaling pathways. Based upon these new findings, mGlu7's potential as a drug target in the treatment of cognitive disorders and learning impairments is primed for exploration.
Collapse
Affiliation(s)
- Harrison H Parent
- Department of Pharmacology (H.H.P., C.M.N.), Warren Center for Neuroscience Drug Discovery (H.H.P., C.M.N.), Vanderbilt Brain Institute (C.M.N.), and Vanderbilt Institute for Chemical Biology (C.M.N.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N.)
| | - Colleen M Niswender
- Department of Pharmacology (H.H.P., C.M.N.), Warren Center for Neuroscience Drug Discovery (H.H.P., C.M.N.), Vanderbilt Brain Institute (C.M.N.), and Vanderbilt Institute for Chemical Biology (C.M.N.), Vanderbilt University, Nashville, Tennessee; and Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, Tennessee (C.M.N.)
| |
Collapse
|
9
|
Yi S, Tang X, Zhang Q, Liang Y, Huang J, Zhang S, Huang L, Yi S, Huang M, Qin Z, Luo J. A nonsense CC2D1A variant is associated with congenital anomalies, motor delay, hypotonia, and slight deformities. Heliyon 2024; 10:e27946. [PMID: 38496842 PMCID: PMC10944275 DOI: 10.1016/j.heliyon.2024.e27946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 03/04/2024] [Accepted: 03/08/2024] [Indexed: 03/19/2024] Open
Abstract
Background Autosomal recessive intellectual developmental disorder-3 is caused by homozygous or compound heterozygous mutations in the CC2D1A gene. The disorder is characterized by intellectual disability (ID) and autism spectrum disorder (ASD). To date, 39 patients from 17 families with CC2D1A -related disorders have been reported worldwide, in whom only six pathogenic or likely pathogenic loss-of-function variants and three variants of uncertain significance (VUS) in the CC2D1A gene have been identified in these patients. Methods We described a patient with ID from a non-consanguineous Chinese family and whole-exome sequencing (WES) was used to identify the causative gene. Results The patient presented with severe ID and ASD, speech impairment, motor delay, hypotonia, slight facial anomalies, and finger deformities. Threatened abortion and abnormal fetal movements occurred during pregnancy with the proband but not his older healthy sister. WES analysis identified a homozygous nonsense variant, c.736C > T (p.Gln246Ter), in the CC2D1A gene. In addition, six novel likely pathogenic CC2D1A variants were identified by a retrospective review of the in-house database. Conclusions This study expands the genetic and clinical spectra of CC2D1A-associated disorders, and may aid in increasing awareness of this rare condition. Our findings have provided new insights into the clinical heterogeneity of the disease and further phenotype-genotype correlation, which could help to offer scope for more accurate genetic testing and counseling to affected families.
Collapse
Affiliation(s)
- Sheng Yi
- Genetic and Metabolic Central Laboratory, Guangxi Birth Defects Research and Prevention Institute, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Clinical Research Center for Pediatric Diseases, Guangxi Key Laboratory of Reproductive Health and Birth Defects Prevention, Guangxi Key Laboratory of Precision Medicine for Genetic Diseases, Guangxi Key Laboratory of Birth Defects and Stem Cell Biobank, Guangxi Key Laboratory of Birth Defects Research and Prevention, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Xianglian Tang
- Genetic and Metabolic Central Laboratory, Guangxi Birth Defects Research and Prevention Institute, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Clinical Research Center for Pediatric Diseases, Guangxi Key Laboratory of Reproductive Health and Birth Defects Prevention, Guangxi Key Laboratory of Precision Medicine for Genetic Diseases, Guangxi Key Laboratory of Birth Defects and Stem Cell Biobank, Guangxi Key Laboratory of Birth Defects Research and Prevention, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Qiang Zhang
- Genetic and Metabolic Central Laboratory, Guangxi Birth Defects Research and Prevention Institute, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Clinical Research Center for Pediatric Diseases, Guangxi Key Laboratory of Reproductive Health and Birth Defects Prevention, Guangxi Key Laboratory of Precision Medicine for Genetic Diseases, Guangxi Key Laboratory of Birth Defects and Stem Cell Biobank, Guangxi Key Laboratory of Birth Defects Research and Prevention, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yu Liang
- Department of Pathology, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jing Huang
- Department of Obstetrics, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Shujie Zhang
- Genetic and Metabolic Central Laboratory, Guangxi Birth Defects Research and Prevention Institute, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Clinical Research Center for Pediatric Diseases, Guangxi Key Laboratory of Reproductive Health and Birth Defects Prevention, Guangxi Key Laboratory of Precision Medicine for Genetic Diseases, Guangxi Key Laboratory of Birth Defects and Stem Cell Biobank, Guangxi Key Laboratory of Birth Defects Research and Prevention, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Limei Huang
- Genetic and Metabolic Central Laboratory, Guangxi Birth Defects Research and Prevention Institute, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Clinical Research Center for Pediatric Diseases, Guangxi Key Laboratory of Reproductive Health and Birth Defects Prevention, Guangxi Key Laboratory of Precision Medicine for Genetic Diseases, Guangxi Key Laboratory of Birth Defects and Stem Cell Biobank, Guangxi Key Laboratory of Birth Defects Research and Prevention, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Shang Yi
- Genetic and Metabolic Central Laboratory, Guangxi Birth Defects Research and Prevention Institute, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Clinical Research Center for Pediatric Diseases, Guangxi Key Laboratory of Reproductive Health and Birth Defects Prevention, Guangxi Key Laboratory of Precision Medicine for Genetic Diseases, Guangxi Key Laboratory of Birth Defects and Stem Cell Biobank, Guangxi Key Laboratory of Birth Defects Research and Prevention, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Minpan Huang
- Genetic and Metabolic Central Laboratory, Guangxi Birth Defects Research and Prevention Institute, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Clinical Research Center for Pediatric Diseases, Guangxi Key Laboratory of Reproductive Health and Birth Defects Prevention, Guangxi Key Laboratory of Precision Medicine for Genetic Diseases, Guangxi Key Laboratory of Birth Defects and Stem Cell Biobank, Guangxi Key Laboratory of Birth Defects Research and Prevention, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Zailong Qin
- Genetic and Metabolic Central Laboratory, Guangxi Birth Defects Research and Prevention Institute, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Clinical Research Center for Pediatric Diseases, Guangxi Key Laboratory of Reproductive Health and Birth Defects Prevention, Guangxi Key Laboratory of Precision Medicine for Genetic Diseases, Guangxi Key Laboratory of Birth Defects and Stem Cell Biobank, Guangxi Key Laboratory of Birth Defects Research and Prevention, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jingsi Luo
- Genetic and Metabolic Central Laboratory, Guangxi Birth Defects Research and Prevention Institute, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Clinical Research Center for Pediatric Diseases, Guangxi Key Laboratory of Reproductive Health and Birth Defects Prevention, Guangxi Key Laboratory of Precision Medicine for Genetic Diseases, Guangxi Key Laboratory of Birth Defects and Stem Cell Biobank, Guangxi Key Laboratory of Birth Defects Research and Prevention, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
10
|
Le TH, Oh JM, Rami FZ, Li L, Chun SK, Chung YC. Effects of Social Defeat Stress on Microtubule Regulating Proteins and Tubulin Polymerization. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2024; 22:129-138. [PMID: 38247419 PMCID: PMC10811395 DOI: 10.9758/cpn.23.1077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/29/2023] [Accepted: 06/21/2023] [Indexed: 01/23/2024]
Abstract
Objective : Microtubule (MT) stability in neurons is vital for brain development; instability is associated with neuropsychiatric disorders. The present study examined the effects of social defeat stress (SDS) on MT-regulating proteins and tubulin polymerization. Methods : After 10 days of SDS, defeated mice were separated into susceptible (Sus) and unsusceptible (Uns) groups based on their performance in a social avoidance test. Using extracted brain tissues, we measured the expression levels of α-tubulin, acetylated α-tubulin, tyrosinated α-tubulin, MT-associated protein-2 (MAP2), stathmin (STMN1), phospho stathmin serine 16 (p-STMN1 [Ser16]), phospho stathmin serine 25 (p-STMN1 [Ser25]), phospho stathmin serine 38 (p-STMN1 [Ser38]), stathmin2 (STMN2), phospho stathmin 2 serine 73 (p-STMN2 [Ser73]), 78-kDa glucose-regulated protein (GRP-78), and CCAAT/enhancer binding protein (C/EBP)-homologous protein (CHOP) using Western blot assay. The tubulin polymerization rate was also measured. Results : We observed increased and decreased expression of acetylated and tyrosinated α-tubulin, respectively, decreased expression of p-STMN1 (Ser16) and increased expression of p-STMN1 (Ser25), p-STMN2 (Ser73) and GRP-78 and CHOP in the prefrontal cortex and/or hippocampus of defeated mice. A reduced tubulin polymerization rate was observed in the Sus group compared to the Uns and Con groups. Conclusion : Our findings suggest that SDS has detrimental effects on MT stability, and a lower tubulin polymerization rate could be a molecular marker for susceptibility to SDS.
Collapse
Affiliation(s)
- Thi-Hung Le
- Department of Psychiatry, Jeonbuk National University Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Jung-Mi Oh
- Department of Physiology, Jeonbuk National University Medical School, Jeonju, Korea
| | - Fatima Zahra Rami
- Department of Psychiatry, Jeonbuk National University Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Ling Li
- Department of Psychiatry, Jeonbuk National University Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Sung-Kun Chun
- Department of Physiology, Jeonbuk National University Medical School, Jeonju, Korea
| | - Young-Chul Chung
- Department of Psychiatry, Jeonbuk National University Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| |
Collapse
|
11
|
Vacharasin JM, Ward JA, McCord MM, Cox K, Imitola J, Lizarraga SB. Neuroimmune mechanisms in autism etiology - untangling a complex problem using human cellular models. OXFORD OPEN NEUROSCIENCE 2024; 3:kvae003. [PMID: 38665176 PMCID: PMC11044813 DOI: 10.1093/oons/kvae003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 01/13/2024] [Accepted: 01/31/2024] [Indexed: 04/28/2024]
Abstract
Autism spectrum disorder (ASD) affects 1 in 36 people and is more often diagnosed in males than in females. Core features of ASD are impaired social interactions, repetitive behaviors and deficits in verbal communication. ASD is a highly heterogeneous and heritable disorder, yet its underlying genetic causes account only for up to 80% of the cases. Hence, a subset of ASD cases could be influenced by environmental risk factors. Maternal immune activation (MIA) is a response to inflammation during pregnancy, which can lead to increased inflammatory signals to the fetus. Inflammatory signals can cross the placenta and blood brain barriers affecting fetal brain development. Epidemiological and animal studies suggest that MIA could contribute to ASD etiology. However, human mechanistic studies have been hindered by a lack of experimental systems that could replicate the impact of MIA during fetal development. Therefore, mechanisms altered by inflammation during human pre-natal brain development, and that could underlie ASD pathogenesis have been largely understudied. The advent of human cellular models with induced pluripotent stem cell (iPSC) and organoid technology is closing this gap in knowledge by providing both access to molecular manipulations and culturing capability of tissue that would be otherwise inaccessible. We present an overview of multiple levels of evidence from clinical, epidemiological, and cellular studies that provide a potential link between higher ASD risk and inflammation. More importantly, we discuss how stem cell-derived models may constitute an ideal experimental system to mechanistically interrogate the effect of inflammation during the early stages of brain development.
Collapse
Affiliation(s)
- Janay M Vacharasin
- Department of Biological Sciences, and Center for Childhood Neurotherapeutics, Univ. of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA
- Department of Biological Sciences, Francis Marion University, 4822 East Palmetto Street, Florence, S.C. 29506, USA
| | - Joseph A Ward
- Department of Molecular Biology, Cell Biology, & Biochemistry, Brown University, 185 Meeting Street, Providence, RI 02912, USA
- Center for Translational Neuroscience, Carney Institute of Brain Science, Brown University, 70 Ship Street, Providence, RI 02903, USA
| | - Mikayla M McCord
- Department of Biological Sciences, and Center for Childhood Neurotherapeutics, Univ. of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA
| | - Kaitlin Cox
- Department of Biological Sciences, and Center for Childhood Neurotherapeutics, Univ. of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA
| | - Jaime Imitola
- Laboratory of Neural Stem Cells and Functional Neurogenetics, UConn Health, Departments of Neuroscience, Neurology, Genetics and Genome Sciences, UConn Health, 263 Farmington Avenue, Farmington, CT 06030-5357, USA
| | - Sofia B Lizarraga
- Department of Molecular Biology, Cell Biology, & Biochemistry, Brown University, 185 Meeting Street, Providence, RI 02912, USA
- Center for Translational Neuroscience, Carney Institute of Brain Science, Brown University, 70 Ship Street, Providence, RI 02903, USA
| |
Collapse
|
12
|
Zhang J, Liu H, Wang M, Xu Y, Zhu D, Yang F. Autosomal recessive intellectual disability caused by compound heterozygous variants of the EEF1D gene in a Chinese family. Mol Genet Genomic Med 2024; 12:e2333. [PMID: 38083972 PMCID: PMC10767685 DOI: 10.1002/mgg3.2333] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 09/23/2023] [Accepted: 11/28/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND Intellectual disability is a prevalent neurodevelopmental disorder, with the majority of affected children exhibiting global developmental delay before the age of 5 years. In recent years, certain children have been found to carry homozygous variations of the EEF1D gene, leading to autosomal recessive intellectual disability. However, the pathogenicity of compound heterozygous variations in this gene remains largely unknown. METHODS Trio whole-exome sequencing and copy number variation sequencing were done for the genetic etiological diagnosis of a 3-year and 11-month-old Chinese boy who presented with brachycephaly, severe to profound global developmental delay, and hypotonia in the lower limbs. RESULTS In this case, compound heterozygous variants of the EEF1D gene were found in the child through trio whole-exome sequencing; one was a splice variant (NM_032378.6:c.1905+1G>A) inherited from his father, and the other was a nonsense variant (NM_032378.6:c.676C>T) inherited from his mother. The nonsense variant leads to the production of a premature termination (p.Gln226*). These variations have the ability to explain the clinical phenotypes of the child. CONCLUSIONS Our study expands the variation spectrum and provides compelling evidence for EEF1D as a candidate gene for autosomal recessive intellectual disability. However, due to the deficient number of reported cases, researchers need to further study EEF1D and supplement the clinical phenotypes and treatment measures.
Collapse
Affiliation(s)
- Jiamei Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research CenterThird Affiliated Hospital and Institute of Neuroscience of Zhengzhou UniversityZhengzhouChina
| | - Hongxing Liu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research CenterThird Affiliated Hospital and Institute of Neuroscience of Zhengzhou UniversityZhengzhouChina
| | - Mingmei Wang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research CenterThird Affiliated Hospital and Institute of Neuroscience of Zhengzhou UniversityZhengzhouChina
| | - Yiran Xu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research CenterThird Affiliated Hospital and Institute of Neuroscience of Zhengzhou UniversityZhengzhouChina
- Commission Key Laboratory of Birth Defects PreventionHenan Key Laboratory of Population Defects PreventionZhengzhouChina
| | - Dengna Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research CenterThird Affiliated Hospital and Institute of Neuroscience of Zhengzhou UniversityZhengzhouChina
| | | |
Collapse
|
13
|
Li H, Zhang Q, Wan R, Zhou L, Xu X, Xu C, Yu Y, Xu Y, Xiang Y, Tang S. PLPPR4 haploinsufficiency causes neurodevelopmental disorders by disrupting synaptic plasticity via mTOR signalling. J Cell Mol Med 2023; 27:3286-3295. [PMID: 37550884 PMCID: PMC10623522 DOI: 10.1111/jcmm.17899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/20/2023] [Accepted: 07/30/2023] [Indexed: 08/09/2023] Open
Abstract
Phospholipid phosphatase related 4 (PLPPR4), a neuron-specific membrane protein located at the postsynaptic density of glutamatergic synapses, is a putative regulator of neuronal plasticity. However, PLPPR4 dysfunction has not been linked to genetic disorders. In this study, we report three unrelated patients with intellectual disability (ID) or autism spectrum disorder (ASD) who harbour a de novo heterozygous copy number loss of PLPPR4 in 1p21.2p21.3, a heterozygous nonsense mutation in PLPPR4 (NM_014839, c.4C > T, p.Gln2*) and a homozygous splice mutation in PLPPR4 (NM_014839: c.408 + 2 T > C), respectively. Bionano single-molecule optical mapping confirmed PLPPR4 deletion contains no additional pathogenic genes. Our results suggested that the loss of function of PLPPR4 is associated with neurodevelopmental disorders. To test the pathogenesis of PLPPR4, peripheral blood mononuclear cells obtained from the patient with heterozygous deletion of PLPPR4 were induced to specific iPSCs (CHWi001-A) and then differentiated into neurons. The neurons carrying the deletion of PLPPR4 displayed the reduced density of dendritic protrusions, shorter neurites and reduced axon length, suggesting the causal role of PLPPR4 in neurodevelopmental disorders. As the mTOR signalling pathway was essential for regulating the axon maturation and function, we found that mTOR signalling was inhibited with a higher level of p-AKT, p-mTOR and p-ERK1/2, decreased p-PI3K in PLPPR4-iPSCs neurons. Additionally, we found silencing PLPPR4 disturbed the mTOR signalling pathway. Our results suggested PLPPR4 modulates neurodevelopment by affecting the plasticity of neurons via the mTOR signalling pathway.
Collapse
Affiliation(s)
- Huanzheng Li
- Human Aging Research InstituteNanchang UniversityNanchangChina
| | | | - Ru Wan
- Wenzhou Central HospitalWenzhouChina
| | - Lili Zhou
- Wenzhou Central HospitalWenzhouChina
| | - Xueqin Xu
- Wenzhou Central HospitalWenzhouChina
| | | | - Yuan Yu
- Wenzhou Central HospitalWenzhouChina
| | - Yunzhi Xu
- Wenzhou Central HospitalWenzhouChina
| | | | - Shaohua Tang
- Wenzhou Central HospitalWenzhouChina
- Pediatric GeneticsZhejiang Provincial People's HospitalHangzhouChina
| |
Collapse
|
14
|
Kharrat M, Issa AB, Tlili A, Jallouli O, Alila-Fersi O, Maalej M, Chouchen J, Ghouylia Y, Kamoun F, Triki C, Fakhfakh F. A Novel Mutation in the MAP7D3 Gene in Two Siblings with Severe Intellectual Disability and Autistic Traits: Concurrent Assessment of BDNF Functional Polymorphism, X-Inactivation and Oxidative Stress to Explain Disease Severity. J Mol Neurosci 2023; 73:853-864. [PMID: 37817054 DOI: 10.1007/s12031-023-02163-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/27/2023] [Indexed: 10/12/2023]
Abstract
Intellectual disabilities (ID) and autism spectrum disorders (ASD) are characterized by extreme genetic and phenotypic heterogeneity. However, understanding this heterogeneity is difficult due to the intricate interplay among multiple interconnected genes, epigenetic factors, oxidative stress, and environmental factors. Employing next-generation sequencing (NGS), we revealed the genetic cause of ID and autistic traits in two patients from a consanguineous family followed by segregation analysis. Furthermore, in silico prediction methods and 3D modeling were conducted to predict the effect of the variants. To establish genotype-phenotype correlation, X-chromosome inactivation using Methylation-specific PCR and oxidative stress markers were also investigated. By analyzing the NGS data of the two patients, we identified a novel frameshift mutation c.2174_2177del (p.Thr725MetfsTer2) in the MAP7D3 gene inherited from their mother along with the functional BDNF Val66Met polymorphism inherited from their father. The 3D modeling demonstrated that the p.Thr725MetfsTer2 variant led to the loss of the C-terminal tail of the MAP7D3 protein. This change could destabilize its structure and impact kinesin-1's binding to microtubules via an allosteric effect. Moreover, the analysis of oxidative stress biomarkers revealed an elevated oxidative stress in the two patients compared to the controls. To the best of our knowledge, this is the first report describing severe ID and autistic traits in familial cases with novel frameshift mutation c.2174_2177del in the MAP7D3 gene co-occurring with the functional polymorphism Val66M in the BDNF gene. Besides, our study underlines the importance of investigating combined genetic variations, X-chromosome inactivation (XCI) patterns, and oxidative stress markers for a better understanding of ID and autism etiology.
Collapse
Affiliation(s)
- Marwa Kharrat
- Laboratory of Molecular and Functional Genetics, Faculty of Science of Sfax University, Sfax, Tunisia.
| | - Abir Ben Issa
- Child Neurology Department, Hedi Chaker Hospital, Sfax, Tunisia
- Research Laboratory (LR19ES15), Sfax Medical School, Sfax University, Sfax, Tunisia
| | - Abdelaziz Tlili
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Olfa Jallouli
- Child Neurology Department, Hedi Chaker Hospital, Sfax, Tunisia
- Research Laboratory (LR19ES15), Sfax Medical School, Sfax University, Sfax, Tunisia
| | - Olfa Alila-Fersi
- Laboratory of Molecular and Functional Genetics, Faculty of Science of Sfax University, Sfax, Tunisia
| | - Marwa Maalej
- Laboratory of Molecular and Functional Genetics, Faculty of Science of Sfax University, Sfax, Tunisia
| | - Jihen Chouchen
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Yosra Ghouylia
- Child Neurology Department, Hedi Chaker Hospital, Sfax, Tunisia
- Research Laboratory (LR19ES15), Sfax Medical School, Sfax University, Sfax, Tunisia
| | - Fatma Kamoun
- Child Neurology Department, Hedi Chaker Hospital, Sfax, Tunisia
- Research Laboratory (LR19ES15), Sfax Medical School, Sfax University, Sfax, Tunisia
| | - Chahnez Triki
- Child Neurology Department, Hedi Chaker Hospital, Sfax, Tunisia
- Research Laboratory (LR19ES15), Sfax Medical School, Sfax University, Sfax, Tunisia
| | - Faiza Fakhfakh
- Laboratory of Molecular and Functional Genetics, Faculty of Science of Sfax University, Sfax, Tunisia.
| |
Collapse
|
15
|
Gawade K, Raczynska KD. Imprinted small nucleolar RNAs: Missing link in development and disease? WILEY INTERDISCIPLINARY REVIEWS. RNA 2023:e1818. [PMID: 37722601 DOI: 10.1002/wrna.1818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/31/2023] [Accepted: 08/31/2023] [Indexed: 09/20/2023]
Abstract
The 14q32.2 (DLK1-DIO3) and 15q11-q13 (SNURF-SNRPN) imprinted gene loci harbor the largest known small nucleolar RNA clusters expressed from the respective maternal and paternal alleles. Recent studies have demonstrated significant roles for the 15q11-q13 located SNORD115-SNORD116 C/D box snoRNAs in Prader-Willi syndrome (PWS), a neurodevelopmental disorder. Even though the effect of SNORD116 deletion is apparent in the PWS phenotype, similar effects of a SNORD113-SNORD114 cluster deletion from the 14q32.2 locus in Kagami-Ogata syndrome (KOS14) and upregulation in Temple syndrome (TS14) remain to be explored. Moreover, apart from their probable involvement in neurodevelopmental disorders, snoRNAs from the SNORD113-SNORD114 cluster have been implicated in multiple biological processes, including pluripotency, development, cancers, and RNA modifications. Here we summarize the current understanding of the system to explore the possibility of a link between developmental disorders and C/D box snoRNA expression from the imprinted 14q32.2 locus. This article is categorized under: RNA in Disease and Development > RNA in Disease RNA in Disease and Development > RNA in Development RNA Processing > Processing of Small RNAs.
Collapse
Affiliation(s)
- Kishor Gawade
- Laboratory of RNA Processing, Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University in Poznan, Poznan, Poland
- Center for Advanced Technology, Adam Mickiewicz University in Poznan, Poznan, Poland
| | - Katarzyna D Raczynska
- Laboratory of RNA Processing, Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University in Poznan, Poznan, Poland
- Center for Advanced Technology, Adam Mickiewicz University in Poznan, Poznan, Poland
| |
Collapse
|
16
|
Liu S, Larsson H, Kuja-Halkola R, Lichtenstein P, Butwicka A, Taylor MJ. Age-related physical health of older autistic adults in Sweden: a longitudinal, retrospective, population-based cohort study. THE LANCET. HEALTHY LONGEVITY 2023; 4:e307-e315. [PMID: 37295448 DOI: 10.1016/s2666-7568(23)00067-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/20/2023] [Accepted: 04/20/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND Research of health outcomes in older autistic adults (≥45 years) is concerningly scarce, and little is known about whether intellectual disability and sex affect the health outcomes of this population. The aim of this study was to investigate the association between autism and physical health conditions in older adults and to examine these associations by intellectual disability and sex. METHODS We conducted a longitudinal, retrospective, population-based cohort study of the Swedish population born between Jan 1, 1932, and Dec 31, 1967, using linked data from the nationwide Total Population Register and the National Patient Register. We excluded individuals who died or emigrated before the age of 45 years, or with any chromosomal abnormalities. Follow-up started at age 45 years for all individuals, and ended at emigration, death, or Dec 31, 2013 (the latest date of available follow-up), whichever was soonest. Diagnoses of autism, intellectual disability, 39 age-related physical conditions, and five types of injury (outcomes) were obtained from the National Patient Register. For each outcome, we calculated 25-year cumulative incidence and used Cox models to estimate hazard ratios (HRs). All analyses were repeated separately by intellectual disability and sex. FINDINGS Of 4 200 887 older adults (2 063 718 women [49·1%] and 2 137 169 men [50·9%]) in the study cohort, 5291 (0·1%) had a diagnosis of autism recorded in the National Patient Register. Older autistic adults (median follow-up 8·4 years [IQR 4·2-14·6]) had higher cumulative incidence and HRs of various physical conditions and injuries than their non-autistic counterparts (median follow-up 16·4 years [8·2-24·4]). In autistic individuals, the highest cumulative incidence was observed for bodily injuries (50·0% [95% CI 47·6-52·4]). Conditions that autistic adults were at higher risk of than were non-autistic adults included heart failure (HR 1·89 [95% CI 1·61-2·22]), cystitis (2·03 [1·66-2·49]), glucose dysregulation (2·96 [2·04-4·29]), iron deficiency anaemia (3·12 [2·65-3·68]), poisoning (4·63 [4·13-5·18]), and self-harm (7·08 [6·24-8·03]). These increased risks mainly persisted regardless of intellectual disability or sex. INTERPRETATION Our data indicate that older autistic adults are at substantially increased risk of age-related physical conditions and injuries compared with non-autistic adults. These findings highlight the need for collaborative efforts from researchers, health services, and policy makers to provide older autistic individuals with the necessary support to attain healthy longevity and a high quality of life. FUNDING Swedish Research Council, Servier Affaires Medicales. TRANSLATION For the Swedish translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Shengxin Liu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Solna, Sweden.
| | - Henrik Larsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Solna, Sweden; School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Ralf Kuja-Halkola
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Solna, Sweden
| | - Paul Lichtenstein
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Solna, Sweden
| | - Agnieszka Butwicka
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Solna, Sweden; Child and Adolescent Psychiatry Stockholm, Stockholm Health Care Services, Stockholm, Sweden; Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland; Department of Child Psychiatry, Medical University of Warsaw, Warsaw, Poland
| | - Mark J Taylor
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
17
|
Iglesias-Ortega L, Megías-Fernández C, Domínguez-Giménez P, Jimeno-González S, Rivero S. Cell consequences of loss of function of the epigenetic factor EHMT1. Cell Signal 2023:110734. [PMID: 37257768 DOI: 10.1016/j.cellsig.2023.110734] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 05/20/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023]
Abstract
EHMT1 is an epigenetic factor with histone methyltransferase activity that appears mutated in Kleefstra syndrome, a neurodevelopmental genetic disorder characterized by developmental delay, intellectual disability, and autistic-like features. Despite recent progress in the study of the function of this gene and the molecular etiology of the disease, our knowledge of how EHMT1 haploinsufficiency causes Kleefstra syndrome is still very limited. Here, we show that EHMT1 depletion in RPE1 cells leads to alterations in the morphology and distribution of different subcellular structures, such as the Golgi apparatus, the lysosomes and different cell adhesion components. EHMT1 downregulation also increases centriolar satellites detection, which may indicate a role for EHMT1 in centrosome functioning. Furthermore, the migration process is also altered in EHMT1 depleted cells, which show reduced migration capacity. We consider that the described phenotypes could open new possibilities for understanding the functional impact of EHMT1 haploinsufficiency in Kleefstra syndrome, helping to elucidate the link between epigenetic regulation and the underlying cellular mechanisms that result in this neurodevelopmental disorder. This knowledge could be relevant not only for the treatment of this syndrome, but also for other neurodevelopmental conditions that could share similar deregulated cellular pathways.
Collapse
Affiliation(s)
- Lucía Iglesias-Ortega
- Departamento de Citología e Histología Normal y Patológica, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Clara Megías-Fernández
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain; Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Sevilla, Spain
| | - Paloma Domínguez-Giménez
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Sevilla, Spain
| | - Silvia Jimeno-González
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain; Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Sevilla, Spain
| | - Sabrina Rivero
- Departamento de Citología e Histología Normal y Patológica, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain; Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, Sevilla, Spain.
| |
Collapse
|
18
|
Abstract
Autism is often considered to reflect categorically 'different brains'. Neuropsychological research on autism spectrum disorder (ASD) however, has struggled to define this difference, or derive clear-cut boundaries between autism and non-autism. Consequently, restructuring or disbanding the ASD diagnosis is becoming increasingly advocated within research. Nonetheless, autism now exists as a salient social construction, of which 'difference' is a key facet. Clinical and educational professionals must influence this cautiously, as changes to autism's social construction may counterproductively affect the quality of life of autistic people. This paper therefore reviews ASD's value as both neuropsychological and social constructs. Although lacking neuropsychological validity, the autism label may be beneficial for autistic self-identity, reduction of stigma, and administering support. Whilst a shift away from case-control ASD research is warranted, lay notions of 'different brains' may be preserved.
Collapse
Affiliation(s)
- Daniel Crawshaw
- School of Psychology, University of Nottingham, University Park, Nottingham, UK
| |
Collapse
|
19
|
Shaffer RC, Reisinger DL, Schmitt LM, Lamy M, Dominick KC, Smith EG, Coffman MC, Esbensen AJ. Systematic Review: Emotion Dysregulation in Syndromic Causes of Intellectual and Developmental Disabilities. J Am Acad Child Adolesc Psychiatry 2023; 62:518-557. [PMID: 36007813 DOI: 10.1016/j.jaac.2022.06.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 06/03/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To summarize the current state of the literature regarding emotion dysregulation (ED) in syndromic intellectual disabilities (S-IDs) in 6 of the most common forms of S-IDs-Down syndrome, fragile X syndrome (FXS), tuberous sclerosis complex, Williams syndrome, Prader-Willi syndrome, and Angelman syndrome-and to determine future research directions for identification and treatment of ED. METHOD PubMed bibliographic database was searched from date of inception to May 2021. PRISMA 2020 guidelines were followed with the flowchart, table of included studies, list of excluded studies, and checklist provided. Filters applied included human research and English. Only original research articles were included in the final set, but review articles were used to identify secondary citations of primary studies. All articles were reviewed for appropriateness by 2 authors and summarized. Inclusion criteria were met by 145 articles (Down syndrome = 29, FXS = 55, tuberous sclerosis complex = 11, Williams syndrome = 18, Prader-Willi syndrome = 24, Angelman syndrome = 8). RESULTS Each syndrome review was summarized separately and further subdivided into articles related to underlying neurobiology, behaviors associated with ED, assessment, and targeted intervention. FXS had the most thorough research base, followed by Down syndrome and Prader-Willi syndrome, with the other syndromes having more limited available research. Very limited research was available regarding intervention for all disorders except FXS. CONCLUSION Core underlying characteristics of S-IDs appear to place youth at higher risk for ED, but further research is needed to better assess and treat ED in S-IDs. Future studies should have a standard assessment measure of ED, such as the Emotion Dysregulation Inventory, and explore adapting established curricula for ED from the neurotypical and autism spectrum disorder fields.
Collapse
Affiliation(s)
- Rebecca C Shaffer
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; University of Cincinnati School of Medicine, Cincinnati, Ohio.
| | | | - Lauren M Schmitt
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; University of Cincinnati School of Medicine, Cincinnati, Ohio
| | - Martine Lamy
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; University of Cincinnati School of Medicine, Cincinnati, Ohio
| | - Kelli C Dominick
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; University of Cincinnati School of Medicine, Cincinnati, Ohio
| | - Elizabeth G Smith
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; University of Cincinnati School of Medicine, Cincinnati, Ohio
| | | | - Anna J Esbensen
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; University of Cincinnati School of Medicine, Cincinnati, Ohio
| |
Collapse
|
20
|
van Balkom ID, Burdeus-Olavarrieta M, Cooke J, de Cuba AG, Turner A. Consensus recommendations on mental health issues in Phelan-McDermid syndrome. Eur J Med Genet 2023; 66:104770. [PMID: 37085014 DOI: 10.1016/j.ejmg.2023.104770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 04/12/2023] [Accepted: 04/18/2023] [Indexed: 04/23/2023]
Abstract
Phelan-McDermid syndrome is a rare genetic condition caused by a deletion encompassing the 22q13.3 region or a pathogenic variant of the gene SHANK3. The clinical presentation is variable, but main characteristics include global developmental delay/intellectual disability (ID), marked speech impairment or delay, along with other features like hypotonia and somatic or psychiatric comorbidities. This publication delineates mental health, developmental and behavioural themes across the lifetime of individuals with PMS as informed by parents/caregivers, experts, and other key professionals involved in PMS care. We put forward several recommendations based on the available literature concerning mental health and behaviour in PMS. Additionally, this article aims to improve our awareness of the importance of considering developmental level of the individual with PMS when assessing mental health and behavioural issues. Understanding how the discrepancy between developmental level and chronological age may impact concerning behaviours offers insight into the meaning of those behaviours and informs care for individuals with PMS, enabling clinicians to address unmet (mental health) care needs and improve quality of life.
Collapse
Affiliation(s)
- Ingrid Dc van Balkom
- Jonx, Department of (Youth) Mental Health and Autism, Lentis Psychiatric Institute, Groningen, the Netherlands; Rob Giel Research Centre, Department of Psychiatry, University Medical Center Groningen, Groningen, the Netherlands.
| | - Monica Burdeus-Olavarrieta
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, IiSGM, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; School of Psychology, Universidad Autónoma, Madrid, Spain
| | - Jennifer Cooke
- Forensic and Neurodevelopmental Sciences Department, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, United Kingdom
| | - A Graciela de Cuba
- Jonx, Department of (Youth) Mental Health and Autism, Lentis Psychiatric Institute, Groningen, the Netherlands
| | - Alison Turner
- Phelan-McDermid Syndrome Foundation UK, 99 Highgate W Hill, London, N6 6NR, United Kingdom
| |
Collapse
|
21
|
Rodrigues EL, Figueiredo PS, Marcelino G, de Cássia Avellaneda Guimarães R, Pott A, Santana LF, Hiane PA, do Nascimento VA, Bogo D, de Cássia Freitas K. Maternal Intake of Polyunsaturated Fatty Acids in Autism Spectrum Etiology and Its Relation to the Gut Microbiota: What Do We Know? Nutrients 2023; 15:nu15071551. [PMID: 37049390 PMCID: PMC10097097 DOI: 10.3390/nu15071551] [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: 01/17/2023] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 04/14/2023] Open
Abstract
Maternal food habits and gut microbiota composition have potential effects on fetal neurodevelopment, impacting Autism Spectrum Disorder (ASD). Our research aims to outline the relationship that ingestion of polyunsaturated fatty acids (PUFAs) and the composition of maternal gut microbiota have with the possible development of ASD in offspring. We suggest that genetic factors could be related to the different conversions between unsaturated fatty acids according to sex and, mainly, the impact of the pregnancy diet on the higher or lower risk of neurological impairments. The proportion of the phyla Firmicutes/Bacteroidetes is high with an increased consumption of linoleic acid (LA, n-6 PUFA), which is associated with maternal intestinal dysbiosis and consequently starts the inflammatory process, harming myelinization. In contrast, the consumption of α-linolenic acid (ALA, n-3 PUFA) tends to re-establish the balance of the maternal microbiota with anti-inflammatory action. Moreover, human observational studies showed a strong correlation between the consumption of n-3 PUFA, mainly above 340 g of fish per week, with beneficial effects on infant neurodevelopment. Therefore, we suggest that the proper intake of foods rich in n-3 PUFAs and their supplementation during pregnancy until lactation has an impact on reducing the development of ASD. Controlled studies with n-3 PUFA supplementation are still necessary to verify the ideal dose and the best form of administration.
Collapse
Affiliation(s)
- Elisana Lima Rodrigues
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Priscila Silva Figueiredo
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Gabriela Marcelino
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Rita de Cássia Avellaneda Guimarães
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Arnildo Pott
- Institute of Biosciences, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Lidiani Figueiredo Santana
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Priscila Aiko Hiane
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Valter Aragão do Nascimento
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Danielle Bogo
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| | - Karine de Cássia Freitas
- Graduate Program in Health and Development in the Central-West Region of Brazil, Federal University of Mato Grosso do Sul-UFMS, Campo Grande 79079-900, MS, Brazil
| |
Collapse
|
22
|
High Performance of a Dominant/X-Linked Gene Panel in Patients with Neurodevelopmental Disorders. Genes (Basel) 2023; 14:genes14030708. [PMID: 36980980 PMCID: PMC10048137 DOI: 10.3390/genes14030708] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/06/2023] [Accepted: 03/12/2023] [Indexed: 03/15/2023] Open
Abstract
Neurodevelopmental disorders (NDDs) affect 2–5% of the population and approximately 50% of cases are due to genetic factors. Since de novo pathogenic variants account for the majority of cases, a gene panel including 460 dominant and X-linked genes was designed and applied to 398 patients affected by intellectual disability (ID)/global developmental delay (GDD) and/or autism (ASD). Pathogenic variants were identified in 83 different genes showing the high genetic heterogeneity of NDDs. A molecular diagnosis was established in 28.6% of patients after high-depth sequencing and stringent variant filtering. Compared to other available gene panel solutions for NDD molecular diagnosis, our panel has a higher diagnostic yield for both ID/GDD and ASD. As reported previously, a significantly higher diagnostic yield was observed: (i) in patients affected by ID/GDD compared to those affected only by ASD, and (ii) in females despite the higher proportion of males among our patients. No differences in diagnostic rates were found between patients affected by different levels of ID severity. Interestingly, patients harboring pathogenic variants presented different phenotypic features, suggesting that deep phenotypic profiling may help in predicting the presence of a pathogenic variant. Despite the high performance of our panel, whole exome-sequencing (WES) approaches may represent a more robust solution. For this reason, we propose the list of genes included in our customized gene panel and the variant filtering procedure presented here as a first-tier approach for the molecular diagnosis of NDDs in WES studies.
Collapse
|
23
|
Chowdhury MAK, Hardin JW, Love BL, Merchant AT, McDermott S. Relationship of Nonsteroidal Anti-Inflammatory Drug Use During Pregnancy with Autism Spectrum Disorder and Intellectual Disability Among Offspring. J Womens Health (Larchmt) 2023; 32:356-365. [PMID: 36576852 DOI: 10.1089/jwh.2022.0113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Objective: This study aimed to examine the association of nonsteroidal anti-inflammatory drug (NSAID) use by pregnant women during pregnancy with autism spectrum disorder (ASD) and intellectual disability (ID) in their children among Medicaid-insured mother-child dyads. Materials and Methods: We conducted a retrospective cohort study linking multiple datasets of South Carolina for the years between 2010 and 2017, in which the main exposure variable was NSAID use during pregnancy and outcome variables were ASD only, ID only, and ASD with ID. We conducted a multinomial logistic regression analysis, controlling for identified risk factors for ASD (mother's age, race, body-mass index, preeclampsia, and gestational diabetes). Results: NSAID use during pregnancy was found to be associated with ID only in both unadjusted and adjusted analyses. Children with mothers who had NSAID prescriptions were 26% more likely to have ID in comparison with children whose mothers did not have NSAID prescriptions (odds ratio: 1.26 [1.10-1.46]). The other risk factors identified for ASD were maternal age, race, preeclampsia, smoking, low birth weight, and obesity. For ID, the risk factors were maternal age, race, smoking, birth weight, overweight, and obesity, all of which were also associated with ASD with ID, except for overweight. Conclusions: NSAID usage during pregnancy was found to be associated with ID only and not with ASD. However, more research is needed to validate the effect of NSAIDs during pregnancy on ASD and ID among children.
Collapse
Affiliation(s)
| | - James W Hardin
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, USA
| | - Bryan L Love
- Department of Clinical Pharmacy and Outcome Sciences, College of Pharmacy, University of South Carolina, Columbia, South Carolina, USA
| | - Anwar T Merchant
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, USA
| | - Suzanne McDermott
- Department of Environmental, Occupational, and Geospatial Health Sciences, CUNY Graduate School of Public Health and Health Policy, New York, New York, USA
| |
Collapse
|
24
|
Shenouda J, Barrett E, Davidow AL, Sidwell K, Lescott C, Halperin W, Silenzio VMB, Zahorodny W. Prevalence and Disparities in the Detection of Autism Without Intellectual Disability. Pediatrics 2023; 151:e2022056594. [PMID: 36700335 DOI: 10.1542/peds.2022-056594] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/04/2022] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Intellectual ability predicts functional outcomes for children with autism spectrum disorder (ASD). It is essential to classify ASD children with and without intellectual disability (ID) to aid etiological research, provide services, and inform evidence-based educational and health planning. METHODS Using a cross-sectional study design, data from 2000 to 2016 active ASD surveillance among 8-year-olds residing in the New York-New Jersey Metropolitan Area were analyzed to determine ASD prevalence with and without ID. Multivariable Poisson regression models were used to identify trends for ASD with ID (ASD-I) and without ID (ASD-N). RESULTS Overall, 4661 8-year-olds were identified with ASD. Those that were ASI-I were 1505 (32.3%) and 2764 (59.3%) were ASD-N. Males were 3794 (81.4%), 946 (20.3%) were non-Hispanic Black (Black), 1230 (26.4%) were Hispanic, and 2114 (45.4%) were non-Hispanic white (white). We observed 2-fold and 5-fold increases in the prevalence of ASD-I and ASD-N, respectively, from 2000-2016. Black children were 30% less likely to be identified with ASD-N compared with white children. Children residing in affluent areas were 80% more likely to be identified with ASD-N compared with children in underserved areas. A greater proportion of children with ASD-I resided in vulnerable areas compared with children with ASD-N. Males had higher prevalence compared with females regardless of ID status; however, male-to-female ratios were slightly lower among ASD-I compared with ASD-N cases. CONCLUSIONS One-in-3 children with ASD had ID. Disparities in the identification of ASD without ID were observed among Black and Hispanic children as well as among children residing in underserved areas.
Collapse
Affiliation(s)
- Josephine Shenouda
- Rutgers School of Public Health, Piscataway, New Jersey
- Rutgers New Jersey Medical School, Newark, New Jersey
| | - Emily Barrett
- Rutgers School of Public Health, Piscataway, New Jersey
| | - Amy L Davidow
- New York University School of Global Public Health, New York, New York
| | - Kate Sidwell
- Rutgers New Jersey Medical School, Newark, New Jersey
| | - Cara Lescott
- Rutgers New Jersey Medical School, Newark, New Jersey
| | | | | | | |
Collapse
|
25
|
Yang J, Shen Y, Tian Y, Peng J, Fu X, Li Y, Ou J. Investigating and comparing the psychometric properties of the Chinese Mandarin version of social responsiveness scale-2 and its shortened version in preschool-age children with autism spectrum disorder. Asian J Psychiatr 2023; 79:103395. [PMID: 36495828 DOI: 10.1016/j.ajp.2022.103395] [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: 07/31/2022] [Revised: 10/11/2022] [Accepted: 10/14/2022] [Indexed: 12/12/2022]
Abstract
We aimed to investigate and compare the psychometric properties of the Chinese Mandarin Social Responsiveness Scale-2 (SRS-2) and its shortened version. The study assessed 670 children with autism spectrum disorder (ASD) aged 30-54 months and 138 typical developmental (TD) children of the same age in mainland China. Our item reliability test revealed that only 36 items of the 65 items in the Chinese Mandarin SRS-2 (Preschool) met the reliability criteria. Moreover, the shortened version of SRS-2 (Preschool) with four subscales and 30 items maintained strong correlations (r = 0.961) with the Chinese Mandarin SRS-2 (Preschool), and demonstrated improved psychometric performance on the 4-week test-retest reliability (intraclass correlations was 0.70), internal consistency (Cronbach's alpha 0.71-0.91), construct validity, and convergent validity with the Autism Diagnostic Observation Schedule, Autism Diagnostic Interview-Revised, and Child Behavior Checklist. Receiver operating characteristics (ROC) analyses showed excellent and comparable discriminant validity of the shortened version with an area under the curve of 0.992. Our data suggested a cutoff ≥ 22.5 for the shortened version, with good accuracy in screening autism symptoms (sensitivity=96.9 %, specificity=94.2 %). Our findings demonstrated that the shortened version of SRS-2 (Preschool) was a reliable and valid instrument for identifying preschoolers with ASD in mainland China.
Collapse
Affiliation(s)
- Jiaxin Yang
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Yidong Shen
- National Clinical Research Center for Mental Disorders, Department of Psychiatry and Hunan Medical Center for Mental Health, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Yusheng Tian
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; National Clinical Research Center for Mental Disorders, Department of Psychiatry and Hunan Medical Center for Mental Health, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Juan Peng
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xi Fu
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Yamin Li
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| | - Jianjun Ou
- National Clinical Research Center for Mental Disorders, Department of Psychiatry and Hunan Medical Center for Mental Health, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
26
|
Ma K, Taylor C, Williamson M, Newton SS, Qin L. Diminished activity-dependent BDNF signaling differentially causes autism-like behavioral deficits in male and female mice. Front Psychiatry 2023; 14:1182472. [PMID: 37205980 PMCID: PMC10189061 DOI: 10.3389/fpsyt.2023.1182472] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/11/2023] [Indexed: 05/21/2023] Open
Abstract
Autism spectrum disorder (ASD) is a group of neurodevelopmental disorders with strong genetic heterogeneity and more prevalent in males than females. Recent human genetic studies have identified multiple high-risk genes for ASD, which produce similar phenotypes, indicating that diverse genetic factors converge to common molecular pathways. We and others have hypothesized that activity-dependent neural signaling is a convergent molecular pathway dysregulated in ASD. However, the causal link between diminished activity-dependent neural signaling and ASD remains unclear. Brain-derived neurotrophic factor (BDNF) is a key molecule mediating activity-dependent neural signaling. We therefore hypothesize that diminished activity-dependent BDNF signaling could confer autism-like behavioral deficits. Here, we investigated the effect of diminished activity-dependent BDNF signaling on autism-like behavioral deficits by using mice with genetic knock-in of a human BDNF methionine (Met) allele, which has decreased activity-dependent BDNF release without altering basal BDNF level. Compared with wild-type (WT) controls, diminished activity-dependent BDNF signaling similarly induced anxiety-like behaviors in male and female mice. Notably, diminished activity-dependent BDNF signaling differentially resulted in autism-like social deficits and increased self-grooming in male and female mice, and male mice were more severe than female mice. Again, sexually dimorphic spatial memory deficits were observed in female BDNF+/Met mice, but not in male BDNF+/Met mice. Our study not only reveals a causal link between diminished activity-dependent BDNF signaling and ASD-like behavioral deficits, but also identifies previously underappreciated sex-specific effect of diminished activity-dependent BDNF signaling in ASD. These mice with genetic knock-in of the human BDNF Met variant provide a distinct mouse model for studying the cellular and molecular mechanisms underlying diminished activity-dependent neural signaling, the common molecular pathway dysregulated in ASD.
Collapse
Affiliation(s)
- Kaijie Ma
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| | - Connie Taylor
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, United States
| | - Mark Williamson
- Biostatistics, Epidemiology, and Research Design Core, University of North Dakota, Grand Forks, ND, United States
| | - Samuel S. Newton
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| | - Luye Qin
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
- *Correspondence: Luye Qin,
| |
Collapse
|
27
|
Badal KK, Puthanveettil SV. Axonal transport deficits in neuropsychiatric disorders. Mol Cell Neurosci 2022; 123:103786. [PMID: 36252719 DOI: 10.1016/j.mcn.2022.103786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/02/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
Axonal transport is a major cellular process that mediates bidirectional signaling between the soma and synapse, enabling both intracellular and intercellular communications. Cellular materials, such as proteins, RNAs, and organelles, are transported by molecular motor proteins along cytoskeletal highways in a highly regulated manner. Several studies have demonstrated that axonal transport is central to normal neuronal function, plasticity, and memory storage. Importantly, disruptions in axonal transport result in neuronal dysfunction and are associated with several neurodegenerative disorders. However, we do not know much about axonal transport deficits in neuropsychiatric disorders. Here, we briefly discuss our current understanding of the role of axonal transport in schizophrenia, bipolar and autism.
Collapse
Affiliation(s)
- Kerriann K Badal
- Department of Neuroscience, UF Scripps Biomedical Research, University of Florida, 130 Scripps Way, Jupiter, FL 33458, USA; Integrative Biology PhD Program, Charles E. Schmidt College of Science, Florida Atlantic University, Jupiter, FL 33458, USA
| | | |
Collapse
|
28
|
Wang T, Kim CN, Bakken TE, Gillentine MA, Henning B, Mao Y, Gilissen C, Nowakowski TJ, Eichler EE. Integrated gene analyses of de novo variants from 46,612 trios with autism and developmental disorders. Proc Natl Acad Sci U S A 2022; 119:e2203491119. [PMID: 36350923 PMCID: PMC9674258 DOI: 10.1073/pnas.2203491119] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 09/28/2022] [Indexed: 08/15/2023] Open
Abstract
Most genetic studies consider autism spectrum disorder (ASD) and developmental disorder (DD) separately despite overwhelming comorbidity and shared genetic etiology. Here, we analyzed de novo variants (DNVs) from 15,560 ASD (6,557 from SPARK) and 31,052 DD trios independently and also combined as broader neurodevelopmental disorders (NDDs) using three models. We identify 615 NDD candidate genes (false discovery rate [FDR] < 0.05) supported by ≥1 models, including 138 reaching Bonferroni exome-wide significance (P < 3.64e-7) in all models. The genes group into five functional networks associating with different brain developmental lineages based on single-cell nuclei transcriptomic data. We find no evidence for ASD-specific genes in contrast to 18 genes significantly enriched for DD. There are 53 genes that show mutational bias, including enrichments for missense (n = 41) or truncating (n = 12) DNVs. We also find 10 genes with evidence of male- or female-bias enrichment, including 4 X chromosome genes with significant female burden (DDX3X, MECP2, WDR45, and HDAC8). This large-scale integrative analysis identifies candidates and functional subsets of NDD genes.
Collapse
Affiliation(s)
- Tianyun Wang
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195
- Department of Medical Genetics, Center for Medical Genetics, Peking University Health Science Center, Beijing, 100191, China
- Neuroscience Research Institute, Peking University, Key Laboratory for Neuroscience, Ministry of Education of China & National Health Commission of China, Beijing, 100191, China
| | - Chang N. Kim
- Department of Anatomy, University of California, San Francisco, CA 94143
| | | | - Madelyn A. Gillentine
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195
| | - Barbara Henning
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195
| | - Yafei Mao
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Christian Gilissen
- Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | | | - Tomasz J. Nowakowski
- Department of Anatomy, University of California, San Francisco, CA 94143
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA 94143
| | - Evan E. Eichler
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195
- HHMI, University of Washington, Seattle, WA 98195
| |
Collapse
|
29
|
Huang R, Potla S, Bhalla S, Al Qabandi Y, Nandula SA, Boddepalli CS, Gutlapalli SD, Lavu VK, Abdelwahab R, Hamid P. The Clinical Implications of the Academic Performance of the Siblings of Individuals With Autism Spectrum Disorder. Cureus 2022; 14:e29116. [PMID: 36258957 PMCID: PMC9559693 DOI: 10.7759/cureus.29116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 09/13/2022] [Indexed: 11/11/2022] Open
Abstract
We all know that autism spectrum disorder (ASD) can affect academic performance. Many children with autism face different challenges at school. However, less attention is paid to the siblings of autistic children, who are at a high risk of ASD or the broad autism phenotype (BAP). Recent data also shows that many siblings of ASD children suffer from neurodevelopmental disorders, mental health problems as well as poor academic performance. This review will look at the possible etiologies of the poor school performance of autistic children's siblings, with an emphasis on the challenges they face. We will also highlight the clinical implications of these findings, and the possible solutions that can help this vulnerable group.
Collapse
|
30
|
Jacob US, Edozie IS, Pillay J. Strategies for enhancing social skills of individuals with intellectual disability: A systematic review. FRONTIERS IN REHABILITATION SCIENCES 2022; 3:968314. [PMID: 36188912 PMCID: PMC9513363 DOI: 10.3389/fresc.2022.968314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/24/2022] [Indexed: 11/24/2022]
Abstract
Individuals with intellectual disability who suffer from comorbid mental health problems are likely to experience difficulties in socialising. Deficits in social skills are also associated with challenging behaviours and self-injury. This paper presents global evidence from a systematic review of literature on such issues as 'interventions'; 'social skills development', and 'individuals with intellectual disability'. A thorough search of various bibliographic databases identified 1 124 academic papers. Ten papers met the inclusion criteria for in-depth analysis concerning the use of interventions to develop social skills among individuals with intellectual disability. The study revealed that the social skills of individuals with intellectual disability had been fostered using different strategies, such as classroom-based intervention, emotional intelligence training, use of a peer network intervention, computer games of emotion regulation, and puppet play therapy. Furthermore, the findings suggest that various aspects like communication, bridging the gap in social skills deficits, emotional recognition and regulation, and adaptive behaviour were fostered using the identified intervention strategy. This review revealed that social skills interventions appeared modestly effective but may not be generalisable to school settings or self-reported social behaviour for individuals with intellectual disability. It is also necessary to increase the sample size in future studies to draw generalisable conclusions.
Collapse
Affiliation(s)
- Udeme Samuel Jacob
- South African Research Chair in Education and Care in Childhood, Faculty of Education, University of Johannesburg, Johannesburg, South Africa
| | - Isioma Sitamalife Edozie
- Department of Special Education, Faculty of Education, University of Ibadan, Ibadan, Nigeria
- Department of Adult and Non - Formal Education, School of Early Childhood Care, Primary, Adult and Non -Formal Education, Federal College of Education (Technical) Asaba, Asaba, Nigeria
| | - Jace Pillay
- South African Research Chair in Education and Care in Childhood, Faculty of Education, University of Johannesburg, Johannesburg, South Africa
| |
Collapse
|
31
|
Huo Y, Lu W, Tian Y, Hou Q, Man HY. Prkn knockout mice show autistic-like behaviors and aberrant synapse formation. iScience 2022; 25:104573. [PMID: 35789851 PMCID: PMC9249611 DOI: 10.1016/j.isci.2022.104573] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 02/26/2022] [Accepted: 06/07/2022] [Indexed: 11/20/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder with high genetic heterogeneity, affecting one in 44 children in the United States. Recent genomic sequencing studies from autistic human individuals indicate that PARK2, a gene that has long been considered in the pathogenesis of Parkinson's disease, is involved in ASD. Here, we report that Prkn knockout (KO) mice demonstrate autistic-like behaviors including impaired social interaction, elevated repetitive behaviors, and deficits in communication. In addition, Prkn KO mice show reduced neuronal activity in the context of sociability in the prelimbic cortex. Cell morphological examination of layer 5 prelimbic cortical neurons shows a reduction in dendritic arborization and spine number. Furthermore, biochemistry and immunocytochemistry analyses reveal alterations in synapse density and the molecular composition of synapses. These findings indicate that Prkn is implicated in brain development and suggest the potential use of the Prkn KO mouse as a model for autism research.
Collapse
Affiliation(s)
- Yuda Huo
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Wen Lu
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Yuan Tian
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Qingming Hou
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA 02215, USA
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA 02118, USA
- Center for Systems Neuroscience, Boston University, Boston, MA 02215, USA
| |
Collapse
|
32
|
Hoff KJ, Aiken JE, Gutierrez MA, Franco SJ, Moore JK. Tubulinopathy mutations in TUBA1A that disrupt neuronal morphogenesis and migration override XMAP215/Stu2 regulation of microtubule dynamics. eLife 2022; 11:76189. [PMID: 35511030 PMCID: PMC9236607 DOI: 10.7554/elife.76189] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Heterozygous, missense mutations in α- or β-tubulin genes are associated with a wide range of human brain malformations, known as tubulinopathies. We seek to understand whether a mutation’s impact at the molecular and cellular levels scale with the severity of brain malformation. Here, we focus on two mutations at the valine 409 residue of TUBA1A, V409I, and V409A, identified in patients with pachygyria or lissencephaly, respectively. We find that ectopic expression of TUBA1A-V409I/A mutants disrupt neuronal migration in mice and promote excessive neurite branching and a decrease in the number of neurite retraction events in primary rat neuronal cultures. These neuronal phenotypes are accompanied by increased microtubule acetylation and polymerization rates. To determine the molecular mechanisms, we modeled the V409I/A mutants in budding yeast and found that they promote intrinsically faster microtubule polymerization rates in cells and in reconstitution experiments with purified tubulin. In addition, V409I/A mutants decrease the recruitment of XMAP215/Stu2 to plus ends in budding yeast and ablate tubulin binding to TOG (tumor overexpressed gene) domains. In each assay tested, the TUBA1A-V409I mutant exhibits an intermediate phenotype between wild type and the more severe TUBA1A-V409A, reflecting the severity observed in brain malformations. Together, our data support a model in which the V409I/A mutations disrupt microtubule regulation typically conferred by XMAP215 proteins during neuronal morphogenesis and migration, and this impact on tubulin activity at the molecular level scales with the impact at the cellular and tissue levels. Proteins are molecules made up of long chains of building blocks called amino acids. When a mutation changes one of these amino acids, it can lead to the protein malfunctioning, which can have many effects at the cell and tissue level. Given that human proteins are made up of 20 different amino acids, each building block in a protein could mutate to any of the other 19 amino acids, and each mutations could have different effects. Tubulins are proteins that form microtubules, thin tubes that help give cells their shape and allow them to migrate. These proteins are added or removed to microtubules depending on the cell’s needs, meaning that microtubules can grow or shrink depending on the situation. Mutations in the tubulin proteins have been linked to malformations of varying severities involving the formation of ridges and folds on the surface of the brain, including lissencephaly, pachygyria or polymicrogyria. Hoff et al. wanted to establish links between tubulin mutations and the effects observed at both cell and tissue level in the brain. They focused on two mutations in the tubulin protein TUBA1A that affect the amino acid in position 409 in the protein, which is normally a valine. One of the mutations turns this valine into an amino acid called isoleucine. This mutation is associated with pachygyria, which leads to the brain developing few ridges that are broad and flat. The second mutation turns the valine into an alanine, and is linked to lissencephaly, a more severe condition in which the brain develops no ridges, appearing smooth. Hoff et al. found that both mutations interfere with the development of the brain by stopping neurons from migrating properly, which prevents them from forming the folds in the brain correctly. At the cellular level, the mutations lead to tubulins becoming harder to remove from microtubules, making microtubules more stable than usual. This results in longer microtubules that are harder for the cell to shorten or destroy as needed. Additionally, Hoff et al. showed that the mutant versions of TUBA1A have weaker interactions with a protein called XMAP215, which controls the addition of tubulin to microtubules. This causes the microtubules to grow uncontrollably. Hoff et al. also established that the magnitude of the effects of each mutation on microtubule growth scale with the severity of the disorder they cause. Specifically, cells in which TUBA1A is not mutated have microtubules that grow at a normal rate, and lead to typical brain development. Meanwhile, cells carrying the mutation that turns a valine into an alanine, which is linked to the more severe condition lissencephaly, have microtubules that grow very fast. Finally, cells in which the valine is mutated to an isoleucine – the mutation associated with the less severe malformation pachygyria – have microtubules that grow at an intermediate rate. These findings provide a link between mutations in tubulin proteins and larger effects on cell movement that lead to brain malformations. Additionally, they also link the severity of the malformation to the severity of the microtubule defect caused by each mutation. Further work could examine whether microtubule stabilization is also seen in other similar diseases, which, in the long term, could reveal ways to detect and treat these illnesses.
Collapse
Affiliation(s)
- Katelyn J Hoff
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Jayne E Aiken
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Mark A Gutierrez
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Santos J Franco
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Jeffrey K Moore
- University of Colorado School of Medicine, Aurora, United States
| |
Collapse
|
33
|
Mignogna ML, Ficarella R, Gelmini S, Marzulli L, Ponzi E, Gabellone A, Peschechera A, Alessio M, Margari L, Gentile M, D’Adamo P. Clinical characterization of a novel RAB39B nonstop mutation in a family with ASD and severe ID causing RAB39B downregulation and study of a Rab39b knock down mouse model. Hum Mol Genet 2022; 31:1389-1406. [PMID: 34761259 PMCID: PMC9071400 DOI: 10.1093/hmg/ddab320] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 11/18/2022] Open
Abstract
Autism spectrum disorder (ASD) and intellectual disability (ID) often exist together in patients. The RAB39B gene has been reported to be mutated in ID patients with additional clinical features ranging from ASD, macrocephaly, seizures and/or early-onset parkinsonism. Here, we describe a novel RAB39B nonstop mutation [Xq28; c.640 T > C; p.(*214Glnext*21)] in a family with ASD, severe ID and poor motor coordination, and we assessed the pathogenicity of the mutation. A heterologous cell system and a Rab39b knockdown (KD) murine model, which mimic the nonstop mutation, were used to validate the deleterious effect of the RAB39B mutation. The mutation led to RAB39B protein instability, resulting in its increased degradation and consequent downregulation. Using a Rab39b KD mouse model, we demonstrated that the downregulation of RAB39B led to increased GluA2 lacking Ca2+-permeable AMPAR composition at the hippocampal neuronal surface and increased dendritic spine density that remained in an immature filopodia-like state. These phenotypes affected behavioural performance in a disease-specific manner. Rab39b KD mice revealed impaired social behaviour but intact social recognition. They also showed normal anxiety-like, exploratory and motivational behaviours but impaired working and associative memories. In conclusion, we found a novel RAB39B nonstop variant that segregated in a family with a clinical phenotype including ID, ASD and poor motor coordination. The pathogenicity of mutations causing the downregulation of RAB39B proteins, impacting AMPAR trafficking and dendritic spine morphogenesis, reinforced the idea that AMPAR modulation and dendritic spine assets could be considered hallmarks of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Maria Lidia Mignogna
- Molecular Genetics of Intellectual Disability, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Romina Ficarella
- Medical Genetics Unit, Department of Reproductive Medicine, ASL Bari, 70132, Bari, Italy
| | - Susanna Gelmini
- Molecular Genetics of Intellectual Disability, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Lucia Marzulli
- Child Neuropsychiatry Unit, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70126, Bari, Italy
| | - Emanuela Ponzi
- Medical Genetics Unit, Department of Reproductive Medicine, ASL Bari, 70132, Bari, Italy
| | - Alessandra Gabellone
- Child Neuropsychiatry Unit, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70126, Bari, Italy
| | - Antonia Peschechera
- Child Neuropsychiatry Unit, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70126, Bari, Italy
| | - Massino Alessio
- Proteome Biochemistry, Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Lucia Margari
- Child Neuropsychiatry Unit, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70126, Bari, Italy
| | - Mattia Gentile
- Medical Genetics Unit, Department of Reproductive Medicine, ASL Bari, 70132, Bari, Italy
| | - Patrizia D’Adamo
- Molecular Genetics of Intellectual Disability, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| |
Collapse
|
34
|
Krami AM, Bouzidi A, Charif M, Amalou G, Charoute H, Rouba H, Roky R, Lenaers G, Barakat A, Nahili H. A homozygous nonsense HECW2 variant is associated with neurodevelopmental delay and intellectual disability. Eur J Med Genet 2022; 65:104515. [PMID: 35487419 DOI: 10.1016/j.ejmg.2022.104515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 03/07/2022] [Accepted: 04/23/2022] [Indexed: 11/17/2022]
Abstract
Intellectual disability is characterized by a significant impaired intellectual and adaptive functioning, affecting approximately 1-3% of the population, which can be caused by a variety of environmental and genetic factors. In this respect, de novo heterozygous HECW2 variants were associated recently with neurodevelopmental disorders associated to hypotonia, seizures, and absent language. HECW2 encodes an E3 ubiquitin-protein ligase that stabilizes and enhances transcriptional activity of p73, a key factor regulating proliferation, apoptosis, and neuronal differentiation, which are together essential for proper brain development. Here, using whole exome sequencing, we identified a homozygous nonsense HECW2 variant: c.736C > T; p.Arg246* in a proband from a Moroccan consanguineous family, with developmental delay, intellectual disability, hypotonia, generalized tonico-clonic seizures and a persistent tilted head. Thus this study describes the first homozygous HECW2 variant, inherited as an autosomal recessive pattern, contrasting with former reported de novo variants found in HECW2 patients.
Collapse
Affiliation(s)
- Al Mehdi Krami
- Genomics and Human Genetics Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco; Laboratory of Physiopathology, Molecular Genetics & Biotechnology, Faculty of Sciences Ain Chock, Health and Biotechnology Research Centre, Hassan II University of Casablanca, Maarif B.P, 5366, Casablanca, Morocco.
| | - Aymane Bouzidi
- Genomics and Human Genetics Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco; Université Angers, MitoLab Team, UMR CNRS 6015, INSERM U1083, Institut MitoVasc, SFR ICAT, Angers, France; Team of Anthropogenetics and Biotechnologies, Faculty of Sciences, Chouaïb Doukkali University, El Jadida, Morocco
| | - Majida Charif
- Genetics and Immuno-Cell Therapy Team, Mohammed First University, Oujda, Morocco
| | - Ghita Amalou
- Genomics and Human Genetics Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco; Université Angers, MitoLab Team, UMR CNRS 6015, INSERM U1083, Institut MitoVasc, SFR ICAT, Angers, France; Team of Anthropogenetics and Biotechnologies, Faculty of Sciences, Chouaïb Doukkali University, El Jadida, Morocco
| | - Hicham Charoute
- Research Unit of Epidemiology, Biostatistics and Bioinfortmatics, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Hassan Rouba
- Genomics and Human Genetics Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Rachida Roky
- Laboratory of Physiopathology, Molecular Genetics & Biotechnology, Faculty of Sciences Ain Chock, Health and Biotechnology Research Centre, Hassan II University of Casablanca, Maarif B.P, 5366, Casablanca, Morocco
| | - Guy Lenaers
- Université Angers, MitoLab Team, UMR CNRS 6015, INSERM U1083, Institut MitoVasc, SFR ICAT, Angers, France; Service de Neurologie, CHU d'Angers, Angers, France
| | - Abdelhamid Barakat
- Genomics and Human Genetics Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Halima Nahili
- Genomics and Human Genetics Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| |
Collapse
|
35
|
Borilli MC, Germano CMR, de Avó LRDS, Pilotto RF, Melo DG. Family quality of life among families who have children with mild intellectual disability associated with mild autism spectrum disorder. ARQUIVOS DE NEURO-PSIQUIATRIA 2022; 80:360-367. [PMID: 35293554 DOI: 10.1590/0004-282x-anp-2020-0537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 06/15/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Intellectual disability (ID) and autism spectrum disorder (ASD) are often concomitant childhood developmental disorders. These disorders can alter family quality of life (FQoL). OBJECTIVE To investigate FQoL among families who have children with mild ID, associated with mild ASD. METHODS Cross-sectional descriptive study with 69 families who have children with mild ID and ASD, ranging from six to 16 years old, and who were provided with disability-related services in Brazil. Data were collected using a family sociodemographic questionnaire, an ID and ASD personal profile form, the Barthel index for activities of daily living and the Beach Center FQoL scale. RESULTS People with ID and ASD had an average score of 88.2±11.5 in the Barthel index, thus indicating moderate dependency in basic activities of daily living. The average total FQoL score (3.56±0.34) was lower than the scores for the "family interaction" (3.91±0.42; p<0.001), "parenting" (3.79±0.35; p<0.001) and "disability-related support" (3.98±0.16; p<0.001) domains; and higher than the scores for the "physical/material well-being" (3.19±0.64; p<0.001) and "emotional wellbeing" (2.75±0.62; p<0.001) domains. Parents' marital condition, monthly family income, family religious practice and effective communication skills among the people with ID and ASD were predictors for FQoL (R2=0.407; p<0.001). CONCLUSIONS FQoL was sustained through factors such as family interaction and parents' care for their children. Improving families' emotional wellbeing and physical and material conditions is likely to positively affect the FQoL of these families.
Collapse
Affiliation(s)
| | - Carla Maria Ramos Germano
- Universidade Federal de São Carlos, Programa de Pós-Graduação em Enfermagem, São Carlos SP, Brazil.,Universidade Federal de São Carlos, Departamento de Medicina, São Carlos SP, Brazil.,Instituto Nacional de Genética Médica Populacional, Porto Alegre RS, Brazil
| | - Lucimar Retto da Silva de Avó
- Universidade Federal de São Carlos, Departamento de Medicina, São Carlos SP, Brazil.,Instituto Nacional de Genética Médica Populacional, Porto Alegre RS, Brazil
| | | | - Débora Gusmão Melo
- Universidade Federal de São Carlos, Programa de Pós-Graduação em Enfermagem, São Carlos SP, Brazil.,Universidade Federal de São Carlos, Departamento de Medicina, São Carlos SP, Brazil.,Instituto Nacional de Genética Médica Populacional, Porto Alegre RS, Brazil
| |
Collapse
|
36
|
Karlsson H, Sjöqvist H, Brynge M, Gardner R, Dalman C. Childhood infections and autism spectrum disorders and/or intellectual disability: a register-based cohort study. J Neurodev Disord 2022; 14:12. [PMID: 35151261 PMCID: PMC8903600 DOI: 10.1186/s11689-022-09422-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 01/26/2022] [Indexed: 12/13/2022] Open
Abstract
Objective To explore the associations between childhood infections and subsequent diagnoses of autism spectrum disorder (ASD), intellectual disability (ID), and their co-occurrence. Methods The association between specialized care for any infection, defined by ICD-codes, and later ASD or ID was investigated in a register-based cohort of 556,732 individuals born 1987–2010, resident in Stockholm County, followed from birth to their 18th birthday or December 31, 2016. We considered as potential confounders children’s characteristics, family socioeconomic factors, obstetric complications, and parental histories of treatment for infection and psychiatric disorders in survival analyses with extended Cox regression models. Residual confounding by shared familial factors was addressed in sibling analyses using within-strata estimation in Cox regression models. Sensitivity analyses with the exclusion of congenital causes of ASD/ID and documented risk for infections were also performed. Results Crude estimates indicated that infections during childhood were associated with later ASD and ID with the largest risks observed for diagnoses involving ID. Inclusion of covariates, exclusion of congenital causes of ASD/ID from the population, and sibling comparisons highlighted the potential for confounding by both heritable and non-heritable factors, though risks remained in all adjusted models. In adjusted sibling comparisons, excluding congenital causes, infections were associated with later “ASD without ID” (HR 1.24, 95%CI 1.15–1.33), “ASD with ID” (1.57, 1.35–1.82), and “ID without ASD” (2.01, 1.76–2.28). Risks associated with infections varied by age at exposure and by age at diagnosis of ASD/ID. Conclusions Infections during childhood may contribute to a later diagnosis of ID and ASD. Supplementary Information The online version contains supplementary material available at 10.1186/s11689-022-09422-4.
Collapse
|
37
|
Fernandes H. Therapeutic Alliance in Cognitive Behavioural Therapy in Child and Adolescent Mental Health-Current Trends and Future Challenges. Front Psychol 2022; 12:610874. [PMID: 35046861 PMCID: PMC8763013 DOI: 10.3389/fpsyg.2021.610874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 11/02/2021] [Indexed: 11/30/2022] Open
Abstract
This extended literature review proposes to present the trends in the therapeutic alliance, outcomes, and measures in the last decade within the premises of individual cognitive behaviour therapy (CBT) and its innovations, used as an interventional measure in the context of child and adolescent mental health setting. A brief background of the rationale for conducting this literature search is presented at the start. This is followed by the methodology and design which incorporates the inclusion and exclusion criteria and the basis for the same. The critical appraisal of the primary studies is presented in the literature review section with a brief description of the summary features of the studies in the study tables followed by the results and discussion of the study findings. To summarise, the literature review of primary studies conducted in the last decade demonstrates the need for further research to be conducted both in the field of CBT in children and therapeutic alliance, competence, and therapy outcomes, integrating perspectives in child development, carer alliance, and the social construct theory in children, to allow for further innovations in CBT in the context of increasing challenges in the current times of exponentially developing technology and its utility without compromising the quality of therapy. In conclusion, recommendations are made as a guideline for future studies and research in this field.
Collapse
Affiliation(s)
- Hazel Fernandes
- Child and Adolescent Psychiatry, Health Service Executive, Dublin, Ireland
| |
Collapse
|
38
|
Wang J, Rappold GA, Fröhlich H. Disrupted Mitochondrial Network Drives Deficits of Learning and Memory in a Mouse Model of FOXP1 Haploinsufficiency. Genes (Basel) 2022; 13:127. [PMID: 35052467 PMCID: PMC8775322 DOI: 10.3390/genes13010127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 11/16/2022] Open
Abstract
Reduced cognitive flexibility, characterized by restricted interests and repetitive behavior, is associated with atypical memory performance in autism spectrum disorder (ASD), suggesting hippocampal dysfunction. FOXP1 syndrome is a neurodevelopmental disorder characterized by ASD, language deficits, global developmental delay, and mild to moderate intellectual disability. Strongly reduced Foxp1 expression has been detected in the hippocampus of Foxp1+/- mice, a brain region required for learning and memory. To investigate learning and memory performance in these animals, fear conditioning tests were carried out, which showed impaired associative learning compared with wild type (WT) animals. To shed light on the underlying mechanism, we analyzed various components of the mitochondrial network in the hippocampus. Several proteins regulating mitochondrial biogenesis (e.g., Foxo1, Pgc-1α, Tfam) and dynamics (Mfn1, Opa1, Drp1 and Fis1) were significantly dysregulated, which may explain the increased mitophagy observed in the Foxp1+/- hippocampus. The reduced activity of complex I and decreased expression of Sod2 most likely increase the production of reactive oxygen species and the expression of the pre-apoptotic proteins Bcl-2 and Bax in this tissue. In conclusion, we provide evidence that a disrupted mitochondrial network and the resulting oxidative stress in the hippocampus contribute to the altered learning and cognitive impairment in Foxp1+/- mice, suggesting that similar alterations also play a major role in patients with FOXP1 syndrome.
Collapse
Affiliation(s)
- Jing Wang
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University Hospital, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany; (J.W.); (G.A.R.)
| | - Gudrun A. Rappold
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University Hospital, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany; (J.W.); (G.A.R.)
- Interdisciplinary Center for Neurosciences, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Henning Fröhlich
- Department of Human Molecular Genetics, Institute of Human Genetics, Heidelberg University Hospital, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany; (J.W.); (G.A.R.)
| |
Collapse
|
39
|
Duman JG, Blanco FA, Cronkite CA, Ru Q, Erikson KC, Mulherkar S, Saifullah AB, Firozi K, Tolias KF. Rac-maninoff and Rho-vel: The symphony of Rho-GTPase signaling at excitatory synapses. Small GTPases 2022; 13:14-47. [PMID: 33955328 PMCID: PMC9707551 DOI: 10.1080/21541248.2021.1885264] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/22/2021] [Accepted: 01/28/2021] [Indexed: 01/15/2023] Open
Abstract
Synaptic connections between neurons are essential for every facet of human cognition and are thus regulated with extreme precision. Rho-family GTPases, molecular switches that cycle between an active GTP-bound state and an inactive GDP-bound state, comprise a critical feature of synaptic regulation. Rho-GTPases are exquisitely controlled by an extensive suite of activators (GEFs) and inhibitors (GAPs and GDIs) and interact with many different signalling pathways to fulfill their roles in orchestrating the development, maintenance, and plasticity of excitatory synapses of the central nervous system. Among the mechanisms that control Rho-GTPase activity and signalling are cell surface receptors, GEF/GAP complexes that tightly regulate single Rho-GTPase dynamics, GEF/GAP and GEF/GEF functional complexes that coordinate multiple Rho-family GTPase activities, effector positive feedback loops, and mutual antagonism of opposing Rho-GTPase pathways. These complex regulatory mechanisms are employed by the cells of the nervous system in almost every step of development, and prominently figure into the processes of synaptic plasticity that underlie learning and memory. Finally, misregulation of Rho-GTPases plays critical roles in responses to neuronal injury, such as traumatic brain injury and neuropathic pain, and in neurodevelopmental and neurodegenerative disorders, including intellectual disability, autism spectrum disorder, schizophrenia, and Alzheimer's Disease. Thus, decoding the mechanisms of Rho-GTPase regulation and function at excitatory synapses has great potential for combatting many of the biggest current challenges in mental health.
Collapse
Affiliation(s)
- Joseph G. Duman
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Francisco A. Blanco
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Integrative Molecular and Biomedical Science Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Christopher A. Cronkite
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | - Qin Ru
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Kelly C. Erikson
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Shalaka Mulherkar
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Ali Bin Saifullah
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Karen Firozi
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Kimberley F. Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
40
|
Bruno LP, Doddato G, Valentino F, Baldassarri M, Tita R, Fallerini C, Bruttini M, Lo Rizzo C, Mencarelli MA, Mari F, Pinto AM, Fava F, Fabbiani A, Lamacchia V, Carrer A, Caputo V, Granata S, Benetti E, Zguro K, Furini S, Renieri A, Ariani F. New Candidates for Autism/Intellectual Disability Identified by Whole-Exome Sequencing. Int J Mol Sci 2021; 22:ijms222413439. [PMID: 34948243 PMCID: PMC8707363 DOI: 10.3390/ijms222413439] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 12/27/2022] Open
Abstract
Intellectual disability (ID) is characterized by impairments in the cognitive processes and in the tasks of daily life. It encompasses a clinically and genetically heterogeneous group of neurodevelopmental disorders often associated with autism spectrum disorder (ASD). Social and communication abilities are strongly compromised in ASD. The prevalence of ID/ASD is 1–3%, and approximately 30% of the patients remain without a molecular diagnosis. Considering the extreme genetic locus heterogeneity, next-generation sequencing approaches have provided powerful tools for candidate gene identification. Molecular diagnosis is crucial to improve outcome, prevent complications, and hopefully start a therapeutic approach. Here, we performed parent–offspring trio whole-exome sequencing (WES) in a cohort of 60 mostly syndromic ID/ASD patients and we detected 8 pathogenic variants in genes already known to be associated with ID/ASD (SYNGAP1, SMAD6, PACS1, SHANK3, KMT2A, KCNQ2, ACTB, and POGZ). We found four de novo disruptive variants of four novel candidate ASD/ID genes: MBP, PCDHA1, PCDH15, PDPR. We additionally selected via bioinformatic tools many variants in unknown genes that alone or in combination can contribute to the phenotype. In conclusion, our data confirm the efficacy of WES in detecting pathogenic variants of known and novel ID/ASD genes.
Collapse
Affiliation(s)
- Lucia Pia Bruno
- Medical Genetics, University of Siena, 53100 Siena, Italy; (L.P.B.); (G.D.); (F.V.); (M.B.); (C.F.); (M.B.); (F.M.); (F.F.); (A.F.); (V.L.); (A.C.); (V.C.); (S.G.); (A.R.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Gabriella Doddato
- Medical Genetics, University of Siena, 53100 Siena, Italy; (L.P.B.); (G.D.); (F.V.); (M.B.); (C.F.); (M.B.); (F.M.); (F.F.); (A.F.); (V.L.); (A.C.); (V.C.); (S.G.); (A.R.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Floriana Valentino
- Medical Genetics, University of Siena, 53100 Siena, Italy; (L.P.B.); (G.D.); (F.V.); (M.B.); (C.F.); (M.B.); (F.M.); (F.F.); (A.F.); (V.L.); (A.C.); (V.C.); (S.G.); (A.R.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Margherita Baldassarri
- Medical Genetics, University of Siena, 53100 Siena, Italy; (L.P.B.); (G.D.); (F.V.); (M.B.); (C.F.); (M.B.); (F.M.); (F.F.); (A.F.); (V.L.); (A.C.); (V.C.); (S.G.); (A.R.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Rossella Tita
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy; (R.T.); (C.L.R.); (M.A.M.); (A.M.P.)
| | - Chiara Fallerini
- Medical Genetics, University of Siena, 53100 Siena, Italy; (L.P.B.); (G.D.); (F.V.); (M.B.); (C.F.); (M.B.); (F.M.); (F.F.); (A.F.); (V.L.); (A.C.); (V.C.); (S.G.); (A.R.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Mirella Bruttini
- Medical Genetics, University of Siena, 53100 Siena, Italy; (L.P.B.); (G.D.); (F.V.); (M.B.); (C.F.); (M.B.); (F.M.); (F.F.); (A.F.); (V.L.); (A.C.); (V.C.); (S.G.); (A.R.)
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy; (R.T.); (C.L.R.); (M.A.M.); (A.M.P.)
| | - Caterina Lo Rizzo
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy; (R.T.); (C.L.R.); (M.A.M.); (A.M.P.)
| | - Maria Antonietta Mencarelli
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy; (R.T.); (C.L.R.); (M.A.M.); (A.M.P.)
| | - Francesca Mari
- Medical Genetics, University of Siena, 53100 Siena, Italy; (L.P.B.); (G.D.); (F.V.); (M.B.); (C.F.); (M.B.); (F.M.); (F.F.); (A.F.); (V.L.); (A.C.); (V.C.); (S.G.); (A.R.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy; (R.T.); (C.L.R.); (M.A.M.); (A.M.P.)
| | - Anna Maria Pinto
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy; (R.T.); (C.L.R.); (M.A.M.); (A.M.P.)
| | - Francesca Fava
- Medical Genetics, University of Siena, 53100 Siena, Italy; (L.P.B.); (G.D.); (F.V.); (M.B.); (C.F.); (M.B.); (F.M.); (F.F.); (A.F.); (V.L.); (A.C.); (V.C.); (S.G.); (A.R.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy; (R.T.); (C.L.R.); (M.A.M.); (A.M.P.)
| | - Alessandra Fabbiani
- Medical Genetics, University of Siena, 53100 Siena, Italy; (L.P.B.); (G.D.); (F.V.); (M.B.); (C.F.); (M.B.); (F.M.); (F.F.); (A.F.); (V.L.); (A.C.); (V.C.); (S.G.); (A.R.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy; (R.T.); (C.L.R.); (M.A.M.); (A.M.P.)
| | - Vittoria Lamacchia
- Medical Genetics, University of Siena, 53100 Siena, Italy; (L.P.B.); (G.D.); (F.V.); (M.B.); (C.F.); (M.B.); (F.M.); (F.F.); (A.F.); (V.L.); (A.C.); (V.C.); (S.G.); (A.R.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy; (R.T.); (C.L.R.); (M.A.M.); (A.M.P.)
| | - Anna Carrer
- Medical Genetics, University of Siena, 53100 Siena, Italy; (L.P.B.); (G.D.); (F.V.); (M.B.); (C.F.); (M.B.); (F.M.); (F.F.); (A.F.); (V.L.); (A.C.); (V.C.); (S.G.); (A.R.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy; (R.T.); (C.L.R.); (M.A.M.); (A.M.P.)
| | - Valentina Caputo
- Medical Genetics, University of Siena, 53100 Siena, Italy; (L.P.B.); (G.D.); (F.V.); (M.B.); (C.F.); (M.B.); (F.M.); (F.F.); (A.F.); (V.L.); (A.C.); (V.C.); (S.G.); (A.R.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy; (R.T.); (C.L.R.); (M.A.M.); (A.M.P.)
| | - Stefania Granata
- Medical Genetics, University of Siena, 53100 Siena, Italy; (L.P.B.); (G.D.); (F.V.); (M.B.); (C.F.); (M.B.); (F.M.); (F.F.); (A.F.); (V.L.); (A.C.); (V.C.); (S.G.); (A.R.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy; (R.T.); (C.L.R.); (M.A.M.); (A.M.P.)
| | - Elisa Benetti
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Kristina Zguro
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Simone Furini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
| | - Alessandra Renieri
- Medical Genetics, University of Siena, 53100 Siena, Italy; (L.P.B.); (G.D.); (F.V.); (M.B.); (C.F.); (M.B.); (F.M.); (F.F.); (A.F.); (V.L.); (A.C.); (V.C.); (S.G.); (A.R.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy; (R.T.); (C.L.R.); (M.A.M.); (A.M.P.)
| | - Francesca Ariani
- Medical Genetics, University of Siena, 53100 Siena, Italy; (L.P.B.); (G.D.); (F.V.); (M.B.); (C.F.); (M.B.); (F.M.); (F.F.); (A.F.); (V.L.); (A.C.); (V.C.); (S.G.); (A.R.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (E.B.); (K.Z.); (S.F.)
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy; (R.T.); (C.L.R.); (M.A.M.); (A.M.P.)
- Correspondence: ; Tel.: +39-0577-233303
| |
Collapse
|
41
|
Baptist J, Shetty P, Singh R, Achappa B. Problems in managing a co-morbid patient with no previous medical records undergoing alternative therapy. Med Leg J 2021; 89:250-253. [PMID: 34590897 DOI: 10.1177/00258172211038095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Individuals with intellectual disability are often uncooperative for complete dental evaluation and treatment. Many of these patients fall within autistic spectrum disease. These patients are also said to be associated with other medically relevant disorders such as seizures, and metabolic and hormonal dysfunction. Undertaking treatment of such patients under general anaesthesia will require complete medical evaluation. As many of the antiepileptic drugs interact with anaesthetic agents, a sound knowledge of drugs taken by the patient is necessary. However, when the patient is under alternative therapy, this problem becomes multi-fold. This paper will discuss the basic, but real problems with the gap of understanding between allopathic and alternative therapy, the importance of reviewing the patients', previous medical records, and its medico-legal consequences. It will also raise the issue of delays in management with increased cost and time of hospitalisation in such patients.
Collapse
Affiliation(s)
- Joanna Baptist
- Department of Oral and Maxillofacial Surgery, Manipal College of Dental Sciences, Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - Premalatha Shetty
- Department of Oral and Maxillofacial Surgery, Manipal College of Dental Sciences, Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - Rica Singh
- Department of Oral and Maxillofacial Surgery, Manipal College of Dental Sciences, Mangalore, Manipal Academy of Higher Education, Manipal, India
| | - Basavaprabhu Achappa
- Department of General Medicine, Kasturba Medical College, Mangalore Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
42
|
Nikam V, Mohammad NS. Tissue-specific DNase I footprint analysis confirms the association of GATAD2B Q470* variant with intellectual disability. J Genet 2021. [DOI: 10.1007/s12041-021-01308-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
43
|
Heyman M, Ledoux Galligan M, Salinas GB, Baker E, Blacher J, Stavropoulos K. Differential diagnosis of autism spectrum disorder, intellectual disability and attention-deficit hyperactivity disorder (ADHD). ADVANCES IN AUTISM 2021. [DOI: 10.1108/aia-01-2021-0002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Purpose
Professionals working with community populations are often presented with complicated cases where it is difficult to determine which diagnosis or diagnoses are appropriate. Differentiating among neurodevelopmental disorders such as autism spectrum disorder (ASD), attention-deficit/hyperactivity disorder and intellectual disability can be a complex process, especially, as these disorders have some overlapping symptoms and often co-occur in young children. This series of case studies aims to present commonly overlapping symptoms in children who present to clinics with developmental concerns.
Design/methodology/approach
This paper presents three case studies that were completed at a free community ASD screening clinic in Southern California.
Findings
The case studies have common presenting behaviors and symptoms (e.g. social communication difficulties) that often co-occur across diagnoses; explanations for the final diagnoses are given in each case.
Research limitations/implications
Conclusions from these three cases cannot generalize to all children being seen in clinics for neurodevelopmental concerns.
Practical implications
This series of case studies highlights commonly overlapping symptoms in children who present for differential diagnosis with social and/or behavioral concerns. Implications for educational placement and intervention are discussed.
Social implications
These cases highlight the challenges involved in the differential and dual diagnostic process for young children with developmental concerns. Diagnostic considerations can affect later educational placement and opportunities for socialization.
Originality/value
This series of case studies provide practical information for clinicians about how to effectively differentiate between commonly occurring neurodevelopmental disorders, particularly given recent changes to the Diagnostic and Statistical Manual, 5th edition (DSM-5).
Collapse
|
44
|
Piergiorge RM, de Vasconcelos ATR, Gonçalves Pimentel MM, Santos-Rebouças CB. Strict network analysis of evolutionary conserved and brain-expressed genes reveals new putative candidates implicated in Intellectual Disability and in Global Development Delay. World J Biol Psychiatry 2021; 22:435-445. [PMID: 32914658 DOI: 10.1080/15622975.2020.1821916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVES Intellectual Disability (ID) and Global Development Delay (GDD) are frequent reasons for referral to genetic services and although they present overlapping phenotypes concerning cognitive, motor, language, or social skills, they are not exactly synonymous. Aiming to better understand independent or shared mechanisms related to these conditions and to identify new candidate genes, we performed a highly stringent protein-protein interaction network based on genes previously related to ID/GDD in the Human Phenotype Ontology portal. METHODS ID/GDD genes were searched for reliable interactions through STRING and clustering analysis was applied to detect biological complexes through the MCL algorithm. Six coding hub genes (TP53, CDC42, RAC1, GNB1, APP, and EP300) were recognised by the Cytoscape NetworkAnalyzer plugin, interacting with 1625 proteins not yet associated with ID or GDD. Genes encoding these proteins were explored by gene ontology, associated diseases, evolutionary conservation, and brain expression. RESULTS One hundred and seventy-two new putative genes playing a role in enriched processes/pathways previously related to ID and GDD were revealed, some of which were already postulated to be linked to ID/GDD in additional databases. CONCLUSIONS Our findings expanded the aetiological genetic landscape of ID/GDD and showed evidence that both conditions are closely related at the molecular and functional levels.
Collapse
Affiliation(s)
- Rafael Mina Piergiorge
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Márcia Mattos Gonçalves Pimentel
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cíntia Barros Santos-Rebouças
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
45
|
El Khouri E, Ghoumid J, Haye D, Giuliano F, Drevillon L, Briand-Suleau A, De La Grange P, Nau V, Gaillon T, Bienvenu T, Jacquemin-Sablon H, Goossens M, Amselem S, Giurgea I. Wnt/β-catenin pathway and cell adhesion deregulation in CSDE1-related intellectual disability and autism spectrum disorders. Mol Psychiatry 2021; 26:3572-3585. [PMID: 33867523 DOI: 10.1038/s41380-021-01072-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 03/08/2021] [Accepted: 03/19/2021] [Indexed: 12/26/2022]
Abstract
Among the genetic factors playing a key role in the etiology of intellectual disabilities (IDs) and autism spectrum disorders (ASDs), several encode RNA-binding proteins (RBPs). In this study, we deciphered the molecular and cellular bases of ID-ASD in a patient followed from birth to the age of 21, in whom we identified a de novo CSDE1 (Cold Shock Domain-containing E1) nonsense variation. CSDE1 encodes an RBP that regulates multiple cellular pathways by monitoring the translation and abundance of target transcripts. Analyses performed on the patient's primary fibroblasts showed that the identified CSDE1 variation leads to haploinsufficiency. We identified through RNA-seq assays the Wnt/β-catenin signaling and cellular adhesion as two major deregulated pathways. These results were further confirmed by functional studies involving Wnt-specific luciferase and substrate adhesion assays. Additional data support a disease model involving APC Down-Regulated-1 (APCDD1) and cadherin-2 (CDH2), two components of the Wnt/β-catenin pathway, CDH2 being also pivotal for cellular adhesion. Our study, which relies on both the deep phenotyping and long-term follow-up of a patient with CSDE1 haploinsufficiency and on ex vivo studies, sheds new light on the CSDE1-dependent deregulated pathways in ID-ASD.
Collapse
Affiliation(s)
- E El Khouri
- Sorbonne Université, INSERM, Maladies génétiques d'expression pédiatrique, Département de Génétique médicale, Assistance Publique Hôpitaux de Paris, Hôpital Trousseau, Paris, France
| | - J Ghoumid
- Département de Génétique, Groupe Hospitalier Henri Mondor, Créteil, France.,Service de Génétique Clinique, Hôpital Jeanne de Flandre, CHU Lille, Lille, France
| | - D Haye
- Service de Génétique Médicale Centre, Hospitalo-Universitaire de Nice, Nice, France
| | - F Giuliano
- Service de Génétique Médicale Centre, Hospitalo-Universitaire de Nice, Nice, France
| | - L Drevillon
- Département de Génétique, Groupe Hospitalier Henri Mondor, Créteil, France.,CHU Caen Normandie, Caen, France
| | - A Briand-Suleau
- Département de Génétique, Groupe Hospitalier Henri Mondor, Créteil, France.,Service de Génétique et Biologie Moléculaires, Hôpital Cochin, INSERM UMR1266 - Institute of Psychiatry and Neuroscience of Paris (IPNP) and University of Paris, Paris, France
| | | | - V Nau
- Sorbonne Université, INSERM, Maladies génétiques d'expression pédiatrique, Département de Génétique médicale, Assistance Publique Hôpitaux de Paris, Hôpital Trousseau, Paris, France
| | - T Gaillon
- Département de Génétique, Groupe Hospitalier Henri Mondor, Créteil, France
| | - T Bienvenu
- Service de Génétique et Biologie Moléculaires, Hôpital Cochin, INSERM UMR1266 - Institute of Psychiatry and Neuroscience of Paris (IPNP) and University of Paris, Paris, France
| | - H Jacquemin-Sablon
- INSERM UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Bordeaux, France
| | - M Goossens
- Département de Génétique, Groupe Hospitalier Henri Mondor, Créteil, France
| | - S Amselem
- Sorbonne Université, INSERM, Maladies génétiques d'expression pédiatrique, Département de Génétique médicale, Assistance Publique Hôpitaux de Paris, Hôpital Trousseau, Paris, France
| | - I Giurgea
- Sorbonne Université, INSERM, Maladies génétiques d'expression pédiatrique, Département de Génétique médicale, Assistance Publique Hôpitaux de Paris, Hôpital Trousseau, Paris, France. .,Département de Génétique, Groupe Hospitalier Henri Mondor, Créteil, France.
| |
Collapse
|
46
|
Brief Report: An Exploration of Cognitive Flexibility of Autistic Adolescents with Low Intelligence Using the Wisconsin Card Sorting Task. J Autism Dev Disord 2021; 53:1726-1732. [PMID: 34114127 DOI: 10.1007/s10803-021-05134-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2021] [Indexed: 10/21/2022]
Abstract
Cognitive flexibility (CF) is the ability to shift between concepts or rules. Difficulty with CF is associated with autism (i.e., ASD) as it contributes to repetitive behaviours. However, little is known about CF skills of autistic adolescents with low intelligence. This study uses the Wisconsin Card Sorting Task (WCST) to assess the CF of 36 adolescents, all with a Weschler full-scale IQ between 50 and 85, 14 of whom had an ASD diagnosis. The results indicated no statistically significant differences in WCST performance between those with and without ASD. It was also found that performance IQ significantly contributed to the WCST performance in the ASD group only, suggesting an autism-specific role of non-verbal cognitive functioning in CF.
Collapse
|
47
|
Liaci C, Camera M, Caslini G, Rando S, Contino S, Romano V, Merlo GR. Neuronal Cytoskeleton in Intellectual Disability: From Systems Biology and Modeling to Therapeutic Opportunities. Int J Mol Sci 2021; 22:ijms22116167. [PMID: 34200511 PMCID: PMC8201358 DOI: 10.3390/ijms22116167] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/25/2021] [Accepted: 06/04/2021] [Indexed: 02/06/2023] Open
Abstract
Intellectual disability (ID) is a pathological condition characterized by limited intellectual functioning and adaptive behaviors. It affects 1–3% of the worldwide population, and no pharmacological therapies are currently available. More than 1000 genes have been found mutated in ID patients pointing out that, despite the common phenotype, the genetic bases are highly heterogeneous and apparently unrelated. Bibliomic analysis reveals that ID genes converge onto a few biological modules, including cytoskeleton dynamics, whose regulation depends on Rho GTPases transduction. Genetic variants exert their effects at different levels in a hierarchical arrangement, starting from the molecular level and moving toward higher levels of organization, i.e., cell compartment and functions, circuits, cognition, and behavior. Thus, cytoskeleton alterations that have an impact on cell processes such as neuronal migration, neuritogenesis, and synaptic plasticity rebound on the overall establishment of an effective network and consequently on the cognitive phenotype. Systems biology (SB) approaches are more focused on the overall interconnected network rather than on individual genes, thus encouraging the design of therapies that aim to correct common dysregulated biological processes. This review summarizes current knowledge about cytoskeleton control in neurons and its relevance for the ID pathogenesis, exploiting in silico modeling and translating the implications of those findings into biomedical research.
Collapse
Affiliation(s)
- Carla Liaci
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
| | - Mattia Camera
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
| | - Giovanni Caslini
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
| | - Simona Rando
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
| | - Salvatore Contino
- Department of Engineering, University of Palermo, Viale delle Scienze Ed. 8, 90128 Palermo, Italy;
| | - Valentino Romano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze Ed. 16, 90128 Palermo, Italy;
| | - Giorgio R. Merlo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
- Correspondence: ; Tel.: +39-0116706449; Fax: +39-0116706432
| |
Collapse
|
48
|
Vermehren-Schmaedick A, Huang JY, Levinson M, Pomaville MB, Reed S, Bellus GA, Gilbert F, Keren B, Heron D, Haye D, Janello C, Makowski C, Danhauser K, Fedorov LM, Haack TB, Wright KM, Cohen MS. Characterization of PARP6 Function in Knockout Mice and Patients with Developmental Delay. Cells 2021; 10:1289. [PMID: 34067418 PMCID: PMC8224619 DOI: 10.3390/cells10061289] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/01/2021] [Accepted: 05/04/2021] [Indexed: 01/08/2023] Open
Abstract
PARP6, a member of a family of enzymes (17 in humans) known as poly-ADP-ribose polymerases (PARPs), is a neuronally enriched PARP. While previous studies from our group show that Parp6 is a regulator of dendrite morphogenesis in rat hippocampal neurons, its function in the nervous system in vivo is poorly understood. Here, we describe the generation of a Parp6 loss-of-function mouse model for examining the function of Parp6 during neurodevelopment in vivo. Using CRISPR-Cas9 mutagenesis, we generated a mouse line that expressed a Parp6 truncated variant (Parp6TR) in place of Parp6WT. Unlike Parp6WT, Parp6TR is devoid of catalytic activity. Homozygous Parp6TR do not exhibit obvious neuromorphological defects during development, but nevertheless die perinatally. This suggests that Parp6 catalytic activity is important for postnatal survival. We also report PARP6 mutations in six patients with several neurodevelopmental disorders, including microencephaly, intellectual disabilities, and epilepsy. The most severe mutation in PARP6 (C563R) results in the loss of catalytic activity. Expression of Parp6C563R in hippocampal neurons decreases dendrite morphogenesis. To gain further insight into PARP6 function in neurons we also performed a BioID proximity labeling experiment in hippocampal neurons and identified several microtubule-binding proteins (e.g., MAP-2) using proteomics. Taken together, our results suggest that PARP6 is an essential microtubule-regulatory gene in mice, and that the loss of PARP6 catalytic activity has detrimental effects on neuronal function in humans.
Collapse
Affiliation(s)
- Anke Vermehren-Schmaedick
- Hospital & Specialty Medicine, VA Portland Health Care System, Portland, OR 97239, USA;
- Translational Oncology Program, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97210, USA
| | - Jeffrey Y. Huang
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97210, USA; (J.Y.H.); (M.L.)
| | - Madison Levinson
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97210, USA; (J.Y.H.); (M.L.)
| | - Matthew B. Pomaville
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA; (M.B.P.); (K.M.W.)
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Sarah Reed
- Clinical Genetics, Geisinger Medical Center, Danville, PA 17822, USA; (S.R.); (G.A.B.)
| | - Gary A. Bellus
- Clinical Genetics, Geisinger Medical Center, Danville, PA 17822, USA; (S.R.); (G.A.B.)
| | - Fred Gilbert
- Department of Pediatrics, Brooklyn Hospital, New York, NY 11201, USA;
| | - Boris Keren
- Department of Genetics, Pitié-Salpêtrière Hospital, 75013 Paris, France; (B.K.); (D.H.)
| | - Delphine Heron
- Department of Genetics, Pitié-Salpêtrière Hospital, 75013 Paris, France; (B.K.); (D.H.)
| | - Damien Haye
- Department of Genetics, Hospices Civils de Lyon, 69002 Lyon, France;
| | - Christine Janello
- Department of Pediatrics, Technical University of Munich, 80804 Munich, Germany; (C.J.); (C.M.)
| | - Christine Makowski
- Department of Pediatrics, Technical University of Munich, 80804 Munich, Germany; (C.J.); (C.M.)
| | - Katharina Danhauser
- Institute of Human Genetics, Technical University of Munich, 81675 Munich, Germany; (K.D.); (T.B.H.)
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, University Hospital, Ludwig Maximilian University of Munich, 80337 Munich, Germany
| | - Lev M. Fedorov
- Transgenic Mouse Models Shared Resource, Oregon Health & Science University, Portland, OR 97239, USA;
| | - Tobias B. Haack
- Institute of Human Genetics, Technical University of Munich, 81675 Munich, Germany; (K.D.); (T.B.H.)
- Institute of Medical Genetics and Applied Genomics, 72076 Tuebingen, Germany
| | - Kevin M. Wright
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA; (M.B.P.); (K.M.W.)
| | - Michael S. Cohen
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97210, USA; (J.Y.H.); (M.L.)
| |
Collapse
|
49
|
Li H, Lou R, Xu X, Xu C, Yu Y, Xu Y, Hu L, Xiang Y, Lin X, Tang S. The variations in human orphan G protein-coupled receptor QRFPR affect PI3K-AKT-mTOR signaling. J Clin Lab Anal 2021; 35:e23822. [PMID: 34018631 DOI: 10.1002/jcla.23822] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND QRFPR is a recently identified member of the G protein-coupled receptor and is an orphan receptor for 26Rfa, which plays important role in the regulation of many physiological functions. METHODS Here, we employed whole exome sequencing (WES) to examine the patients with intellectual disability (ID) and difficulty in feeding. We performed SIFT and PolyPhen2 predictions for the variants. The structure model was built from scratch by I-TASSER. Here, results derived from a number of cell-based functional assays, including shRNA experiment, intracellular Ca2+ measurement, the expression of PI3 K-AKT-mTOR, and phosphorylation. The functional effect of QRFPR variants on PI3K-AKT-mTOR signaling was evaluated in vitro transfection experiments. RESULT Here, we identified two QRFPR variants at c.202 T>C (p.Y68H) and c.1111C>T (p.R371W) in 2 unrelated individuals. Structural analysis revealed that p.Y68H and p.R371W variants may affect the side chain structure of adjacent amino acids causing reduced binding of QRFPR to 26Rfa. The results show that QRFPR stimulated by 26Rfa leading to the transient rise of intracellular Ca2+ . The QRFPR variations p.Y68H and p.R371 W can reduce the mobilization of intracellular Ca2+ . The phosphorylation levels of the PI3K, Akt, and mTOR were significantly up- or downregulated by QRFPR overexpression or silencing, respectively. The QRFPR variations inhibited PI3K-AKT-mTOR signaling, resulting in downregulation of p-mTOR. CONCLUSIONS Our findings suggest that QRFPR acts as important role in neurodevelopment, and the effects of QRFPR are likely to be mediated by the Ca2+ -dependent PI3K-AKT-mTOR pathways. Importantly, these findings provide a foundation for future elucidation of GPCR-mediated signaling and the physiological implications.
Collapse
Affiliation(s)
- Huanzheng Li
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China.,Human Aging Research Institute, Nanchang University, Nanchang, China
| | - Ran Lou
- Department of Acupuncture, Wenzhou Central Hospital, Wenzhou, China
| | - Xueqin Xu
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Chenyang Xu
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Yuan Yu
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Yunzhi Xu
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Lin Hu
- Department of Blood Transfusion, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yanbao Xiang
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Xuan Lin
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Shaohua Tang
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
50
|
Upadhyay J, Patra J, Tiwari N, Salankar N, Ansari MN, Ahmad W. Dysregulation of Multiple Signaling Neurodevelopmental Pathways during Embryogenesis: A Possible Cause of Autism Spectrum Disorder. Cells 2021; 10:958. [PMID: 33924211 PMCID: PMC8074600 DOI: 10.3390/cells10040958] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 04/17/2021] [Accepted: 04/19/2021] [Indexed: 12/24/2022] Open
Abstract
Understanding the autistic brain and the involvement of genetic, non-genetic, and numerous signaling pathways in the etiology and pathophysiology of autism spectrum disorder (ASD) is complex, as is evident from various studies. Apart from multiple developmental disorders of the brain, autistic subjects show a few characteristics like impairment in social communications related to repetitive, restricted, or stereotypical behavior, which suggests alterations in neuronal circuits caused by defects in various signaling pathways during embryogenesis. Most of the research studies on ASD subjects and genetic models revealed the involvement of mutated genes with alterations of numerous signaling pathways like Wnt, hedgehog, and Retinoic Acid (RA). Despite significant improvement in understanding the pathogenesis and etiology of ASD, there is an increasing awareness related to it as well as a need for more in-depth research because no effective therapy has been developed to address ASD symptoms. Therefore, identifying better therapeutic interventions like "novel drugs for ASD" and biomarkers for early detection and disease condition determination are required. This review article investigated various etiological factors as well as the signaling mechanisms and their alterations to understand ASD pathophysiology. It summarizes the mechanism of signaling pathways, their significance, and implications for ASD.
Collapse
Affiliation(s)
- Jyoti Upadhyay
- Department of Pharmaceutical Sciences, School of Health Sciences, University of Petroleum and Energy Studies, Energy Acre Campus Bidholi, Dehradun 248007, Uttarakhand, India; (J.U.); (J.P.)
| | - Jeevan Patra
- Department of Pharmaceutical Sciences, School of Health Sciences, University of Petroleum and Energy Studies, Energy Acre Campus Bidholi, Dehradun 248007, Uttarakhand, India; (J.U.); (J.P.)
| | - Nidhi Tiwari
- Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organisation, Delhi 110054, India;
| | - Nilima Salankar
- School of Computer Sciences, University of Petroleum and Energy Studies, Energy Acre Campus Bidholi, Dehradun 248007, Uttarakhand, India;
| | - Mohd Nazam Ansari
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Wasim Ahmad
- Department of Pharmacy, Mohammed Al-Mana College for Medical Sciences, Dammam 34222, Saudi Arabia;
| |
Collapse
|