1
|
Siqueiros-Sanchez M, Serur Y, McGhee CA, Smith TF, Green T. Social Communication in Ras Pathway Disorders: A Comprehensive Review From Genetics to Behavior in Neurofibromatosis Type 1 and Noonan Syndrome. Biol Psychiatry 2025; 97:461-498. [PMID: 39366539 DOI: 10.1016/j.biopsych.2024.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 09/10/2024] [Accepted: 09/22/2024] [Indexed: 10/06/2024]
Abstract
Neurofibromatosis type 1 (NF1) and Noonan syndrome (NS) are neurogenetic syndromes caused by pathogenetic variants encoding components of the Ras-ERK-MAPK (Ras/extracellular signal-regulated kinase/mitogen-activated protein kinase) signaling pathway (Ras pathway). NF1 and NS are associated with differences in social communication and related neuropsychiatric risks. During the last decade, there has been growing interest in Ras-linked syndromes as models to understand social communication deficits and autism spectrum disorder. We systematically review the literature between 2010 and 2023 focusing on the social communication construct of the Research Domain Criteria framework. We provide an integrative summary of the research on facial and nonfacial social communication processes in NF1 and NS across molecular, cellular, neural circuitry, and behavioral domains. At the molecular and cellular levels, dysregulation in the Ras pathway is intricately tied to variations in social communication through changes in GABAergic (gamma-aminobutyric acidergic), glutamatergic, and serotonergic transmission, as well as inhibitory/excitatory imbalance. Neural circuitry typically associated with learning, attention, and memory in NF1 and NS (e.g., corticostriatal connectivity) is also implicated in social communication. We highlight less-researched potential mechanisms for social communication, such as white matter connectivity and the default mode network. Finally, key gaps in NF1 and NS literature are identified, and a roadmap for future research is provided. By leveraging genetic syndrome research, we can understand the mechanisms associated with behaviors and psychiatric disorders.
Collapse
Affiliation(s)
- Monica Siqueiros-Sanchez
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California; Division of Interdisciplinary Brain Sciences, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California.
| | - Yaffa Serur
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California; Division of Interdisciplinary Brain Sciences, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California
| | - Chloe A McGhee
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California; Division of Interdisciplinary Brain Sciences, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California
| | - Taylor F Smith
- Department of Psychology and Child Development, California Polytechnic State University, San Luis Obispo, California
| | - Tamar Green
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California; Division of Interdisciplinary Brain Sciences, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
2
|
Cole JJ, Ferner RE, Gutmann DH. Neurofibromatosis type 1. ROSENBERG'S MOLECULAR AND GENETIC BASIS OF NEUROLOGICAL AND PSYCHIATRIC DISEASE 2025:231-249. [DOI: 10.1016/b978-0-443-19176-3.00017-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
3
|
Hou Y, Yu L, Liu D, Wilson-Lemoine E, Wu X, Moreira JP, Mujica BF, Mukhopadhyay ES, Novotney AN, Payne JM. Systematic Review and Meta-Analysis: Attention-Deficit/Hyperactivity Disorder Symptoms in Children With Neurofibromatosis Type 1. J Am Acad Child Adolesc Psychiatry 2024:S0890-8567(24)02059-8. [PMID: 39709008 DOI: 10.1016/j.jaac.2024.09.011] [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: 03/29/2024] [Revised: 08/30/2024] [Accepted: 12/12/2024] [Indexed: 12/23/2024]
Abstract
OBJECTIVE This meta-analysis aimed to robustly estimate differences in attention-deficit/hyperactivity disorder (ADHD) symptoms between children and adolescents with and without neurofibromatosis type 1 (NF1). METHOD Systematic literature searches were conducted in Scopus, PsycINFO, Web of Science, PubMed, and ProQuest in September 2022, with a supplemental search conducted in Google Scholar in February 2023. The searches identified 2,153 unique articles. Screening identified 114 academic journal articles that assessed parent/caregiver- or teacher-reported ADHD symptoms for children/adolescents with NF1. Two researchers independently screened articles and extracted data. The primary outcome was group differences in ADHD symptoms between children/adolescents with and without NF1 (Hedges g). Data were analyzed using robust variance estimation and random-effects models. RESULTS The meta-analysis included 70 studies (138 effect sizes), involving 3,653 children/adolescents with NF1 (46% female; mean age = 9.69 years, SD = 2.60 years) and 4,895 children/adolescents without NF1 (48% female; mean age = 10.03 years, SD = 3.10 years). According to parent/caregiver reports, children/adolescents with NF1 exhibited more severe inattentive symptoms (g = 1.20; 95% CI = 1.06-1.35), hyperactive/impulsive symptoms (g = 0.85; 95% CI = 0. 68-1.03), and combined ADHD symptoms (g = 1.02; 95% CI = 0.87-1.17) than unaffected controls. Inattentive ADHD symptoms were more elevated than hyperactivity/impulsivity for children/adolescents with NF1. Larger effect sizes for inattention and hyperactivity/impulsivity were associated with older age, lower intelligence quotient (IQ), and parent/caregiver vs teacher reports. CONCLUSION NF1 is a monogenic condition that has strong associations with elevated ADHD symptoms. Findings highlight the importance of early intervention and targeted support for ADHD-related problems in children with NF1. STUDY PREREGISTRATION INFORMATION Compare the ADHD problems between NF1 and control groups; https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=462063.
Collapse
Affiliation(s)
- Yang Hou
- Florida State University, Tallahassee, Florida, USA.
| | - Liyan Yu
- Florida State University, Tallahassee, Florida, USA
| | - Dan Liu
- Florida State University, Tallahassee, Florida, USA
| | | | - Xian Wu
- University of Kentucky, Lexington, Kentucky, USA
| | | | | | | | | | - Jonathan M Payne
- Murdoch Children's Research Institute, Australia and The University of Melbourne, Australia
| |
Collapse
|
4
|
Routier L, Querné L, Fontaine C, Berquin P, Le Moing AG. Distinct attentional and executive profiles in neurofibromatosis type 1: Is there difference with primary attention deficit-hyperactivity disorder? Eur J Paediatr Neurol 2024; 51:93-99. [PMID: 38905883 DOI: 10.1016/j.ejpn.2024.05.016] [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: 12/01/2023] [Revised: 04/03/2024] [Accepted: 05/31/2024] [Indexed: 06/23/2024]
Abstract
PURPOSE Attentional and executive dysfunctions are the most frequent cognitive disorders in neurofibromatosis type 1 (NF1), with a high prevalence of attention deficit-hyperactivity disorder (ADHD). We (i) compared attentional profiles between NF1 children with and without ADHD and children with primary ADHD criteria and (ii) investigated the possible relationship between attentional disorders and "unidentified bright objects" (UBOs) in NF1. METHODS This retrospective study included 47 NF1 children, 25 with ADHD criteria (NF1+adhd group), matched for age, sex, and cognitive level with 47 children with primary ADHD (ADHD group). We collected computer task (sustained-attention, visuomotor-decision, inhibition, and cognitive-flexibility tasks) scores normalized for age and sex, and brain magnetic resonance imaging data. RESULTS (i) Working memory was impaired in all groups. (ii) Omissions (p < 0.002) and response-time variability (p < 0.05) in sustained-attention and visuomotor-decision tasks and errors (p < 0.02) in the cognitive-flexibility task were lower for the NFI+adhd and ADHD groups than for the NF1-no-adhd group. (iii) The NF1+adhd group had slower response times (p ≤ 0.02) for inhibition and visuomotor-decision tasks than the other groups. (iv) We found no relevant association between cognitive performance and UBOs. CONCLUSIONS NF1 children with ADHD have an attentional and executive functions deficit profile similar to that of children with primary ADHD, but with a slower response-time, increasing learning difficulties. The atypical connectivity of fronto-striatal pathways, poorer dopamine homeostasis, and increased GABA inhibition observed in NF1 renders vulnerable the development of the widely distributed neural networks that support attentional, working-memory, and executive functions.
Collapse
Affiliation(s)
- Laura Routier
- Pediatric Neurology Department, Amiens-Picardie University Hospital, 1 rue du Professeur Christian Cabrol, 80054, Amiens Cedex, France; INSERM UMR 1105, Research Group on Multimodal Analysis of Brain Function, University of Picardie Jules Verne, rue René Laennec, 80054, Amiens, Cedex, France; Pediatric Neurophysiology Unit, Amiens-Picardie University Hospital, 1 rue du Professeur Christian Cabrol, 80054, Amiens Cedex, France.
| | - Laurent Querné
- Pediatric Neurology Department, Amiens-Picardie University Hospital, 1 rue du Professeur Christian Cabrol, 80054, Amiens Cedex, France; INSERM UMR 1105, Research Group on Multimodal Analysis of Brain Function, University of Picardie Jules Verne, rue René Laennec, 80054, Amiens, Cedex, France
| | - Cécile Fontaine
- Pediatric Neurology Department, Amiens-Picardie University Hospital, 1 rue du Professeur Christian Cabrol, 80054, Amiens Cedex, France
| | - Patrick Berquin
- Pediatric Neurology Department, Amiens-Picardie University Hospital, 1 rue du Professeur Christian Cabrol, 80054, Amiens Cedex, France; INSERM UMR 1105, Research Group on Multimodal Analysis of Brain Function, University of Picardie Jules Verne, rue René Laennec, 80054, Amiens, Cedex, France
| | - Anne-Gaëlle Le Moing
- Pediatric Neurology Department, Amiens-Picardie University Hospital, 1 rue du Professeur Christian Cabrol, 80054, Amiens Cedex, France; INSERM UMR 1105, Research Group on Multimodal Analysis of Brain Function, University of Picardie Jules Verne, rue René Laennec, 80054, Amiens, Cedex, France
| |
Collapse
|
5
|
Hocking DR, Sun X, Haebich K, Darke H, North KN, Vivanti G, Payne JM. Delineating Visual Habituation Profiles in Preschoolers with Neurofibromatosis Type 1 and Autism Spectrum Disorder: A Cross-Syndrome Study. J Autism Dev Disord 2024; 54:1998-2011. [PMID: 36877426 DOI: 10.1007/s10803-023-05913-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2023] [Indexed: 03/07/2023]
Abstract
Atypical habituation to repetitive information has been commonly reported in Autism Spectrum Disorder (ASD) but it is not yet clear whether similar abnormalities are present in Neurofibromatosis Type 1 (NF1). We employed a cross-syndrome design using a novel eye tracking paradigm to measure habituation in preschoolers with NF1, children with idiopathic ASD and typically developing (TD) children. Eye movements were recorded to examine fixation duration to simultaneously presented repeating and novel stimuli. Children with NF1 showed a bias for longer look durations to repeating stimuli at the expense of novel stimuli, and slower habituation in NF1 was associated with elevated ASD traits. These findings could indicate aberrant modulation of bottom-up attentional networks that interact with the emergence of ASD phenotypes.
Collapse
Affiliation(s)
- Darren R Hocking
- School of Psychology and Public Health, La Trobe University, Melbourne, Australia.
| | - Xiaoyun Sun
- School of Psychology and Public Health, La Trobe University, Melbourne, Australia
| | - Kristina Haebich
- Murdoch Children's Research Institute, Parkville, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Hayley Darke
- Murdoch Children's Research Institute, Parkville, Australia
| | - Kathryn N North
- Murdoch Children's Research Institute, Parkville, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Giacomo Vivanti
- A.J. Drexel Autism Institute, Drexel University, 3020 Market Street, Suite 560, 19104-3734, Philadelphia, PA, USA
| | - Jonathan M Payne
- Murdoch Children's Research Institute, Parkville, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
6
|
McCullough KB, Titus A, Reardon K, Conyers S, Dougherty JD, Ge X, Garbow JR, Dickson P, Yuede CM, Maloney SE. Characterization of early markers of disease in the mouse model of mucopolysaccharidosis IIIB. J Neurodev Disord 2024; 16:16. [PMID: 38632525 PMCID: PMC11022360 DOI: 10.1186/s11689-024-09534-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 04/01/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Mucopolysaccharidosis (MPS) IIIB, also known as Sanfilippo Syndrome B, is a devastating childhood disease. Unfortunately, there are currently no available treatments for MPS IIIB patients. Yet, animal models of lysosomal storage diseases have been valuable tools in identifying promising avenues of treatment. Enzyme replacement therapy, gene therapy, and bone marrow transplant have all shown efficacy in the MPS IIIB model systems. A ubiquitous finding across rodent models of lysosomal storage diseases is that the best treatment outcomes resulted from intervention prior to symptom onset. Therefore, the aim of the current study was to identify early markers of disease in the MPS IIIB mouse model as well as examine clinically-relevant behavioral domains not yet explored in this model. METHODS Using the MPS IIIB mouse model, we explored early developmental trajectories of communication and gait, and later social behavior, fear-related startle and conditioning, and visual capabilities. In addition, we examined brain structure and function via magnetic resonance imaging and diffusion tensor imaging. RESULTS We observed reduced maternal isolation-induced ultrasonic vocalizations in MPS IIIB mice relative to controls, as well as disruption in a number of the spectrotemporal features. MPS IIIB also exhibited disrupted thermoregulation during the first two postnatal weeks without any differences in body weight. The developmental trajectories of gait were largely normal. In early adulthood, we observed intact visual acuity and sociability yet a more submissive phenotype, increased aggressive behavior, and decreased social sniffing relative to controls. MPS IIIB mice showed greater inhibition of startle in response to a pretone with a decrease in overall startle response and reduced cued fear memory. MPS IIIB also weighed significantly more than controls throughout adulthood and showed larger whole brain volumes and normalized regional volumes with intact tissue integrity as measured with magnetic resonance and diffusion tensor imaging, respectively. CONCLUSIONS Together, these results indicate disease markers are present as early as the first two weeks postnatal in this model. Further, this model recapitulates social, sensory and fear-related clinical features. Our study using a mouse model of MPS IIIB provides essential baseline information that will be useful in future evaluations of potential treatments.
Collapse
Affiliation(s)
- Katherine B McCullough
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Amanda Titus
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Kate Reardon
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sara Conyers
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Joseph D Dougherty
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Xia Ge
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Joel R Garbow
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Patricia Dickson
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Carla M Yuede
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Susan E Maloney
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
7
|
Hernandez DP, Cruz DM, Martinez CS, Garcia LM, Figueroa A, Villarreal M, Manoj LM, Lopez S, López-Lorenzo KD, López-Juárez A. Gender-Specific Fine Motor Skill Learning Is Impaired by Myelin-Targeted Neurofibromatosis Type 1 Gene Mutation. Cancers (Basel) 2024; 16:477. [PMID: 38339230 PMCID: PMC10854893 DOI: 10.3390/cancers16030477] [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: 01/04/2024] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
Neurofibromatosis type 1 (NF1) is caused by mutations in the NF1 gene. The clinical presentation of NF1 includes diverse neurological issues in pediatric and adult patients, ranging from learning disabilities, motor skill issues, and attention deficit disorder, to increased risk of depression and dementia. Preclinical research suggests that abnormal neuronal signaling mediates spatial learning and attention issues in NF1; however, drugs that improve phenotypes in models show inconclusive results in clinical trials, highlighting the need for a better understanding of NF1 pathophysiology and broader therapeutic options. Most NF1 patients show abnormalities in their brain white matter (WM) and myelin, and links with NF1 neuropathophysiology have been suggested; however, no current data can clearly support or refute this idea. We reported that myelin-targeted Nf1 mutation impacts oligodendrocyte signaling, myelin ultrastructure, WM connectivity, and sensory-motor behaviors in mice; however, any impact on learning and memory remains unknown. Here, we adapted a voluntary running test-the complex wheel (CW; a wheel with unevenly spaced rungs)-to delineate fine motor skill learning curves following induction of an Nf1 mutation in pre-existing myelinating cells (pNf1 mice). We found that pNf1 mutant females experience delayed or impaired learning in the CW, while proper learning in pNf1 males is predominantly disrupted; these phenotypes add complexity to the gender-dependent learning differences in the mouse strain used. No broad differences in memory of acquired CW skills were detected in any gender, but gene-dose effects were observed at the studied time points. Finally, nitric oxide signaling regulation differentially impacted learning in wild type (WT)/pNf1, male/female mice. Our results provide evidence for fine motor skill learning issues upon induction of an Nf1 mutation in mature myelinating cells. Together with previous connectivity, cellular, and molecular analyses, these results diversify the potential treatments for neurological issues in NF1.
Collapse
Affiliation(s)
- Daniella P. Hernandez
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Daniela M. Cruz
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Celeste S. Martinez
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Larisa M. Garcia
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Ashley Figueroa
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Marisol Villarreal
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Liya M. Manoj
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Saul Lopez
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | | | - Alejandro López-Juárez
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| |
Collapse
|
8
|
de Blank P, Nishiyama A, López-Juárez A. A new era for myelin research in Neurofibromatosis type 1. Glia 2023; 71:2701-2719. [PMID: 37382486 PMCID: PMC10592420 DOI: 10.1002/glia.24432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 06/12/2023] [Accepted: 06/12/2023] [Indexed: 06/30/2023]
Abstract
Evidence for myelin regulating higher-order brain function and disease is rapidly accumulating; however, defining cellular/molecular mechanisms remains challenging partially due to the dynamic brain physiology involving deep changes during development, aging, and in response to learning and disease. Furthermore, as the etiology of most neurological conditions remains obscure, most research models focus on mimicking symptoms, which limits understanding of their molecular onset and progression. Studying diseases caused by single gene mutations represents an opportunity to understand brain dys/function, including those regulated by myelin. Here, we discuss known and potential repercussions of abnormal central myelin on the neuropathophysiology of Neurofibromatosis Type 1 (NF1). Most patients with this monogenic disease present with neurological symptoms diverse in kind, severity, and onset/decline, including learning disabilities, autism spectrum disorders, attention deficit and hyperactivity disorder, motor coordination issues, and increased risk for depression and dementia. Coincidentally, most NF1 patients show diverse white matter/myelin abnormalities. Although myelin-behavior links were proposed decades ago, no solid data can prove or refute this idea yet. A recent upsurge in myelin biology understanding and research/therapeutic tools provides opportunities to address this debate. As precision medicine moves forward, an integrative understanding of all cell types disrupted in neurological conditions becomes a priority. Hence, this review aims to serve as a bridge between fundamental cellular/molecular myelin biology and clinical research in NF1.
Collapse
Affiliation(s)
- Peter de Blank
- Department of Pediatrics, The Cure Starts Now Brain Tumor Center, University of Cincinnati and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Akiko Nishiyama
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut, USA
| | - Alejandro López-Juárez
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| |
Collapse
|
9
|
Gupta S, Jinka SKA, Khanal S, Bhavnani N, Almashhori F, Lallo J, Mathias A, Al-Rhayyel Y, Herman D, Holden JG, Fleming SM, Raman P. Cognitive dysfunction and increased phosphorylated tau are associated with reduced O-GlcNAc signaling in an aging mouse model of metabolic syndrome. J Neurosci Res 2023; 101:1324-1344. [PMID: 37031439 DOI: 10.1002/jnr.25196] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 12/15/2022] [Accepted: 03/21/2023] [Indexed: 04/10/2023]
Abstract
Metabolic syndrome (MetS), characterized by hyperglycemia, obesity, and hyperlipidemia, can increase the risk of developing late-onset dementia. Recent studies in patients and mouse models suggest a putative link between hyperphosphorylated tau, a component of Alzheimer's disease-related dementia (ADRD) pathology, and cerebral glucose hypometabolism. Impaired glucose metabolism reduces glucose flux through the hexosamine metabolic pathway triggering attenuated O-linked N-acetylglucosamine (O-GlcNAc) protein modification. The goal of the current study was to investigate the link between cognitive function, tau pathology, and O-GlcNAc signaling in an aging mouse model of MetS, agouti KKAy+/- . Male and female C57BL/6, non-agouti KKAy-/- , and agouti KKAy+/- mice were aged 12-18 months on standard chow diet. Body weight, blood glucose, total cholesterol, and triglyceride were measured to confirm the MetS phenotype. Cognition, sensorimotor function, and emotional reactivity were assessed for each genotype followed by plasma and brain tissue collection for biochemical and molecular analyses. Body weight, blood glucose, total cholesterol, and triglyceride levels were significantly elevated in agouti KKAy+/- mice versus C57BL/6 controls and non-agouti KKAy-/- . Behaviorally, agouti KKAy+/- revealed impairments in sensorimotor and cognitive function versus age-matched C57BL/6 and non-agouti KKAy-/- mice. Immunoblotting demonstrated increased phosphorylated tau accompanied with reduced O-GlcNAc protein expression in hippocampal-associated dorsal midbrain of female agouti KKAy+/- versus C57BL/6 control mice. Together, these data demonstrate that impaired cognitive function and AD-related pathology are associated with reduced O-GlcNAc signaling in aging MetS KKAy+/- mice. Overall, our study suggests that interaction of tau pathology with O-GlcNAc signaling may contribute to MetS-induced cognitive dysfunction in aging.
Collapse
Affiliation(s)
- Shreya Gupta
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
- Biomedical Sciences Graduate Program, Kent State University, Kent, Ohio, USA
| | - Sanjay K A Jinka
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Saugat Khanal
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
- Biomedical Sciences Graduate Program, Kent State University, Kent, Ohio, USA
| | - Neha Bhavnani
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
- Biomedical Sciences Graduate Program, Kent State University, Kent, Ohio, USA
| | - Fayez Almashhori
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
- Biomedical Sciences Graduate Program, Kent State University, Kent, Ohio, USA
| | - Jason Lallo
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Amy Mathias
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Yasmine Al-Rhayyel
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Danielle Herman
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - John G Holden
- Department of Psychology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Sheila M Fleming
- Biomedical Sciences Graduate Program, Kent State University, Kent, Ohio, USA
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Priya Raman
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
- Biomedical Sciences Graduate Program, Kent State University, Kent, Ohio, USA
| |
Collapse
|
10
|
Carotenuto M, Messina G, Esposito M, Santoro C, Iacono D, Spruyt K. Polysomnographic study in pediatric neurofibromatosis type 1. Front Neurol 2023; 14:1213430. [PMID: 37538252 PMCID: PMC10394094 DOI: 10.3389/fneur.2023.1213430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/29/2023] [Indexed: 08/05/2023] Open
Abstract
Background Neurofibromatosis type 1 (NF1) is a genetic disease that alters neurodevelopment. We aimed to analyze the sleep macrostructure of a sample of children affected by NF1 without neurocognitive co-morbidities and MRI reports of unidentified bright objects (UBOs). Methods A 100 pre-pubertal children participated in the cross-sectional study: 50 subjects were children diagnosed with NF1 and 50 subjects were typically developing healthy children (TDC). All participants underwent polysomnographic evaluation through which conventional sleep parameters were collected: Total sleep time (TST), Sleep latency (SOL), first REM latency (FRL), number of stage shifts/h (SS/h), number of awakenings/h (AWN/h), wake after sleep onset (WASO%), sleep efficiency percentage (SE%), percentage of sleep time spent in sleep stages 1 (N1%) and 2 (N2%), slow-wave sleep (N3%), and REM sleep (REM%). Additionally, nocturnal respiratory events such as apnea/hypopnea index (AHI), oxygen desaturation index (ODI), and periodic limb movement index (PLMI) were recorded. Results Neurofibromatosis type 1 children showed a reduction in sleep duration parameters (TST; p < 0.001), sleep efficiency (SE%; p < 0.001), and stage N2% (p < 0.001). Moreover, the number of awakenings per hour (AWN/h), wake after sleep onset (WASO%), and respiratory events such as AHI, ODI, and PLMI resulted higher in NF1 vs. TDC children. Conclusion The data showed that the sleep macrostructure differs between NF1 and TDC children. These findings suggest that the evaluation of sleep may provide useful support in corroborating the diagnosis and offers additional therapeutic management perspectives in NF1 and genetic neurodevelopmental disorders in general.
Collapse
Affiliation(s)
- Marco Carotenuto
- Sleep Lab for Developmental Age, Clinic of Child and Adolescent Neuropsychiatry, Department of Mental and Physical Health and Preventive Medicine, Child and Adolescent Neuropsychiatry Clinic, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giovanni Messina
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Maria Esposito
- Sleep Lab for Developmental Age, Clinic of Child and Adolescent Neuropsychiatry, Department of Mental and Physical Health and Preventive Medicine, Child and Adolescent Neuropsychiatry Clinic, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Claudia Santoro
- Sleep Lab for Developmental Age, Clinic of Child and Adolescent Neuropsychiatry, Department of Mental and Physical Health and Preventive Medicine, Child and Adolescent Neuropsychiatry Clinic, University of Campania "Luigi Vanvitelli", Naples, Italy
- Department of Women's and Children's Health, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Diego Iacono
- Neuropathology Research, Biomedical Research Institute of New Jersey, BRInj, Cedar Knolls, NJ, United States
- Department of Pediatrics, Neuropathology Research, Mid-Atlantic Neonatology Associates (MANA), Atlantic Health System (AHS), Morristown, NJ, United States
| | - Karen Spruyt
- NeuroDiderot INSERM, Université de Paris, Paris, France
| |
Collapse
|
11
|
Lalancette E, Charlebois-Poirier AR, Agbogba K, Knoth IS, Jones EJH, Mason L, Perreault S, Lippé S. Steady-state visual evoked potentials in children with neurofibromatosis type 1: associations with behavioral rating scales and impact of psychostimulant medication. J Neurodev Disord 2022; 14:42. [PMID: 35869419 PMCID: PMC9306184 DOI: 10.1186/s11689-022-09452-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 07/11/2022] [Indexed: 11/20/2022] Open
Abstract
Background Neurofibromatosis type 1 (NF1) is a genetic disorder often associated with cognitive dysfunctions, including a high occurrence of deficits in visuoperceptual skills. The neural underpinnings of these visuoperceptual deficits are not fully understood. We used steady-state visual evoked potentials (SSVEPs) to investigate possible alterations in the synchronization of neural activity in the occipital cortex of children with NF1. Methods SSVEPs were measured using electroencephalography and compared between children with NF1 (n = 28) and neurotypical controls (n = 28) aged between 4 and 13 years old. SSVEPs were recorded during visual stimulation with coloured icons flickering at three different frequencies (6 Hz, 10 Hz, and 15 Hz) and analyzed in terms of signal-to-noise ratios. A mixed design ANCOVA was performed to compare SSVEP responses between groups at the three stimulation frequencies. Pearson’s correlations with levels of intellectual functioning as well as with symptoms of ADHD, ASD and emotional/behavioral problems were performed. The impact of psychostimulant medication on the SSVEP responses was analyzed in a subset of the NF1 group (n = 8) with paired t-tests. Results We observed reduced signal-to-noise ratios of the SSVEP responses in children with NF1. The SSVEP responses were negatively correlated with symptoms of inattention and with symptoms of emotional/behavioral problems in the NF1 group. The SSVEP response generated by the lowest stimulation frequency (i.e., 6 Hz) was rescued with the intake of psychostimulant medication. Conclusions Impaired processing of rhythmic visual stimulation was evidenced in children with NF1 through measures of SSVEP responses. Those responses seem to be more reduced in children with NF1 who exhibit more symptoms of inattention and emotional/behavioral problems in their daily life. SSVEPs are potentially sensitive electrophysiological markers that could be included in future studies investigating the impact of medication on brain activity and cognitive functioning in children with NF1. Supplementary Information The online version contains supplementary material available at 10.1186/s11689-022-09452-y.
Collapse
|
12
|
Miller AH, Halloran MC. Mechanistic insights from animal models of neurofibromatosis type 1 cognitive impairment. Dis Model Mech 2022; 15:276464. [PMID: 36037004 PMCID: PMC9459395 DOI: 10.1242/dmm.049422] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal-dominant neurogenetic disorder caused by mutations in the gene neurofibromin 1 (NF1). NF1 predisposes individuals to a variety of symptoms, including peripheral nerve tumors, brain tumors and cognitive dysfunction. Cognitive deficits can negatively impact patient quality of life, especially the social and academic development of children. The neurofibromin protein influences neural circuits via diverse cellular signaling pathways, including through RAS, cAMP and dopamine signaling. Although animal models have been useful in identifying cellular and molecular mechanisms that regulate NF1-dependent behaviors, translating these discoveries into effective treatments has proven difficult. Clinical trials measuring cognitive outcomes in patients with NF1 have mainly targeted RAS signaling but, unfortunately, resulted in limited success. In this Review, we provide an overview of the structure and function of neurofibromin, and evaluate several cellular and molecular mechanisms underlying neurofibromin-dependent cognitive function, which have recently been delineated in animal models. A better understanding of neurofibromin roles in the development and function of the nervous system will be crucial for identifying new therapeutic targets for the various cognitive domains affected by NF1. Summary: Neurofibromin influences neural circuits through RAS, cAMP and dopamine signaling. Exploring the mechanisms underlying neurofibromin-dependent behaviors in animal models might enable future treatment of the various cognitive deficits that are associated with neurofibromatosis type 1.
Collapse
Affiliation(s)
- Andrew H Miller
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI 53706, USA.,Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA.,Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Mary C Halloran
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI 53706, USA.,Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
13
|
Crow AJD, Janssen JM, Marshall C, Moffit A, Brennan L, Kohler CG, Roalf DR, Moberg PJ. A systematic review and meta-analysis of intellectual, neuropsychological, and psychoeducational functioning in neurofibromatosis type 1. Am J Med Genet A 2022; 188:2277-2292. [PMID: 35546306 PMCID: PMC9302478 DOI: 10.1002/ajmg.a.62773] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 02/27/2022] [Accepted: 04/06/2022] [Indexed: 01/07/2023]
Abstract
Neurofibromatosis Type 1 (NF1) is a common genetic disorder frequently associated with cognitive deficits. Despite cognitive deficits being a key feature of NF1, the profile of such impairments in NF1 has been shown to be heterogeneous. Thus, we sought to quantitatively synthesize the extant literature on cognitive functioning in NF1. A random-effects meta-analysis of cross-sectional studies was carried out comparing cognitive functioning of patients with NF1 to typically developing or unaffected sibling comparison subjects of all ages. Analyses included 50 articles (Total NNF1 = 1,522; MAge = 15.70 years, range = 0.52-69.60), yielding 460 effect sizes. Overall moderate deficits were observed [g = -0.64, 95% CI = (-0.69, -0.60)] wherein impairments differed at the level of cognitive domain. Deficits ranged from large [general intelligence: g = -0.95, 95% CI = (-1.12, -0.79)] to small [emotion: g = -0.37, 95% CI = (-0.63, -0.11)]. Moderation analyses revealed nonsignificant contributions of age, sex, educational attainment, and parental level of education to outcomes. These results illustrate that cognitive impairments are diffuse and salient across the lifespan in NF1. Taken together, these results further demonstrate efforts should be made to evaluate and address cognitive morbidity in patients with NF1 in conjunction with existing best practices.
Collapse
Affiliation(s)
- Andrew J D Crow
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jennica M Janssen
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA.,Department of Psychology, Drexel University, Philadelphia, Pennsylvania, USA
| | - Carolina Marshall
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA.,Department of Psychology, Hope College, Holland, Michigan, USA
| | - Anne Moffit
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA.,Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, USA
| | | | - Christian G Kohler
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA.,Lifespan Brain Institute (LiBI), Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - David R Roalf
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA.,Lifespan Brain Institute (LiBI), Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Paul J Moberg
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
14
|
Anastasaki C, Orozco P, Gutmann DH. RAS and beyond: the many faces of the neurofibromatosis type 1 protein. Dis Model Mech 2022; 15:274437. [PMID: 35188187 PMCID: PMC8891636 DOI: 10.1242/dmm.049362] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Neurofibromatosis type 1 is a rare neurogenetic syndrome, characterized by pigmentary abnormalities, learning and social deficits, and a predisposition for benign and malignant tumor formation caused by germline mutations in the NF1 gene. With the cloning of the NF1 gene and the recognition that the encoded protein, neurofibromin, largely functions as a negative regulator of RAS activity, attention has mainly focused on RAS and canonical RAS effector pathway signaling relevant to disease pathogenesis and treatment. However, as neurofibromin is a large cytoplasmic protein the RAS regulatory domain of which occupies only 10% of its entire coding sequence, both canonical and non-canonical RAS pathway modulation, as well as the existence of potential non-RAS functions, are becoming apparent. In this Special article, we discuss our current understanding of neurofibromin function.
Collapse
Affiliation(s)
- Corina Anastasaki
- Department of Neurology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Paola Orozco
- Department of Neurology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St Louis, MO 63110, USA
| |
Collapse
|
15
|
Feedback-Based Learning of Timing in Attention-Deficit/Hyperactivity Disorder and Neurofibromatosis Type 1. J Int Neuropsychol Soc 2022; 28:12-21. [PMID: 33573707 DOI: 10.1017/s1355617721000072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Patients with Neurofibromatosis Type 1 (NF1) frequently display symptoms resembling those of Attention Deficit/Hyperactivity Disorder (ADHD). Importantly, these disorders are characterised by distinct changes in the dopaminergic system, which plays an important role in timing performance and feedback-based adjustments in timing performance. In a transdiagnostic approach, we examine how far NF1 and ADHD show distinct or comparable profiles of timing performance and feedback-based adjustments in timing. METHOD We examined time estimation and learning processes in healthy control children (HC), children with ADHD with predominantly inattentive symptoms and those with NF1 using a feedback-based time estimation paradigm. RESULTS Healthy controls consistently responded closer to the correct time window than both patient groups, were less variable in their reaction times and displayed intact learning-based adjustments across time. The patient groups did not differ from each other regarding the number of in-time responses. In ADHD patients, the performance was rather unstable across time. No performance changes could be observed in patients with NF1 across the entire task. CONCLUSIONS Children with ADHD and NF1 differ in feedback learning-based adjustments of time estimation processes. ADHD is characterised by behavioural fluctuations during the learning process. These are likely to be associated with inefficiencies in the dopaminergic system. NF1 is characterised by impairments of feedback learning which could be due to various neurotransmitter alterations occurring in addition to deficits in dopamine synthesis. Results show that despite the strong overlap in clinical phenotype and neuropsychological deficits between NF1 and ADHD, the underlying cognitive mechanisms are different.
Collapse
|
16
|
Houpt AC, Schwartz SE, Coover RA. Assessing Psychiatric Comorbidity and Pharmacologic Treatment Patterns Among Patients With Neurofibromatosis Type 1. Cureus 2021; 13:e20244. [PMID: 35004058 PMCID: PMC8735883 DOI: 10.7759/cureus.20244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2021] [Indexed: 11/17/2022] Open
Abstract
Background and objective Neurofibromatosis 1 (NF1) is a genetic disorder that is accompanied by psychiatric comorbidities such as depression, anxiety, and attention-deficit hyperactivity disorder (ADHD) in more than half of the patients. However, there are limited data describing optimal treatment strategies for these conditions. This study aimed to address that gap in understanding and explore the neurobiological basis of psychiatric comorbidities in NF1. Materials and methods A retrospective cohort study was conducted among NF1 patients with a comorbid diagnosis of depression, anxiety, and/or ADHD. These disease states were chosen based on their relatively high reported prevalence in NF1 and shared pathophysiological mechanisms via monoaminergic dysfunction. Information regarding demographics, psychotherapeutic medication use, and clinical outcomes was gathered from electronic medical records. Relationships between patient- and medication-related factors and outcome measures were assessed using statistical analysis. Results The study population (n = 82) consisted of NF1 patients with a comorbid diagnosis of depression (76.8%), anxiety (53.7%), and/or ADHD (23.2%). The use of second-generation antipsychotic agent augmentation therapy or hydroxyzine monotherapy was associated with significantly more behavioral health (BH)-related emergency department (ED) visits, admissions, and inpatient days in the study population. Conversely, the use of bupropion augmentation therapy, buspirone augmentation therapy, and stimulants was associated with improved clinical outcomes, though these results were not statistically significant. Conclusions Based on our findings in this real-world study setting, patients with NF1 and psychiatric comorbidities appear to experience significant benefits from medications that enhance dopaminergic neurotransmission (e.g., bupropion, stimulants) when compared to drugs that oppose it (e.g., second-generation antipsychotics).
Collapse
|
17
|
Hardy KK, Berger C, Griffin D, Walsh KS, Sharkey CM, Weisman H, Gioia A, Packer RJ, Acosta MT. Computerized Working Memory Training for Children With Neurofibromatosis Type 1 (NF1): A Pilot Study. J Child Neurol 2021; 36:1078-1085. [PMID: 34472416 DOI: 10.1177/08830738211038083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
INTRODUCTION The present study aimed to evaluate the feasibility and efficacy of CogmedRM, a computerized, home-based working memory (WM) training program, in children with NF1. METHOD A pre-post design was used to evaluate changes in performance-based measures of attention and WM, and parent-completed ratings of executive functioning. Children meeting eligibility criteria completed CogmedRM over 9 weeks. Primary outcomes included compliance statistics and change in attention and WM scores. RESULTS Thirty-one children (52% male; M age = 10.97 ± 2.51), aged 8-15, were screened for participation; 27 children (87%) evidenced WM difficulties and participated in CogmedRM training. On average, participants completed 19.7 out of 25 prescribed sessions, with an adherence rate of 69%. Participants demonstrated improvements in short-term memory, attention, and executive functioning (all Ps < .05). CONCLUSION Results suggest that computerized, home-based WM training programs may be both feasible and efficacious for children with NF1 and cognitive deficits.
Collapse
Affiliation(s)
- Kristina K Hardy
- Children's National Hospital, Washington, DC, USA.,Department of Pediatrics and Neurology, The George Washington University School of Medicine, Washington, DC, USA
| | - Carly Berger
- Children's National Hospital, Washington, DC, USA
| | | | - Karin S Walsh
- Children's National Hospital, Washington, DC, USA.,Department of Pediatrics and Neurology, The George Washington University School of Medicine, Washington, DC, USA
| | - Christina M Sharkey
- Children's National Hospital, Washington, DC, USA.,Department of Pediatrics and Neurology, The George Washington University School of Medicine, Washington, DC, USA
| | | | | | - Roger J Packer
- Children's National Hospital, Washington, DC, USA.,Department of Pediatrics and Neurology, The George Washington University School of Medicine, Washington, DC, USA
| | - Maria T Acosta
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
18
|
Payne JM, Haebich KM, MacKenzie R, Walsh KS, Hearps SJC, Coghill D, Barton B, Pride NA, Ullrich NJ, Tonsgard JH, Viskochil D, Schorry EK, Klesse L, Fisher MJ, Gutmann DH, Rosser T, Packer RJ, Korf B, Acosta MT, Bellgrove MA, North KN. Cognition, ADHD Symptoms, and Functional Impairment in Children and Adolescents With Neurofibromatosis Type 1. J Atten Disord 2021; 25:1177-1186. [PMID: 31838937 DOI: 10.1177/1087054719894384] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Objective: We examined the contribution of attention and executive cognitive processes to ADHD symptomatology in NF1, as well as the relationships between cognition and ADHD symptoms with functional outcomes. Methods: The study sample consisted of 141 children and adolescents with NF1. Children were administered neuropsychological tests that assessed attention and executive function, from which latent cognitive variables were derived. ADHD symptomatology, adaptive skills, and quality of life (QoL) were assessed using parent-rated questionnaires. Path analyses were conducted to test relationships among cognitive functioning, ADHD symptomatology, and functional outcomes. Results: Significant deficits were observed on all outcome variables. Cognitive variables did not predict ADHD symptomatology. Neither did they predict functional outcomes. However, elevated ADHD symptomatology significantly predicted functional outcomes. Conclusion: Irrespective of cognitive deficits, elevated ADHD symptoms in children with NF1 negatively impact daily functioning and emphasize the importance of interventions aimed at minimizing ADHD symptoms in NF1.
Collapse
Affiliation(s)
- Jonathan M Payne
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia.,University of Melbourne, Parkville, Victoria, Australia
| | - Kristina M Haebich
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | | | - Karin S Walsh
- Children's National Health System, Washington, DC, USA
| | - Stephen J C Hearps
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - David Coghill
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia.,University of Melbourne, Parkville, Victoria, Australia
| | - Belinda Barton
- The Children's Hospital at Westmead, New South Wales, Australia.,The University of Sydney, New South Wales, Australia
| | - Natalie A Pride
- The Children's Hospital at Westmead, New South Wales, Australia.,The University of Sydney, New South Wales, Australia
| | | | - James H Tonsgard
- The University of Chicago Medicine Comer Children's Hospital, IL, USA
| | | | | | - Laura Klesse
- University of Texas Southwestern Medical Center, Dallas, USA
| | | | | | | | | | - Bruce Korf
- The University of Alabama at Birmingham, USA
| | | | | | - Kathryn N North
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia.,University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
19
|
Importance of Thalamostriatal Pathway Associated With Neurocognitive Dysfunctions in Children With Neurofibromatosis Type 1: Diffusion Tensor Imaging Findings. J Comput Assist Tomogr 2021; 45:294-299. [PMID: 33661154 DOI: 10.1097/rct.0000000000001134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To determine whether there is a difference between healthy control group and children with neurofibromatosis type 1 (NF1) in terms of apparent diffusion coefficient (ADC) and fractional anisotropy (FA) values in different regions of the brain associated with neurocognitive functions and to investigate the correlation between diffusion tensor imaging parameters and neurocognitive dysfunctions. METHODS The study included 28 children with NF1 and 21 controls. Nine distinct areas related to cognitive functions were selected for the analysis. The ADC and FA values were compared. RESULTS There was a significant difference between NF1 and healthy control in terms of ADC values obtained from all areas. The ADC values at obtained from thalamus and striatum were positively correlated with the full-scale intelligence quotient (IQ), verbal IQ, and performance IQ. CONCLUSIONS We are speculated that the development of microstructural damage in the thalamostriatal pathway may lead to neurocognitive dysfunction.
Collapse
|
20
|
Lion-François L, Herbillon V, Peyric E, Mercier C, Gérard D, Ginhoux T, Coutinho V, Kemlin I, Kassai B, Desportes V, Michael GA. Attention and Executive Disorders in Neurofibromatosis 1: Comparison Between NF1 With ADHD Symptomatology (NF1 + ADHD) and ADHD Per Se. J Atten Disord 2020; 24:1807-1823. [PMID: 28587546 DOI: 10.1177/1087054717707579] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Objective: To compare children with Neurofibromatosis type 1 and associated ADHD symptomatology (NF1 + ADHD) with children having received a diagnosis of ADHD without NF1. The idea was that performance differences in tasks of attention between these two groups would be attributable not to the ADHD symptomatology, but to NF1 alone. Method: One group of children with NF1 + ADHD (N = 32), one group of children with ADHD (N = 31), and one group of healthy controls (N = 40) participated in a set of computerized tasks assessing intensive, selective, and executive aspects of attention. Results: Differences were found between the two groups of patients in respect of several aspects of attention. Children with NF1 + ADHD did not always perform worse than children with ADHD. Several double dissociations can be established between the two groups of patients. Conclusion: ADHD symptomatology in NF1 does not contribute to all attention deficits, and ADHD cannot account for all attention impairments in NF1.
Collapse
Affiliation(s)
- Laurence Lion-François
- Hospices Civils de Lyon, Service de Neurologie pédiatrique, Hôpital Femme Mère Enfant, Lyon, France
| | - Vania Herbillon
- Hospices Civils de Lyon, Epilepsie, sommeil et explorations fonctionnelles neuropédiatriques, Hôpital Femme Mère Enfant, Lyon, France
| | - Emeline Peyric
- Hospices Civils de Lyon, Service de Neurologie pédiatrique, Hôpital Femme Mère Enfant, Lyon, France
| | - Catherine Mercier
- Laboratoire Biostatistique-Santé, UMR CNRS 5558, Université de Lyon, France
| | - Daniel Gérard
- Service de Psychiatrie infantile, Hôpital Pierre Wertheimer, Lyon, France
| | | | - Virginie Coutinho
- Service de Neuropédiatrie, Hôpital Trousseau La Roche-Guyon, Hôpitaux Universitaires Est Parisien, Paris, France
| | - Isabelle Kemlin
- Service de Neuropédiatrie, Hôpital Trousseau La Roche-Guyon, Hôpitaux Universitaires Est Parisien, Paris, France
| | - Behrouz Kassai
- Service de Pharmacologie Clinique, Inserm EPICIME-CIC 1407, CHU Lyon, Bron, France.,Laboratoire de Biométrie et Biologie Evolutive, CNRS, UMR 5558, Université Lyon 1, Villeurbanne, France
| | - Vincent Desportes
- Hospices Civils de Lyon, Service de Neurologie pédiatrique, Hôpital Femme Mère Enfant, Lyon, France
| | - George A Michael
- Laboratoire d'Étude des Mécanismes Cognitifs, EA 3082, Université de Lyon, Université Lumière-Lyon 2, Lyon, France
| |
Collapse
|
21
|
Bergoug M, Doudeau M, Godin F, Mosrin C, Vallée B, Bénédetti H. Neurofibromin Structure, Functions and Regulation. Cells 2020; 9:cells9112365. [PMID: 33121128 PMCID: PMC7692384 DOI: 10.3390/cells9112365] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/21/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022] Open
Abstract
Neurofibromin is a large and multifunctional protein encoded by the tumor suppressor gene NF1, mutations of which cause the tumor predisposition syndrome neurofibromatosis type 1 (NF1). Over the last three decades, studies of neurofibromin structure, interacting partners, and functions have shown that it is involved in several cell signaling pathways, including the Ras/MAPK, Akt/mTOR, ROCK/LIMK/cofilin, and cAMP/PKA pathways, and regulates many fundamental cellular processes, such as proliferation and migration, cytoskeletal dynamics, neurite outgrowth, dendritic-spine density, and dopamine levels. The crystallographic structure has been resolved for two of its functional domains, GRD (GAP-related (GTPase-activating protein) domain) and SecPH, and its post-translational modifications studied, showing it to be localized to several cell compartments. These findings have been of particular interest in the identification of many therapeutic targets and in the proposal of various therapeutic strategies to treat the symptoms of NF1. In this review, we provide an overview of the literature on neurofibromin structure, function, interactions, and regulation and highlight the relationships between them.
Collapse
|
22
|
An executive functioning perspective in neurofibromatosis type 1: from ADHD and autism spectrum disorder to research domains. Childs Nerv Syst 2020; 36:2321-2332. [PMID: 32617712 DOI: 10.1007/s00381-020-04745-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 06/11/2020] [Indexed: 01/31/2023]
Abstract
PURPOSE Neurofibromatosis type 1 (NF1) is a rare monogenic disorder associated with executive function (EF) deficits and heightened risk for attention-deficit/hyperactivity disorder (ADHD) and autism spectrum disorder (ASD). The goal of this paper is to understand how EFs provide a common foundation to understand vulnerabilities for ADHD and ASD within NF1. METHODS A literature review and synthesis was conducted. RESULTS EF difficulties in working memory, inhibitory control, cognitive flexibility, and planning are evident in NF1, ADHD, and ASD. However, relatively little is known about the heterogeneity of EFs and ADHD and ASD outcomes in NF1. Assessment of ADHD and ASD in NF1 is based on behavioral symptoms without understanding neurobiological contributions. Recent efforts are promoting the use of dimensional and multidisciplinary methods to better understand normal and abnormal behavior, including integrating information from genetics to self-report measures. CONCLUSION NF1 is a monogenic disease with well-developed molecular and phenotypic research as well as complementary animal models. NF1 presents an excellent opportunity to advance our understanding of the neurobiological impact of known pathogenic variation in normal and abnormal neural pathways implicated in human psychopathology. EFs are core features of NF1, ADHD, and ASD, and these neurodevelopmental outcomes are highly prevalent in NF1. We propose a multilevel approach for understanding EFs in patients with NF1.This is essential to advance targeted interventions for NF1 patients and to advance the exciting field of research in this condition.
Collapse
|
23
|
Brain-wide structural and functional disruption in mice with oligodendrocyte-specific Nf1 deletion is rescued by inhibition of nitric oxide synthase. Proc Natl Acad Sci U S A 2020; 117:22506-22513. [PMID: 32839340 PMCID: PMC7486714 DOI: 10.1073/pnas.2008391117] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
This study assessed the effects of myelin decompaction on motor behavior and brain-wide structural and functional connectivity, and the effect of nitric oxide synthase inhibition by N-nitro-l-arginine methyl ester (L-NAME) on these imaging measures. We report that inducible oligodendrocyte-specific inactivation of the Nf1 gene, which causes myelin decompaction, results in reduced initial motor coordination. Using diffusion-based magnetic resonance imaging (MRI), we show reduced myelin integrity, and using functional MRI, we show reduced functional connectivity in awake passive mice. L-NAME administration results in rescue of the pathology at the mesoscopic level, as measured using imaging procedures that can be directly applied to humans to study treatment efficacy in clinical trials. Neurofibromin gene (NF1) mutation causes neurofibromatosis type 1 (NF1), a disorder in which brain white matter deficits identified by neuroimaging are common, yet of unknown cellular etiology. In mice, Nf1 loss in adult oligodendrocytes causes myelin decompaction and increases oligodendrocyte nitric oxide (NO) levels. Nitric oxide synthase (NOS) inhibitors rescue this pathology. Whether oligodendrocyte pathology is sufficient to affect brain-wide structure and account for NF1 imaging findings is unknown. Here we show that Nf1 gene inactivation in adult oligodendrocytes (Plp-Nf1fl/+ mice) results in a motor coordination deficit. Magnetic resonance imaging in awake mice showed that fractional anisotropy is reduced in Plp-Nf1fl/+ corpus callosum and that interhemispheric functional connectivity in the motor cortex is also reduced, consistent with disrupted myelin integrity. Furthermore, NOS-specific inhibition rescued both measures. These results suggest that oligodendrocyte defects account for aspects of brain dysfunction in NF1 that can be identified by neuroimaging and ameliorated by NOS inhibition.
Collapse
|
24
|
Neurofibromatosis type 1: New developments in genetics and treatment. J Am Acad Dermatol 2020; 84:1667-1676. [PMID: 32771543 DOI: 10.1016/j.jaad.2020.07.105] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 07/26/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022]
Abstract
Neurofibromatosis type 1 is the most common neurocutaneous syndrome, with a frequency of 1 in 2500 persons. Diagnosis is paramount in the pretumor stage to provide proper anticipatory guidance for a number of neoplasms, both benign and malignant. Loss-of-function mutations in the NF1 gene result in truncated and nonfunctional production of neurofibromin, a tumor suppressor protein involved in downregulating the RAS signaling pathway. New therapeutic and preventive options include tyrosine kinase inhibitors, mTOR inhibitors, interferons, and radiofrequency therapy. This review summarizes recent updates in genetics, mutation analysis assays, and treatment options targeting aberrant genetic pathways. We also propose modified diagnostic criteria and provide an algorithm for surveillance of patients with neurofibromatosis type 1.
Collapse
|
25
|
Kopp N, McCullough K, Maloney SE, Dougherty JD. Gtf2i and Gtf2ird1 mutation do not account for the full phenotypic effect of the Williams syndrome critical region in mouse models. Hum Mol Genet 2020; 28:3443-3465. [PMID: 31418010 DOI: 10.1093/hmg/ddz176] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 06/04/2019] [Accepted: 06/27/2019] [Indexed: 12/31/2022] Open
Abstract
Williams syndrome (WS) is a neurodevelopmental disorder caused by a 1.5-1.8 Mbp deletion on chromosome 7q11.23, affecting the copy number of 26-28 genes. Phenotypes of WS include cardiovascular problems, craniofacial dysmorphology, deficits in visual-spatial cognition and a characteristic hypersocial personality. There are still no genes in the region that have been consistently linked to the cognitive and behavioral phenotypes, although human studies and mouse models have led to the current hypothesis that the general transcription factor 2 I family of genes, GTF2I and GTF2IRD1, are responsible. Here we test the hypothesis that these two transcription factors are sufficient to reproduce the phenotypes that are caused by deletion of the WS critical region (WSCR). We compare a new mouse model with loss of function mutations in both Gtf2i and Gtf2ird1 to an established mouse model lacking the complete WSCR. We show that the complete deletion (CD) model has deficits across several behavioral domains including social communication, motor functioning and conditioned fear that are not explained by loss of function mutations in Gtf2i and Gtf2ird1. Furthermore, transcriptome profiling of the hippocampus shows changes in synaptic genes in the CD model that are not seen in the double mutants. Thus, we have thoroughly defined a set of molecular and behavioral consequences of complete WSCR deletion and shown that genes or combinations of genes beyond Gtf2i and Gtf2ird1 are necessary to produce these phenotypic effects.
Collapse
Affiliation(s)
- Nathan Kopp
- Department of Genetics.,Department of Psychiatry
| | | | - Susan E Maloney
- Department of Psychiatry.,Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joseph D Dougherty
- Department of Genetics.,Department of Psychiatry.,Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
26
|
Wulaer B, Kunisawa K, Hada K, Jaya Suento W, Kubota H, Iida T, Kosuge A, Nagai T, Yamada K, Nitta A, Yamamoto Y, Saito K, Mouri A, Nabeshima T. Shati/Nat8l deficiency disrupts adult neurogenesis and causes attentional impairment through dopaminergic neuronal dysfunction in the dentate gyrus. J Neurochem 2020; 157:642-655. [PMID: 32275776 DOI: 10.1111/jnc.15022] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/25/2020] [Accepted: 03/25/2020] [Indexed: 12/18/2022]
Abstract
Successful completion of daily activities relies on the ability to select the relevant features of the environment for memory and recall. Disruption to these processes can lead to various disorders, such as attention-deficit hyperactivity disorder (ADHD). Dopamine is a neurotransmitter implicated in the regulation of several processes, including attention. In addition to the higher-order brain function, dopamine is implicated in the regulation of adult neurogenesis. Previously, we generated mice lacking Shati, an N-acetyltransferase-8-like protein on a C57BL/6J genetic background (Shati/Nat8l-/- ). These mice showed a series of changes in the dopamine system and ADHD-like behavioral phenotypes. Therefore, we hypothesized that deficiency of Shati/Nat8l would affect neurogenesis and attentional behavior in mice. We found aberrant morphology of neurons and impaired neurogenesis in the dentate gyrus of Shati/Nat8l-/- mice. Additionally, research has suggested that impaired neurogenesis might be because of the reduction of dopamine in the hippocampus. Galantamine (GAL) attenuated the attentional impairment observed in the object-based attention test via increasing the dopamine release in the hippocampus of Shati/Nat8l-/- mice. The α7 nicotinic acetylcholine receptor antagonist, methyllycaconitine, and dopamine D1 receptor antagonist, SCH23390, blocked the ameliorating effect of GAL on attentional impairment in Shati/Nat8l-/- mice. These results suggest that the ameliorating effect of GAL on Shati/Nat8l-/- attentional impairment is associated with activation of D1 receptors following increased dopamine release in the hippocampus via α7 nicotinic acetylcholine receptor. In summary, Shati/Nat8l is important in both morphogenesis and neurogenesis in the dentate gyrus and attention, possible via modulation of dopaminergic transmission. Cover Image for this issue: https://doi.org/10.1111/jnc.15061.
Collapse
Affiliation(s)
- Bolati Wulaer
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Aichi, Japan.,Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Science, Aichi, Japan
| | - Kazuo Kunisawa
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Aichi, Japan
| | - Kazuhiro Hada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Willy Jaya Suento
- Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Science, Aichi, Japan.,Department of Psychiatry, Hasanuddin University, South Sulawesi, Indonesia
| | - Hisayoshi Kubota
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Aichi, Japan
| | - Tsubasa Iida
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Aichi, Japan
| | - Aika Kosuge
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Aichi, Japan
| | - Taku Nagai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Aichi, Japan.,Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| | - Atsumi Nitta
- Department of Pharmaceutical Therapy and Neuropharmacology, Graduate School of Pharmaceutical Sciences, University of Toyama, Toyama, Japan.,Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| | - Yasuko Yamamoto
- Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Science, Aichi, Japan
| | - Kuniaki Saito
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Aichi, Japan.,Department of Disease Control and Prevention, Fujita Health University Graduate School of Health Science, Aichi, Japan.,Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| | - Akihiro Mouri
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Health Science, Aichi, Japan.,Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Aichi, Japan.,Japanese Drug Organization of Appropriate Use and Research, Aichi, Japan
| |
Collapse
|
27
|
Neurofibromatosis Type 1 Implicates Ras Pathways in the Genetic Architecture of Neurodevelopmental Disorders. Behav Genet 2020; 50:191-202. [DOI: 10.1007/s10519-020-09991-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 01/04/2020] [Indexed: 01/12/2023]
|
28
|
Lukkes JL, Drozd HP, Fitz SD, Molosh AI, Clapp DW, Shekhar A. Guanfacine treatment improves ADHD phenotypes of impulsivity and hyperactivity in a neurofibromatosis type 1 mouse model. J Neurodev Disord 2020; 12:2. [PMID: 31941438 PMCID: PMC6961243 DOI: 10.1186/s11689-019-9304-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 12/16/2019] [Indexed: 02/07/2023] Open
Abstract
Background Neurofibromatosis type 1 (NF1) is an autosomal dominant disorder with a mutation in one copy of the neurofibromin gene (NF1+/−). Even though approximately 40–60% of children with NF1 meet the criteria for attention deficit hyperactivity disorder (ADHD), very few preclinical studies, if any, have investigated alterations in impulsivity and risk-taking behavior. Mice with deletion of a single NF1 gene (Nf1+/−) recapitulate many of the phenotypes of NF1 patients. Methods We compared wild-type (WT) and Nf1+/− mouse strains to investigate differences in impulsivity and hyperactivity using the delay discounting task (DDT), cliff avoidance reaction (CAR) test, and open field. We also investigated whether treatment with the clinically effective alpha-2A adrenergic receptor agonist, guanfacine (0.3 mg/kg, i.p.), would reverse deficits observed in behavioral inhibition. Results Nf1+/− mice chose a higher percentage of smaller rewards when both 10- and 20-s delays were administered compared to WT mice, suggesting Nf1+/− mice are more impulsive. When treated with guanfacine (0.3 mg/kg, i.p.), Nf1+/− mice exhibited decreased impulsive choice by waiting for the larger, delayed reward. Nf1+/− mice also exhibited deficits in behavioral inhibition compared to WT mice in the CAR test by repetitively entering the outer edge of the platform where they risk falling. Treatment with guanfacine ameliorated these deficits. In addition, Nf1+/− mice exhibited hyperactivity as increased distance was traveled compared to WT controls in the open field. This hyperactivity in Nf1+/− mice was reduced with guanfacine pre-treatment. Conclusions Overall, our study confirms that Nf1+/− mice exhibit deficits in behavioral inhibition in multiple contexts, a key feature of ADHD, and can be used as a model system to identify alterations in neural circuitry associated with symptoms of ADHD in children with NF1.
Collapse
Affiliation(s)
- J L Lukkes
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, Indianapolis, IN, 46202, USA.
| | - H P Drozd
- Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, Indianapolis, IN, 46202, USA.,Program in Medical Neurosciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - S D Fitz
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, Indianapolis, IN, 46202, USA
| | - A I Molosh
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, Indianapolis, IN, 46202, USA
| | - D W Clapp
- Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, Indianapolis, IN, 46202, USA.,Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A Shekhar
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, Indianapolis, IN, 46202, USA.,Program in Medical Neurosciences, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana Clinical and Translation Sciences Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
29
|
Attention Deficit Predicts Intellectual Functioning in Children with Neurofibromatosis Type 1. Int J Pediatr 2019; 2019:9493837. [PMID: 31915440 PMCID: PMC6930769 DOI: 10.1155/2019/9493837] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 07/10/2019] [Accepted: 08/23/2019] [Indexed: 11/18/2022] Open
Abstract
Aims Attention deficit hyperactivity disorder (ADHD) is one of the most frequent neurocognitive impairments in neurofibromatosis type 1 (NF1) and a well-known risk factor for intellectual dysfunction in general. Since NF1 is per se associated with intellectual difficulties, this comorbidity may be crucial for the cognitive development of affected patients. In our study, we investigated if attention deficits are associated with intellectual functioning in NF1 and if children with NF1 plus ADHD differ in their intellectual and attention profiles from children affected by NF1-only or ADHD only. Methods 111 children aged between 6 and 12 years (53 NF1 plus ADHD, 28 NF1-only, 30 ADHD-only) performed the German version of the intelligence test WISC-IV and a continuous performance test (T.O.V.A.) to assess attention functions. Parents completed questionnaires about everyday attention and executive functions (Conners 3®, BRIEF). Results Children with NF1 plus ADHD showed significantly lower intelligence test scores (full-scale IQ: 89.39 [1.40]) than patients with NF1-only (full-scale IQ: 101.14 [1.98]; p < .001), and intellectual functioning correlated significantly with attention performance in NF1 (p < .001). As compared to NF1-only, attention, and executive functioning were impaired on several dimensions (T.O.V.A., Conners 3® and BRIEF) in NF1 plus ADHD. ADHD-only was associated with significantly higher problem scores regarding hyperactivity/impulsivity and inattention (Conners 3®). NF1-only was associated with inattentiveness when compared to the normative sample of the T.O.V.A. Conclusion NF1 is associated with variable attention problems. Severe attention deficits appear to be a risk factor for intellectual dysfunction in NF1, more than NF1 without attention deficit. NF1 plus ADHD presents a specific cognitive profile, which differs from that of NF1 and from neurotypical ADHD.
Collapse
|
30
|
Payne JM, Hearps SJC, Walsh KS, Paltin I, Barton B, Ullrich NJ, Haebich KM, Coghill D, Gioia GA, Cantor A, Cutter G, Tonsgard JH, Viskochil D, Rey-Casserly C, Schorry EK, Ackerson JD, Klesse L, Fisher MJ, Gutmann DH, Rosser T, Packer RJ, Korf B, Acosta MT, North KN. Reproducibility of cognitive endpoints in clinical trials: lessons from neurofibromatosis type 1. Ann Clin Transl Neurol 2019; 6:2555-2565. [PMID: 31797581 PMCID: PMC6917317 DOI: 10.1002/acn3.50952] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/31/2019] [Accepted: 11/01/2019] [Indexed: 11/10/2022] Open
Abstract
Objective Rapid developments in understanding the molecular mechanisms underlying cognitive deficits in neurodevelopmental disorders have increased expectations for targeted, mechanism‐based treatments. However, translation from preclinical models to human clinical trials has proven challenging. Poor reproducibility of cognitive endpoints may provide one explanation for this finding. We examined the suitability of cognitive outcomes for clinical trials in children with neurofibromatosis type 1 (NF1) by examining test‐retest reliability of the measures and the application of data reduction techniques to improve reproducibility. Methods Data were analyzed from the STARS clinical trial (n = 146), a multi‐center double‐blind placebo‐controlled phase II trial of lovastatin, conducted by the NF Clinical Trials Consortium. Intra‐class correlation coefficients were generated between pre‐ and post‐performances (16‐week interval) on neuropsychological endpoints in the placebo group to determine test‐retest reliabilities. Confirmatory factor analysis was used to reduce data into cognitive domains and account for measurement error. Results Test‐retest reliabilities were highly variable, with most endpoints demonstrating unacceptably low reproducibility. Data reduction confirmed four distinct neuropsychological domains: executive functioning/attention, visuospatial ability, memory, and behavior. Test‐retest reliabilities of latent factors improved to acceptable levels for clinical trials. Applicability and utility of our model was demonstrated by homogeneous effect sizes in the reanalyzed efficacy data. Interpretation These data demonstrate that single observed endpoints are not appropriate to determine efficacy, partly accounting for the poor test‐retest reliability of cognitive outcomes in clinical trials in neurodevelopmental disorders. Recommendations to improve reproducibility are outlined to guide future trial design.
Collapse
Affiliation(s)
- Jonathan M Payne
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia.,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Stephen J C Hearps
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Karin S Walsh
- Center for Neuroscience and Behavioral Medicine, Children's National Health System, Washington, DC
| | - Iris Paltin
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Belinda Barton
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Westmead, New South Wales, Australia.,Children's Hospital Education Research Institute, The Children's Hospital at Westmead, Westmead, New South Wales, Australia.,The University of Sydney Children's Hospital Westmead Clinical School, University of Sydney, Westmead, New South Wales, Australia
| | - Nicole J Ullrich
- Department of Neurology, Boston Children's Hospital, Boston, Massachusetts
| | - Kristina M Haebich
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - David Coghill
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia.,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Gerard A Gioia
- Center for Neuroscience and Behavioral Medicine, Children's National Health System, Washington, DC
| | - Alan Cantor
- Department of Preventative Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Gary Cutter
- School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama
| | - James H Tonsgard
- Division of Neurology, The University of Chicago Medicine Comer Children's Hospital, Chicago, Illinois
| | - David Viskochil
- Department of Genetics, University of Utah, Salt Lake City, Utah
| | | | - Elizabeth K Schorry
- Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Joseph D Ackerson
- Department of Psychology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Laura Klesse
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Michael J Fisher
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri
| | - Tena Rosser
- Department of Neurology, Children's Hospital of Los Angeles, Los Angeles, California
| | - Roger J Packer
- Center for Neuroscience and Behavioral Medicine, Children's National Health System, Washington, DC
| | - Bruce Korf
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Maria T Acosta
- Center for Neuroscience and Behavioral Medicine, Children's National Health System, Washington, DC.,National Institutes of Health, National Human Genome Research Institute, Bethesda, Maryland
| | - Kathryn N North
- Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia.,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | | |
Collapse
|
31
|
Maloney SE, Creeley CE, Hartman RE, Yuede CM, Zorumski CF, Jevtovic-Todorovic V, Dikranian K, Noguchi KK, Farber NB, Wozniak DF. Using animal models to evaluate the functional consequences of anesthesia during early neurodevelopment. Neurobiol Learn Mem 2019; 165:106834. [PMID: 29550366 PMCID: PMC6179938 DOI: 10.1016/j.nlm.2018.03.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/16/2018] [Accepted: 03/13/2018] [Indexed: 12/15/2022]
Abstract
Fifteen years ago Olney and colleagues began using animal models to evaluate the effects of anesthetic and sedative agents (ASAs) on neurodevelopment. The results from ongoing studies indicate that, under certain conditions, exposure to these drugs during development induces an acute elevated apoptotic neurodegenerative response in the brain and long-term functional impairments. These animal models have played a significant role in bringing attention to the possible adverse effects of exposing the developing brain to ASAs when few concerns had been raised previously in the medical community. The apoptotic degenerative response resulting from neonatal exposure to ASAs has been replicated in many studies in both rodents and non-human primates, suggesting that a similar effect may occur in humans. In both rodents and non-human primates, significantly increased levels of apoptotic degeneration are often associated with functional impairments later in life. However, behavioral deficits following developmental ASA exposure have not been consistently reported even when significantly elevated levels of apoptotic degeneration have been documented in animal models. In the present work, we review this literature and propose a rodent model for assessing potential functional deficits following neonatal ASA exposure with special reference to experimental design and procedural issues. Our intent is to improve test sensitivity and replicability for detecting subtle behavioral effects, and thus enhance the translational significance of ASA models.
Collapse
Affiliation(s)
- Susan E Maloney
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Intellectual and Developmental Disabilities Research Center, Washington University, St. Louis, MO, USA
| | - Catherine E Creeley
- Department of Psychology, The State University of New York at Fredonia, Fredonia, NY 14063, USA
| | - Richard E Hartman
- Department of Psychology, Loma Linda University, 11130 Anderson St., Loma Linda, CA 92354, USA
| | - Carla M Yuede
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Charles F Zorumski
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Krikor Dikranian
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
| | - Kevin K Noguchi
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Intellectual and Developmental Disabilities Research Center, Washington University, St. Louis, MO, USA
| | - Nuri B Farber
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Intellectual and Developmental Disabilities Research Center, Washington University, St. Louis, MO, USA
| | - David F Wozniak
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA; Intellectual and Developmental Disabilities Research Center, Washington University, St. Louis, MO, USA.
| |
Collapse
|
32
|
Karouzaki S, Peta C, Tsirimonaki E, Mangoura D. PKCε-dependent H-Ras activation encompasses the recruitment of the RasGEF SOS1 and of the RasGAP neurofibromin in the lipid rafts of embryonic neurons. Neurochem Int 2019; 131:104582. [PMID: 31629778 DOI: 10.1016/j.neuint.2019.104582] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 09/27/2019] [Accepted: 10/15/2019] [Indexed: 01/07/2023]
Abstract
The spatial organization of plasma membrane proteins is a key factor in the generation of distinct signal outputs, especially for PKC/Ras/ERK signalling. Regulation of activation of the membrane-bound Ras, critical for neuronal differentiation and highly specialized functions, is controlled by exchanges in nucleotides catalyzed by nucleotide exchange factors (GEFs) for GTP loading and Ras activation, and by Ras GTPase Activated Proteins (RasGAPs) that lead to activation of the intrinsic GTPase activity of Ras and thus its inactivation. PKCs are potent Ras activators yet the mechanistic details of these interactions, or the involvement of specific PKC isoforms are now beginning to be addressed. Even less known is the topology where RasGAPs terminate Ras activation. Towards this aim, we isolated lipid rafts from chick embryo neural tissue and primary neuronal cultures when PKCε is the prominent isoform and in combination with in vitro kinase assays, we now show that, in response the PKCε-specific activating peptide ψεRACK, an activated PKCε is recruited to lipid rafts; similar mobility was established when PKCε was physiologically activated with the Cannabinoid receptor 1 (CB1) agonist methanandamide. Activation of H-Ras for both agents was then established for the first time using in vivo RasGAP activity assays, which showed similar temporal profiles of activation and lateral mobility. Moreover, we found that the GEF SOS1, and the major neuronal RasGAP neurofibromin, a specific PKCε substrate, were both transiently significantly enriched in the rafts. Finally, our in silico analysis revealed a highly probable, conserved palmitoylation site adjacent to a CARC motif on neurofibromin, both of which are included only in the RasGAP related domain type I (GRDI) with the known high H-RasGAP activity. Taken together, these results suggest that PKCε activation regulates the spatial plasma membrane enrichments of both SOS1 and neurofibromin, thus controlling the output of activated H-Ras available for downstream signalling in neurons.
Collapse
Affiliation(s)
- Sophia Karouzaki
- Basic Research Center, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou, Athens, 11527, Greece
| | - Charoula Peta
- Basic Research Center, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou, Athens, 11527, Greece
| | - Emmanouella Tsirimonaki
- Basic Research Center, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou, Athens, 11527, Greece
| | - Dimitra Mangoura
- Basic Research Center, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou, Athens, 11527, Greece.
| |
Collapse
|
33
|
Sutton LP, Muntean BS, Ostrovskaya O, Zucca S, Dao M, Orlandi C, Song C, Xie K, Martemyanov KA. NF1-cAMP signaling dissociates cell type-specific contributions of striatal medium spiny neurons to reward valuation and motor control. PLoS Biol 2019; 17:e3000477. [PMID: 31600280 PMCID: PMC6805008 DOI: 10.1371/journal.pbio.3000477] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 10/22/2019] [Accepted: 09/17/2019] [Indexed: 12/11/2022] Open
Abstract
The striatum plays a fundamental role in motor learning and reward-related behaviors that are synergistically shaped by populations of D1 dopamine receptor (D1R)- and D2 dopamine receptor (D2R)-expressing medium spiny neurons (MSNs). How various neurotransmitter inputs converging on common intracellular pathways are parsed out to regulate distinct behavioral outcomes in a neuron-specific manner is poorly understood. Here, we reveal that distinct contributions of D1R-MSNs and D2R-MSNs towards reward and motor behaviors are delineated by the multifaceted signaling protein neurofibromin 1 (NF1). Using genetic mouse models, we show that NF1 in D1R-MSN modulates opioid reward, whereas loss of NF1 in D2R-MSNs delays motor learning by impeding the formation and consolidation of repetitive motor sequences. We found that motor learning deficits upon NF1 loss were associated with the disruption in dopamine signaling to cAMP in D2R-MSN. Restoration of cAMP levels pharmacologically or chemogenetically rescued the motor learning deficits seen upon NF1 loss in D2R-MSN. Our findings illustrate that multiplex signaling capabilities of MSNs are deployed at the level of intracellular pathways to achieve cell-specific control over behavioral outcomes. A mouse genetic study reveals that the multifaceted signaling protein neurofibromin (known for its role in the human genetic disease neurofibromatosis type 1) plays a key role in differential routing of motor and reward signals in populations of striatal medium spiny neurons.
Collapse
Affiliation(s)
- Laurie P. Sutton
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Brian S. Muntean
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Olga Ostrovskaya
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Stefano Zucca
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Maria Dao
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Cesare Orlandi
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Chenghui Song
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Keqiang Xie
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Kirill A. Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
- * E-mail:
| |
Collapse
|
34
|
Selective impairment of the executive attentional network in adult patients with neurofibromatosis type 1. Neuroreport 2019; 30:921-926. [PMID: 31469719 DOI: 10.1097/wnr.0000000000001275] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Cognitive dysfunction accompanied by neurofibromatosis type 1 is one of the significant characteristics of this neurocutaneous disorder and has a serious impact on patients' quality of life. Although studies on cognitive function in children with neurofibromatosis type 1 have revealed that attentional impairment is a key deficit in these patients, few studies have examined their neuropsychological profile, especially whether the attentional function is also abnormal and specific in adult patients with neurofibromatosis type 1. In this study, we used the revised attention network test to examine the function of three attentional networks-alerting, orienting and executive control-in 20 adult patients with neurofibromatosis type 1 in comparison to 20 normal controls. Adult patients with neurofibromatosis type 1 showed significant greater conflict effect for the executive control network, but no significant differences were found for alerting and orienting network relative to normal controls. These results provide evidence that there is an attentional deficit which is specifically associated with the executive control network in adult patients with neurofibromatosis type 1.
Collapse
|
35
|
Robinson JE, Coughlin GM, Hori AM, Cho JR, Mackey ED, Turan Z, Patriarchi T, Tian L, Gradinaru V. Optical dopamine monitoring with dLight1 reveals mesolimbic phenotypes in a mouse model of neurofibromatosis type 1. eLife 2019; 8:e48983. [PMID: 31545171 PMCID: PMC6819083 DOI: 10.7554/elife.48983] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 09/21/2019] [Indexed: 12/12/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant disorder whose neurodevelopmental symptoms include impaired executive function, attention, and spatial learning and could be due to perturbed mesolimbic dopaminergic circuitry. However, these circuits have never been directly assayed in vivo. We employed the genetically encoded optical dopamine sensor dLight1 to monitor dopaminergic neurotransmission in the ventral striatum of NF1 mice during motivated behavior. Additionally, we developed novel systemic AAV vectors to facilitate morphological reconstruction of dopaminergic populations in cleared tissue. We found that NF1 mice exhibit reduced spontaneous dopaminergic neurotransmission that was associated with excitation/inhibition imbalance in the ventral tegmental area and abnormal neuronal morphology. NF1 mice also had more robust dopaminergic and behavioral responses to salient visual stimuli, which were independent of learning, and rescued by optogenetic inhibition of non-dopaminergic neurons in the VTA. Overall, these studies provide a first in vivo characterization of dopaminergic circuit function in the context of NF1 and reveal novel pathophysiological mechanisms.
Collapse
Affiliation(s)
- J Elliott Robinson
- Division of Biology and Biological EngineeringCalifornia Institute of TechnologyPasadenaUnited States
| | - Gerard M Coughlin
- Division of Biology and Biological EngineeringCalifornia Institute of TechnologyPasadenaUnited States
| | - Acacia M Hori
- Division of Biology and Biological EngineeringCalifornia Institute of TechnologyPasadenaUnited States
| | - Jounhong Ryan Cho
- Division of Biology and Biological EngineeringCalifornia Institute of TechnologyPasadenaUnited States
| | - Elisha D Mackey
- Division of Biology and Biological EngineeringCalifornia Institute of TechnologyPasadenaUnited States
| | - Zeynep Turan
- Division of Biology and Biological EngineeringCalifornia Institute of TechnologyPasadenaUnited States
| | - Tommaso Patriarchi
- Department of Biochemistry and Molecular MedicineUniversity of California, DavisDavisUnited States
| | - Lin Tian
- Department of Biochemistry and Molecular MedicineUniversity of California, DavisDavisUnited States
| | - Viviana Gradinaru
- Division of Biology and Biological EngineeringCalifornia Institute of TechnologyPasadenaUnited States
| |
Collapse
|
36
|
Characterization of Motor and Non-Motor Behavioral Alterations in the Dj-1 (PARK7) Knockout Rat. J Mol Neurosci 2019; 69:298-311. [PMID: 31250274 DOI: 10.1007/s12031-019-01358-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 06/18/2019] [Indexed: 02/03/2023]
Abstract
Parkinson's disease is a neurodegenerative disorder that encompasses a constellation of motor and non-motor symptoms. The etiology of the disease is still poorly understood because of complex interactions between environmental and genetic risk factors. Using animal models to assess these risk factors may lead to a better understanding of disease manifestation. In this study, we assessed the Dj-1 knockout (KO) genetic rat model in a battery of motor and non-motor behaviors. We tested the Dj-1 KO rat, as well as age-matched wild-type (WT) control rats, in several sensorimotor tests at 2, 4, 7, and 13 months of age. The Dj-1-deficient rats were found to rear and groom less, and to have a shorter stride length than their WT counterparts, but to take more forelimb and hindlimb steps. In non-motor behavioral tasks, performed at several different ages, we evaluated the following: olfactory function, anxiety-like behavior, short-term memory, anhedonia, and stress coping behavior. Non-motor testing was conducted as early as 4.5 months and as late as 17 months of age. We found that Dj-1 KO animals displayed deficits in short-term spatial memory as early as 4.5 months of age during place preference testing, as well as impaired coping strategies in the forced swim test, which are consistent with a parkinsonian-like phenotype. In some instances, effects of chronic stress were evaluated in the Dj-1-deficient rats, as an initial test of an environmental challenge combined with a genetic disposition for PD. Although some of the results were mixed with differential effects across several of the behaviors, the combination of the changes we observed indicates that the Dj-1 KO rat may be a promising model for the assessment of the prodromal stage of Parkinson's disease, but further evaluation is necessary.
Collapse
|
37
|
Shofty B, Bergmann E, Zur G, Asleh J, Bosak N, Kavushansky A, Castellanos FX, Ben-Sira L, Packer RJ, Vezina GL, Constantini S, Acosta MT, Kahn I. Autism-associated Nf1 deficiency disrupts corticocortical and corticostriatal functional connectivity in human and mouse. Neurobiol Dis 2019; 130:104479. [PMID: 31128207 PMCID: PMC6689441 DOI: 10.1016/j.nbd.2019.104479] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/11/2019] [Accepted: 05/21/2019] [Indexed: 10/26/2022] Open
Abstract
Children with the autosomal dominant single gene disorder, neurofibromatosis type 1 (NF1), display multiple structural and functional changes in the central nervous system, resulting in neuropsychological cognitive abnormalities. Here we assessed the pathological functional organization that may underlie the behavioral impairments in NF1 using resting-state functional connectivity MRI. Coherent spontaneous fluctuations in the fMRI signal across the entire brain were used to interrogate the pattern of functional organization of corticocortical and corticostriatal networks in both NF1 pediatric patients and mice with a heterozygous mutation in the Nf1 gene (Nf1+/-). Children with NF1 demonstrated abnormal organization of cortical association networks and altered posterior-anterior functional connectivity in the default network. Examining the contribution of the striatum revealed that corticostriatal functional connectivity was altered. NF1 children demonstrated reduced functional connectivity between striatum and the frontoparietal network and increased striatal functional connectivity with the limbic network. Awake passive mouse functional connectivity MRI in Nf1+/- mice similarly revealed reduced posterior-anterior connectivity along the cingulate cortex as well as disrupted corticostriatal connectivity. The striatum of Nf1+/- mice showed increased functional connectivity to somatomotor and frontal cortices and decreased functional connectivity to the auditory cortex. Collectively, these results demonstrate similar alterations across species, suggesting that NF1 pathogenesis is linked to striatal dysfunction and disrupted corticocortical connectivity in the default network.
Collapse
Affiliation(s)
- Ben Shofty
- Department of Neuroscience, Rappaport Faculty of Medicine and Institute, Technion - Israel Institute of Technology, Haifa, Israel; The Gilbert Israeli NF Center, Department of Pediatric Neurosurgery, Dana Children's Hospital, Tel Aviv Medical Center, and Tel Aviv University, Tel Aviv, Israel
| | - Eyal Bergmann
- Department of Neuroscience, Rappaport Faculty of Medicine and Institute, Technion - Israel Institute of Technology, Haifa, Israel
| | - Gil Zur
- Department of Neuroscience, Rappaport Faculty of Medicine and Institute, Technion - Israel Institute of Technology, Haifa, Israel
| | - Jad Asleh
- Department of Neuroscience, Rappaport Faculty of Medicine and Institute, Technion - Israel Institute of Technology, Haifa, Israel
| | - Noam Bosak
- Department of Neuroscience, Rappaport Faculty of Medicine and Institute, Technion - Israel Institute of Technology, Haifa, Israel
| | - Alexandra Kavushansky
- Department of Neuroscience, Rappaport Faculty of Medicine and Institute, Technion - Israel Institute of Technology, Haifa, Israel
| | - F Xavier Castellanos
- Department of Child and Adolescent Psychiatry, Hassenfeld Children's Hospital at NYU Langone, New York, NY, USA; Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Liat Ben-Sira
- The Gilbert Israeli NF Center, Department of Pediatric Neurosurgery, Dana Children's Hospital, Tel Aviv Medical Center, and Tel Aviv University, Tel Aviv, Israel
| | - Roger J Packer
- The Gilbert Family Neurofibromatosis Institute, Children's National Health System, Department of Neurology and Pediatrics, George Washington University, Washington, DC, USA
| | - Gilbert L Vezina
- Department of Diagnostic Imaging and Radiology, Children's National Health System, Washington, DC, USA
| | - Shlomi Constantini
- The Gilbert Israeli NF Center, Department of Pediatric Neurosurgery, Dana Children's Hospital, Tel Aviv Medical Center, and Tel Aviv University, Tel Aviv, Israel
| | - Maria T Acosta
- The Gilbert Family Neurofibromatosis Institute, Children's National Health System, Department of Neurology and Pediatrics, George Washington University, Washington, DC, USA; National Human Genome Research Institute, National Institutes of Health, Bethesda, MD USA
| | - Itamar Kahn
- Department of Neuroscience, Rappaport Faculty of Medicine and Institute, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
38
|
Anastasaki C, Rensing N, Johnson KJ, Wong M, Gutmann DH. Neurofibromatosis type 1 (Nf1)-mutant mice exhibit increased sleep fragmentation. J Sleep Res 2019; 28:e12816. [PMID: 30609083 DOI: 10.1111/jsr.12816] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 11/21/2018] [Accepted: 11/22/2018] [Indexed: 12/16/2022]
Abstract
Neurofibromatosis type 1 (NF1) is a neurodevelopmental disorder in which affected children and adults are at a higher risk of sleep disorders. In an effort to identify potential sleep disturbances in a small animal model, we used a previously reported Nf1 conditional knockout (Nf1CKO ) mouse strain. In contrast to Nf1 mutant flies, the distribution of vigilance states was intact in Nf1CKO mice. However, Nf1CKO mice exhibited increased non-REM sleep (NREM)-to-wake and wake-to-NREM transitions. This sleep disruption was accompanied by decreased bout durations during awake and NREM sleep states under both light and dark conditions. Moreover, Nf1CKO mice have higher percentage delta power during awake and NREM sleep states under all light conditions. Taken together, Nf1CKO mice phenocopy some of the sleep disturbances observed in NF1 patients and provide a tractable platform to explore the molecular mechanisms governing sleep abnormalities in NF1.
Collapse
Affiliation(s)
- Corina Anastasaki
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Nicholas Rensing
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Kevin J Johnson
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Michael Wong
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
39
|
Cheng C, Deng PY, Ikeuchi Y, Yuede C, Li D, Rensing N, Huang J, Baldridge D, Maloney SE, Dougherty JD, Constantino J, Jahani-Asl A, Wong M, Wozniak DF, Wang T, Klyachko VA, Bonni A. Characterization of a Mouse Model of Börjeson-Forssman-Lehmann Syndrome. Cell Rep 2018; 25:1404-1414.e6. [PMID: 30403997 PMCID: PMC6261530 DOI: 10.1016/j.celrep.2018.10.043] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 09/18/2018] [Accepted: 10/11/2018] [Indexed: 01/10/2023] Open
Abstract
Mutations of the transcriptional regulator PHF6 cause the X-linked intellectual disability disorder Börjeson-Forssman-Lehmann syndrome (BFLS), but the pathogenesis of BFLS remains poorly understood. Here, we report a mouse model of BFLS, generated using a CRISPR-Cas9 approach, in which cysteine 99 within the PHD domain of PHF6 is replaced with phenylalanine (C99F). Mice harboring the patient-specific C99F mutation display deficits in cognitive functions, emotionality, and social behavior, as well as reduced threshold to seizures. Electrophysiological studies reveal that the intrinsic excitability of entorhinal cortical stellate neurons is increased in PHF6 C99F mice. Transcriptomic analysis of the cerebral cortex in C99F knockin mice and PHF6 knockout mice show that PHF6 promotes the expression of neurogenic genes and represses synaptic genes. PHF6-regulated genes are also overrepresented in gene signatures and modules that are deregulated in neurodevelopmental disorders of cognition. Our findings advance our understanding of the mechanisms underlying BFLS pathogenesis.
Collapse
Affiliation(s)
- Cheng Cheng
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA; Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Pan-Yue Deng
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63110, USA; Department of Cell Biology and Physiology, Washington University, St. Louis, MO 63110, USA
| | - Yoshiho Ikeuchi
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carla Yuede
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Daofeng Li
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63108, USA; Department of Genetics, Washington University School of Medicine, 4515 McKinley Ave., St. Louis, MO 63108, USA
| | - Nicholas Rensing
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ju Huang
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dustin Baldridge
- Department of Pediatrics, Division of Newborn Medicine, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Susan E Maloney
- Department of Genetics, Washington University School of Medicine, 4515 McKinley Ave., St. Louis, MO 63108, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Joseph D Dougherty
- Department of Genetics, Washington University School of Medicine, 4515 McKinley Ave., St. Louis, MO 63108, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - John Constantino
- Department of Psychiatry, Division of Child Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Arezu Jahani-Asl
- Department of Oncology, Faculty of Medicine, McGill University, Montreal, QC H3T 1E2, Canada; Lady Davis Research Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Michael Wong
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David F Wozniak
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Ting Wang
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63108, USA; Department of Genetics, Washington University School of Medicine, 4515 McKinley Ave., St. Louis, MO 63108, USA
| | - Vitaly A Klyachko
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63110, USA; Department of Cell Biology and Physiology, Washington University, St. Louis, MO 63110, USA
| | - Azad Bonni
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA; Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
40
|
Molosh AI, Shekhar A. Neurofibromatosis type 1 as a model system to study molecular mechanisms of autism spectrum disorder symptoms. PROGRESS IN BRAIN RESEARCH 2018; 241:37-62. [PMID: 30447756 DOI: 10.1016/bs.pbr.2018.09.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Neurofibromatosis type 1 (NF1) is monogenic neurodevelopmental disorder caused by mutation of NF1 gene, which leads to increased susceptibility to various tumors formations. Additionally, majority of patients with NF1 are experience high incidence of cognitive deficits. Particularly, we review the growing number of reports demonstrated a higher incidence of autism spectrum disorder (ASD) in individuals with NF1. In this review we also discuss face validity of preclinical Nf1 mouse models. Then we describe discoveries from these animal models that have uncovered the deficiencies in the regulation of Ras and other intracellular pathways as critical mechanisms underlying the Nf1 cognitive problems. We also summarize and interpret recent preclinical and clinical studies that point toward potential pharmacological therapies for NF1 patients.
Collapse
Affiliation(s)
- Andrei I Molosh
- Department of Psychiatry, Institute of Psychiatric Research, IU School of Medicine, Indianapolis, IN, United States; Stark Neurosciences Research Institute, IU School of Medicine, Indianapolis, IN, United States.
| | - Anantha Shekhar
- Department of Psychiatry, Institute of Psychiatric Research, IU School of Medicine, Indianapolis, IN, United States; Stark Neurosciences Research Institute, IU School of Medicine, Indianapolis, IN, United States; Department of Pharmacology & Toxicology, IU School of Medicine, Indianapolis, IN, United States; Indiana Clinical and Translational Institute, IU School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
41
|
Pride NA, Barton B, Hutchins P, Coghill DR, Korgaonkar MS, Hearps SJC, Rouel M, Malarbi S, North KN, Payne JM. Effects of methylphenidate on cognition and behaviour in children with neurofibromatosis type 1: a study protocol for a randomised placebo-controlled crossover trial. BMJ Open 2018; 8:e021800. [PMID: 30166301 PMCID: PMC6119452 DOI: 10.1136/bmjopen-2018-021800] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION Dopamine dysregulation has been identified as a key modulator of behavioural impairment in neurofibromatosis type 1 (NF1) and a potential therapeutic target. Preclinical research demonstrates reduced dopamine in the brains of genetically engineered NF1 mouse strains is associated with reduced spatial-learning and attentional dysfunction. Methylphenidate, a stimulant medication that increases dopaminergic and noradrenergic neurotransmission, rescued the behavioural and dopamine abnormalities. Although preliminary clinical trials have demonstrated that methylphenidate is effective in treating attention deficit hyperactivity disorder (ADHD) symptoms in children with NF1, its therapeutic effect on cognitive performance is unclear. The primary aim of this clinical trial is to assess the efficacy of methylphenidate for reducing attention deficits, spatial working memory impairments and ADHD symptoms in children with NF1. METHODS AND ANALYSIS A randomised, double-blind, placebo-controlled trial of methylphenidate with a two period crossover design. Thirty-six participants with NF1 aged 7-16 years will be randomised to one of two treatment sequences: 6 weeks of methylphenidate followed by 6 weeks of placebo or; 6 weeks of placebo followed by 6 weeks of methylphenidate. Neurocognitive and behavioural outcomes as well as neuroimaging measures will be completed at baseline and repeated at the end of each treatment condition (week 6, week 12). Primary outcome measures are omission errors on the Conners Continuous Performance Test-II (attention), between-search errors on the Spatial Working Memory task from the Cambridge Neuropsychological Test Automated Battery (spatial working memory) and the Inattentive and Hyperactivity/Impulsivity Symptom Scales on the Conners 3-Parent. Secondary outcomes will examine the effect of methylphenidate on executive functions, attention, visuospatial skills, behaviour, fine-motor skills, language, social skills and quality of life. ETHICS AND DISSEMINATION This trial has hospital ethics approval and the results will be disseminated through peer-reviewed publications and international conferences. TRIAL REGISTRATION NUMBER ACTRN12611000765921.
Collapse
Affiliation(s)
- Natalie A Pride
- Institute for Neuroscience and Muscle Research, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Discipline of Child and Adolescent Health, Sydney Medical School, Universtiy of Sydney, Sydney, New South Wales, Australia
| | - Belinda Barton
- Institute for Neuroscience and Muscle Research, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Discipline of Child and Adolescent Health, Sydney Medical School, Universtiy of Sydney, Sydney, New South Wales, Australia
- Children's Hosptial Education Research Institute, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Paul Hutchins
- Children's Hosptial Education Research Institute, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - David R Coghill
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Science, University of Melbourne, Melbourne, Victoria, Australia
- Department of Psychiatry, Faculty of Medicine, Dentistry and Health Science, University of Melbourne, Melbourne, Victoria, Australia
| | - Mayuresh S Korgaonkar
- Brain Dynamics Centre, Westmead Institute for Medical Research, University of Sydney, Westmead, New South Wales, Australia
| | - Stephen J C Hearps
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
| | - Melissa Rouel
- Institute for Neuroscience and Muscle Research, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Stephanie Malarbi
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Science, University of Melbourne, Melbourne, Victoria, Australia
| | - Kathryn N North
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Science, University of Melbourne, Melbourne, Victoria, Australia
| | - Jonathan M Payne
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Science, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
42
|
Dard L, Bellance N, Lacombe D, Rossignol R. RAS signalling in energy metabolism and rare human diseases. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2018; 1859:845-867. [PMID: 29750912 DOI: 10.1016/j.bbabio.2018.05.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/12/2018] [Accepted: 05/03/2018] [Indexed: 02/07/2023]
Abstract
The RAS pathway is a highly conserved cascade of protein-protein interactions and phosphorylation that is at the heart of signalling networks that govern proliferation, differentiation and cell survival. Recent findings indicate that the RAS pathway plays a role in the regulation of energy metabolism via the control of mitochondrial form and function but little is known on the participation of this effect in RAS-related rare human genetic diseases. Germline mutations that hyperactivate the RAS pathway have been discovered and linked to human developmental disorders that are known as RASopathies. Individuals with RASopathies, which are estimated to affect approximately 1/1000 human birth, share many overlapping characteristics, including cardiac malformations, short stature, neurocognitive impairment, craniofacial dysmorphy, cutaneous, musculoskeletal, and ocular abnormalities, hypotonia and a predisposition to developing cancer. Since the identification of the first RASopathy, type 1 neurofibromatosis (NF1), which is caused by the inactivation of neurofibromin 1, several other syndromes have been associated with mutations in the core components of the RAS-MAPK pathway. These syndromes include Noonan syndrome (NS), Noonan syndrome with multiple lentigines (NSML), which was formerly called LEOPARD syndrome, Costello syndrome (CS), cardio-facio-cutaneous syndrome (CFC), Legius syndrome (LS) and capillary malformation-arteriovenous malformation syndrome (CM-AVM). Here, we review current knowledge about the bioenergetics of the RASopathies and discuss the molecular control of energy homeostasis and mitochondrial physiology by the RAS pathway.
Collapse
Affiliation(s)
- L Dard
- Bordeaux University, 33000 Bordeaux, France; INSERM U1211, 33000 Bordeaux, France
| | - N Bellance
- Bordeaux University, 33000 Bordeaux, France; INSERM U1211, 33000 Bordeaux, France
| | - D Lacombe
- Bordeaux University, 33000 Bordeaux, France; INSERM U1211, 33000 Bordeaux, France; CHU de Bordeaux, Service de Génétique Médicale, F-33076 Bordeaux, France
| | - R Rossignol
- Bordeaux University, 33000 Bordeaux, France; INSERM U1211, 33000 Bordeaux, France; CELLOMET, CGFB-146 Rue Léo Saignat, Bordeaux, France.
| |
Collapse
|
43
|
Abstract
INTRODUCTION Neurofibromatosis type 1 (NF1) is an autosomal dominantly inherited tumor predisposition syndrome with an incidence of one in 3000-4000 individuals with no currently effective therapies. The NF1 gene encodes neurofibromin, which functions as a negative regulator of RAS. NF1 is a chronic multisystem disorder affecting many different tissues. Due to cell-specific complexities of RAS signaling, therapeutic approaches for NF1 will likely have to focus on a particular tissue and manifestation of the disease. Areas covered: We discuss the multisystem nature of NF1 and the signaling pathways affected due to neurofibromin deficiency. We explore the cell-/tissue-specific molecular and cellular consequences of aberrant RAS signaling in NF1 and speculate on their potential as therapeutic targets for the disease. We discuss recent genomic, transcriptomic, and proteomic studies combined with molecular, cellular, and biochemical analyses which have identified several targets for specific NF1 manifestations. We also consider the possibility of patient-specific gene therapy approaches for NF1. Expert opinion: The emergence of NF1 genotype-phenotype correlations, characterization of cell-specific signaling pathways affected in NF1, identification of novel biomarkers, and the development of sophisticated animal models accurately reflecting human pathology will continue to provide opportunities to develop therapeutic approaches to combat this multisystem disorder.
Collapse
Affiliation(s)
- James A Walker
- a Center for Genomic Medicine , Massachusetts General Hospital, Harvard Medical School , Boston , MA , USA
| | - Meena Upadhyaya
- b Division of Cancer and Genetics , Cardiff University , Cardiff , UK
| |
Collapse
|
44
|
Beste C, Adelhöfer N, Gohil K, Passow S, Roessner V, Li SC. Dopamine Modulates the Efficiency of Sensory Evidence Accumulation During Perceptual Decision Making. Int J Neuropsychopharmacol 2018; 21:649-655. [PMID: 29618012 PMCID: PMC6030879 DOI: 10.1093/ijnp/pyy019] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/28/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Perceptual decision making is the process through which available sensory information is gathered and processed to guide our choices. However, the neuropsychopharmacological basis of this important cognitive function is largely elusive. Yet, theoretical considerations suggest that the dopaminergic system may play an important role. METHODS In a double-blind, randomized, placebo-controlled study design, we examined the effect of methylphenidate in 2 dosages (0.25 mg/kg and 0.5 mg/kg body weight) in separate groups of healthy young adults. We used a moving dots task in which the coherency of the direction of moving dots stimuli was manipulated in 3 levels (5%, 15%, and 35%). Drift diffusion modelling was applied to behavioral data to capture subprocesses of perceptual decision making. RESULTS The findings show that only the drift rate (v), reflecting the efficiency of sensory evidence accumulation, but not the decision criterion threshold (a) or the duration of nondecisional processes (Ter), is affected by methylphenidate vs placebo administration. Compared with placebo, administering 0.25 mg/kg methylphenidate increased v, but only in the 35% coherence condition. Administering 0.5 mg/kg methylphenidate did not induce modulations. CONCLUSIONS The data suggest that dopamine selectively modulates the efficacy of evidence accumulation during perceptual decision making. This modulation depends on 2 factors: (1) the degree to which the dopaminergic system is modulated using methylphenidate (i.e., methylphenidate dosage) and (2) the signal-to-noise ratio of the visual information. Dopamine affects sensory evidence accumulation only when dopamine concentration is not shifted beyond an optimal level and the incoming information is less noisy.
Collapse
Affiliation(s)
- Christian Beste
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universitat Dresden, Dresden, Germany,Correspondence: Christian Beste, PhD, Faculty of Medicine Carl Gustav Carus, TU Dresden, Department of Child and Adolescent Psychiatry, Fetscherstrasse 74, 01307 Dresden, Germany ()
| | - Nico Adelhöfer
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universitat Dresden, Dresden, Germany
| | - Krutika Gohil
- Faculty of Psychology, Chair of Lifespan Developmental Neuroscience, Technische Universitat Dresden, Dresden, Germany
| | - Susanne Passow
- Faculty of Psychology, Chair of Lifespan Developmental Neuroscience, Technische Universitat Dresden, Dresden, Germany
| | - Veit Roessner
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universitat Dresden, Dresden, Germany
| | - Shu-Chen Li
- Faculty of Psychology, Chair of Lifespan Developmental Neuroscience, Technische Universitat Dresden, Dresden, Germany
| |
Collapse
|
45
|
Robinson JE, Gradinaru V. Dopaminergic dysfunction in neurodevelopmental disorders: recent advances and synergistic technologies to aid basic research. Curr Opin Neurobiol 2018; 48:17-29. [PMID: 28850815 PMCID: PMC5825239 DOI: 10.1016/j.conb.2017.08.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 08/03/2017] [Indexed: 12/19/2022]
Abstract
Neurodevelopmental disorders (NDDs) represent a diverse group of syndromes characterized by abnormal development of the central nervous system and whose symptomatology includes cognitive, emotional, sensory, and motor impairments. The identification of causative genetic defects has allowed for creation of transgenic NDD mouse models that have revealed pathophysiological mechanisms of disease phenotypes in a neural circuit- and cell type-specific manner. Mouse models of several syndromes, including Rett syndrome, Fragile X syndrome, Angelman syndrome, Neurofibromatosis type 1, etc., exhibit abnormalities in the structure and function of dopaminergic circuitry, which regulates motivation, motor behavior, sociability, attention, and executive function. Recent advances in technologies for functional circuit mapping, including tissue clearing, viral vector-based tracing methods, and optical readouts of neural activity, have refined our knowledge of dopaminergic circuits in unperturbed states, yet these tools have not been widely applied to NDD research. Here, we will review recent findings exploring dopaminergic function in NDD models and discuss the promise of new tools to probe NDD pathophysiology in these circuits.
Collapse
Affiliation(s)
- J Elliott Robinson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
46
|
Abstract
Neurofibromatosis type 1 and type 2, affecting both children and adults, often results in devastating complications. The rapid unravelling of the genetic underpinnings of these unique disorders has led to the development of novel therapies, especially molecular-targeted therapies. To facilitate clinical trial development, the Neurofibromatosis Clinical Trial Consortium (NFCTC) was established in 2006 by the Department of Defense. Over the past decade, the Consortium has successfully completed studies for children and adults with neurofibromatosis type 1 and plexiform neurofibromas, neurocognitive challenges, low-grade gliomas, and malignant peripheral nerve sheath tumors. In addition, a study for children and adults with neurofibromatosis type 2 and acoustic schwannomas is near completion. The NFCTC has now been expanded to 19 sites in the United Stated and Australia. Mechanisms have been put in place to work closely with other consortia, foundations, and industry to expeditiously translate preclinical discoveries into clinical trials.
Collapse
Affiliation(s)
- Roger J Packer
- 1 From the Center for Neuroscience and Behavioral Medicine, Children's National Health System, Washington, DC, USA.,2 Gilbert Neurofibromatosis Family Institute, Children's National Health System, Washington, DC, USA.,3 Brain Tumor Institute, Children's National Health System, Washington, DC, USA
| | - Michael J Fisher
- 4 Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Gary Cutter
- 5 University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Bruce R Korf
- 5 University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
47
|
Haas-Lude K, Heimgärtner M, Winter S, Mautner VF, Krägeloh-Mann I, Lidzba K. Motor dysfunction in NF1: Mediated by attention deficit or inherent to the disorder? Eur J Paediatr Neurol 2018; 22:164-169. [PMID: 29111114 DOI: 10.1016/j.ejpn.2017.10.005] [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: 07/20/2017] [Revised: 09/29/2017] [Accepted: 10/12/2017] [Indexed: 10/18/2022]
Abstract
AIM Attention deficit and compromised motor skills are both prevalent in Neurofibromatosis type 1 (NF1), but the relationship is unclear. We investigated motor function in children with NF1 and in children with Attention Deficit/Hyperactivity Disorder (ADHD), and explored if, in patients with NF1, attention deficit influences motor performance. METHODS Motor performance was measured using the Movement Assessment Battery for Children (M-ABC) in 71 children (26 with NF1 plus ADHD, 14 with NF1 without ADHD, and 31 with ADHD without NF1) aged 6-12 years. RESULTS There was a significant effect of group on motor performance. Both NF1 groups scored below children with ADHD without NF1. Attention performance mediated motor performance in children with ADHD without NF1, but not in children with NF1. CONCLUSIONS Motor function is not mediated by attention performance in children with NF1. While in ADHD, attention deficit influences motor performance, motor problems in NF1 seem to be independent from attention deficit. This argues for different pathomechanisms in these two groups of developmental disorders.
Collapse
Affiliation(s)
- Karin Haas-Lude
- University Children's Hospital Tübingen, Department of Pediatric Neurology and Developmental Medicine, Hoppe-Seyler-Straße 1, 72076, Tübingen, Germany.
| | - Magdalena Heimgärtner
- University Children's Hospital Tübingen, Department of Pediatric Neurology and Developmental Medicine, Hoppe-Seyler-Straße 1, 72076, Tübingen, Germany
| | - Sarah Winter
- University Children's Hospital Tübingen, Department of Pediatric Neurology and Developmental Medicine, Hoppe-Seyler-Straße 1, 72076, Tübingen, Germany
| | - Victor-Felix Mautner
- University Hospital Hamburg-Eppendorf, Neurofibromatosis Outpatient Department, Martinistraße 52, 20246, Hamburg, Germany
| | - Ingeborg Krägeloh-Mann
- University Children's Hospital Tübingen, Department of Pediatric Neurology and Developmental Medicine, Hoppe-Seyler-Straße 1, 72076, Tübingen, Germany
| | - Karen Lidzba
- University Children's Hospital Tübingen, Department of Pediatric Neurology and Developmental Medicine, Hoppe-Seyler-Straße 1, 72076, Tübingen, Germany
| |
Collapse
|
48
|
|
49
|
Vogel AC, Gutmann DH, Morris SM. Neurodevelopmental disorders in children with neurofibromatosis type 1. Dev Med Child Neurol 2017; 59:1112-1116. [PMID: 28845518 DOI: 10.1111/dmcn.13526] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/18/2017] [Indexed: 12/19/2022]
Abstract
Over the past several decades, neurofibromatosis type 1 (NF1) has become increasingly recognized as a neurodevelopmental disorder conferring increased risk for several important neurodevelopmental problems. In this review, we summarize the specific neurodevelopmental problems encountered in the context of NF1. These include impairments in general cognitive function, deficits in specific cognitive domains such as executive function and visuospatial processing and risk for specific learning disorders, impairments in attention and social skills and the overlap with attention-deficit-hyperactivity disorder and autism spectrum disorder, and the risk of developing other psychiatric conditions including anxiety and depression. Early recognition of these developmental impairments is important for the effective treatment of children with NF1, and further characterization is essential to improve our understanding of how mutations in the NF1 gene create the diversity of clinical neuropsychiatric symptomatology observed in this at-risk population.
Collapse
Affiliation(s)
- Alecia C Vogel
- Departments of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Stephanie M Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
50
|
Borrie SC, Brems H, Legius E, Bagni C. Cognitive Dysfunctions in Intellectual Disabilities: The Contributions of the Ras-MAPK and PI3K-AKT-mTOR Pathways. Annu Rev Genomics Hum Genet 2017; 18:115-142. [DOI: 10.1146/annurev-genom-091416-035332] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Sarah C. Borrie
- Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Hilde Brems
- Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Eric Legius
- Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Claudia Bagni
- Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
- Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00173 Rome, Italy
| |
Collapse
|