1
|
Morin-Parent F, Champigny C, Côté S, Mohamad T, Hasani SA, Çaku A, Corbin F, Lepage JF. Neurophysiological effects of a combined treatment of lovastatin and minocycline in patients with fragile X syndrome: Ancillary results of the LOVAMIX randomized clinical trial. Autism Res 2024; 17:1944-1956. [PMID: 39248107 DOI: 10.1002/aur.3222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/09/2024] [Indexed: 09/10/2024]
Abstract
Fragile X syndrome (FXS) is the primary hereditary cause of intellectual disability and autism spectrum disorder. It is characterized by exacerbated neuronal excitability, and its correction is considered an objective measure of treatment response in animal models, a marker albeit rarely used in clinical trials. Here, we used an extensive transcranial magnetic stimulation (TMS) battery to assess the neurophysiological effects of a therapy combining two disease-modifying drugs, lovastatin (40 mg) and minocycline (100 mg), administered alone for 8 weeks and in combination for 12 weeks, in 19 patients (mean age of 23.58 ± 1.51) with FXS taking part in the LOVAmix trial. The TMS battery, which included the resting motor threshold, short-interval intracortical inhibition, long-interval intracortical inhibition, corticospinal silent period, and intracortical facilitation, was completed at baseline after 8 weeks of monotherapy (visit 2 of the clinical trial) and after 12 weeks of dual therapy (visit 4 of the clinical trial). Repeated measure ANOVAs were performed between baseline and visit 2 (monotherapy) and visit 3 (dual therapy) with interactions for which monotherapy the participants received when they began the clinical trial. Results showed that dual therapy was associated with reduced cortical excitability after 20 weeks. This was reflected by a significant increase in the resting-motor threshold after dual therapy compared to baseline. There was a tendency for enhanced short-intracortical inhibition, a marker of GABAa-mediated inhibition after 8 weeks of monotherapy compared to baseline. Together, these results suggest that a combined therapy of minocycline and lovastatin might act on the core neurophysiopathology of FXS. This trial was registered at clinicaltrials.gov (NCT02680379).
Collapse
Affiliation(s)
- Florence Morin-Parent
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Canada
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada
| | - Camille Champigny
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences Sherbrooke University, Sherbrooke, Canada
| | - Samantha Côté
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Canada
| | - Teddy Mohamad
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Canada
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada
| | - Seyede Anis Hasani
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Canada
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada
| | - Artuela Çaku
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences Sherbrooke University, Sherbrooke, Canada
| | - François Corbin
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences Sherbrooke University, Sherbrooke, Canada
| | - Jean-François Lepage
- Department of Pediatrics, Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Canada
- Sherbrooke University Hospital Research Center, Sherbrooke, Canada
| |
Collapse
|
2
|
Protic D, Hagerman R. State-of-the-art therapies for fragile X syndrome. Dev Med Child Neurol 2024; 66:863-871. [PMID: 38385885 PMCID: PMC11144093 DOI: 10.1111/dmcn.15885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/23/2024]
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by a full mutation (> 200 CGG repeats) in the FMR1 gene. FXS is the leading cause of inherited intellectual disabilities and the most commonly known genetic cause of autism spectrum disorder. Children with FXS experience behavioral and sleep problems, anxiety, inattention, learning difficulties, and speech and language delays. There are no approved medications for FXS; however, there are several interventions and treatments aimed at managing the symptoms and improving the quality of life of individuals with FXS. A combination of non-pharmacological therapies and pharmacotherapy is currently the most effective treatment for FXS. Currently, several targeted treatments, such as metformin, sertraline, and cannabidiol, can be used by clinicians to treat FXS. Gene therapy is rapidly developing and holds potential as a prospective treatment option. Soon its efficacy and safety in patients with FXS will be demonstrated. WHAT THIS PAPER ADDS: Targeted treatment of fragile X syndrome (FXS) is the best current therapeutic approach. Gene therapy holds potential as a prospective treatment for FXS in the future.
Collapse
Affiliation(s)
- Dragana Protic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine University of Belgrade, Belgrade, Serbia
- Fragile X Clinic, Special Hospital for Cerebral Palsy and Developmental Neurology, Belgrade, Serbia
| | - Randi Hagerman
- Medical Investigation of Neurodevelopmental Disorders Institute, University of California, Davis, CA, USA
- Department of Pediatrics, University of California, Davis School of Medicine, Sacramento, CA, USA
| |
Collapse
|
3
|
Jonak CR, Assad SA, Garcia TA, Sandhu MS, Rumschlag JA, Razak KA, Binder DK. Phenotypic analysis of multielectrode array EEG biomarkers in developing and adult male Fmr1 KO mice. Neurobiol Dis 2024; 195:106496. [PMID: 38582333 DOI: 10.1016/j.nbd.2024.106496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024] Open
Abstract
Fragile X Syndrome (FXS) is a leading known genetic cause of intellectual disability with symptoms that include increased anxiety and social and sensory processing deficits. Recent electroencephalographic (EEG) studies in humans with FXS have identified neural oscillation deficits that include increased resting state gamma power, increased amplitude of auditory evoked potentials, and reduced phase locking of sound-evoked gamma oscillations. Similar EEG phenotypes are present in mouse models of FXS, but very little is known about the development of such abnormal responses. In the current study, we employed a 30-channel mouse multielectrode array (MEA) system to record and analyze resting and stimulus-evoked EEG signals in male P21 and P91 WT and Fmr1 KO mice. This led to several novel findings. First, P91, but not P21, Fmr1 KO mice have significantly increased resting EEG power in the low- and high-gamma frequency bands. Second, both P21 and P91 Fmr1 KO mice have markedly attenuated inter-trial phase coherence (ITPC) to spectrotemporally dynamic auditory stimuli as well as to 40 Hz and 80 Hz auditory steady-state response (ASSR) stimuli. This suggests abnormal temporal processing from early development that may lead to abnormal speech and language function in FXS. Third, we found hemispheric asymmetry of fast temporal processing in the mouse auditory cortex in WT but not Fmr1 KO mice. Together, these findings define a set of EEG phenotypes in young and adult mice that can serve as translational targets for genetic and pharmacological manipulation in phenotypic rescue studies.
Collapse
Affiliation(s)
- Carrie R Jonak
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, United States of America
| | - Samantha A Assad
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, United States of America
| | - Terese A Garcia
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, United States of America
| | - Manbir S Sandhu
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, United States of America
| | - Jeffrey A Rumschlag
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, SC, United States of America
| | - Khaleel A Razak
- Neuroscience Graduate Program, University of California, Riverside, CA, United States of America; Department of Psychology, University of California, Riverside, CA, United States of America
| | - Devin K Binder
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, United States of America; Neuroscience Graduate Program, University of California, Riverside, CA, United States of America.
| |
Collapse
|
4
|
Erickson CA, Shaffer RC, Will M, Schmitt LM, Horn P, Hirst K, Pedapati EV, Ober N, Tumuluru RV, Handen BL, Beversdorf DQ. Brief Report: A Double-Blind, Placebo-Controlled, Crossover, Proof-of-Concept Study of Minocycline in Autism Spectrum Disorder. J Autism Dev Disord 2023:10.1007/s10803-023-06132-1. [PMID: 38102393 DOI: 10.1007/s10803-023-06132-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2023] [Indexed: 12/17/2023]
Abstract
Neuroinflammatory mechanisms have been implicated in the pathophysiology of autism spectrum disorder (ASD). Minocycline is a matrix metalloproteinase inhibitor 9 (MMP9) inhibitor tetracycline antibiotic with known anti-inflammatory properties. In preclinical animal models of ASD, minocycline has demonstrated potential positive effects on phenotypes that may have relevance to ASD. We conducted the first placebo-controlled study of minocycline in ASD. This double-blind, placebo-controlled crossover trial employed four week treatment periods with a two week washout period. Twenty-four 12-22 year olds (mean age 17.4 years; range 12.9-22.5 years) with ASD were enrolled. Overall minocycline was well tolerated. No minocycline-associated clinical changes were noted with treatment on any performance or clinician or caregiver completed measures were noted. We hypothesize that either minocycline does not have potential therapeutic effects in ASD or our project was underpowered to define potential subject subgroups who may potentially respond positively to this drug.
Collapse
Affiliation(s)
- Craig A Erickson
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 4002, Cincinnati, OH, 45229, USA.
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Rebecca C Shaffer
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Behavioral Medicine and Clinical Psychology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Meredith Will
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Behavioral Medicine and Clinical Psychology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Lauren M Schmitt
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Behavioral Medicine and Clinical Psychology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Paul Horn
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kathy Hirst
- Thompson Center for Autism and Neurodevelopmental Disorders, University of Missouri, Columbia, MO, USA
| | - Ernest V Pedapati
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 4002, Cincinnati, OH, 45229, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Nicole Ober
- Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | | | - Benjamin L Handen
- Psychiatry, Pediatrics, Psychology, and Education Departments, University of Pittsburgh, Pittsburgh, USA
| | - David Q Beversdorf
- Thompson Center for Autism and Neurodevelopmental Disorders, University of Missouri, Columbia, MO, USA
- Radiology, Neurology, and Psychological Sciences, William and Nancy Thompson Endowed Chair in Radiology, University of Missouri, Columbia, MO, USA
| |
Collapse
|
5
|
Martinez LA, Born HA, Harris S, Regnier-Golanov A, Grieco JC, Weeber EJ, Anderson AE. Quantitative EEG Analysis in Angelman Syndrome: Candidate Method for Assessing Therapeutics. Clin EEG Neurosci 2023; 54:203-212. [PMID: 33203220 DOI: 10.1177/1550059420973095] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The goal of these studies was to use quantitative (q)EEG techniques on data from children with Angelman syndrome (AS) using spectral power analysis, and to evaluate this as a potential biomarker and quantitative method to evaluate therapeutics. Although characteristic patterns are evident in visual inspection, using qEEG techniques has the potential to provide quantitative evidence of treatment efficacy. We first assessed spectral power from baseline EEG recordings collected from children with AS compared to age-matched neurotypical controls, which corroborated the previously reported finding of increased total power driven by elevated delta power in children with AS. We then retrospectively analyzed data collected during a clinical trial evaluating the safety and tolerability of minocycline (3 mg/kg/d) to compare pretreatment recordings from children with AS (4-12 years of age) to EEG activity at the end of treatment and following washout for EEG spectral power and epileptiform events. At baseline and during minocycline treatment, the AS subjects demonstrated increased delta power; however, following washout from minocycline treatment the AS subjects had significantly reduced EEG spectral power and epileptiform activity. Our findings support the use of qEEG analysis in evaluating AS and suggest that this technique may be useful to evaluate therapeutic efficacy in AS. Normalizing EEG power in AS therefore may become an important metric in screening therapeutics to gauge overall efficacy. As therapeutics transition from preclinical to clinical studies, it is vital to establish outcome measures that can quantitatively evaluate putative treatments for AS and neurological disorders with distinctive EEG patterns.
Collapse
Affiliation(s)
- Luis A Martinez
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,The Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.,The Gordon and Mary Cain Pediatric Neurology Research Foundation Laboratories, Texas Children's Hospital, Houston, TX, USA
| | - Heather A Born
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,The Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.,The Gordon and Mary Cain Pediatric Neurology Research Foundation Laboratories, Texas Children's Hospital, Houston, TX, USA
| | - Sarah Harris
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,The Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.,The Gordon and Mary Cain Pediatric Neurology Research Foundation Laboratories, Texas Children's Hospital, Houston, TX, USA
| | - Angelique Regnier-Golanov
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,The Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.,The Gordon and Mary Cain Pediatric Neurology Research Foundation Laboratories, Texas Children's Hospital, Houston, TX, USA
| | - Joseph C Grieco
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | - Edwin J Weeber
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | - Anne E Anderson
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,The Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.,The Gordon and Mary Cain Pediatric Neurology Research Foundation Laboratories, Texas Children's Hospital, Houston, TX, USA.,Departments of Neuroscience and Neurology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
6
|
Jonak CR, Pedapati EV, Schmitt LM, Assad SA, Sandhu MS, DeStefano L, Ethridge L, Razak KA, Sweeney JA, Binder DK, Erickson CA. Baclofen-associated neurophysiologic target engagement across species in fragile X syndrome. J Neurodev Disord 2022; 14:52. [PMID: 36167501 PMCID: PMC9513876 DOI: 10.1186/s11689-022-09455-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 08/03/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is the most common inherited form of neurodevelopmental disability. It is often characterized, especially in males, by intellectual disability, anxiety, repetitive behavior, social communication deficits, delayed language development, and abnormal sensory processing. Recently, we identified electroencephalographic (EEG) biomarkers that are conserved between the mouse model of FXS (Fmr1 KO mice) and humans with FXS. METHODS In this report, we evaluate small molecule target engagement utilizing multielectrode array electrophysiology in the Fmr1 KO mouse and in humans with FXS. Neurophysiologic target engagement was evaluated using single doses of the GABAB selective agonist racemic baclofen (RBAC). RESULTS In Fmr1 KO mice and in humans with FXS, baclofen use was associated with suppression of elevated gamma power and increase in low-frequency power at rest. In the Fmr1 KO mice, a baclofen-associated improvement in auditory chirp synchronization was also noted. CONCLUSIONS Overall, we noted synchronized target engagement of RBAC on resting state electrophysiology, in particular the reduction of aberrant high frequency gamma activity, across species in FXS. This finding holds promise for translational medicine approaches to drug development for FXS, synchronizing treatment study across species using well-established EEG biological markers in this field. TRIAL REGISTRATION The human experiments are registered under NCT02998151.
Collapse
Affiliation(s)
- Carrie R. Jonak
- grid.266097.c0000 0001 2222 1582Division of Biomedical Sciences, School of Medicine, University of California, Riverside, USA
| | - Ernest V. Pedapati
- grid.239573.90000 0000 9025 8099Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.239573.90000 0000 9025 8099Division of Neurology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.24827.3b0000 0001 2179 9593Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Lauren M. Schmitt
- grid.239573.90000 0000 9025 8099Division of Developmental and Behavioral Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.24827.3b0000 0001 2179 9593Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Samantha A. Assad
- grid.266097.c0000 0001 2222 1582Division of Biomedical Sciences, School of Medicine, University of California, Riverside, USA
| | - Manbir S. Sandhu
- grid.266097.c0000 0001 2222 1582Division of Biomedical Sciences, School of Medicine, University of California, Riverside, USA
| | - Lisa DeStefano
- grid.239573.90000 0000 9025 8099Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.266900.b0000 0004 0447 0018Department of Psychology, University of Oklahoma, Norman, OK USA
| | - Lauren Ethridge
- grid.266900.b0000 0004 0447 0018Department of Psychology, University of Oklahoma, Norman, OK USA ,grid.266902.90000 0001 2179 3618Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Khaleel A. Razak
- grid.266097.c0000 0001 2222 1582Neuroscience Graduate Program, University of California, Riverside, USA ,grid.266097.c0000 0001 2222 1582Psychology Graduate Program, University of California, Riverside, USA
| | - John A. Sweeney
- grid.24827.3b0000 0001 2179 9593Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH USA
| | - Devin K. Binder
- grid.266097.c0000 0001 2222 1582Division of Biomedical Sciences, School of Medicine, University of California, Riverside, USA ,grid.266097.c0000 0001 2222 1582Neuroscience Graduate Program, University of California, Riverside, USA
| | - Craig A. Erickson
- grid.239573.90000 0000 9025 8099Division of Child and Adolescent Psychiatry, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.24827.3b0000 0001 2179 9593Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH USA
| |
Collapse
|
7
|
Blok LER, Boon M, van Reijmersdal B, Höffler KD, Fenckova M, Schenck A. Genetics, molecular control and clinical relevance of habituation learning. Neurosci Biobehav Rev 2022; 143:104883. [PMID: 36152842 DOI: 10.1016/j.neubiorev.2022.104883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/08/2022] [Accepted: 08/30/2022] [Indexed: 11/29/2022]
Abstract
Habituation is the most fundamental form of learning. As a firewall that protects our brain from sensory overload, it is indispensable for cognitive processes. Studies in humans and animal models provide increasing evidence that habituation is affected in autism and related monogenic neurodevelopmental disorders (NDDs). An integrated application of habituation assessment in NDDs and their animal models has unexploited potential for neuroscience and medical care. With the aim to gain mechanistic insights, we systematically retrieved genes that have been demonstrated in the literature to underlie habituation. We identified 258 evolutionarily conserved genes across species, describe the biological processes they converge on, and highlight regulatory pathways and drugs that may alleviate habituation deficits. We also summarize current habituation paradigms and extract the most decisive arguments that support the crucial role of habituation for cognition in health and disease. We conclude that habituation is a conserved, quantitative, cognition- and disease-relevant process that can connect preclinical and clinical work, and hence is a powerful tool to advance research, diagnostics, and treatment of NDDs.
Collapse
Affiliation(s)
- Laura Elisabeth Rosalie Blok
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Marina Boon
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Boyd van Reijmersdal
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Kira Daniela Höffler
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Michaela Fenckova
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands; Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia in Ceske Budejovice, Branisovska 31, 37005, Ceske Budejovice, Czech Republic.
| | - Annette Schenck
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| |
Collapse
|
8
|
Laroui A, Galarneau L, Abolghasemi A, Benachenhou S, Plantefève R, Bouchouirab FZ, Lepage JF, Corbin F, Çaku A. Clinical significance of matrix metalloproteinase-9 in Fragile X Syndrome. Sci Rep 2022; 12:15386. [PMID: 36100610 PMCID: PMC9470743 DOI: 10.1038/s41598-022-19476-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/30/2022] [Indexed: 11/12/2022] Open
Abstract
High plasma matrix metalloproteases-9 (MMP-9) levels have been reported in Fragile X Syndrome in a limited number of animal and human studies. Since the results obtained are method-dependent and not directly comparable, the clinical utility of MMP-9 measurement in FXS remains unclear. This study aimed to compare quantitative gel zymography and ELISA and to determine which method better discriminates abnormal MMP-9 levels of individuals with FXS from healthy controls and correlates with the clinical profile. The active and total forms of MMP-9 were quantified respectively, by gel zymography and ELISA in a cohort of FXS (n = 23) and healthy controls (n = 20). The clinical profile was assessed for the FXS group using the Aberrant Behavior Checklist FXS adapted version (ABC-CFX), Adaptive Behavior Assessment System (ABAS), Social Communication Questionnaire (SCQ), and Anxiety Depression and Mood Scale questionnaires. Method comparison showed a disagreement between gel zymography and ELISA with a constant error of − 0.18 [95% CI: − 0.35 to − 0.02] and a proportional error of 2.31 [95% CI: 1.53 to 3.24]. Plasma level of MMP-9 active form was significantly higher in FXS (n = 12) as compared to their age-sex and BMI matched controls (n = 12) (p = 0.039) and correlated with ABC-CFX (rs = 0.60; p = 0.039) and ADAMS (rs = 0.57; p = 0.043) scores. As compared to the plasma total form, the plasma MMP-9 active form better enables the discrimination of individuals with FXS from controls and correlates with the clinical profile. Our results highlight the importance of choosing the appropriate method to quantify plasma MMP-9 in future FXS clinical studies.
Collapse
|
9
|
Targeted therapy of cognitive deficits in fragile X syndrome. Mol Psychiatry 2022; 27:2766-2776. [PMID: 35354925 PMCID: PMC7612812 DOI: 10.1038/s41380-022-01527-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 03/03/2022] [Accepted: 03/14/2022] [Indexed: 11/08/2022]
Abstract
Breaking an impasse in finding mechanism-based therapies of neuropsychiatric disorders requires a strategic shift towards alleviating individual symptoms. Here we present a symptom and circuit-specific approach to rescue deficits of reward learning in Fmr1 knockout mice, a model of Fragile X syndrome (FXS), the most common monogenetic cause of inherited mental disability and autism. We use high-throughput, ecologically-relevant automated tests of cognition and social behavior to assess effectiveness of the circuit-targeted injections of designer nanoparticles, loaded with TIMP metalloproteinase inhibitor 1 protein (TIMP-1). Further, to investigate the impact of our therapeutic strategy on neuronal plasticity we perform long-term potentiation recordings and high-resolution electron microscopy. We show that central amygdala-targeted delivery of TIMP-1 designer nanoparticles reverses impaired cognition in Fmr1 knockouts, while having no impact on deficits of social behavior, hence corroborating symptom-specificity of the proposed approach. Moreover, we elucidate the neural correlates of the highly specific behavioral rescue by showing that the applied therapeutic intervention restores functional synaptic plasticity and ultrastructure of neurons in the central amygdala. Thus, we present a targeted, symptom-specific and mechanism-based strategy to remedy cognitive deficits in Fragile X syndrome.
Collapse
|
10
|
Auger E, Berry-Kravis EM, Ethridge LE. Independent evaluation of the harvard automated processing pipeline for Electroencephalography 1.0 using multi-site EEG data from children with Fragile X Syndrome. J Neurosci Methods 2022; 371:109501. [PMID: 35182604 PMCID: PMC8962770 DOI: 10.1016/j.jneumeth.2022.109501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND The Harvard Automatic Processing Pipeline for Electroencephalography (HAPPE) is a computerized EEG data processing pipeline designed for multiple site analysis of populations with neurodevelopmental disorders. This pipeline has been validated in-house by the developers but external testing using real-world datasets remains to be done. NEW METHOD Resting and auditory event-related EEG data from 29 children ages 3-6 years with Fragile X Syndrome as well as simulated EEG data was used to evaluate HAPPE's noise reduction techniques, data standardization features, and data integration compared to traditional manualized processing. RESULTS For the real EEG data, HAPPE pipeline showed greater trials retained, greater variance retained through independent component analysis (ICA) component removal, and smaller kurtosis than the manual pipeline; the manual pipeline had a significantly larger signal-to-noise ratio (SNR). For simulated EEG data, correlation between the pure signal and processed data was significantly higher for manually-processed data compared to HAPPE-processed data. Hierarchical linear modeling showed greater signal recovery in the manual pipeline with the exception of the gamma band signal which showed mixed results. COMPARISON WITH EXISTING METHODS SNR and simulated signal retention was significantly greater in the manually-processed data than the HAPPE-processed data. Signal reduction may negatively affect outcome measures. CONCLUSIONS The HAPPE pipeline benefits from less active processing time and artifact reduction without removing segments. However, HAPPE may bias toward elimination of noise at the cost of signal. Recommended implementation of the HAPPE pipeline for neurodevelopmental populations depends on the goals and priorities of the research.
Collapse
Affiliation(s)
- Emma Auger
- Department of Psychology, University of Oklahoma, Norman, OK 73019-2007, USA
| | - Elizabeth M Berry-Kravis
- Department of Pediatrics, Neurological Sciences, and Biochemistry, Rush University Medical Center, Chicago, IL 60612, USA
| | - Lauren E Ethridge
- Department of Psychology, University of Oklahoma, Norman, OK 73019-2007, USA; Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
11
|
Marquez-Legorreta E, Constantin L, Piber M, Favre-Bulle IA, Taylor MA, Blevins AS, Giacomotto J, Bassett DS, Vanwalleghem GC, Scott EK. Brain-wide visual habituation networks in wild type and fmr1 zebrafish. Nat Commun 2022; 13:895. [PMID: 35173170 PMCID: PMC8850451 DOI: 10.1038/s41467-022-28299-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 01/12/2022] [Indexed: 11/09/2022] Open
Abstract
Habituation is a form of learning during which animals stop responding to repetitive stimuli, and deficits in habituation are characteristic of several psychiatric disorders. Due to technical challenges, the brain-wide networks mediating habituation are poorly understood. Here we report brain-wide calcium imaging during larval zebrafish habituation to repeated visual looming stimuli. We show that different functional categories of loom-sensitive neurons are located in characteristic locations throughout the brain, and that both the functional properties of their networks and the resulting behavior can be modulated by stimulus saliency and timing. Using graph theory, we identify a visual circuit that habituates minimally, a moderately habituating midbrain population proposed to mediate the sensorimotor transformation, and downstream circuit elements responsible for higher order representations and the delivery of behavior. Zebrafish larvae carrying a mutation in the fmr1 gene have a systematic shift toward sustained premotor activity in this network, and show slower behavioral habituation.
Collapse
Affiliation(s)
- Emmanuel Marquez-Legorreta
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia.,Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Lena Constantin
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Marielle Piber
- School of Medicine, Medical Sciences, and Nutrition, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Itia A Favre-Bulle
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia.,School of Mathematics and Physics, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Michael A Taylor
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Ann S Blevins
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jean Giacomotto
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia.,Queensland Centre for Mental Health Research, West Moreton Hospital and Health Service, Wacol, QLD, 4076, Australia.,Griffith Institute for Drug Discovery, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia.,Discovery Biology, Griffith University, Brisbane, QLD, 4111, Australia
| | - Dani S Bassett
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Departments of Electrical & Systems Engineering, Physics & Astronomy, Neurology, Psychiatry, University of Pennsylvania, Philadelphia, PA, 19104, USA.,Santa Fe Institute, Santa Fe, NM, 87501, USA
| | - Gilles C Vanwalleghem
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia. .,Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.
| | - Ethan K Scott
- The Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia.
| |
Collapse
|
12
|
Protic DD, Aishworiya R, Salcedo-Arellano MJ, Tang SJ, Milisavljevic J, Mitrovic F, Hagerman RJ, Budimirovic DB. Fragile X Syndrome: From Molecular Aspect to Clinical Treatment. Int J Mol Sci 2022; 23:1935. [PMID: 35216055 PMCID: PMC8875233 DOI: 10.3390/ijms23041935] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 02/01/2023] Open
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by the full mutation as well as highly localized methylation of the fragile X mental retardation 1 (FMR1) gene on the long arm of the X chromosome. Children with FXS are commonly co-diagnosed with Autism Spectrum Disorder, attention and learning problems, anxiety, aggressive behavior and sleep disorder, and early interventions have improved many behavior symptoms associated with FXS. In this review, we performed a literature search of original and review articles data of clinical trials and book chapters using MEDLINE (1990-2021) and ClinicalTrials.gov. While we have reviewed the biological importance of the fragile X mental retardation protein (FMRP), the FXS phenotype, and current diagnosis techniques, the emphasis of this review is on clinical interventions. Early non-pharmacological interventions in combination with pharmacotherapy and targeted treatments aiming to reverse dysregulated brain pathways are the mainstream of treatment in FXS. Overall, early diagnosis and interventions are fundamental to achieve optimal clinical outcomes in FXS.
Collapse
Affiliation(s)
- Dragana D. Protic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia
| | - Ramkumar Aishworiya
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA; (R.A.); (M.J.S.-A.); (S.J.T.); (R.J.H.)
- Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, 5 Lower Kent Ridge Road, Singapore 119074, Singapore
| | - Maria Jimena Salcedo-Arellano
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA; (R.A.); (M.J.S.-A.); (S.J.T.); (R.J.H.)
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA 95817, USA
- Department of Pathology and Laboratory Medicine, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Si Jie Tang
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA; (R.A.); (M.J.S.-A.); (S.J.T.); (R.J.H.)
| | - Jelena Milisavljevic
- Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (J.M.); (F.M.)
| | - Filip Mitrovic
- Faculty of Medicine, University of Belgrade, 11129 Belgrade, Serbia; (J.M.); (F.M.)
| | - Randi J. Hagerman
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDH, University of California Davis, 2825 50th Street, Sacramento, CA 95817, USA; (R.A.); (M.J.S.-A.); (S.J.T.); (R.J.H.)
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Dejan B. Budimirovic
- Department of Psychiatry, Fragile X Clinic, Kennedy Krieger Institute, Baltimore, MD 21205, USA
- Department of Psychiatry & Behavioral Sciences-Child Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
13
|
Kenny A, Wright D, Stanfield AC. EEG as a translational biomarker and outcome measure in fragile X syndrome. Transl Psychiatry 2022; 12:34. [PMID: 35075104 PMCID: PMC8786970 DOI: 10.1038/s41398-022-01796-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 12/01/2021] [Accepted: 01/12/2022] [Indexed: 01/08/2023] Open
Abstract
Targeted treatments for fragile X syndrome (FXS) have frequently failed to show efficacy in clinical testing, despite success at the preclinical stages. This has highlighted the need for more effective translational outcome measures. EEG differences observed in FXS, including exaggerated N1 ERP amplitudes, increased resting gamma power and reduced gamma phase-locking in the sensory cortices, have been suggested as potential biomarkers of the syndrome. These abnormalities are thought to reflect cortical hyper excitability resulting from an excitatory (glutamate) and inhibitory (GABAergic) imbalance in FXS, which has been the target of several pharmaceutical remediation studies. EEG differences observed in humans also show similarities to those seen in laboratory models of FXS, which may allow for greater translational equivalence and better predict clinical success of putative therapeutics. There is some evidence from clinical trials showing that treatment related changes in EEG may be associated with clinical improvements, but these require replication and extension to other medications. Although the use of EEG characteristics as biomarkers is still in the early phases, and further research is needed to establish its utility in clinical trials, the current research is promising and signals the emergence of an effective translational biomarker.
Collapse
Affiliation(s)
- Aisling Kenny
- Patrick Wild Centre, Division of Psychiatry, Kennedy Tower, Royal Edinburgh Hospital, University of Edinburgh, EH10 5HF, Edinburgh, UK.
| | - Damien Wright
- grid.4305.20000 0004 1936 7988Patrick Wild Centre, Division of Psychiatry, Kennedy Tower, Royal Edinburgh Hospital, University of Edinburgh, EH10 5HF Edinburgh, UK
| | - Andrew C. Stanfield
- grid.4305.20000 0004 1936 7988Patrick Wild Centre, Division of Psychiatry, Kennedy Tower, Royal Edinburgh Hospital, University of Edinburgh, EH10 5HF Edinburgh, UK
| |
Collapse
|
14
|
Prospective cohort study reveals MMP-9, a neuroplasticity regulator, as a prediction marker of cochlear implantation outcome in prelingual deafness treatment. Mol Neurobiol 2022; 59:2190-2203. [PMID: 35061219 PMCID: PMC9262127 DOI: 10.1007/s12035-022-02732-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 01/04/2022] [Indexed: 11/11/2022]
Abstract
Because of vast variability of cochlear implantation outcomes in
prelingual deafness treatment, identification of good and poor performers remains a
challenging task. To address this issue, we investigated genetic variants of matrix
metalloproteinase 9 (MMP9) and brain-derived
neurotrophic factor (BDNF) and plasma levels of
MMP-9, BDNF, and pro-BDNF that have all been implicated in neuroplasticity after
sensory deprivation in the auditory pathway. We recruited a cohort of prelingually
deaf children, all implanted before the age of 2, and carried out a prospective
observation (N = 61). Next, we analyzed the
association between (i) functional MMP9 (rs20544,
rs3918242, rs2234681) and BDNF (rs6265) gene
variants (and their respective protein levels) and (ii) the child’s auditory
development as measured with the LittlEARS Questionnaire (LEAQ) before cochlear
implant (CI) activation and at 8 and 18 months post-CI activation. Statistical
analyses revealed that the plasma level of MMP-9 measured at implantation in
prelingually deaf children was significantly correlated with the LEAQ score
18 months after CI activation. In the subgroup of DFNB1-related deafness (N = 40), rs3918242 of MMP9 was significantly associated with LEAQ score at 18 months after
CI activation; also, according to a multiple regression model, the ratio of plasma
levels of pro-BDNF/BDNF measured at implantation was a significant predictor of
overall LEAQ score at follow-up. In the subgroup with DFNB1-related deafness, who
had CI activation after 1 year old (N = 22), a
multiple regression model showed that rs3918242 of MMP9 was a significant predictor of overall LEAQ score at
follow-up.
Collapse
|
15
|
An WW, Nelson CA, Wilkinson CL. Neural response to repeated auditory stimuli and its association with early language ability in male children with Fragile X syndrome. Front Integr Neurosci 2022; 16:987184. [PMID: 36452884 PMCID: PMC9702328 DOI: 10.3389/fnint.2022.987184] [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: 07/05/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
Abstract
Background Fragile X syndrome (FXS) is the most prevalent form of inherited intellectual disability and is commonly associated with autism. Previous studies have linked the structural and functional alterations in FXS with impaired sensory processing and sensory hypersensitivity, which may hinder the early development of cognitive functions such as language comprehension. In this study, we compared the P1 response of the auditory evoked potential and its habituation to repeated auditory stimuli in male children (2-7 years old) with and without FXS, and examined their association with clinical measures in these two groups. Methods We collected high-density electroencephalography (EEG) data in an auditory oddball paradigm from 12 male children with FXS and 11 age- and sex-matched typically developing (TD) children. After standardized EEG pre-processing, we conducted a spatial principal component (PC) analysis and identified two major PCs-a frontal PC and a temporal PC. Within each PC, we compared the P1 amplitude and inter-trial phase coherence (ITPC) between the two groups, and performed a series of linear regression analysis to study the association between these EEG measures and several clinical measures, including assessment scores for language abilities, non-verbal skills, and sensory hypersensitivity. Results At the temporal PC, both early and late standard stimuli evoked a larger P1 response in FXS compared to TD participants. For temporal ITPC, the TD group showed greater habituation than the FXS group. However, neither group showed significant habituation of the frontal or temporal P1 response. Despite lack of habituation, exploratory analysis of brain-behavior associations observed that within the FXS group, reduced frontal P1 response to late standard stimuli, and increased frontal P1 habituation were both associated with better language scores. Conclusion We identified P1 amplitude and ITPC in the temporal region as a contrasting EEG phenotype between the FXS and the TD groups. However, only frontal P1 response and habituation were associated with language measures. Larger longitudinal studies are required to determine whether these EEG measures could be used as biomarkers for language development in patients with FXS.
Collapse
Affiliation(s)
- Winko W An
- Division of Developmental Medicine, Boston Children's Hospital, Boston, MA, United States.,Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Charles A Nelson
- Division of Developmental Medicine, Boston Children's Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States.,Harvard Graduate School of Education, Cambridge, MA, United States
| | - Carol L Wilkinson
- Division of Developmental Medicine, Boston Children's Hospital, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| |
Collapse
|
16
|
Rais M, Lovelace JW, Shuai XS, Woodard W, Bishay S, Estrada L, Sharma AR, Nguy A, Kulinich A, Pirbhoy PS, Palacios AR, Nelson DL, Razak KA, Ethell IM. Functional consequences of postnatal interventions in a mouse model of Fragile X syndrome. Neurobiol Dis 2021; 162:105577. [PMID: 34871737 DOI: 10.1016/j.nbd.2021.105577] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 10/22/2021] [Accepted: 12/02/2021] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is a leading genetic cause of autism and intellectual disability with cortical hyperexcitability and sensory hypersensitivity attributed to loss and hypofunction of inhibitory parvalbumin-expressing (PV) cells. Our studies provide novel insights into the role of excitatory neurons in abnormal development of PV cells during a postnatal period of inhibitory circuit refinement. METHODS To achieve Fragile X mental retardation gene (Fmr1) deletion and re-expression in excitatory neurons during the postnatal day (P)14-P21 period, we generated CreCaMKIIa/Fmr1Flox/y (cOFF) and CreCaMKIIa/Fmr1FloxNeo/y (cON) mice, respectively. Cortical phenotypes were evaluated in adult mice using biochemical, cellular, clinically relevant electroencephalogram (EEG) and behavioral tests. RESULTS We found that similar to global Fmr1 KO mice, the density of PV-expressing cells, their activation, and sound-evoked gamma synchronization were impaired in cOFF mice, but the phenotypes were improved in cON mice. cOFF mice also showed enhanced cortical gelatinase activity and baseline EEG gamma power, which were reduced in cON mice. In addition, TrkB phosphorylation and PV levels were lower in cOFF mice, which also showed increased locomotor activity and anxiety-like behaviors. Remarkably, when FMRP levels were restored in only excitatory neurons during the P14-P21 period, TrkB phosphorylation and mouse behaviors were also improved. CONCLUSIONS These results indicate that postnatal deletion or re-expression of FMRP in excitatory neurons is sufficient to elicit or ameliorate structural and functional cortical deficits, and abnormal behaviors in mice, informing future studies about appropriate treatment windows and providing fundamental insights into the cellular mechanisms of cortical circuit dysfunction in FXS.
Collapse
Affiliation(s)
- Maham Rais
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Jonathan W Lovelace
- Department of Psychology, University of California Riverside, Riverside, CA 92521, USA
| | - Xinghao S Shuai
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Walker Woodard
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Steven Bishay
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Leo Estrada
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Ashwin R Sharma
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Austin Nguy
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Anna Kulinich
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Patricia S Pirbhoy
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Arnold R Palacios
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | | | - Khaleel A Razak
- Department of Psychology, University of California Riverside, Riverside, CA 92521, USA; Neuroscience Graduate Program, University of California Riverside, Riverside, CA 92521, USA
| | - Iryna M Ethell
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA; Neuroscience Graduate Program, University of California Riverside, Riverside, CA 92521, USA.
| |
Collapse
|
17
|
Razak KA, Binder DK, Ethell IM. Neural Correlates of Auditory Hypersensitivity in Fragile X Syndrome. Front Psychiatry 2021; 12:720752. [PMID: 34690832 PMCID: PMC8529206 DOI: 10.3389/fpsyt.2021.720752] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/16/2021] [Indexed: 01/20/2023] Open
Abstract
The mechanisms underlying the common association between autism spectrum disorders (ASD) and sensory processing disorders (SPD) are unclear, and treatment options to reduce atypical sensory processing are limited. Fragile X Syndrome (FXS) is a leading genetic cause of intellectual disability and ASD behaviors. As in most children with ASD, atypical sensory processing is a common symptom in FXS, frequently manifesting as sensory hypersensitivity. Auditory hypersensitivity is a highly debilitating condition in FXS that may lead to language delays, social anxiety and ritualized repetitive behaviors. Animal models of FXS, including Fmr1 knock out (KO) mouse, also show auditory hypersensitivity, providing a translation relevant platform to study underlying pathophysiological mechanisms. The focus of this review is to summarize recent studies in the Fmr1 KO mouse that identified neural correlates of auditory hypersensitivity. We review results of electroencephalography (EEG) recordings in the Fmr1 KO mice and highlight EEG phenotypes that are remarkably similar to EEG findings in humans with FXS. The EEG phenotypes associated with the loss of FMRP include enhanced resting EEG gamma band power, reduced cross frequency coupling, reduced sound-evoked synchrony of neural responses at gamma band frequencies, increased event-related potential amplitudes, reduced habituation of neural responses and increased non-phase locked power. In addition, we highlight the postnatal period when the EEG phenotypes develop and show a strong association of the phenotypes with enhanced matrix-metalloproteinase-9 (MMP-9) activity, abnormal development of parvalbumin (PV)-expressing inhibitory interneurons and reduced formation of specialized extracellular matrix structures called perineuronal nets (PNNs). Finally, we discuss how dysfunctions of inhibitory PV interneurons may contribute to cortical hyperexcitability and EEG abnormalities observed in FXS. Taken together, the studies reviewed here indicate that EEG recordings can be utilized in both pre-clinical studies and clinical trials, while at the same time, used to identify cellular and circuit mechanisms of dysfunction in FXS. New therapeutic approaches that reduce MMP-9 activity and restore functions of PV interneurons may succeed in reducing FXS sensory symptoms. Future studies should examine long-lasting benefits of developmental vs. adult interventions on sensory phenotypes.
Collapse
Affiliation(s)
- Khaleel A. Razak
- Department of Psychology, University of California, Riverside, Riverside, CA, United States
- Graduate Neuroscience Program, University of California, Riverside, Riverside, CA, United States
| | - Devin K. Binder
- Graduate Neuroscience Program, University of California, Riverside, Riverside, CA, United States
- Division of Biomedical Sciences and Graduate Biomedical Sciences Program, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Iryna M. Ethell
- Graduate Neuroscience Program, University of California, Riverside, Riverside, CA, United States
- Division of Biomedical Sciences and Graduate Biomedical Sciences Program, School of Medicine, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
18
|
Hagerman RJ, Hagerman PJ. Fragile X Syndrome: Lessons Learned and What New Treatment Avenues Are on the Horizon. Annu Rev Pharmacol Toxicol 2021; 62:365-381. [PMID: 34499526 DOI: 10.1146/annurev-pharmtox-052120-090147] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Fragile X syndrome (FXS) is the most common form of inherited intellectual disability and the leading single-gene form of autism spectrum disorder, encompassing cognitive, behavioral, and physical forms of clinical involvement. FXS is caused by large expansions of a noncoding CGG repeat (>200 repeats) in the FMR1 gene, at which point the gene is generally silenced. Absence of FMR1 protein (FMRP), important for synaptic development and maintenance, gives rise to the neurodevelopmental disorder. There is, at present, no therapeutic approach that directly reverses the loss of FMRP; however, there is an increasing number of potential treatments that target the pathways dysregulated in FXS, including those that address the enhanced activity of the mGluR5 pathway and deficits in GABA pathways. Based on studies of targeted therapeutics to date, the prospects are good for one or more effective therapies for FXS in the near future. Expected final online publication date for the Annual Review of Pharmacology and Toxicology, Volume 62 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Randi J Hagerman
- Department of Pediatrics, University of California, Davis, School of Medicine, Sacramento, California 95817, USA; .,MIND Institute, University of California Davis Health, Sacramento, California 95817, USA
| | - Paul J Hagerman
- MIND Institute, University of California Davis Health, Sacramento, California 95817, USA.,Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Davis, California 95616, USA;
| |
Collapse
|
19
|
A white paper on a neurodevelopmental framework for drug discovery in autism and other neurodevelopmental disorders. Eur Neuropsychopharmacol 2021; 48:49-88. [PMID: 33781629 DOI: 10.1016/j.euroneuro.2021.02.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/08/2021] [Accepted: 02/15/2021] [Indexed: 12/20/2022]
Abstract
In the last decade there has been a revolution in terms of genetic findings in neurodevelopmental disorders (NDDs), with many discoveries critical for understanding their aetiology and pathophysiology. Clinical trials in single-gene disorders such as fragile X syndrome highlight the challenges of investigating new drug targets in NDDs. Incorporating a developmental perspective into the process of drug development for NDDs could help to overcome some of the current difficulties in identifying and testing new treatments. This paper provides a summary of the proceedings of the 'New Frontiers Meeting' on neurodevelopmental disorders organised by the European College of Neuropsychopharmacology in conjunction with the Innovative Medicines Initiative-sponsored AIMS-2-TRIALS consortium. It brought together experts in developmental genetics, autism, NDDs, and clinical trials from academia and industry, regulators, patient and family associations, and other stakeholders. The meeting sought to provide a platform for focused communication on scientific insights, challenges, and methodologies that might be applicable to the development of CNS treatments from a neurodevelopmental perspective. Multidisciplinary translational consortia to develop basic and clinical research in parallel could be pivotal to advance knowledge in the field. Although implementation of clinical trials for NDDs in paediatric populations is widely acknowledged as essential, safety concerns should guide each aspect of their design. Industry and academia should join forces to improve knowledge of the biology of brain development, identify the optimal timing of interventions, and translate these findings into new drugs, allowing for the needs of users and families, with support from regulatory agencies.
Collapse
|
20
|
McCracken JT, Anagnostou E, Arango C, Dawson G, Farchione T, Mantua V, McPartland J, Murphy D, Pandina G, Veenstra-VanderWeele J. Drug development for Autism Spectrum Disorder (ASD): Progress, challenges, and future directions. Eur Neuropsychopharmacol 2021; 48:3-31. [PMID: 34158222 PMCID: PMC10062405 DOI: 10.1016/j.euroneuro.2021.05.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/13/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022]
Abstract
In 2017, facing lack of progress and failures encountered in targeted drug development for Autism Spectrum Disorder (ASD) and related neurodevelopmental disorders, the ISCTM with the ECNP created the ASD Working Group charged to identify barriers to progress and recommending research strategies for the field to gain traction. Working Group international academic, regulatory and industry representatives held multiple in-person meetings, teleconferences, and subgroup communications to gather a wide range of perspectives on lessons learned from extant studies, current challenges, and paths for fundamental advances in ASD therapeutics. This overview delineates the barriers identified, and outlines major goals for next generation biomedical intervention development in ASD. Current challenges for ASD research are many: heterogeneity, lack of validated biomarkers, need for improved endpoints, prioritizing molecular targets, comorbidities, and more. The Working Group emphasized cautious but unwavering optimism for therapeutic progress for ASD core features given advances in the basic neuroscience of ASD and related disorders. Leveraging genetic data, intermediate phenotypes, digital phenotyping, big database discovery, refined endpoints, and earlier intervention, the prospects for breakthrough treatments are substantial. Recommendations include new priorities for expanded research funding to overcome challenges in translational clinical ASD therapeutic research.
Collapse
Affiliation(s)
- James T McCracken
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024, United States.
| | | | - Celso Arango
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Univesitario Gregorio Maranon, and School of Medicine, Universidad Complutense de Madrid, CIBERSAM, Madrid, Spain
| | - Geraldine Dawson
- Duke University Medical Center, Durham, North Carolina, United States
| | - Tiffany Farchione
- Food and Drug Administration, Silver Spring, Maryland, United States
| | - Valentina Mantua
- Food and Drug Administration, Silver Spring, Maryland, United States
| | | | - Declan Murphy
- Institute of Psychiatry, Psychology and Neuroscience, King's College De Crespigny Park, Denmark Hill, London SE5 8AF, United Kingdom
| | - Gahan Pandina
- Neuroscience Therapeutic Area, Janssen Research & Development, Pennington, New Jersey, United States
| | | |
Collapse
|
21
|
Armstrong C, Marsh ED. Electrophysiological Biomarkers in Genetic Epilepsies. Neurotherapeutics 2021; 18:1458-1467. [PMID: 34642905 PMCID: PMC8609056 DOI: 10.1007/s13311-021-01132-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2021] [Indexed: 02/04/2023] Open
Abstract
Precision treatments for epilepsy targeting the underlying genetic diagnoses are becoming a reality. Historically, the goal of epilepsy treatments was to reduce seizure frequency. In the era of precision medicine, however, outcomes such as prevention of epilepsy progression or even improvements in cognitive functions are both aspirational targets for any intervention. Developing methods, both in clinical trial design and in novel endpoints, will be necessary for measuring, not only seizures, but also the other neurodevelopmental outcomes that are predicted to be targeted by precision treatments. Biomarkers that quantitatively measure disease progression or network level changes are needed to allow for unbiased measurements of the effects of any gene-level treatments. Here, we discuss some of the promising electrophysiological biomarkers that may be of use in clinical trials of precision therapies, as well as the difficulties in implementing them.
Collapse
Affiliation(s)
- Caren Armstrong
- Division of Neurology and Pediatric Epilepsy Program, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Eric D Marsh
- Division of Neurology and Pediatric Epilepsy Program, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.
- Department of Pediatrics and Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
22
|
Lovelace JW, Rais M, Palacios AR, Shuai XS, Bishay S, Popa O, Pirbhoy PS, Binder DK, Nelson DL, Ethell IM, Razak KA. Deletion of Fmr1 from Forebrain Excitatory Neurons Triggers Abnormal Cellular, EEG, and Behavioral Phenotypes in the Auditory Cortex of a Mouse Model of Fragile X Syndrome. Cereb Cortex 2021; 30:969-988. [PMID: 31364704 DOI: 10.1093/cercor/bhz141] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 05/08/2019] [Accepted: 05/29/2019] [Indexed: 12/13/2022] Open
Abstract
Fragile X syndrome (FXS) is a leading genetic cause of autism with symptoms that include sensory processing deficits. In both humans with FXS and a mouse model [Fmr1 knockout (KO) mouse], electroencephalographic (EEG) recordings show enhanced resting state gamma power and reduced sound-evoked gamma synchrony. We previously showed that elevated levels of matrix metalloproteinase-9 (MMP-9) may contribute to these phenotypes by affecting perineuronal nets (PNNs) around parvalbumin (PV) interneurons in the auditory cortex of Fmr1 KO mice. However, how different cell types within local cortical circuits contribute to these deficits is not known. Here, we examined whether Fmr1 deletion in forebrain excitatory neurons affects neural oscillations, MMP-9 activity, and PV/PNN expression in the auditory cortex. We found that cortical MMP-9 gelatinase activity, mTOR/Akt phosphorylation, and resting EEG gamma power were enhanced in CreNex1/Fmr1Flox/y conditional KO (cKO) mice, whereas the density of PV/PNN cells was reduced. The CreNex1/Fmr1Flox/y cKO mice also show increased locomotor activity, but not the anxiety-like behaviors. These results indicate that fragile X mental retardation protein changes in excitatory neurons in the cortex are sufficient to elicit cellular, electrophysiological, and behavioral phenotypes in Fmr1 KO mice. More broadly, these results indicate that local cortical circuit abnormalities contribute to sensory processing deficits in autism spectrum disorders.
Collapse
Affiliation(s)
| | - Maham Rais
- Division of Biomedical Sciences, School of Medicine
| | | | | | | | - Otilia Popa
- Division of Biomedical Sciences, School of Medicine
| | | | - Devin K Binder
- Division of Biomedical Sciences, School of Medicine.,Graduate Neuroscience Program, University of California Riverside, Riverside, CA 92521,USA
| | - David L Nelson
- Molecular and Human Genetics, Baylor College of Medicine , Houston, TX 77030, USA
| | - Iryna M Ethell
- Division of Biomedical Sciences, School of Medicine.,Graduate Neuroscience Program, University of California Riverside, Riverside, CA 92521,USA
| | - Khaleel A Razak
- Department of Psychology.,Graduate Neuroscience Program, University of California Riverside, Riverside, CA 92521,USA
| |
Collapse
|
23
|
Jonak CR, Sandhu MS, Assad SA, Barbosa JA, Makhija M, Binder DK. The PDE10A Inhibitor TAK-063 Reverses Sound-Evoked EEG Abnormalities in a Mouse Model of Fragile X Syndrome. Neurotherapeutics 2021; 18:1175-1187. [PMID: 33594533 PMCID: PMC8423959 DOI: 10.1007/s13311-021-01005-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2021] [Indexed: 10/22/2022] Open
Abstract
Fragile X syndrome (FXS) is a genetic neurodevelopmental syndrome characterized by increased anxiety, repetitive behaviors, social communication deficits, delayed language development, and abnormal sensory processing. Recently, we have identified electroencephalographic (EEG) biomarkers that are conserved between the mouse model of FXS (Fmr1 KO mice) and humans with FXS. In this study, we test a specific candidate mechanism for engagement of multielectrode array (MEA) EEG biomarkers in the FXS mouse model. We administered TAK-063, a potent, selective, and orally active phosphodiesterase 10A (PDE10A) inhibitor, to Fmr1 KO mice, and examined its effects on MEA EEG biomarkers. We demonstrate significant dose-related amelioration of inter-trial phase coherence (ITPC) to temporally modulated auditory stimuli by TAK-063 in Fmr1 KO mice. Our data suggest that TAK-063 improves cortical auditory stimulus processing in Fmr1 KO mice, without significantly depressing baseline EEG power or causing any noticeable sedation or behavioral side effects. Thus, the PDE10A inhibitor TAK-063 has salutary effects on normalizing EEG biomarkers in a mouse model of FXS and should be pursued in further translational treatment development.
Collapse
Affiliation(s)
- Carrie R Jonak
- Division of Biomedical Sciences, School of Medicine, University of California, 900 University Avenue, Riverside, CA, 92521, USA
| | - Manbir S Sandhu
- Division of Biomedical Sciences, School of Medicine, University of California, 900 University Avenue, Riverside, CA, 92521, USA
| | - Samantha A Assad
- Division of Biomedical Sciences, School of Medicine, University of California, 900 University Avenue, Riverside, CA, 92521, USA
| | - Jacqueline A Barbosa
- Division of Biomedical Sciences, School of Medicine, University of California, 900 University Avenue, Riverside, CA, 92521, USA
| | - Mahindra Makhija
- Takeda International - UK, Rare Diseases Therapeutic Area Unit, 1 Kingdom Street, London, W2 6BD, UK
| | - Devin K Binder
- Division of Biomedical Sciences, School of Medicine, University of California, 900 University Avenue, Riverside, CA, 92521, USA.
- Neuroscience Graduate Program, University of California, Riverside, CA, USA.
| |
Collapse
|
24
|
Wilkinson CL, Nelson CA. Increased aperiodic gamma power in young boys with Fragile X Syndrome is associated with better language ability. Mol Autism 2021; 12:17. [PMID: 33632320 PMCID: PMC7908768 DOI: 10.1186/s13229-021-00425-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/16/2021] [Indexed: 02/17/2023] Open
Abstract
Background The lack of robust and reliable clinical biomarkers in Fragile X Syndrome (FXS), the most common inherited form of intellectual disability, has limited the successful translation of bench-to-bedside therapeutics. While numerous drugs have shown promise in reversing synaptic and behavioral phenotypes in mouse models of FXS, none have demonstrated clinical efficacy in humans. Electroencephalographic (EEG) measures have been identified as candidate biomarkers as EEG recordings of both adults with FXS and mouse models of FXS consistently exhibit alterations in resting state and task-related activity. However, the developmental timing of these EEG differences is not known as thus far EEG studies have not focused on young children with FXS. Further, understanding how EEG differences are associated with core symptoms of FXS is crucial to successful use of EEG as a biomarker, and may improve our understanding of the disorder. Methods Resting-state EEG was collected from FXS boys with full mutation of Fmr1 (2.5–7 years old, n = 11) and compared with both age-matched (n = 12) and cognitive-matched (n = 12) typically developing boys. Power spectra (including aperiodic and periodic components) were compared using non-parametric cluster-based permutation testing. Associations between 30 and 50 Hz gamma power and cognitive, language, and behavioral measures were evaluated using Pearson correlation and linear regression with age as a covariate. Results FXS participants showed increased power in the beta/gamma range (~ 25–50 Hz) across multiple brain regions. Both a reduction in the aperiodic (1/f) slope and increase in beta/gamma periodic activity contributed to the significant increase in high-frequency power. Increased gamma power, driven by the aperiodic component, was associated with better language ability in the FXS group. No association was observed between gamma power and parent report measures of behavioral challenges, sensory hypersensitivities, or adaptive behaviors. Limitations The study sample size was small, although comparable to other human studies in rare-genetic disorders. Findings are also limited to males in the age range studied. Conclusions Resting-state EEG measures from this study in young boys with FXS identified similar increases in gamma power previously reported in adults and mouse models. The observed positive association between resting state aperiodic gamma power and language development supports hypotheses that alterations in some EEG measures may reflect ongoing compensatory mechanisms. Supplementary Information The online version contains supplementary material available at 10.1186/s13229-021-00425-x.
Collapse
Affiliation(s)
- Carol L Wilkinson
- Division of Developmental Medicine, Boston Children's Hospital, 1 Autumn Street, 6th Floor, Boston, MA, 02115, USA.
| | - Charles A Nelson
- Division of Developmental Medicine, Boston Children's Hospital, 1 Autumn Street, 6th Floor, Boston, MA, 02115, USA
| |
Collapse
|
25
|
Proteau-Lemieux M, Knoth IS, Agbogba K, Côté V, Barlahan Biag HM, Thurman AJ, Martin CO, Bélanger AM, Rosenfelt C, Tassone F, Abbeduto LJ, Jacquemont S, Hagerman R, Bolduc F, Hessl D, Schneider A, Lippé S. EEG Signal Complexity Is Reduced During Resting-State in Fragile X Syndrome. Front Psychiatry 2021; 12:716707. [PMID: 34858220 PMCID: PMC8632368 DOI: 10.3389/fpsyt.2021.716707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/06/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction: Fragile X syndrome (FXS) is a genetic disorder caused by a mutation of the fragile X mental retardation 1 gene (FMR1). FXS is associated with neurophysiological abnormalities, including cortical hyperexcitability. Alterations in electroencephalogram (EEG) resting-state power spectral density (PSD) are well-defined in FXS and were found to be linked to neurodevelopmental delays. Whether non-linear dynamics of the brain signal are also altered remains to be studied. Methods: In this study, resting-state EEG power, including alpha peak frequency (APF) and theta/beta ratio (TBR), as well as signal complexity using multi-scale entropy (MSE) were compared between 26 FXS participants (ages 5-28 years), and 7 neurotypical (NT) controls with a similar age distribution. Subsequently a replication study was carried out, comparing our cohort to 19 FXS participants independently recorded at a different site. Results: PSD results confirmed the increased gamma, decreased alpha power and APF in FXS participants compared to NT controls. No alterations in TBR were found. Importantly, results revealed reduced signal complexity in FXS participants, specifically in higher scales, suggesting that altered signal complexity is sensitive to brain alterations in this population. The replication study mostly confirmed these results and suggested critical points of stagnation in the neurodevelopmental curve of FXS. Conclusion: Signal complexity is a powerful feature that can be added to the electrophysiological biomarkers of brain maturation in FXS.
Collapse
Affiliation(s)
- Mélodie Proteau-Lemieux
- Department of Psychology, University of Montreal, Montreal, QC, Canada.,Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada
| | - Inga Sophia Knoth
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada
| | - Kristian Agbogba
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada
| | - Valérie Côté
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada
| | - Hazel Maridith Barlahan Biag
- University of California Davis Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, Sacramento, CA, United States
| | - Angela John Thurman
- University of California Davis Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, Sacramento, CA, United States
| | | | - Anne-Marie Bélanger
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada
| | - Cory Rosenfelt
- Department of Pediatric Neurology, University of Alberta, Edmonton, AB, Canada
| | - Flora Tassone
- University of California Davis Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, Sacramento, CA, United States.,Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Leonard J Abbeduto
- University of California Davis Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, Sacramento, CA, United States.,Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Sébastien Jacquemont
- Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada.,Department of Pediatrics, University of Montreal, Montreal, QC, Canada
| | - Randi Hagerman
- University of California Davis Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, Sacramento, CA, United States
| | - François Bolduc
- Department of Pediatric Neurology, University of Alberta, Edmonton, AB, Canada
| | - David Hessl
- University of California Davis Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, Sacramento, CA, United States.,Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Andrea Schneider
- University of California Davis Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, Sacramento, CA, United States.,California North State University, College of Psychology, Rancho Cordova, CA, United States
| | - Sarah Lippé
- Department of Psychology, University of Montreal, Montreal, QC, Canada.,Research Center of the Sainte-Justine University Hospital, Montreal, QC, Canada
| |
Collapse
|
26
|
Champigny C, Morin-Parent F, Bellehumeur-Lefebvre L, Çaku A, Lepage JF, Corbin F. Combining Lovastatin and Minocycline for the Treatment of Fragile X Syndrome: Results From the LovaMiX Clinical Trial. Front Psychiatry 2021; 12:762967. [PMID: 35058813 PMCID: PMC8763805 DOI: 10.3389/fpsyt.2021.762967] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/29/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Limited success of previous clinical trials for Fragile X syndrome (FXS) has led researchers to consider combining different drugs to correct the pleiotropic consequences caused by the absence of the Fragile X mental retardation protein (FMRP). Here, we report the results of the LovaMiX clinical trial, the first trial for FXS combining two disease-modifying drugs, lovastatin, and minocycline, which have both shown positive effects when used independently. Aim: The main goals of the study were to assess the safety and efficacy of a treatment combining lovastatin and minocycline for patients with FXS. Design: Pilot Phase II open-label clinical trial. Patients with a molecular diagnostic of FXS were first randomized to receive, in two-step titration either lovastatin or minocycline for 8 weeks, followed by dual treatment with lovastatin 40 mg and minocycline 100 mg for 2 weeks. Clinical assessments were performed at the beginning, after 8 weeks of monotherapy, and at week 20 (12 weeks of combined therapy). Outcome Measures: The primary outcome measure was the Aberrant Behavior Checklist-Community (ABC-C) global score. Secondary outcome measures included subscales of the FXS specific ABC-C (ABC-CFX), the Anxiety, Depression, and Mood Scale (ADAMS), the Social Responsiveness Scale (SRS), the Behavior Rating Inventory of Executive Functions (BRIEF), and the Vineland Adaptive Behavior Scale second edition (VABS-II). Results: Twenty-one individuals out of 22 completed the trial. There were no serious adverse events related to the use of either drugs alone or in combination, suggesting good tolerability and safety profile of the combined therapy. Significant improvement was noted on the primary outcome measure with a 40% decrease on ABC-C global score with the combined therapy. Several outcome measures also showed significance. Conclusion: The combination of lovastatin and minocycline is safe in patients for FXS individuals and appears to improve several elements of the behavior. These results set the stage for a larger, placebo-controlled double-blind clinical trial to confirm the beneficial effects of the combined therapy.
Collapse
Affiliation(s)
- Camille Champigny
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.,Centre de Recherche du CHUS (CRCHUS), Sherbrooke, QC, Canada
| | | | - Laurence Bellehumeur-Lefebvre
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.,Centre de Recherche du CHUS (CRCHUS), Sherbrooke, QC, Canada
| | - Artuela Çaku
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.,Centre de Recherche du CHUS (CRCHUS), Sherbrooke, QC, Canada
| | - Jean-François Lepage
- Centre de Recherche du CHUS (CRCHUS), Sherbrooke, QC, Canada.,Faculty of Medicine and Health Sciences, Department of Pediatrics, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - François Corbin
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.,Centre de Recherche du CHUS (CRCHUS), Sherbrooke, QC, Canada
| |
Collapse
|
27
|
Gamma power abnormalities in a Fmr1-targeted transgenic rat model of fragile X syndrome. Sci Rep 2020; 10:18799. [PMID: 33139785 PMCID: PMC7608556 DOI: 10.1038/s41598-020-75893-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022] Open
Abstract
Fragile X syndrome (FXS) is characteristically displayed intellectual disability, hyperactivity, anxiety, and abnormal sensory processing. Electroencephalography (EEG) abnormalities are also observed in subjects with FXS, with many researchers paying attention to these as biomarkers. Despite intensive preclinical research using Fmr1 knock out (KO) mice, an effective treatment for FXS has yet to be developed. Here, we examined Fmr1-targeted transgenic rats (Fmr1-KO rats) as an alternative preclinical model of FXS. We characterized the EEG phenotypes of Fmr1-KO rats by measuring basal EEG power and auditory steady state response (ASSR) to click trains of stimuli at a frequency of 10–80 Hz. Fmr1-KO rats exhibited reduced basal alpha power and enhanced gamma power, and these rats showed enhanced locomotor activity in novel environment. While ASSR clearly peaked at around 40 Hz, both inter-trial coherence (ITC) and event-related spectral perturbation (ERSP) were significantly reduced at the gamma frequency band in Fmr1-KO rats. Fmr1-KO rats showed gamma power abnormalities and behavioral hyperactivity that were consistent with observations reported in mouse models and subjects with FXS. These results suggest that gamma power abnormalities are a translatable biomarker among species and demonstrate the utility of Fmr1-KO rats for investigating drugs for the treatment of FXS.
Collapse
|
28
|
Ethridge L, Thaliath A, Kraff J, Nijhawan K, Berry-Kravis E. Development of Neural Response to Novel Sounds in Fragile X Syndrome: Potential Biomarkers. AMERICAN JOURNAL ON INTELLECTUAL AND DEVELOPMENTAL DISABILITIES 2020; 125:449-464. [PMID: 33211818 PMCID: PMC8631234 DOI: 10.1352/1944-7558-125.6.449] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 05/22/2020] [Indexed: 06/11/2023]
Abstract
Auditory processing abnormalities in fragile X syndrome (FXS) may contribute to difficulties with language development, pattern identification, and contextual updating. Participants with FXS (N = 41) and controls (N = 27) underwent auditory event-related potentials during presentation of an oddball paradigm. Data was adequate for analysis for 33 participants with FXS and 27 controls (age 4-51 y, 13 females [FXS]; 4-54 y, 11 females [control]). Participants with FXS showed larger N1 and P2 amplitudes, abnormal lack of modulation of P1 and P2 amplitudes and P2 latency in response to oddball stimuli ) relative to controls: Females with FXS were more similar to controls. Participants with FXS showed a marginal speeding of the P2 latency, suggesting potentiation to oddball stimuli rather than habituation. Participants with FXS showed a heightened N1 habituation effect compared to controls. Gamma power was significantly higher for participants with FXS. Groups did not differ on mismatch negativity. Both controls and participants with FXS showed similar developmental trajectories in P1 and N1 amplitude, P2 latency, and gamma power, but not for P2 amplitude. One month retest analyses performed in 14 participants suggest strong test-retest reliability for most measures. Individuals with FXS show previously demonstrated increased response amplitude and high frequency neural activity. Despite an overall normal developmental trajectory for most measures, individuals with FXS show age-independent but gender-dependent decreases in complex processing of novel stimuli. Many markers show strong retest reliability even in children and thus are potential biomarkers for clinical trials in FXS.
Collapse
Affiliation(s)
- Lauren Ethridge
- Lauren Ethridge, University of Oklahoma Health Sciences Center
| | - Andrew Thaliath
- Andrew Thaliath, Jeremy Kraff, Karan Nijhawan, and Elizabeth Berry-Kravis, Rush University Medical Center, Chicago
| | - Jeremy Kraff
- Andrew Thaliath, Jeremy Kraff, Karan Nijhawan, and Elizabeth Berry-Kravis, Rush University Medical Center, Chicago
| | - Karan Nijhawan
- Andrew Thaliath, Jeremy Kraff, Karan Nijhawan, and Elizabeth Berry-Kravis, Rush University Medical Center, Chicago
| | - Elizabeth Berry-Kravis
- Andrew Thaliath, Jeremy Kraff, Karan Nijhawan, and Elizabeth Berry-Kravis, Rush University Medical Center, Chicago
| |
Collapse
|
29
|
Lovelace JW, Ethell IM, Binder DK, Razak KA. Minocycline Treatment Reverses Sound Evoked EEG Abnormalities in a Mouse Model of Fragile X Syndrome. Front Neurosci 2020; 14:771. [PMID: 32848552 PMCID: PMC7417521 DOI: 10.3389/fnins.2020.00771] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/30/2020] [Indexed: 01/19/2023] Open
Abstract
Fragile X Syndrome (FXS) is a leading known genetic cause of intellectual disability. Many symptoms of FXS overlap with those in autism including repetitive behaviors, language delays, anxiety, social impairments and sensory processing deficits. Electroencephalogram (EEG) recordings from humans with FXS and an animal model, the Fmr1 knockout (KO) mouse, show remarkably similar phenotypes suggesting that EEG phenotypes can serve as biomarkers for developing treatments. This includes enhanced resting gamma band power and sound evoked total power, and reduced fidelity of temporal processing and habituation of responses to repeated sounds. Given the therapeutic potential of the antibiotic minocycline in humans with FXS and animal models, it is important to determine sensitivity and selectivity of EEG responses to minocycline. Therefore, in this study, we examined if a 10-day treatment of adult Fmr1 KO mice with minocycline (oral gavage, 30 mg/kg per day) would reduce EEG abnormalities. We tested if minocycline treatment has specific effects based on the EEG measurement type (e.g., resting versus sound-evoked) from the frontal and auditory cortex of the Fmr1 KO mice. We show increased resting EEG gamma power and reduced phase locking to time varying stimuli as well as the 40 Hz auditory steady state response in the Fmr1 KO mice in the pre-drug condition. Minocycline treatment increased gamma band phase locking in response to auditory stimuli, and reduced sound-evoked power of auditory event related potentials (ERP) in Fmr1 KO mice compared to vehicle treatment. Minocycline reduced resting EEG gamma power in Fmr1 KO mice, but this effect was similar to vehicle treatment. We also report frequency band-specific effects on EEG responses. Taken together, these data indicate that sound-evoked EEG responses may serve as more sensitive measures, compared to resting EEG measures, to isolate minocycline effects from placebo in humans with FXS. Given the use of minocycline and EEG recordings in a number of neurodegenerative and neurodevelopmental conditions, these findings may be more broadly applicable in translational neuroscience.
Collapse
Affiliation(s)
- Jonathan W Lovelace
- Department of Psychology and Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States
| | - Iryna M Ethell
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States.,Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Devin K Binder
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States.,Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Khaleel A Razak
- Department of Psychology and Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States.,Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
30
|
Telias M. Pharmacological Treatments for Fragile X Syndrome Based on Synaptic Dysfunction. Curr Pharm Des 2020; 25:4394-4404. [PMID: 31682210 DOI: 10.2174/1381612825666191102165206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 10/31/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND Fragile X syndrome (FXS) is the most common form of monogenic hereditary cognitive impairment, including intellectual disability, autism, hyperactivity, and epilepsy. METHODS This article reviews the literature pertaining to the role of synaptic dysfunction in FXS. RESULTS In FXS, synaptic dysfunction alters the excitation-inhibition ratio, dysregulating molecular and cellular processes underlying cognition, learning, memory, and social behavior. Decades of research have yielded important hypotheses that could explain, at least in part, the development of these neurological disorders in FXS patients. However, the main goal of translating lab research in animal models to pharmacological treatments in the clinic has been so far largely unsuccessful, leaving FXS a still incurable disease. CONCLUSION In this concise review, we summarize and analyze the main hypotheses proposed to explain synaptic dysregulation in FXS, by reviewing the scientific evidence that led to pharmaceutical clinical trials and their outcome.
Collapse
Affiliation(s)
- Michael Telias
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, United States
| |
Collapse
|
31
|
Pirbhoy PS, Rais M, Lovelace JW, Woodard W, Razak KA, Binder DK, Ethell IM. Acute pharmacological inhibition of matrix metalloproteinase-9 activity during development restores perineuronal net formation and normalizes auditory processing in Fmr1 KO mice. J Neurochem 2020; 155:538-558. [PMID: 32374912 DOI: 10.1111/jnc.15037] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/31/2020] [Accepted: 04/30/2020] [Indexed: 12/13/2022]
Abstract
Individuals with Fragile X Syndrome (FXS) and autism spectrum disorder (ASD) exhibit cognitive impairments, social deficits, increased anxiety, and sensory hyperexcitability. Previously, we showed that elevated levels of matrix metalloproteinase-9 (MMP-9) may contribute to abnormal development of parvalbumin (PV) interneurons and perineuronal nets (PNNs) in the developing auditory cortex (AC) of Fmr1 knock-out (KO) mice, which likely underlie auditory hypersensitivity. Thus, MMP-9 may serve as a potential target for treatment of auditory hypersensitivity in FXS. Here, we used the MMP-2/9 inhibitor, SB-3CT, to pharmacologically inhibit MMP-9 activity during a specific developmental period and to test whether inhibition of MMP-9 activity reverses neural oscillation deficits and behavioral impairments by enhancing PNN formation around PV cells in Fmr1 KO mice. Electroencephalography (EEG) was used to measure resting state and sound-evoked electrocortical activity in auditory and frontal cortices of postnatal day (P)22-23 male mice before and one-day after treatment with SB-3CT (25 mg/kg) or vehicle. At P27-28, animal behaviors were tested to measure the effects of the treatment on anxiety and hyperactivity. Results show that acute inhibition of MMP-9 activity improved evoked synchronization to auditory stimuli and ameliorated mouse behavioral deficits. MMP-9 inhibition enhanced PNN formation, increased PV levels and TrkB phosphorylation yet reduced Akt phosphorylation in the AC of Fmr1 KO mice. Our results show that MMP-9 inhibition during early postnatal development is beneficial in reducing some auditory processing deficits in the FXS mouse model and may serve as a candidate therapeutic for reversing sensory hypersensitivity in FXS and possibly other ASDs.
Collapse
Affiliation(s)
- Patricia S Pirbhoy
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Maham Rais
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Jonathan W Lovelace
- Department of Psychology, University of California Riverside, Riverside, CA, USA
| | - Walker Woodard
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Khaleel A Razak
- Department of Psychology, University of California Riverside, Riverside, CA, USA
| | - Devin K Binder
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Iryna M Ethell
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| |
Collapse
|
32
|
Razak KA, Dominick KC, Erickson CA. Developmental studies in fragile X syndrome. J Neurodev Disord 2020; 12:13. [PMID: 32359368 PMCID: PMC7196229 DOI: 10.1186/s11689-020-09310-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 02/13/2020] [Indexed: 01/27/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common single gene cause of autism and intellectual disabilities. Humans with FXS exhibit increased anxiety, sensory hypersensitivity, seizures, repetitive behaviors, cognitive inflexibility, and social behavioral impairments. The main purpose of this review is to summarize developmental studies of FXS in humans and in the mouse model, the Fmr1 knockout mouse. The literature presents considerable evidence that a number of early developmental deficits can be identified and that these early deficits chart a course of altered developmental experience leading to symptoms well characterized in adolescents and adults. Nevertheless, a number of critical issues remain unclear or untested regarding the development of symptomology and underlying mechanisms. First, what is the role of FMRP, the protein product of Fmr1 gene, during different developmental ages? Does the absence of FMRP during early development lead to irreversible changes, or could reintroduction of FMRP or therapeutics aimed at FMRP-interacting proteins/pathways hold promise when provided in adults? These questions have implications for clinical trial designs in terms of optimal treatment windows, but few studies have systematically addressed these issues in preclinical and clinical work. Published studies also point to complex trajectories of symptom development, leading to the conclusion that single developmental time point studies are unlikely to disambiguate effects of genetic mutation from effects of altered developmental experience and compensatory plasticity. We conclude by suggesting a number of experiments needed to address these major gaps in the field.
Collapse
Affiliation(s)
- Khaleel A Razak
- Department of Psychology and Graduate Neuroscience Program, University of California, Riverside, USA
| | - Kelli C Dominick
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA.,Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 4002, Cincinnati, OH, 45229, USA
| | - Craig A Erickson
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA. .,Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 4002, Cincinnati, OH, 45229, USA.
| |
Collapse
|
33
|
Nguyen AO, Binder DK, Ethell IM, Razak KA. Abnormal development of auditory responses in the inferior colliculus of a mouse model of Fragile X Syndrome. J Neurophysiol 2020; 123:2101-2121. [PMID: 32319849 DOI: 10.1152/jn.00706.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Sensory processing abnormalities are frequently associated with autism spectrum disorders, but the underlying mechanisms are unclear. Here we studied auditory processing in a mouse model of Fragile X Syndrome (FXS), a leading known genetic cause of autism and intellectual disability. Both humans with FXS and the Fragile X mental retardation gene (Fmr1) knockout (KO) mouse model show auditory hypersensitivity, with the latter showing a strong propensity for audiogenic seizures (AGS) early in development. Because midbrain abnormalities cause AGS, we investigated whether the inferior colliculus (IC) of the Fmr1 KO mice shows abnormal auditory processing compared with wild-type (WT) controls at specific developmental time points. Using antibodies against neural activity marker c-Fos, we found increased density of c-Fos+ neurons in the IC, but not auditory cortex, of Fmr1 KO mice at P21 and P34 following sound presentation. In vivo single-unit recordings showed that IC neurons of Fmr1 KO mice are hyperresponsive to tone bursts and amplitude-modulated tones during development and show broader frequency tuning curves. There were no differences in rate-level responses or phase locking to amplitude-modulated tones in IC neurons between genotypes. Taken together, these data provide evidence for the development of auditory hyperresponsiveness in the IC of Fmr1 KO mice. Although most human and mouse work in autism and sensory processing has centered on the forebrain, our new findings, along with recent work on the lower brainstem, suggest that abnormal subcortical responses may underlie auditory hypersensitivity in autism spectrum disorders.NEW & NOTEWORTHY Autism spectrum disorders (ASD) are commonly associated with sensory sensitivity issues, but the underlying mechanisms are unclear. This study presents novel evidence for neural correlates of auditory hypersensitivity in the developing inferior colliculus (IC) in Fmr1 knockout (KO) mouse, a mouse model of Fragile X Syndrome (FXS), a leading genetic cause of ASD. Responses begin to show genotype differences between postnatal days 14 and 21, suggesting an early developmental treatment window.
Collapse
Affiliation(s)
- Anna O Nguyen
- Bioengineering Program, University of California, Riverside, California
| | - Devin K Binder
- Graduate Neuroscience Program, University of California, Riverside, California.,Division of Biomedical Sciences, University of California, Riverside, California
| | - Iryna M Ethell
- Graduate Neuroscience Program, University of California, Riverside, California.,Division of Biomedical Sciences, University of California, Riverside, California
| | - Khaleel A Razak
- Graduate Neuroscience Program, University of California, Riverside, California.,Psychology Department, University of California, Riverside, California
| |
Collapse
|
34
|
McCullagh EA, Rotschafer SE, Auerbach BD, Klug A, Kaczmarek LK, Cramer KS, Kulesza RJ, Razak KA, Lovelace JW, Lu Y, Koch U, Wang Y. Mechanisms underlying auditory processing deficits in Fragile X syndrome. FASEB J 2020; 34:3501-3518. [PMID: 32039504 DOI: 10.1096/fj.201902435r] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/31/2019] [Accepted: 01/18/2020] [Indexed: 01/14/2023]
Abstract
Autism spectrum disorders (ASD) are strongly associated with auditory hypersensitivity or hyperacusis (difficulty tolerating sounds). Fragile X syndrome (FXS), the most common monogenetic cause of ASD, has emerged as a powerful gateway for exploring underlying mechanisms of hyperacusis and auditory dysfunction in ASD. This review discusses examples of disruption of the auditory pathways in FXS at molecular, synaptic, and circuit levels in animal models as well as in FXS individuals. These examples highlight the involvement of multiple mechanisms, from aberrant synaptic development and ion channel deregulation of auditory brainstem circuits, to impaired neuronal plasticity and network hyperexcitability in the auditory cortex. Though a relatively new area of research, recent discoveries have increased interest in auditory dysfunction and mechanisms underlying hyperacusis in this disorder. This rapidly growing body of data has yielded novel research directions addressing critical questions regarding the timing and possible outcomes of human therapies for auditory dysfunction in ASD.
Collapse
Affiliation(s)
- Elizabeth A McCullagh
- Department of Physiology and Biophysics, University of Colorado Anschutz, Aurora, CO, USA.,Department of Integrative Biology, Oklahoma State University, Stillwater, OK, USA
| | - Sarah E Rotschafer
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA.,Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA
| | - Benjamin D Auerbach
- Center for Hearing and Deafness, Department of Communicative Disorders & Sciences, SUNY at Buffalo, Buffalo, NY, USA
| | - Achim Klug
- Department of Physiology and Biophysics, University of Colorado Anschutz, Aurora, CO, USA
| | - Leonard K Kaczmarek
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
| | - Karina S Cramer
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Randy J Kulesza
- Department of Anatomy, Lake Erie College of Osteopathic Medicine, Erie, PA, USA
| | - Khaleel A Razak
- Department of Psychology, University of California, Riverside, CA, USA
| | | | - Yong Lu
- Department of Anatomy and Neurobiology, College of Medicine, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Ursula Koch
- Institute of Biology, Neurophysiology, Freie Universität Berlin, Berlin, Germany
| | - Yuan Wang
- Department of Biomedical Sciences, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| |
Collapse
|
35
|
Jonak CR, Lovelace JW, Ethell IM, Razak KA, Binder DK. Multielectrode array analysis of EEG biomarkers in a mouse model of Fragile X Syndrome. Neurobiol Dis 2020; 138:104794. [PMID: 32036032 DOI: 10.1016/j.nbd.2020.104794] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/27/2020] [Accepted: 02/05/2020] [Indexed: 02/06/2023] Open
Abstract
Fragile X Syndrome (FXS) is a leading known genetic cause of intellectual disability with symptoms that include increased anxiety and social and sensory processing deficits. Recent EEG studies in humans with FXS have identified neural oscillation deficits that include increased resting state gamma power, increased amplitude of auditory evoked potentials, and reduced inter-trial phase coherence of sound-evoked gamma oscillations. Identification of comparable EEG biomarkers in mouse models of FXS could facilitate the pre-clinical to clinical therapeutic pipeline. However, while human EEG studies have involved 128-channel scalp EEG acquisition, no mouse studies have been performed with more than three EEG channels. In the current study, we employed a recently developed 30-channel mouse multielectrode array (MEA) system to record and analyze resting and stimulus-evoked EEG signals in WT vs. Fmr1 KO mice. Using this system, we now report robust MEA-derived phenotypes including higher resting EEG power, altered event-related potentials (ERPs) and reduced inter-trial phase coherence to auditory chirp stimuli in Fmr1 KO mice that are remarkably similar to those reported in humans with FXS. We propose that the MEA system can be used for: (i) derivation of higher-level EEG parameters; (ii) EEG biomarkers for drug testing; and (ii) mechanistic studies of FXS pathophysiology.
Collapse
Affiliation(s)
- Carrie R Jonak
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, United States of America
| | - Jonathan W Lovelace
- Department of Psychology, University of California, Riverside, United States of America
| | - Iryna M Ethell
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, United States of America; Neuroscience Graduate Program, University of California, Riverside, United States of America
| | - Khaleel A Razak
- Neuroscience Graduate Program, University of California, Riverside, United States of America; Department of Psychology, University of California, Riverside, United States of America
| | - Devin K Binder
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, United States of America; Neuroscience Graduate Program, University of California, Riverside, United States of America.
| |
Collapse
|
36
|
Hyperexcitability and impaired intracortical inhibition in patients with fragile-X syndrome. Transl Psychiatry 2019; 9:312. [PMID: 31748507 PMCID: PMC6868148 DOI: 10.1038/s41398-019-0650-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 10/08/2019] [Accepted: 11/01/2019] [Indexed: 01/10/2023] Open
Abstract
Fragile-X syndrome (FXS) is characterized by neurological and psychiatric problems symptomatic of cortical hyperexcitability. Recent animal studies identified deficient γ-aminobutyricacid (GABA) inhibition as a key mechanism for hyperexcitability in FXS, but the GABA system remains largely unexplored in humans with the disorder. The primary objective of this study was to assess GABA-mediated inhibition and its relationship with hyperexcitability in patients with FXS. Transcranial magnetic stimulation (TMS) was used to assess cortical and corticospinal inhibitory and excitatory mechanisms in 18 patients with a molecular diagnosis of FXS and 18 healthy controls. GABA-mediated inhibition was measured with short-interval intracortical inhibition (GABAA), long-interval intracortical inhibition (GABAB), and the corticospinal silent period (GABAA+B). Net intracortical facilitation involving glutamate was assessed with intracortical facilitation, and corticospinal excitability was measured with the resting motor threshold. Results showed that FXS patients had significantly reduced short-interval intracortical inhibition, increased long-interval intracortical inhibition, and increased intracortical facilitation compared to healthy controls. In the FXS group, reduced short-interval intracortical inhibition was associated with heightened intracortical facilitation. Taken together, these results suggest that reduced GABAA inhibition is a plausible mechanism underlying cortical hyperexcitability in patients with FXS. These findings closely match those observed in animal models, supporting the translational validity of these markers for clinical research.
Collapse
|
37
|
Kulinich AO, Reinhard SM, Rais M, Lovelace JW, Scott V, Binder DK, Razak KA, Ethell IM. Beneficial effects of sound exposure on auditory cortex development in a mouse model of Fragile X Syndrome. Neurobiol Dis 2019; 134:104622. [PMID: 31698054 DOI: 10.1016/j.nbd.2019.104622] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/22/2019] [Accepted: 09/23/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is the most common genetic cause of autism and intellectual disability. Fragile X mental retardation gene (Fmr1) knock-out (KO) mice display core deficits of FXS, including abnormally increased sound-evoked responses, and show a delayed development of parvalbumin (PV) cells. Here, we present the surprising result that sound exposure during early development reduces correlates of auditory hypersensitivity in Fmr1 KO mice. METHODS Fmr1 KO and wild-type (WT) mice were raised in a sound-attenuated environment (AE) or sound-exposed (SE) to 14 kHz tones (5 Hz repetition rate) from P9 until P21. At P21-P23, event-related potentials (ERPs), dendritic spine density, PV expression and phosphorylation of tropomyosin receptor kinase B (TrkB) were analyzed in the auditory cortex of AE and SE mice. RESULTS Enhanced N1 amplitude of ERPs, impaired PV cell development, and increased spine density in layers (L) 2/3 and L5/6 excitatory neurons were observed in AE Fmr1 KO compared to WT mice. In contrast, developmental sound exposure normalized ERP N1 amplitude, density of PV cells and dendritic spines in SE Fmr1 KO mice. Finally, TrkB phosphorylation was reduced in AE Fmr1 KO, but was enhanced in SE Fmr1 KO mice, suggesting that BDNF-TrkB signaling may be regulated by sound exposure to influence PV cell development. CONCLUSIONS Our results demonstrate that sound exposure, but not attenuation, during early developmental window restores molecular, cellular and functional properties in the auditory cortex of Fmr1 KO mice, and suggest this approach as a potential treatment for sensory phenotypes in FXS.
Collapse
Affiliation(s)
- Anna O Kulinich
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Sarah M Reinhard
- Psychology Department, University of California, Riverside, CA, USA
| | - Maham Rais
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA; Biomedical Sciences Graduate Program, University of California, Riverside, CA, USA
| | | | - Veronica Scott
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Devin K Binder
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA; Neuroscience Graduate Program, University of California, Riverside, CA, USA; Biomedical Sciences Graduate Program, University of California, Riverside, CA, USA
| | - Khaleel A Razak
- Psychology Department, University of California, Riverside, CA, USA; Neuroscience Graduate Program, University of California, Riverside, CA, USA; Biomedical Sciences Graduate Program, University of California, Riverside, CA, USA.
| | - Iryna M Ethell
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA; Neuroscience Graduate Program, University of California, Riverside, CA, USA; Biomedical Sciences Graduate Program, University of California, Riverside, CA, USA.
| |
Collapse
|
38
|
Beroun A, Mitra S, Michaluk P, Pijet B, Stefaniuk M, Kaczmarek L. MMPs in learning and memory and neuropsychiatric disorders. Cell Mol Life Sci 2019; 76:3207-3228. [PMID: 31172215 PMCID: PMC6647627 DOI: 10.1007/s00018-019-03180-8] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 05/27/2019] [Accepted: 05/29/2019] [Indexed: 12/20/2022]
Abstract
Matrix metalloproteinases (MMPs) are a group of over twenty proteases, operating chiefly extracellularly to cleave components of the extracellular matrix, cell adhesion molecules as well as cytokines and growth factors. By virtue of their expression and activity patterns in animal models and clinical investigations, as well as functional studies with gene knockouts and enzyme inhibitors, MMPs have been demonstrated to play a paramount role in many physiological and pathological processes in the brain. In particular, they have been shown to influence learning and memory processes, as well as major neuropsychiatric disorders such as schizophrenia, various kinds of addiction, epilepsy, fragile X syndrome, and depression. A possible link connecting all those conditions is either physiological or aberrant synaptic plasticity where some MMPs, e.g., MMP-9, have been demonstrated to contribute to the structural and functional reorganization of excitatory synapses that are located on dendritic spines. Another common theme linking the aforementioned pathological conditions is neuroinflammation and MMPs have also been shown to be important mediators of immune responses.
Collapse
Affiliation(s)
- Anna Beroun
- BRAINCITY, Nencki Institute, Pasteura 3, 02-093, Warsaw, Poland
| | | | - Piotr Michaluk
- BRAINCITY, Nencki Institute, Pasteura 3, 02-093, Warsaw, Poland
| | - Barbara Pijet
- BRAINCITY, Nencki Institute, Pasteura 3, 02-093, Warsaw, Poland
| | | | - Leszek Kaczmarek
- BRAINCITY, Nencki Institute, Pasteura 3, 02-093, Warsaw, Poland.
| |
Collapse
|
39
|
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder that causes intellectual disability. It is a leading known genetic cause of autism. In addition to cognitive, social, and communication deficits, humans with FXS demonstrate abnormal sensory processing including sensory hypersensitivity. Sensory hypersensitivity commonly manifests as auditory, tactile, or visual defensiveness or avoidance. Clinical, behavioral, and electrophysiological studies consistently show auditory hypersensitivity, impaired habituation to repeated sounds, and reduced auditory attention in humans with FXS. Children with FXS also exhibit significant visuospatial impairments. Studies in infants and toddlers with FXS have documented impairments in processing texture-defined motion stimuli, temporal flicker, perceiving ordinal numerical sequence, and the ability to maintain the identity of dynamic object information during occlusion. Consistent with the observations in humans with FXS, fragile X mental retardation 1 ( Fmr1) gene knockout (KO) rodent models of FXS also show seizures, abnormal visual-evoked responses, auditory hypersensitivity, and abnormal processing at multiple levels of the auditory system, including altered acoustic startle responses. Among other sensory symptoms, individuals with FXS exhibit tactile defensiveness. Fmr1 KO mice also show impaired encoding of tactile stimulation frequency and larger size of receptive fields in the somatosensory cortex. Since sensory deficits are relatively more tractable from circuit mechanisms and developmental perspectives than more complex social behaviors, the focus of this review is on clinical, functional, and structural studies that outline the auditory, visual, and somatosensory processing deficits in FXS. The similarities in sensory phenotypes between humans with FXS and animal models suggest a likely conservation of basic sensory processing circuits across species and may provide a translational platform to not just develop biomarkers but also to understand underlying mechanisms. We argue that preclinical studies in animal models of FXS can facilitate the ongoing search for new therapeutic approaches in FXS by understanding mechanisms of basic sensory processing circuits and behaviors that are conserved across species.
Collapse
Affiliation(s)
- Maham Rais
- 1 Division of Biomedical Sciences, University of California Riverside School of Medicine, CA, USA.,2 Biomedical Sciences Graduate Program, University of California Riverside, CA, USA
| | - Devin K Binder
- 1 Division of Biomedical Sciences, University of California Riverside School of Medicine, CA, USA.,2 Biomedical Sciences Graduate Program, University of California Riverside, CA, USA.,3 Neuroscience Graduate Program, University of California Riverside, CA, USA
| | - Khaleel A Razak
- 2 Biomedical Sciences Graduate Program, University of California Riverside, CA, USA.,3 Neuroscience Graduate Program, University of California Riverside, CA, USA.,4 Psychology Department, University of California Riverside, CA, USA
| | - Iryna M Ethell
- 1 Division of Biomedical Sciences, University of California Riverside School of Medicine, CA, USA.,2 Biomedical Sciences Graduate Program, University of California Riverside, CA, USA.,3 Neuroscience Graduate Program, University of California Riverside, CA, USA
| |
Collapse
|
40
|
Reversal of ultrasonic vocalization deficits in a mouse model of Fragile X Syndrome with minocycline treatment or genetic reduction of MMP-9. Behav Brain Res 2019; 372:112068. [PMID: 31271818 DOI: 10.1016/j.bbr.2019.112068] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/29/2019] [Accepted: 06/30/2019] [Indexed: 01/07/2023]
Abstract
Fragile X Syndrome (FXS) is a leading genetic cause of autism and intellectual disabilities. The Fmr1 knockout (KO) mouse is a commonly studied pre-clinical model of FXS. Adult male Fmr1 KO mice produce fewer ultrasonic vocalizations (USVs) during mating, suggestive of abnormal social communication. Minocycline treatment for 2 months from birth alleviates a number of FXS phenotypes in mice, including USV call rate deficits. In the current study, we investigated if treatment initiated past the early developmental period would be effective, given that in many cases, individuals with FXS are treated during later developmental periods. Wildtype (WT) and Fmr1 KO mice were treated with minocycline between postnatal day (P) 30 and P58. Mating-related USVs were then recorded from these mice between P75 and P90 and analyzed for call rate, duration, bandwidth, and peak frequency. Untreated Fmr1 KO mice call at a significantly reduced rate compared to untreated WT mice. After minocycline treatment from 1 to 2 months of age, WT and Fmr1 KO mice exhibited similar call rates, due to an increase in calling in the latter group. Minocycline is thought to be effective in reducing FXS symptoms by lowering matrix-metalloproteinase-9 (MMP-9) levels. To determine whether abnormal MMP-9 levels underlie USV deficits, we characterized USVs in Fmr1 KO mice which were heterozygous for MMP-9 (MMP-9+/-/Fmr1 KO). The MMP-9+/-/Fmr1 KO mice were between P75 and P90 at the time of recording. MMP-9+/-/Fmr1 KO mice exhibited significantly increased USV call rates, at times even exceeding WT rates. Taken together, these results suggest that minocycline may reverse USV call rate deficits in Fmr1 KO mice through attenuation of MMP-9 levels. These data suggest targeting MMP-9, even in late development, may reduce FXS symptoms.
Collapse
|
41
|
Kokash J, Alderson EM, Reinhard SM, Crawford CA, Binder DK, Ethell IM, Razak KA. Genetic reduction of MMP-9 in the Fmr1 KO mouse partially rescues prepulse inhibition of acoustic startle response. Brain Res 2019; 1719:24-29. [PMID: 31128097 DOI: 10.1016/j.brainres.2019.05.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/26/2019] [Accepted: 05/22/2019] [Indexed: 12/31/2022]
Abstract
Sensory processing abnormalities are consistently associated with autism, but the underlying mechanisms and treatment options are unclear. Fragile X Syndrome (FXS) is the leading known genetic cause of intellectual disabilities and autism. One debilitating symptom of FXS is hypersensitivity to sensory stimuli. Sensory hypersensitivity is seen in both humans with FXS and FXS mouse model, the Fmr1 knock out (Fmr1 KO) mouse. Abnormal sensorimotor gating may play a role in the hypersensitivity to sensory stimuli. Humans with FXS and Fmr1 KO mice show abnormalities in acoustic startle response (ASR) and prepulse inhibition (PPI) of startle, responses commonly used to quantify sensorimotor gating. Recent studies have suggested high levels of matrix metalloproteinase-9 (MMP-9) as a potential mechanism of sensory abnormalities in FXS. Here we tested the hypothesis that genetic reduction of MMP-9 in Fmr1 KO mice rescues ASR and PPI phenotypes in adult Fmr1 KO mice. We measured MMP-9 levels in the inferior colliculus (IC), an integral region of the PPI circuit, of WT and Fmr1 KO mice at P7, P12, P18, and P40. MMP-9 levels were higher in the IC of Fmr1 KO mice during early development (P7, P12), but not in adults. We compared ASR and PPI responses in young (P23-25) and adult (P50-80) Fmr1 KO mice to their age-matched wildtype (WT) controls. We found that both ASR and PPI were reduced in the young Fmr1 KO mice compared to age-matched WT mice. There was no genotype difference for ASR in the adult mice, but PPI was significantly reduced in the adult Fmr1 KO mice. The adult mouse data are similar to those observed in humans with FXS. Genetic reduction of MMP-9 in the Fmr1 KO mice resulted in a rescue of adult PPI responses to WT levels. Taken together, these results show sensorimotor gating abnormalities in Fmr1 KO mice, and suggest the potential for MMP-9 regulation as a therapeutic target to reduce sensory hypersensitivity.
Collapse
Affiliation(s)
- Jamiela Kokash
- Graduate Neuroscience Program, University of California, Riverside, United States
| | - Erin M Alderson
- Dept. of Psychology, University of California, Riverside, United States
| | - Sarah M Reinhard
- Dept. of Psychology, University of California, Riverside, United States
| | - Cynthia A Crawford
- Psychology Dept. California State University, San Bernardino, United States
| | - Devin K Binder
- Graduate Neuroscience Program, University of California, Riverside, United States; Biomedical Sciences Division, School of Medicine, University of California, Riverside, United States
| | - Iryna M Ethell
- Graduate Neuroscience Program, University of California, Riverside, United States; Biomedical Sciences Division, School of Medicine, University of California, Riverside, United States
| | - Khaleel A Razak
- Graduate Neuroscience Program, University of California, Riverside, United States; Dept. of Psychology, University of California, Riverside, United States.
| |
Collapse
|
42
|
Jalnapurkar I, Cochran DM, Frazier JA. New Therapeutic Options for Fragile X Syndrome. Curr Treat Options Neurol 2019; 21:12. [PMID: 30809735 DOI: 10.1007/s11940-019-0551-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE OF REVIEW The purpose of this review is to provide an overview of current research and clinical practice guidelines in fragile X syndrome (FXS) with regard to therapeutic approaches in the management of this condition. The authors summarize and discuss findings from relevant preclinical studies and results from clinical trials in human subjects with FXS. Additionally, we provide an outline of the basic framework for understanding and providing educational and psychosocial supports for these individuals. RECENT FINDINGS Current treatments in FXS are largely symptom based and focused on managing associated psychiatric and behavioral co-morbidities. While data from animal studies has been promising in providing targeted treatments to correct the underlying deficits at the cellular level, there have not been as robust findings in human trials. There are several targeted treatments for FXS currently under development. Individuals with FXS present with several behavioral challenges including anxiety, social withdrawal, ADHD, hyperarousal, self-injury, and aggression. Therapeutic services are often necessary, such as behavioral intervention, speech and language therapy, occupational therapy, and individualized educational support; adjunctive psychopharmacologic treatment is often helpful as well. It is important to address these symptoms and weigh the evidence for the use of medications that target the underlying neurobiology and pathophysiology of the syndrome.
Collapse
Affiliation(s)
- Isha Jalnapurkar
- Eunice Kennedy Shriver Center, University of Massachusetts Medical School, 55 Lake Ave N, Worcester, MA, 01655, USA. .,Department of Psychiatry, University of Massachusetts Medical School, 55 Lake Ave N, Worcester, MA, 01655, USA.
| | - David M Cochran
- Eunice Kennedy Shriver Center, University of Massachusetts Medical School, 55 Lake Ave N, Worcester, MA, 01655, USA.,Department of Psychiatry, University of Massachusetts Medical School, 55 Lake Ave N, Worcester, MA, 01655, USA
| | - Jean A Frazier
- Eunice Kennedy Shriver Center, University of Massachusetts Medical School, 55 Lake Ave N, Worcester, MA, 01655, USA.,Department of Psychiatry, University of Massachusetts Medical School, 55 Lake Ave N, Worcester, MA, 01655, USA
| |
Collapse
|
43
|
Goswami S, Cavalier S, Sridhar V, Huber KM, Gibson JR. Local cortical circuit correlates of altered EEG in the mouse model of Fragile X syndrome. Neurobiol Dis 2019; 124:563-572. [PMID: 30639292 DOI: 10.1016/j.nbd.2019.01.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 01/01/2019] [Accepted: 01/05/2019] [Indexed: 12/18/2022] Open
Abstract
Electroencephalogram (EEG) recordings in Fragile X syndrome (FXS) patients have revealed enhanced sensory responses, enhanced resting "gamma frequency" (30-100 Hz) activity, and a decreased ability for sensory stimuli to modulate cortical activity at gamma frequencies. Similar changes are observed in the FXS model mouse - the Fmr1 knockout. These alterations may become effective biomarkers for diagnosis and treatment of FXS. Therefore, it is critical to better understand what circuit properties underlie these changes. We employed Channelrhodopsin2 to optically activate local circuits in the auditory cortical region in brain slices to examine how changes in local circuit function may be related to EEG changes. We focused on layers 2/3 and 5 (L2/3 and L5). In Fmr1 knockout mice, light-driven excitation of L2/3 revealed hyperexcitability and increased gamma frequency power in both local L2/3 and L5 circuits. Moreover, there is increased synchrony in the gamma frequency band between L2/3 and L5. Hyperexcitability and increased gamma power were not observed in L5 with L5 light-driven excitation, indicating that these changes were layer-specific. A component of L2/3 network hyperexcitability is independent of ionotropic receptor mediated synaptic transmission and may be mediated by increased intrinsic excitability of L2/3 neurons. Finally, lovastatin, a candidate therapeutic compound for FXS that targets ERK signaling did not normalize changes in gamma activity. In conclusion, hyperactivity and increased gamma activity in local neocortical circuits, together with increased gamma synchrony between circuits, provide a putative substrate for EEG alterations observed in both FXS patients and the FXS mouse model.
Collapse
Affiliation(s)
- Sonal Goswami
- Department of Basic Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Sheridan Cavalier
- Department of Basic Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Vinay Sridhar
- Department of Basic Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Kimberly M Huber
- Department of Basic Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States.
| | - Jay R Gibson
- Department of Basic Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States.
| |
Collapse
|
44
|
Best Practices in Fragile X Syndrome Treatment Development. Brain Sci 2018; 8:brainsci8120224. [PMID: 30558274 PMCID: PMC6315698 DOI: 10.3390/brainsci8120224] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/10/2018] [Accepted: 12/12/2018] [Indexed: 01/14/2023] Open
Abstract
Preclinical studies using animal models of fragile X syndrome have yielded several agents that rescue a wide variety of phenotypes. However, translation of these treatments to humans with the disorder has not yet been successful, shedding light on a variety of limitations with both animal models and human trial design. As members of the Clinical Trials Committee of the National Fragile X Foundation, we have discussed a variety of recommendations at the level of preclinical development, transition from preclinical to human projects, family involvement, and multi-site trial planning. Our recommendations are made with the vision that effective new treatment will lie at the intersection of innovation, rigorous and reproducible research, and stakeholder involvement.
Collapse
|
45
|
Wen TH, Lovelace JW, Ethell IM, Binder DK, Razak KA. Developmental Changes in EEG Phenotypes in a Mouse Model of Fragile X Syndrome. Neuroscience 2018; 398:126-143. [PMID: 30528856 DOI: 10.1016/j.neuroscience.2018.11.047] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 11/27/2018] [Accepted: 11/29/2018] [Indexed: 01/29/2023]
Abstract
Fragile X Syndrome (FXS) is a leading genetic cause of autism and intellectual disabilities. Sensory-processing deficits are common in humans with FXS and an animal model, the Fmr1 knockout (KO) mouse, manifesting in the auditory system as debilitating hypersensitivity and abnormal electroencephalographic (EEG) and event-related potential (ERP) phenotypes. FXS is a neurodevelopmental disorder, but how EEG/ERP phenotypes change during development is unclear. Therefore, we characterized baseline and stimulus-evoked EEG in auditory and frontal cortex of developing (postnatal day (P) 21 and P30) and adult (P60) wildtype (WT) and Fmr1 KO mice with the FVB genetic background. We found that baseline gamma-band power and N1 amplitude of auditory ERP were increased in frontal cortex of Fmr1 KO mice during development and in adults. Baseline gamma power was increased in auditory cortex at P30. Genotype differences in stimulus-evoked gamma power were present in both cortical regions, but the direction and strength of the changes were age-dependent. These findings suggest that cortical deficits are present during early development and may contribute to sensory-processing deficits in FXS, which in turn may lead to anxiety and delayed language. Developmental changes in EEG measures indicate that observations at a single time-point during development are not reflective of FXS disease progression and highlight the need to identify developmental trajectories and optimal windows for treatment.
Collapse
Affiliation(s)
- Teresa H Wen
- Neuroscience Graduate Program, University of California Riverside, Riverside, CA 92521, USA
| | - Jonathan W Lovelace
- Psychology Department and Psychology Graduate Program, University of California Riverside, Riverside, CA 92521, USA
| | - Iryna M Ethell
- Neuroscience Graduate Program, University of California Riverside, Riverside, CA 92521, USA; Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Devin K Binder
- Neuroscience Graduate Program, University of California Riverside, Riverside, CA 92521, USA; Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Khaleel A Razak
- Neuroscience Graduate Program, University of California Riverside, Riverside, CA 92521, USA; Psychology Department and Psychology Graduate Program, University of California Riverside, Riverside, CA 92521, USA.
| |
Collapse
|
46
|
Jonak CR, Lovelace JW, Ethell IM, Razak KA, Binder DK. Reusable Multielectrode Array Technique for Electroencephalography in Awake Freely Moving Mice. Front Integr Neurosci 2018; 12:53. [PMID: 30416434 PMCID: PMC6213968 DOI: 10.3389/fnint.2018.00053] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 10/08/2018] [Indexed: 11/13/2022] Open
Abstract
Translational comparison of rodent models of neurological and neuropsychiatric diseases to human electroencephalography (EEG) biomarkers in these conditions will require multisite rodent EEG on the skull surface, rather than local area electrocorticography (ECoG) or multisite local field potential (LFP) recording. We have developed a technique for planar multielectrode array (MEA) implantation on the mouse skull surface, which enables multisite EEG in awake and freely moving mice and reusability of the MEA probes. With this method, we reliably obtain 30-channel low-noise EEG from awake mice. Baseline and stimulus-evoked EEG recordings can be readily obtained and analyzed. For example, we have demonstrated EEG responses to auditory stimuli. Broadband noise elicits reliable 30-channel auditory event-related potentials (ERPs), and chirp stimuli induce phase-locked EEG responses just as in human sound presentation paradigms. This method is unique in achieving chronic implantation of novel MEA technology onto the mouse skull surface for chronic multisite EEG recordings. Furthermore, we demonstrate a reliable method for reusing MEA probes for multiple serial implantations without loss of EEG quality. This skull surface MEA methodology can be used to obtain simultaneous multisite EEG recordings and to test EEG biomarkers in diverse mouse models of human neurological and neuropsychiatric diseases. Reusability of the MEA probes makes it more cost-effective to deploy this system for various studies.
Collapse
Affiliation(s)
- Carrie R Jonak
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Jonathan W Lovelace
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States.,Department of Psychology, University of California, Riverside, Riverside, CA, United States
| | - Iryna M Ethell
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States.,Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States
| | - Khaleel A Razak
- Department of Psychology, University of California, Riverside, Riverside, CA, United States.,Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States
| | - Devin K Binder
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States.,Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
47
|
Wen TH, Binder DK, Ethell IM, Razak KA. The Perineuronal 'Safety' Net? Perineuronal Net Abnormalities in Neurological Disorders. Front Mol Neurosci 2018; 11:270. [PMID: 30123106 PMCID: PMC6085424 DOI: 10.3389/fnmol.2018.00270] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 07/17/2018] [Indexed: 12/22/2022] Open
Abstract
Perineuronal nets (PNN) are extracellular matrix (ECM) assemblies that preferentially ensheath parvalbumin (PV) expressing interneurons. Converging evidence indicates that PV cells and PNN are impaired in a variety of neurological disorders. PNN development and maintenance is necessary for a number of processes within the CNS, including regulation of GABAergic cell function, protection of neurons from oxidative stress, and closure of developmental critical period plasticity windows. Understanding PNN functions may be essential for characterizing the mechanisms of altered cortical excitability observed in neurodegenerative and neurodevelopmental disorders. Indeed, PNN abnormalities have been observed in post-mortem brain tissues of patients with schizophrenia and Alzheimer’s disease. There is impaired development of PNNs and enhanced activity of its key regulator matrix metalloproteinase-9 (MMP-9) in Fragile X Syndrome, a common genetic cause of autism. MMP-9, a protease that cleaves ECM, is differentially regulated in a number of these disorders. Despite this, few studies have addressed the interactions between PNN expression, MMP-9 activity and neuronal excitability. In this review, we highlight the current evidence for PNN abnormalities in CNS disorders associated with altered network function and MMP-9 levels, emphasizing the need for future work targeting PNNs in pathophysiology and therapeutic treatment of neurological disorders.
Collapse
Affiliation(s)
- Teresa H Wen
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States
| | - Devin K Binder
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States.,Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Iryna M Ethell
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States.,Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Khaleel A Razak
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States.,Psychology Graduate Program, Department of Psychology, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
48
|
Lovelace JW, Ethell IM, Binder DK, Razak KA. Translation-relevant EEG phenotypes in a mouse model of Fragile X Syndrome. Neurobiol Dis 2018; 115:39-48. [PMID: 29605426 PMCID: PMC5969806 DOI: 10.1016/j.nbd.2018.03.012] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 02/16/2018] [Accepted: 03/28/2018] [Indexed: 01/29/2023] Open
Abstract
Identification of comparable biomarkers in humans and validated animal models will facilitate pre-clinical to clinical therapeutic pipelines to treat neurodevelopmental disorders. Fragile X Syndrome (FXS) is a leading known genetic cause of intellectual disability with symptoms that include increased anxiety, social and sensory processing deficits. Recent EEG studies in humans with FXS have identified neural oscillation deficits that include enhanced resting state gamma power and reduced inter-trial coherence of sound evoked gamma oscillations. To determine if analogous phenotypes are present in an animal model of FXS, we recorded EEGs in awake, freely moving Fmr1 knock out (KO) mice using similar stimuli as in the human studies. We report remarkably similar neural oscillation phenotypes in the Fmr1 KO mouse including enhanced resting state gamma power and reduced evoked gamma synchronization. The gamma band inter-trial coherence of neural response was reduced in both auditory and frontal cortex of Fmr1 KO mice stimulated with a sound whose envelope was modulated from 1 to 100 Hz, similar to that seen in humans with FXS. These deficits suggest a form of enhanced 'resting state noise' that interferes with the ability of the circuit to mount a synchronized response to sensory input, predicting specific sensory and cognitive deficits in FXS. The abnormal gamma oscillations are consistent with parvalbumin neuron and perineuronal net deficits seen in the Fmr1 KO mouse auditory cortex indicating that the EEG biomarkers are not only clinically relevant, but could also be used to probe cellular and circuit mechanisms of sensory hypersensitivity in FXS.
Collapse
Affiliation(s)
| | - Iryna M Ethell
- Neuroscience Graduate Program, University of California, Riverside, USA; Division of Biomedical Sciences, School of Medicine, University of California, Riverside, USA
| | - Devin K Binder
- Neuroscience Graduate Program, University of California, Riverside, USA; Division of Biomedical Sciences, School of Medicine, University of California, Riverside, USA
| | - Khaleel A Razak
- Department of Psychology, University of California, Riverside, USA; Neuroscience Graduate Program, University of California, Riverside, USA.
| |
Collapse
|
49
|
Tranfaglia MR, Thibodeaux C, Mason DJ, Brown D, Roberts I, Smith R, Guilliams T, Cogram P. Repurposing available drugs for neurodevelopmental disorders: The fragile X experience. Neuropharmacology 2018; 147:74-86. [PMID: 29792283 DOI: 10.1016/j.neuropharm.2018.05.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/30/2018] [Accepted: 05/02/2018] [Indexed: 10/17/2022]
Abstract
Many available drugs have been repurposed as treatments for neurodevelopmental disorders. In the specific case of fragile X syndrome, many clinical trials of available drugs have been conducted with the goal of disease modification. In some cases, detailed understanding of basic disease mechanisms has guided the choice of drugs for clinical trials, and several notable successes in fragile X clinical trials have led to common use of drugs such as minocycline in routine medical practice. Newer technologies like Disease-Gene Expression Matching (DGEM) may allow for more rapid identification of promising repurposing candidates. A DGEM study predicted that sulindac could be therapeutic for fragile X, and subsequent preclinical validation studies have shown promising results. The use of combinations of available drugs and nutraceuticals has the potential to greatly expand the options for repurposing, and may even be a viable business strategy. This article is part of the Special Issue entitled 'Drug Repurposing: old molecules, new ways to fast track drug discovery and development for CNS disorders'.
Collapse
Affiliation(s)
| | - Clare Thibodeaux
- Cures Within Reach, 125 S. Clark Street, 17th Floor, Chicago, IL 60603, USA.
| | - Daniel J Mason
- Healx Ltd., Park House, Castle Park, Cambridge, CB3 0DU, United Kingdom.
| | - David Brown
- Healx Ltd., Park House, Castle Park, Cambridge, CB3 0DU, United Kingdom
| | - Ian Roberts
- Healx Ltd., Park House, Castle Park, Cambridge, CB3 0DU, United Kingdom
| | - Richard Smith
- Healx Ltd., Park House, Castle Park, Cambridge, CB3 0DU, United Kingdom
| | - Tim Guilliams
- Healx Ltd., Park House, Castle Park, Cambridge, CB3 0DU, United Kingdom
| | - Patricia Cogram
- FRAXA-DVI, IEB, Las Encinas 3370, Ñuñoa, Santiago, Chile; Laboratory of Molecular Neuropsychiatry, Institute of Cognitive and Translational Neuroscience (INCyT), INECO Foundation, Favaloro University, National Scientific and Technical Research Council (CONICET), Pacheco de Melo 1854, CP 1126, Ciudad de Buenos Aires, Argentina; Institute of Ecology and Biodiversity, Faculty of Science, University of Chile, Las Palmeras 3425, Ñuñoa, Santiago, Chile.
| |
Collapse
|
50
|
Green J, Garg S. Annual Research Review: The state of autism intervention science: progress, target psychological and biological mechanisms and future prospects. J Child Psychol Psychiatry 2018; 59:424-443. [PMID: 29574740 DOI: 10.1111/jcpp.12892] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/18/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND There has been recent systematic review of key evidence in psychosocial intervention in autism but little review of biological treatments. METHODS We analyse the current literature from the perspective of intervention and mechanism targets across social and biological development. RESULTS The overall quality of trials evidence in autism intervention remains relatively low, despite some recent progress. Many treatments in common use have little or no evidence base. This is very concerning in such an important disorder. A variety of psychosocial interventions can show effect to improve some short-term effects on children's immediate dyadic social interactions, for instance with caregivers. But showing true effectiveness in this developmental disorder requires generalisation of such effects into wider social contexts, on autism symptoms and in long-term progress in development. Only a few interventions so far have begun to show this. A number of early phase interventions on biological targets have shown real promise, but none has yet progressed to larger scale effectiveness trials on behavioural or symptom outcomes. CONCLUSIONS There has been enough progress in psychosocial intervention research now to be able to begin to identify some evidence-based practice in autism treatment. To consolidate and improve outcomes, the next phase of intervention research needs improved trial design, and an iterative approach building on success. It may also include the testing of potential synergies between promising biological and psychosocial interventions.
Collapse
Affiliation(s)
- Jonathan Green
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK.,Manchester University NHS Foundation Trust, Manchester, UK.,Greater Manchester Mental Health NHS Foundation Trust, Manchester, UK
| | - Shruti Garg
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, Manchester, UK.,Manchester University NHS Foundation Trust, Manchester, UK.,Greater Manchester Mental Health NHS Foundation Trust, Manchester, UK
| |
Collapse
|