1
|
de Diego-Otero Y, El Bekay R, García-Guirado F, Sánchez-Salido L, Giráldez-Pérez RM. Apocynin, a Selective NADPH Oxidase (Nox2) Inhibitor, Ameliorates Behavioural and Learning Deficits in the Fragile X Syndrome Mouse Model. Biomedicines 2024; 12:2887. [PMID: 39767793 PMCID: PMC11673502 DOI: 10.3390/biomedicines12122887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/13/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Fragile X Syndrome (FXS) is associated with intellectual disability, hyperactivity, social anxiety and signs of autism. Hyperactivation of NADPH oxidase has been previously described in the brain of the male Fmr1-KO mouse. This work aims to demonstrate the efficacy of Apocynin, a specific NADPH oxidase inhibitor, in treating Fragile X mouse hallmarks. Methods: Free radicals, lipid and protein oxidation markers and behavioural and learning paradigms were measured after chronic treatment with orally administered vehicle, 10 mg/kg/day or 30 mg/kg/day of Apocynin. Results: The results revealed a reduction in testis weight, an increase in peritoneal fat, and no variation in body weight after chronic treatment. Furthermore, a reduction in hyperactivity was detected in Apocynin-treated male Fmr1-KO mice. Additionally, the higher dose of 30 mg/kg/day also improves behaviour and learning in the male Fmr1-KO mice, normalising free radical production and oxidative parameters. Moreover, a reduction in phospho-EKR1 and P47-Phox protein signals was observed in specific brain areas. Conclusions: Thus, chronic treatment with Apocynin could lead to a new therapeutic option for the Fragile X Syndrome.
Collapse
Affiliation(s)
- Yolanda de Diego-Otero
- Cellular Biology, Physiology and Immunology Department, University of Córdoba, 14014 Córdoba, Spain;
| | - Rajaa El Bekay
- Research Laboratory, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIO-NAND, Hospital Civil, 29009 Malaga, Spain
- Endocrinology and Nutrition Clinic Unit, Regional University Hospital of Málaga, 29009 Málaga, Spain
- CIBER of Physiopathology of Obesity and Nutrition (CIBERobn), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Francisco García-Guirado
- Research Laboratory, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIO-NAND, Hospital Civil, 29009 Malaga, Spain
| | - Lourdes Sánchez-Salido
- Research Laboratory, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIO-NAND, Hospital Civil, 29009 Malaga, Spain
| | - Rosa María Giráldez-Pérez
- Cellular Biology, Physiology and Immunology Department, University of Córdoba, 14014 Córdoba, Spain;
| |
Collapse
|
2
|
Kos J, Langiu M, Hellyer SD, Gregory KJ. Pharmacology, Signaling and Therapeutic Potential of Metabotropic Glutamate Receptor 5 Negative Allosteric Modulators. ACS Pharmacol Transl Sci 2024; 7:3671-3690. [PMID: 39698283 PMCID: PMC11651194 DOI: 10.1021/acsptsci.4c00213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/18/2024] [Accepted: 07/01/2024] [Indexed: 12/20/2024]
Abstract
Metabotropic glutamate receptors are a family of eight class C G protein-coupled receptors regulating higher order brain functions including cognition and motion. Metabotropic glutamate receptors have thus been heavily investigated as potential drug targets for treating neurological disorders. Drug discovery efforts directed toward metabotropic glutamate receptor subtype 5 (mGlu5) have been particularly fruitful, with a wealth of drug candidates and pharmacological tools identified. mGlu5 negative allosteric modulators (NAMs) are promising novel therapeutics for developmental, neuropsychiatric and neurodegenerative disorders (e.g., Alzheimer's Disease, Huntington's Disease, Parkinson's Disease, amyotrophic lateral sclerosis, autism spectrum disorders, substance use disorders, stroke, anxiety and depression) and show promise in ameliorating adverse effects induced by other medications (e.g., L-dopa induced dyskinesia in Parkinson's Disease). However, despite preclinical success, mGlu5 NAMs are yet to reach the market due to poor safety and efficacy profiles in clinical trials. Herein, we review the physiology and signal transduction of mGlu5. We provide a comprehensive critique of therapeutic options with respect to mGlu5 inhibitors, spanning from orthosteric antagonists to NAMs. Finally, we address the challenges associated with drug development and highlight future directions to guide rational drug discovery of safe and effective novel therapeutics.
Collapse
Affiliation(s)
- Jackson
A. Kos
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences and
Department of Pharmacology, Monash University, Parkville, VIC 3052, Australia
| | - Monica Langiu
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences and
Department of Pharmacology, Monash University, Parkville, VIC 3052, Australia
| | - Shane D. Hellyer
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences and
Department of Pharmacology, Monash University, Parkville, VIC 3052, Australia
| | - Karen J. Gregory
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences and
Department of Pharmacology, Monash University, Parkville, VIC 3052, Australia
- ARC
Centre for Cryo-electron Microscopy of Membrane Proteins, Monash University, Parkville, VIC 3052, Australia
| |
Collapse
|
3
|
Hakon J, Quattromani MJ, Sjölund C, Talhada D, Kim B, Moyanova S, Mastroiacovo F, Di Menna L, Olsson R, Englund E, Nicoletti F, Ruscher K, Bauer AQ, Wieloch T. Inhibiting metabotropic glutamate receptor 5 after stroke restores brain function and connectivity. Brain 2024; 147:186-200. [PMID: 37656990 PMCID: PMC10766240 DOI: 10.1093/brain/awad293] [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: 07/26/2022] [Revised: 06/12/2023] [Accepted: 08/04/2023] [Indexed: 09/03/2023] Open
Abstract
Stroke results in local neural disconnection and brain-wide neuronal network dysfunction leading to neurological deficits. Beyond the hyper-acute phase of ischaemic stroke, there is no clinically-approved pharmacological treatment that alleviates sensorimotor impairments. Functional recovery after stroke involves the formation of new or alternative neuronal circuits including existing neural connections. The type-5 metabotropic glutamate receptor (mGluR5) has been shown to modulate brain plasticity and function and is a therapeutic target in neurological diseases outside of stroke. We investigated whether mGluR5 influences functional recovery and network reorganization rodent models of focal ischaemia. Using multiple behavioural tests, we observed that treatment with negative allosteric modulators (NAMs) of mGluR5 (MTEP, fenobam and AFQ056) for 12 days, starting 2 or 10 days after stroke, restored lost sensorimotor functions, without diminishing infarct size. Recovery was evident within hours after initiation of treatment and progressed over the subsequent 12 days. Recovery was prevented by activation of mGluR5 with the positive allosteric modulator VU0360172 and accelerated in mGluR5 knock-out mice compared with wild-type mice. After stroke, multisensory stimulation by enriched environments enhanced recovery, a result prevented by VU0360172, implying a role of mGluR5 in enriched environment-mediated recovery. Additionally, MTEP treatment in conjunction with enriched environment housing provided an additive recovery enhancement compared to either MTEP or enriched environment alone. Using optical intrinsic signal imaging, we observed brain-wide disruptions in resting-state functional connectivity after stroke that were prevented by mGluR5 inhibition in distinct areas of contralesional sensorimotor and bilateral visual cortices. The levels of mGluR5 protein in mice and in tissue samples of stroke patients were unchanged after stroke. We conclude that neuronal circuitry subserving sensorimotor function after stroke is depressed by a mGluR5-dependent maladaptive plasticity mechanism that can be restored by mGluR5 inhibition. Post-acute stroke treatment with mGluR5 NAMs combined with rehabilitative training may represent a novel post-acute stroke therapy.
Collapse
Affiliation(s)
- Jakob Hakon
- Division of Neurosurgery, Department of Clinical Sciences, Laboratory for Experimental Brain Research, Lund University, Lund 221 84, Sweden
| | - Miriana J Quattromani
- Division of Neurosurgery, Department of Clinical Sciences, Laboratory for Experimental Brain Research, Lund University, Lund 221 84, Sweden
| | - Carin Sjölund
- Division of Neurosurgery, Department of Clinical Sciences, Laboratory for Experimental Brain Research, Lund University, Lund 221 84, Sweden
| | - Daniela Talhada
- Division of Neurosurgery, Department of Clinical Sciences, Laboratory for Experimental Brain Research, Lund University, Lund 221 84, Sweden
| | - Byungchan Kim
- Department of Radiology, Washington University, Saint Louis, MO 63110, USA
| | - Slavianka Moyanova
- Department of Molecular Pathology, IRCCS Neuromed, 86077 Pozzilli, Italy
| | | | - Luisa Di Menna
- Department of Molecular Pathology, IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Roger Olsson
- Department of Experimental Medical Sciences, Chemical Biology & Therapeutics, Lund University, Lund 221 84, Sweden
| | - Elisabet Englund
- Division of Pathology, Department of Clinical Sciences, Lund University, Lund 221 84, Sweden
| | - Ferdinando Nicoletti
- Department of Molecular Pathology, IRCCS Neuromed, 86077 Pozzilli, Italy
- Department of Physiology and Pharmacology, University of Rome La Sapienza, 00185 Rome, Italy
| | - Karsten Ruscher
- Division of Neurosurgery, Department of Clinical Sciences, Laboratory for Experimental Brain Research, Lund University, Lund 221 84, Sweden
| | - Adam Q Bauer
- Department of Radiology, Washington University, Saint Louis, MO 63110, USA
| | - Tadeusz Wieloch
- Division of Neurosurgery, Department of Clinical Sciences, Laboratory for Experimental Brain Research, Lund University, Lund 221 84, Sweden
| |
Collapse
|
4
|
D'Antoni S, Schiavi S, Buzzelli V, Giuffrida S, Feo A, Ascone F, Busceti CL, Nicoletti F, Trezza V, Catania MV. Group I and group II metabotropic glutamate receptors are upregulated in the synapses of infant rats prenatally exposed to valproic acid. Psychopharmacology (Berl) 2023; 240:2617-2629. [PMID: 37707611 PMCID: PMC10640443 DOI: 10.1007/s00213-023-06457-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 08/23/2023] [Indexed: 09/15/2023]
Abstract
RATIONALE Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impaired social interaction and restricted/stereotyped behavior. Prenatal exposure to valproic acid (VPA) is associated with an increased risk of developing ASD in humans and autistic-like behaviors in rodents. Increasing evidence indicates that dysfunctions of glutamate receptors at synapses are associated with ASD. In the VPA rat model, an involvement of glutamate receptors in autism-like phenotypes has been suggested; however, few studies were carried out on metabotropic glutamate (mGlu) receptors. OBJECTIVES We examined the protein expression levels of group I (mGlu1 and mGlu5) and group II (mGlu2/3) mGlu receptors in rats prenatally exposed to VPA and evaluated the effect of mGlu receptor modulation on an early autism-like phenotype in these animals. METHODS We used western blotting analysis on synaptosomes obtained from forebrain of control and VPA rats at different ages (postnatal day P13, 35, 90) and carried out ultrasonic vocalization (USV) emission test in infant control and VPA rats. RESULTS The expression levels of all these receptors were significantly increased in infant VPA rats. No changes were detected in adolescent and adult rats. An acute treatment with the preferential mGlu2/3 antagonist, LY341495, attenuated the impairment in the USV emission in VPA rats. No effect was observed after a treatment with the mGlu5 selective antagonist, MTEP. CONCLUSIONS Our findings demonstrate that the expression of group I and group II mGlu receptors is upregulated at synapses of infant VPA rats and suggest that mGlu2/3 receptor modulation may have a therapeutic potential in ASD.
Collapse
Affiliation(s)
- Simona D'Antoni
- Institute for Biomedical Research and Innovation, National Research Council (IRIB-CNR), Catania, Italy
| | - Sara Schiavi
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
| | - Valeria Buzzelli
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
| | - Samuele Giuffrida
- Institute for Biomedical Research and Innovation, National Research Council (IRIB-CNR), Catania, Italy
| | - Alessandro Feo
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
| | - Fabrizio Ascone
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
| | | | - Ferdinando Nicoletti
- IRCCS Neuromed, Pozzilli, Italy
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Viviana Trezza
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
- Neuroendocrinology, Metabolism and Neuropharmacology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Maria Vincenza Catania
- Institute for Biomedical Research and Innovation, National Research Council (IRIB-CNR), Catania, Italy.
| |
Collapse
|
5
|
Takeda R, Ishii R, Parvin S, Shiozawa A, Nogi T, Sasaki Y. Novel presynaptic assay system revealed that metformin ameliorates exaggerated synaptic release and Munc18-1 accumulation in presynapses of neurons from Fragile X syndrome mouse model. Neurosci Lett 2023; 810:137317. [PMID: 37286070 DOI: 10.1016/j.neulet.2023.137317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/13/2023] [Accepted: 05/30/2023] [Indexed: 06/09/2023]
Abstract
Fragile X syndrome (FXS) is a developmental disorder characterized by intellectual disability and autistic-like behaviors. These symptoms are supposed to result from dysregulated translation in pre- and postsynapses, resulting in aberrant synaptic plasticity. Although most drug development research on FXS has focused on aberrant postsynaptic functions by excess translation in postsynapses, the effect of drug candidates on FXS in presynaptic release is largely unclear. In this report, we developed a novel assay system using neuron ball culture with beads to induce presynapse formation, allowing for the analysis of presynaptic phenotypes, including presynaptic release. Metformin, which is shown to rescue core phenotypes in FXS mouse model by normalizing dysregulated translation, ameliorated the exaggerated presynaptic release of neurons of FXS model mouse using this assay system. Furthermore, metformin suppressed the excess accumulation of the active zone protein Munc18-1, which is supposed to be locally translated in presynapses. These results suggest that metformin rescues both postsynaptic and presynaptic phenotypes by inhibiting excess translation in FXS neurons.
Collapse
Affiliation(s)
- Renoma Takeda
- Functional Structure Science Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, 1-7-29 Suehiro-cho, Tsurumi-ward, Yokohama 230-0045, Japan
| | - Rie Ishii
- Functional Structure Science Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, 1-7-29 Suehiro-cho, Tsurumi-ward, Yokohama 230-0045, Japan
| | - Shumaia Parvin
- Functional Structure Science Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, 1-7-29 Suehiro-cho, Tsurumi-ward, Yokohama 230-0045, Japan
| | - Aki Shiozawa
- Structural Biology Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, 1-7-29 Suehiro-cho, Tsurumi-ward, Yokohama 230-0045, Japan
| | - Terukazu Nogi
- Structural Biology Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, 1-7-29 Suehiro-cho, Tsurumi-ward, Yokohama 230-0045, Japan
| | - Yukio Sasaki
- Functional Structure Science Laboratory, Department of Medical Life Science, Yokohama City University Graduate School of Medical Life Science, 1-7-29 Suehiro-cho, Tsurumi-ward, Yokohama 230-0045, Japan.
| |
Collapse
|
6
|
Abstract
The fragile X-related disorders are an important group of hereditary disorders that are caused by expanded CGG repeats in the 5' untranslated region of the FMR1 gene or by mutations in the coding sequence of this gene. Two categories of pathological CGG repeats are associated with these disorders, full mutation alleles and shorter premutation alleles. Individuals with full mutation alleles develop fragile X syndrome, which causes autism and intellectual disability, whereas those with premutation alleles, which have shorter CGG expansions, can develop fragile X-associated tremor/ataxia syndrome, a progressive neurodegenerative disease. Thus, fragile X-related disorders can manifest as neurodegenerative or neurodevelopmental disorders, depending on the size of the repeat expansion. Here, we review mouse models of fragile X-related disorders and discuss how they have informed our understanding of neurodegenerative and neurodevelopmental disorders. We also assess the translational value of these models for developing rational targeted therapies for intellectual disability and autism disorders.
Collapse
Affiliation(s)
- Rob Willemsen
- Department of Clinical Genetics, Erasmus University Medical Center, 3015 CN Rotterdam, the Netherlands. Department of Medical Genetics, University of Antwerp, 2000 Antwerp, Belgium
| | - R. Frank Kooy
- Department of Clinical Genetics, Erasmus University Medical Center, 3015 CN Rotterdam, the Netherlands. Department of Medical Genetics, University of Antwerp, 2000 Antwerp, Belgium
| |
Collapse
|
7
|
Witkin JM, Pandey KP, Smith JL. Clinical investigations of compounds targeting metabotropic glutamate receptors. Pharmacol Biochem Behav 2022; 219:173446. [PMID: 35987339 DOI: 10.1016/j.pbb.2022.173446] [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: 04/28/2022] [Revised: 06/22/2022] [Accepted: 08/08/2022] [Indexed: 11/15/2022]
Abstract
Pharmacological modulation of glutamate has long been considered to be of immense therapeutic utility. The metabotropic glutamate receptors (mGluRs) are potential targets for safely altering glutamate-driven excitation. Data support the potential therapeutic use of mGluR modulators in the treatment of anxiety, depression, schizophrenia, and other psychiatric disorders, pain, epilepsy, as well as neurodegenerative and neurodevelopmental disorders. For each of the three mGluR groups, compounds have been constructed that produce either potentiation or functional blockade. PET ligands for mGlu5Rs have been studied in a range of patient populations and several mGlu5R antagonists have been tested for potential efficacy in patients including mavoglurant, diploglurant, basimglurant, GET 73, and ADX10059. Efficacy with mGlu5R antagonists has been reported in trials with patients with gastroesophageal reflux disease; data from patients with Parkinson's disease or Fragile X syndrome have not been as robust as hoped. Fenobam was approved for use as an anxiolytic prior to its recognition as an mGlu5R antagonist. mGlu2/3R agonists (pomaglumated methionil) and mGlu2R agonists (JNJ-40411813, AZD 8529, and LY2979165) have been studied in patients with schizophrenia with promising but mixed results. Antagonists of mGlu2/3Rs (decoglurant and TS-161) have been studied in depression where TS-161 has advanced into a planned Phase 2 study in treatment-resistant depression. The Group III mGluRs are the least developed of the mGluR receptor targets. The mGlu4R potentiator, foliglurax, did not meet its primary endpoint in patients with Parkinson's disease. Ongoing efforts to develop mGluR-targeted compounds continue to promise these glutamate modulators as medicines for psychiatric and neurological disorders.
Collapse
Affiliation(s)
- Jeffrey M Witkin
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent, Indianapolis, IN, USA; Department of Chemistry & Biochemistry, Milwaukee Institute of Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, WI, USA; RespireRx Pharmaceuticals Inc, Glen Rock, NJ, USA.
| | - Kamal P Pandey
- Department of Chemistry & Biochemistry, Milwaukee Institute of Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Jodi L Smith
- Laboratory of Antiepileptic Drug Discovery, Ascension St. Vincent, Indianapolis, IN, USA
| |
Collapse
|
8
|
Jiang CC, Lin LS, Long S, Ke XY, Fukunaga K, Lu YM, Han F. Signalling pathways in autism spectrum disorder: mechanisms and therapeutic implications. Signal Transduct Target Ther 2022; 7:229. [PMID: 35817793 PMCID: PMC9273593 DOI: 10.1038/s41392-022-01081-0] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/19/2022] [Accepted: 06/23/2022] [Indexed: 02/06/2023] Open
Abstract
Autism spectrum disorder (ASD) is a prevalent and complex neurodevelopmental disorder which has strong genetic basis. Despite the rapidly rising incidence of autism, little is known about its aetiology, risk factors, and disease progression. There are currently neither validated biomarkers for diagnostic screening nor specific medication for autism. Over the last two decades, there have been remarkable advances in genetics, with hundreds of genes identified and validated as being associated with a high risk for autism. The convergence of neuroscience methods is becoming more widely recognized for its significance in elucidating the pathological mechanisms of autism. Efforts have been devoted to exploring the behavioural functions, key pathological mechanisms and potential treatments of autism. Here, as we highlight in this review, emerging evidence shows that signal transduction molecular events are involved in pathological processes such as transcription, translation, synaptic transmission, epigenetics and immunoinflammatory responses. This involvement has important implications for the discovery of precise molecular targets for autism. Moreover, we review recent insights into the mechanisms and clinical implications of signal transduction in autism from molecular, cellular, neural circuit, and neurobehavioural aspects. Finally, the challenges and future perspectives are discussed with regard to novel strategies predicated on the biological features of autism.
Collapse
Affiliation(s)
- Chen-Chen Jiang
- International Joint Laboratory for Drug Target of Critical Illnesses; Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Li-Shan Lin
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Sen Long
- Department of Pharmacy, Hangzhou Seventh People's Hospital, Mental Health Center Zhejiang University School of Medicine, Hangzhou, 310013, China
| | - Xiao-Yan Ke
- Child Mental Health Research Center, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Kohji Fukunaga
- Department of CNS Drug Innovation, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980-8578, Japan
| | - Ying-Mei Lu
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China.
| | - Feng Han
- International Joint Laboratory for Drug Target of Critical Illnesses; Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
- Institute of Brain Science, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, China.
- Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215002, China.
| |
Collapse
|
9
|
Hunt JFV, Li M, Risgaard R, Ananiev GE, Wildman S, Zhang F, Bugni TS, Zhao X, Bhattacharyya A. High Throughput Small Molecule Screen for Reactivation of FMR1 in Fragile X Syndrome Human Neural Cells. Cells 2021; 11:cells11010069. [PMID: 35011630 PMCID: PMC8750025 DOI: 10.3390/cells11010069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 11/16/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common inherited cause of autism and intellectual disability. The majority of FXS cases are caused by transcriptional repression of the FMR1 gene due to epigenetic changes that are not recapitulated in current animal disease models. FXS patient induced pluripotent stem cell (iPSC)-derived gene edited reporter cell lines enable novel strategies to discover reactivators of FMR1 expression in human cells on a much larger scale than previously possible. Here, we describe the workflow using FXS iPSC-derived neural cell lines to conduct a massive, unbiased screen for small molecule activators of the FMR1 gene. The proof-of-principle methodology demonstrates the utility of human stem-cell-based methodology for the untargeted discovery of reactivators of the human FMR1 gene that can be applied to other diseases.
Collapse
Affiliation(s)
- Jack F. V. Hunt
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; (J.F.V.H.); (M.L.); (R.R.)
| | - Meng Li
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; (J.F.V.H.); (M.L.); (R.R.)
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ryan Risgaard
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; (J.F.V.H.); (M.L.); (R.R.)
| | - Gene E. Ananiev
- Carbone Cancer Center Drug Discovery Core, University of Wisconsin-Madison, Madison, WI 53705, USA; (G.E.A.); (S.W.)
| | - Scott Wildman
- Carbone Cancer Center Drug Discovery Core, University of Wisconsin-Madison, Madison, WI 53705, USA; (G.E.A.); (S.W.)
| | - Fan Zhang
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA; (F.Z.); (T.S.B.)
| | - Tim S. Bugni
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA; (F.Z.); (T.S.B.)
| | - Xinyu Zhao
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; (J.F.V.H.); (M.L.); (R.R.)
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Correspondence: (X.Z.); (A.B.); Tel.: +1-(608)-263-9906 (X.Z.); +1-(608)-265-6142 (A.B.)
| | - Anita Bhattacharyya
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; (J.F.V.H.); (M.L.); (R.R.)
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Correspondence: (X.Z.); (A.B.); Tel.: +1-(608)-263-9906 (X.Z.); +1-(608)-265-6142 (A.B.)
| |
Collapse
|
10
|
Kurosaki T, Sakano H, Pröschel C, Wheeler J, Hewko A, Maquat LE. NMD abnormalities during brain development in the Fmr1-knockout mouse model of fragile X syndrome. Genome Biol 2021; 22:317. [PMID: 34784943 PMCID: PMC8597091 DOI: 10.1186/s13059-021-02530-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 10/28/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is an intellectual disability attributable to loss of fragile X protein (FMRP). We previously demonstrated that FMRP binds mRNAs targeted for nonsense-mediated mRNA decay (NMD) and that FMRP loss results in hyperactivated NMD and inhibition of neuronal differentiation in human stem cells. RESULTS We show here that NMD is hyperactivated during the development of the cerebral cortex, hippocampus, and cerebellum in the Fmr1-knockout (KO) mouse during embryonic and early postnatal periods. Our findings demonstrate that NMD regulates many neuronal mRNAs that are important for mouse brain development. CONCLUSIONS We reveal the abnormal regulation of these mRNAs in the Fmr1-KO mouse, a model of FXS, and highlight the importance of early intervention.
Collapse
Affiliation(s)
- Tatsuaki Kurosaki
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642 USA
- Center for RNA Biology, University of Rochester, Rochester, NY 14642 USA
| | - Hitomi Sakano
- Center for RNA Biology, University of Rochester, Rochester, NY 14642 USA
- Department of Otolaryngology, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642 USA
| | - Christoph Pröschel
- Department of Biomedical Genetics, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642 USA
- Stem Cell and Regenerative Medicine Institute, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642 USA
| | - Jason Wheeler
- Center for RNA Biology, University of Rochester, Rochester, NY 14642 USA
- Department of Otolaryngology, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642 USA
| | - Alexander Hewko
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642 USA
- Center for RNA Biology, University of Rochester, Rochester, NY 14642 USA
| | - Lynne E. Maquat
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642 USA
- Center for RNA Biology, University of Rochester, Rochester, NY 14642 USA
| |
Collapse
|
11
|
Zangrandi L, Schmuckermair C, Ghareh H, Castaldi F, Heilbronn R, Zernig G, Ferraguti F, Ramos-Prats A. Loss of mGluR5 in D1 Receptor-Expressing Neurons Improves Stress Coping. Int J Mol Sci 2021; 22:ijms22157826. [PMID: 34360592 PMCID: PMC8346057 DOI: 10.3390/ijms22157826] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/11/2021] [Accepted: 07/19/2021] [Indexed: 11/16/2022] Open
Abstract
The metabotropic glutamate receptor type 5 (mGluR5) has been proposed to play a crucial role in the selection and regulation of cognitive, affective, and emotional behaviors. However, the mechanisms by which these receptors mediate these effects remain largely unexplored. Here, we studied the role of mGluR5 located in D1 receptor-expressing (D1) neurons in the manifestation of different behavioral expressions. Mice with conditional knockout (cKO) of mGluR5 in D1 neurons (mGluR5D1 cKO) and littermate controls displayed similar phenotypical profiles in relation to memory expression, anxiety, and social behaviors. However, mGluR5D1 cKO mice presented different coping mechanisms in response to acute escapable or inescapable stress. mGluR5D1 cKO mice adopted an enhanced active stress coping strategy upon exposure to escapable stress in the two-way active avoidance (TWA) task and a greater passive strategy upon exposure to inescapable stress in the forced swim test (FST). In summary, this work provides evidence for a functional integration of the dopaminergic and glutamatergic system to mediate control over internal states upon stress exposure and directly implicates D1 neurons and mGluR5 as crucial mediators of behavioral stress responses.
Collapse
Affiliation(s)
- Luca Zangrandi
- Department of Neurology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (L.Z.); (R.H.)
- Institute of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (C.S.); (F.C.); (F.F.)
| | - Claudia Schmuckermair
- Institute of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (C.S.); (F.C.); (F.F.)
| | - Hussein Ghareh
- Department of Psychiatry 1, Medical University of Innsbruck, 6020 Innsbruck, Austria; (H.G.); (G.Z.)
| | - Federico Castaldi
- Institute of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (C.S.); (F.C.); (F.F.)
| | - Regine Heilbronn
- Department of Neurology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (L.Z.); (R.H.)
| | - Gerald Zernig
- Department of Psychiatry 1, Medical University of Innsbruck, 6020 Innsbruck, Austria; (H.G.); (G.Z.)
| | - Francesco Ferraguti
- Institute of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (C.S.); (F.C.); (F.F.)
| | - Arnau Ramos-Prats
- Institute of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (C.S.); (F.C.); (F.F.)
- Correspondence:
| |
Collapse
|
12
|
Gregory KJ, Goudet C. International Union of Basic and Clinical Pharmacology. CXI. Pharmacology, Signaling, and Physiology of Metabotropic Glutamate Receptors. Pharmacol Rev 2020; 73:521-569. [PMID: 33361406 DOI: 10.1124/pr.119.019133] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Metabotropic glutamate (mGlu) receptors respond to glutamate, the major excitatory neurotransmitter in the mammalian brain, mediating a modulatory role that is critical for higher-order brain functions such as learning and memory. Since the first mGlu receptor was cloned in 1992, eight subtypes have been identified along with many isoforms and splice variants. The mGlu receptors are transmembrane-spanning proteins belonging to the class C G protein-coupled receptor family and represent attractive targets for a multitude of central nervous system disorders. Concerted drug discovery efforts over the past three decades have yielded a wealth of pharmacological tools including subtype-selective agents that competitively block or mimic the actions of glutamate or act allosterically via distinct sites to enhance or inhibit receptor activity. Herein, we review the physiologic and pathophysiological roles for individual mGlu receptor subtypes including the pleiotropic nature of intracellular signal transduction arising from each. We provide a comprehensive analysis of the in vitro and in vivo pharmacological properties of prototypical and commercially available orthosteric agonists and antagonists as well as allosteric modulators, including ligands that have entered clinical trials. Finally, we highlight emerging areas of research that hold promise to facilitate rational design of highly selective mGlu receptor-targeting therapeutics in the future. SIGNIFICANCE STATEMENT: The metabotropic glutamate receptors are attractive therapeutic targets for a range of psychiatric and neurological disorders. Over the past three decades, intense discovery efforts have yielded diverse pharmacological tools acting either competitively or allosterically, which have enabled dissection of fundamental biological process modulated by metabotropic glutamate receptors and established proof of concept for many therapeutic indications. We review metabotropic glutamate receptor molecular pharmacology and highlight emerging areas that are offering new avenues to selectively modulate neurotransmission.
Collapse
Affiliation(s)
- Karen J Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.) and Institut de Génomique Fonctionnelle (IGF), University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Sante et de la Recherche Medicale (INSERM), Montpellier, France (C.G.)
| | - Cyril Goudet
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.) and Institut de Génomique Fonctionnelle (IGF), University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Sante et de la Recherche Medicale (INSERM), Montpellier, France (C.G.)
| |
Collapse
|
13
|
Neurobiological Mechanisms of Autism Spectrum Disorder and Epilepsy, Insights from Animal Models. Neuroscience 2020; 445:69-82. [DOI: 10.1016/j.neuroscience.2020.02.043] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/22/2020] [Accepted: 02/21/2020] [Indexed: 02/09/2023]
|
14
|
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: 0.6] [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
|
15
|
Jmaeff S, Sidorova Y, Lippiatt H, Barcelona PF, Nedev H, Saragovi LM, Hancock MA, Saarma M, Saragovi HU. Small-Molecule Ligands that Bind the RET Receptor Activate Neuroprotective Signals Independent of but Modulated by Coreceptor GFR α1. Mol Pharmacol 2020; 98:1-12. [PMID: 32362584 DOI: 10.1124/mol.119.118950] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 04/17/2020] [Indexed: 12/25/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) binds the GFRα1 receptor, and the GDNF-GFRα1 complex binds to and activates the transmembrane RET tyrosine kinase to signal through intracellular Akt/Erk pathways. To dissect the GDNF-GFRα1-RET signaling complex, agents that bind and activate RET directly and independently of GFRα1 expression are valuable tools. In a focused naphthalenesulfonic acid library from the National Cancer Institute database, we identified small molecules that are genuine ligands binding to the RET extracellular domain. These ligands activate RET tyrosine kinase and afford trophic signals irrespective of GFRα1 coexpression. However, RET activation by these ligands is constrained by GFRα1, likely via an allosteric mechanism that can be overcome by increasing RET ligand concentration. In a mouse model of retinitis pigmentosa, monotherapy with a small-molecule RET agonist activates survival signals and reduces neuronal death significantly better than GDNF, suggesting therapeutic potential. SIGNIFICANCE STATEMENT: A genuine ligand of RET receptor ectodomain was identified, which acts as an agonist. Binding and agonism are independent of a coreceptor glial cell line-derived neurotrophic factor family receptor α, which is required by the natural growth factor glial cell line-derived neurotrophic factor, and are selective for cells expressing RET. The lead agent protects neurons from death in vivo. This work validates RET receptor as a druggable therapeutic target and provides for potential leads to evaluate in neurodegenerative states. We also report problems that arise when screening chemical libraries.
Collapse
Affiliation(s)
- Sean Jmaeff
- Lady Davis Institute - Jewish General Hospital (S.J., H.L., P.F.B., H.N., L.M.S., H.U.S.), Pharmacology and Therapeutics (S.J., H.U.S.), and SPR-MS Facility (M.H.), McGill University, Montreal, Canada; and Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland (Y.S., M.S.)
| | - Yulia Sidorova
- Lady Davis Institute - Jewish General Hospital (S.J., H.L., P.F.B., H.N., L.M.S., H.U.S.), Pharmacology and Therapeutics (S.J., H.U.S.), and SPR-MS Facility (M.H.), McGill University, Montreal, Canada; and Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland (Y.S., M.S.)
| | - Hayley Lippiatt
- Lady Davis Institute - Jewish General Hospital (S.J., H.L., P.F.B., H.N., L.M.S., H.U.S.), Pharmacology and Therapeutics (S.J., H.U.S.), and SPR-MS Facility (M.H.), McGill University, Montreal, Canada; and Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland (Y.S., M.S.)
| | - Pablo F Barcelona
- Lady Davis Institute - Jewish General Hospital (S.J., H.L., P.F.B., H.N., L.M.S., H.U.S.), Pharmacology and Therapeutics (S.J., H.U.S.), and SPR-MS Facility (M.H.), McGill University, Montreal, Canada; and Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland (Y.S., M.S.)
| | - Hinyu Nedev
- Lady Davis Institute - Jewish General Hospital (S.J., H.L., P.F.B., H.N., L.M.S., H.U.S.), Pharmacology and Therapeutics (S.J., H.U.S.), and SPR-MS Facility (M.H.), McGill University, Montreal, Canada; and Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland (Y.S., M.S.)
| | - Lucia M Saragovi
- Lady Davis Institute - Jewish General Hospital (S.J., H.L., P.F.B., H.N., L.M.S., H.U.S.), Pharmacology and Therapeutics (S.J., H.U.S.), and SPR-MS Facility (M.H.), McGill University, Montreal, Canada; and Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland (Y.S., M.S.)
| | - Mark A Hancock
- Lady Davis Institute - Jewish General Hospital (S.J., H.L., P.F.B., H.N., L.M.S., H.U.S.), Pharmacology and Therapeutics (S.J., H.U.S.), and SPR-MS Facility (M.H.), McGill University, Montreal, Canada; and Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland (Y.S., M.S.)
| | - Mart Saarma
- Lady Davis Institute - Jewish General Hospital (S.J., H.L., P.F.B., H.N., L.M.S., H.U.S.), Pharmacology and Therapeutics (S.J., H.U.S.), and SPR-MS Facility (M.H.), McGill University, Montreal, Canada; and Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland (Y.S., M.S.)
| | - H Uri Saragovi
- Lady Davis Institute - Jewish General Hospital (S.J., H.L., P.F.B., H.N., L.M.S., H.U.S.), Pharmacology and Therapeutics (S.J., H.U.S.), and SPR-MS Facility (M.H.), McGill University, Montreal, Canada; and Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland (Y.S., M.S.)
| |
Collapse
|
16
|
Arsova A, Møller TC, Vedel L, Hansen JL, Foster SR, Gregory KJ, Bräuner-Osborne H. Detailed In Vitro Pharmacological Characterization of Clinically Tested Negative Allosteric Modulators of the Metabotropic Glutamate Receptor 5. Mol Pharmacol 2020; 98:49-60. [PMID: 32358164 DOI: 10.1124/mol.119.119032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 04/10/2020] [Indexed: 12/14/2022] Open
Abstract
Negative allosteric modulation of the metabotropic glutamate 5 (mGlu5) receptor has emerged as a potential strategy for the treatment of neurologic disorders. Despite the success in preclinical studies, many mGlu5 negative allosteric modulators (NAMs) that have reached clinical trials failed due to lack of efficacy. In this study, we provide a detailed in vitro pharmacological characterization of nine clinically and preclinically tested NAMs. We evaluated inhibition of l-glutamate-induced signaling with Ca2+ mobilization, inositol monophosphate (IP1) accumulation, extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation, and real-time receptor internalization assays on rat mGlu5 expressed in HEK293A cells. Moreover, we determined association rates (kon) and dissociation rates (koff), as well as NAM affinities with [3H]methoxy-PEPy binding experiments. kon and koff values varied greatly between the nine NAMs (34- and 139-fold, respectively) resulting in long receptor residence times (>400 min) for basimglurant and mavoglurant, medium residence times (10-30 min) for AZD2066, remeglurant, and (RS)-remeglurant, and low residence times (<10 mins) for dipraglurant, F169521, F1699611, and STX107. We found that all NAMs inhibited l-glutamate-induced mGlu5 receptor internalization, generally with a similar potency to IP1 accumulation and ERK1/2 phosphorylation, whereas Ca2+ mobilization was less potently inhibited. Operational model of allosterism analyses revealed that dipraglurant and (RS)-remeglurant were biased toward (affinity) receptor internalization and away (cooperativity) from the ERK1/2 phosphorylation pathway, respectively. Our study is the first to measure mGlu5 NAM binding kinetics and negative allosteric modulation of mGlu5 receptor internalization and adds significant new knowledge about the molecular pharmacology of a diverse range of clinically relevant NAMs. SIGNIFICANCE STATEMENT: The metabotropic glutamate 5 (mGlu5) receptor is important in many brain functions and implicated in several neurological pathologies. Negative allosteric modulators (NAMs) have shown promising results in preclinical models but have so far failed in human clinical trials. Here we provide the most comprehensive and comparative molecular pharmacological study to date of nine preclinically/clinically tested NAMs at the mGlu5 receptor, which is also the first study to measure ligand binding kinetics and negative allosteric modulation of mGlu5 receptor internalization.
Collapse
Affiliation(s)
- Angela Arsova
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.A., T.C.M., L.V., S.R.F., H.B.-O.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.); and Cardiovascular Research, Novo Nordisk A/S, Måløv, Denmark (J.L.H.)
| | - Thor C Møller
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.A., T.C.M., L.V., S.R.F., H.B.-O.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.); and Cardiovascular Research, Novo Nordisk A/S, Måløv, Denmark (J.L.H.)
| | - Line Vedel
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.A., T.C.M., L.V., S.R.F., H.B.-O.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.); and Cardiovascular Research, Novo Nordisk A/S, Måløv, Denmark (J.L.H.)
| | - Jakob Lerche Hansen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.A., T.C.M., L.V., S.R.F., H.B.-O.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.); and Cardiovascular Research, Novo Nordisk A/S, Måløv, Denmark (J.L.H.)
| | - Simon R Foster
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.A., T.C.M., L.V., S.R.F., H.B.-O.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.); and Cardiovascular Research, Novo Nordisk A/S, Måløv, Denmark (J.L.H.)
| | - Karen J Gregory
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.A., T.C.M., L.V., S.R.F., H.B.-O.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.); and Cardiovascular Research, Novo Nordisk A/S, Måløv, Denmark (J.L.H.)
| | - Hans Bräuner-Osborne
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (A.A., T.C.M., L.V., S.R.F., H.B.-O.); Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.); and Cardiovascular Research, Novo Nordisk A/S, Måløv, Denmark (J.L.H.)
| |
Collapse
|
17
|
Medishetti R, Rani R, Kavati S, Mahilkar A, Akella V, Saxena U, Kulkarni P, Sevilimedu A. A DNAzyme based knockdown model for Fragile-X syndrome in zebrafish reveals a critical window for therapeutic intervention. J Pharmacol Toxicol Methods 2019; 101:106656. [PMID: 31734279 DOI: 10.1016/j.vascn.2019.106656] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 11/05/2019] [Accepted: 11/10/2019] [Indexed: 11/16/2022]
Abstract
INTRODUCTION FXS is the leading cause of intellectual disabilities in males and a major monogenic cause of ASD (Autism spectrum disorders). It occurs due to the loss of FMRP, whose role in early development is not well understood. In this study, we have used a novel DNAzyme based approach to create a larval model of FXS in zebrafish with specific focus on the early developmental window. METHODS Fmr1specific DNAzymes were electroporated into embryos to create the knockdown. Changes in RNA and protein levels of FMRP and relevant biomarkers were measured in the 0-7dpf window. Behavioral tests to measure anxiety, cognitive impairments and irritability in the larvae were conducted at the 7dpf stage. Drug treatment was carried out at various time points in the 0-7dpf window to identify the critical window for pharmacological intervention. RESULTS The DNAzyme based knockdown approach led to a significant knockdown of FMRP in the zebrafish embryos, accompanied by increased anxiety, irritability and cognitive impairments at 7dpf, thus creating a robust larval model of FXS. Treatment with the Mavoglurant was able to rescue the behavioral phenotypes in the FXS larvae, and found to be more efficacious in the 0-3dpf window. DISCUSSION The results from this study have revealed that a) a DNAzyme based knockdown approach can be used to create robust larval zebrafish model of disease, in a high-throughput manner and b) optimal window for therapeutic intervention for FXS as well as other pediatric diseases with a monogenic cause can be identified using such a model.
Collapse
Affiliation(s)
- Raghavender Medishetti
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana, 500046, India
| | - Rita Rani
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana, 500046, India
| | - Srinivas Kavati
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana, 500046, India
| | - Anjali Mahilkar
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana, 500046, India
| | - Venkateswarlu Akella
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana, 500046, India
| | - Uday Saxena
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana, 500046, India
| | - Pushkar Kulkarni
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana, 500046, India.
| | - Aarti Sevilimedu
- Center for Innovation in Molecular and Pharmaceutical Sciences (CIMPS), Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Gachibowli, Hyderabad, Telangana, 500046, India.
| |
Collapse
|
18
|
Cannabidiol (CBD) reduces anxiety-related behavior in mice via an FMRP-independent mechanism. Pharmacol Biochem Behav 2019; 181:93-100. [DOI: 10.1016/j.pbb.2019.05.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/21/2019] [Accepted: 05/01/2019] [Indexed: 11/18/2022]
|
19
|
Zerbi V, Markicevic M, Gasparini F, Schroeter A, Rudin M, Wenderoth N. Inhibiting mGluR5 activity by AFQ056/Mavoglurant rescues circuit-specific functional connectivity in Fmr1 knockout mice. Neuroimage 2019; 191:392-402. [DOI: 10.1016/j.neuroimage.2019.02.051] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 02/11/2019] [Accepted: 02/19/2019] [Indexed: 12/12/2022] Open
|
20
|
Zafarullah M, Tassone F. Molecular Biomarkers in Fragile X Syndrome. Brain Sci 2019; 9:E96. [PMID: 31035599 PMCID: PMC6562871 DOI: 10.3390/brainsci9050096] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/22/2019] [Accepted: 04/24/2019] [Indexed: 01/01/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common inherited form of intellectual disability (ID) and a known monogenic cause of autism spectrum disorder (ASD). It is a trinucleotide repeat disorder, in which more than 200 CGG repeats in the 5' untranslated region (UTR) of the fragile X mental retardation 1 (FMR1) gene causes methylation of the promoter with consequent silencing of the gene, ultimately leading to the loss of the encoded fragile X mental retardation 1 protein, FMRP. FMRP is an RNA binding protein that plays a primary role as a repressor of translation of various mRNAs, many of which are involved in the maintenance and development of neuronal synaptic function and plasticity. In addition to intellectual disability, patients with FXS face several behavioral challenges, including anxiety, hyperactivity, seizures, repetitive behavior, and problems with executive and language performance. Currently, there is no cure or approved medication for the treatment of the underlying causes of FXS, but in the past few years, our knowledge about the proteins and pathways that are dysregulated by the loss of FMRP has increased, leading to clinical trials and to the path of developing molecular biomarkers for identifying potential targets for therapies. In this paper, we review candidate molecular biomarkers that have been identified in preclinical studies in the FXS mouse animal model and are now under validation for human applications or have already made their way to clinical trials.
Collapse
Affiliation(s)
- Marwa Zafarullah
- Department of Biochemistry and Molecular Medicine, University of California Davis, School of Medicine, Sacramento, 95817 CA, USA.
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine, University of California Davis, School of Medicine, Sacramento, 95817 CA, USA.
- MIND Institute, University of California Davis Medical Center, Sacramento, 95817 CA, USA.
| |
Collapse
|
21
|
Ramos-Prats A, Kölldorfer J, Paolo E, Zeidler M, Schmid G, Ferraguti F. An Appraisal of the Influence of the Metabotropic Glutamate 5 (mGlu5) Receptor on Sociability and Anxiety. Front Mol Neurosci 2019; 12:30. [PMID: 30873001 PMCID: PMC6401637 DOI: 10.3389/fnmol.2019.00030] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 01/24/2019] [Indexed: 02/04/2023] Open
Abstract
Amongst the many neurotransmitter systems causally linked to the expression of social behavior, glutamate appears to play a pivotal role. In particular, metabotropic glutamate 5 (mGlu5) receptors have received much attention as its altered function has been reported in several mouse models of autism spectrum disorders and mental retardation. Inhibition of the activity of mGlu5 receptors by means of genetic or pharmacological manipulations improved social deficits in some of these animal models. However, in normal wild-type (WT) mice, pharmacological blockade of mGlu5 receptors yielded inconsistent results. The aim of our study was to investigate the actual contribution of decreased or absent mGlu5 receptor function in sociability and anxiety-like behavior as well as to explore the impact of mGlu5 receptor ablation on the pattern of brain activation upon social exposure. Here we show that Grm5-/- mice display higher social preference indexes compared to age-matched WT mice in the three-chambered social task. However, this effect was accompanied by a decreased exploratory activity during the test and increased anxiety-like behavior. Contrary to mGlu5 receptor ablation, the mGlu5 receptor negative allosteric modulator 3-((2-methyl-1,4-thiazolyl)ethynyl)pyridine (MTEP) induced anxiolytic effects without affecting social preference in WT mice. By mapping c-Fos expression in 21 different brain regions known to be involved in social interaction, we detected a specific activation of the prefrontal cortex and dorsolateral septum in Grm5-/- mice following social interaction. C-Fos expression correlation-based network and graph theoretical analyses further suggested dysfunctional connectivity and disruption of the functional brain network generated during social interaction in Grm5-/- mice. The lack of mGlu5 receptors resulted in profound rearrangements of the functional impact of prefrontal and hippocampal regions in the social interaction network. In conclusion, this work reveals a complex contribution of mGlu5 receptors in sociability and anxiety and points to the importance of these receptors in regulating brain functional connectivity during social interaction.
Collapse
Affiliation(s)
- Arnau Ramos-Prats
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Julia Kölldorfer
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Elena Paolo
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Maximilian Zeidler
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Gabriele Schmid
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Francesco Ferraguti
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
22
|
Hessl D, Harvey D, Sansone S, Crestodina C, Chin J, Joshi R, Hagerman RJ, Berry‐Kravis E. Effects of mavoglurant on visual attention and pupil reactivity while viewing photographs of faces in Fragile X Syndrome. PLoS One 2019; 14:e0209984. [PMID: 30653533 PMCID: PMC6336311 DOI: 10.1371/journal.pone.0209984] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 12/14/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Numerous preclinical studies have supported the theory that enhanced activation of mGluR5 signaling, due to the absence or reduction of the FMR1 protein, contributes to cognitive and behavioral deficits in patients with fragile X syndrome (FXS). However multiple phase 2 controlled trials in patients with FXS have failed to demonstrate efficacy of compounds that negatively modulate mGluR5, including two phase 2b randomized controlled trials (RCT) of mavoglurant (AFQ056, Novartis Pharma AG), when the primary measures of interest were behavioral ratings. This has cast some doubt onto the translation of the mGluR5 theory from animal models to humans with the disorder. METHODS We evaluated social gaze behavior-a key phenotypic feature of the disorder-and sympathetic nervous system influence on pupil size using a previously-validated eye tracking paradigm as a biobehavioral probe, in 57 adolescent or adult patients with FXS at baseline and following three months of blinded treatment with one of three doses of mavoglurant or placebo, within the context of the AFQ056 RCTs. RESULTS Patients with FXS treated with mavoglurant demonstrated increased total absolute looking time and number of fixations to the eye region while viewing human faces relative to baseline, and compared to those treated with placebo. In addition, patients had greater pupil reactivity to faces relative to baseline following mavoglurant treatment compared to placebo. DISCUSSION The study shows that negative modulation of mGluR5 activity improves eye gaze behavior and alters sympathetically-driven reactivity to faces in patients with FXS, providing preliminary evidence of this drug's impact on behavior in humans with the disorder.
Collapse
Affiliation(s)
- David Hessl
- MIND Institute, University of California Davis Medical Center, Sacramento, California, United States of America
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Danielle Harvey
- Division of Biostatistics, Department of Public Health Sciences, University of California Davis School of Medicine, Davis, CA, United States of America
| | - Stephanie Sansone
- MIND Institute, University of California Davis Medical Center, Sacramento, California, United States of America
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Crystal Crestodina
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, United States of America
| | - Jamie Chin
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, United States of America
| | - Reshma Joshi
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, United States of America
| | - Randi J. Hagerman
- MIND Institute, University of California Davis Medical Center, Sacramento, California, United States of America
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, California, United States of America
| | - Elizabeth Berry‐Kravis
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, United States of America
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States of America
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, United States of America
| |
Collapse
|
23
|
Mavoglurant in Fragile X Syndrome: Results of two open-label, extension trials in adults and adolescents. Sci Rep 2018; 8:16970. [PMID: 30451888 PMCID: PMC6242849 DOI: 10.1038/s41598-018-34978-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 10/22/2018] [Indexed: 02/06/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common monogenic cause of inherited intellectual and developmental disabilities. Mavoglurant, a selective metabotropic glutamate receptor subtype-5 antagonist, has shown positive neuronal and behavioral effects in preclinical studies, but failed to demonstrate any behavioral benefits in two 12-week, randomized, placebo-controlled, double-blind, phase IIb studies in adults and adolescents with FXS. Here we report the long-term safety (primary endpoint) and efficacy (secondary endpoint) results of the open-label extensions. Adolescent (n = 119, aged 12–19 years) and adult (n = 148, aged 18–45 years) participants received up to 100 mg bid mavoglurant for up to 34 months. Both extension studies were terminated prematurely due to lack of proven efficacy in the core studies. Mavoglurant was well tolerated with no new safety signal. Five percent of adults and 16.9 percent of adolescents discontinued treatment due to adverse events. Gradual and consistent behavioral improvements as measured by the ABC-CFX scale were observed, which were numerically superior to those seen in the placebo arm of the core studies. These two extension studies confirm the long-term safety of mavoglurant in FXS, but further investigations are required to determine whether and under which conditions the significant preclinical results obtained with mGluR5 inhibition can translate to humans.
Collapse
|
24
|
Westmark PR, Dekundy A, Gravius A, Danysz W, Westmark CJ. Rescue of Fmr1 KO phenotypes with mGluR 5 inhibitors: MRZ-8456 versus AFQ-056. Neurobiol Dis 2018; 119:190-198. [PMID: 30125640 DOI: 10.1016/j.nbd.2018.08.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 08/08/2018] [Accepted: 08/13/2018] [Indexed: 12/23/2022] Open
Abstract
Metabotropic glutamate receptor 5 (mGluR5) is a drug target for central nervous system disorders such as fragile X syndrome that involve excessive glutamate-induced excitation. We tested the efficacy of a novel negative allosteric modulator of mGluR5 developed by Merz Pharmaceuticals, MRZ-8456, in comparison to MPEP and AFQ-056 (Novartis, a.k.a. mavoglurant) in both in vivo and in vitro assays in a mouse model of fragile X syndrome, Fmr1KO mice. The in vivo assays included susceptibility to audiogenic-induced seizures and pharmacokinetic measurements of drug availability. The in vitro assays included dose response assessments of biomarker expression and dendritic spine length and density in cultured primary neurons. Both MRZ-8456 and AFQ-056 attenuated wild running and audiogenic-induced seizures in Fmr1KO mice with similar pharmacokinetic profiles. Both drugs significantly reduced dendritic expression of amyloid-beta protein precursor (APP) and rescued the ratio of mature to immature dendritic spines. These findings demonstrate that MRZ-8456, a drug being developed for the treatment of motor complications of L-DOPA in Parkinson's disease and which completed a phase I clinical trial, is effective in attenuating both well-established (seizures and dendritic spine maturity) and exploratory biomarker (APP expression) phenotypes in a mouse model of fragile X syndrome.
Collapse
Affiliation(s)
- Pamela R Westmark
- University of Wisconsin-Madison, Department of Neurology, Madison, WI, USA; University of Wisconsin-Madison, Department of Medicine, Madison, WI, USA
| | - Andrzej Dekundy
- Merz Pharmaceuticals GmbH, Eckenheimer Landstrasse 100, 60318 Frankfurt am Main, Germany
| | - Andreas Gravius
- Merz Pharmaceuticals GmbH, Eckenheimer Landstrasse 100, 60318 Frankfurt am Main, Germany
| | - Wojciech Danysz
- Merz Pharmaceuticals GmbH, Eckenheimer Landstrasse 100, 60318 Frankfurt am Main, Germany
| | - Cara J Westmark
- University of Wisconsin-Madison, Department of Neurology, Madison, WI, USA.
| |
Collapse
|
25
|
Martin HGS, Lassalle O, Manzoni OJ. Differential Adulthood Onset mGlu5 Signaling Saves Prefrontal Function in the Fragile X Mouse. Cereb Cortex 2018; 27:5592-5602. [PMID: 27797833 DOI: 10.1093/cercor/bhw328] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Indexed: 12/13/2022] Open
Abstract
The final maturation of the prefrontal cortex (PFC) continues into early adulthood and is delayed compared with other forebrain structures. However, how these late onset changes in the PFC relate to neurodevelopment disorders is poorly understood. Fragile X syndrome (FXS) is a prevalent neurogenetic disorder linked to deficits in PFC function. mGlu5 is an important molecular hub in the etiology of FXS. Thus we have examined changes in mGlu5 function in the PFC in a mouse model of FXS (Fmr1 knockout) during early adulthood and subsequent maturity. An unusual endophenotype was identified; during early adulthood (2-month-old) Fmr1 knockout mice show a severe deficit in mGlu5 dependent eCB synaptic plasticity; however, in 1-year-old this deficit self rectifies. This adulthood onset correction in mGlu5 function is linked to an engagement of TRPV1 receptors in 1-year-old mice. In 2-month-old Fmr1 knockout mice, mGlu5 mediated synaptic plasticity could be recovered with eCB system targeted drugs, but also by direct enhancement of mGlu5 function with a positive allosteric modulator. These results point to further refinements to the role of mGlu5 in FXS. Furthermore our findings suggest when studying neurodevelopmental disorders with a significant PFC phenotype consideration of late onset changes may be important.
Collapse
Affiliation(s)
- Henry G S Martin
- INSERM U901, Marseille 13009, France.,INMED, Marseille 13009, France.,Université de Aix-Marseille, UMR S901, Marseille 13009, France
| | - Olivier Lassalle
- INSERM U901, Marseille 13009, France.,INMED, Marseille 13009, France.,Université de Aix-Marseille, UMR S901, Marseille 13009, France
| | - Olivier J Manzoni
- INSERM U901, Marseille 13009, France.,INMED, Marseille 13009, France.,Université de Aix-Marseille, UMR S901, Marseille 13009, France
| |
Collapse
|
26
|
Saxena K, Webster J, Hallas-Potts A, Mackenzie R, Spooner PA, Thomson D, Kind P, Chattarji S, Morris RGM. Experiential contributions to social dominance in a rat model of fragile-X syndrome. Proc Biol Sci 2018; 285:20180294. [PMID: 29899064 PMCID: PMC6015851 DOI: 10.1098/rspb.2018.0294] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/18/2018] [Indexed: 11/12/2022] Open
Abstract
Social withdrawal is one phenotypic feature of the monogenic neurodevelopmental disorder fragile-X. Using a 'knockout' rat model of fragile-X, we examined whether deletion of the Fmr1 gene that causes this condition would affect the ability to form and express a social hierarchy as measured in a tube test. Male fragile-X 'knockout' rats living together could successfully form a social dominance hierarchy, but were significantly subordinate to wild-type animals in mixed group cages. Over 10 days of repeated testing, the fragile-X mutant rats gradually showed greater variance and instability of rank during their tube-test encounters. This affected the outcome of future encounters with stranger animals from other cages, with the initial phenotype of wild-type dominance lost to a more complex picture that reflected, regardless of genotype, the prior experience of winning or losing. Our findings offer a novel insight into the complex dynamics of social interactions between laboratory living groups of fragile-X and wild-type rats. Even though this is a monogenic condition, experience has an impact upon future interactions with other animals. Gene/environment interactions should therefore be considered in the development of therapeutics.
Collapse
Affiliation(s)
- K Saxena
- Simons Initiative for the Developing Brain, Edinburgh Neuroscience, 1 George Square, Edinburgh EH8 9JZ, UK
- The Patrick Wild Centre, Edinburgh Neuroscience, 1 George Square, Edinburgh EH8 9JZ, UK
- Centre for Discovery Brain Sciences, Edinburgh Neuroscience, 1 George Square, Edinburgh EH8 9JZ, UK
- Centre for Cognitive and Neural Systems, Edinburgh Neuroscience, 1 George Square, Edinburgh EH8 9JZ, UK
- Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, 560065, India
| | - J Webster
- Centre for Cognitive and Neural Systems, Edinburgh Neuroscience, 1 George Square, Edinburgh EH8 9JZ, UK
| | - A Hallas-Potts
- Centre for Cognitive and Neural Systems, Edinburgh Neuroscience, 1 George Square, Edinburgh EH8 9JZ, UK
| | - R Mackenzie
- Centre for Cognitive and Neural Systems, Edinburgh Neuroscience, 1 George Square, Edinburgh EH8 9JZ, UK
| | - P A Spooner
- Centre for Discovery Brain Sciences, Edinburgh Neuroscience, 1 George Square, Edinburgh EH8 9JZ, UK
- Centre for Cognitive and Neural Systems, Edinburgh Neuroscience, 1 George Square, Edinburgh EH8 9JZ, UK
| | - D Thomson
- Centre for Discovery Brain Sciences, Edinburgh Neuroscience, 1 George Square, Edinburgh EH8 9JZ, UK
- Centre for Cognitive and Neural Systems, Edinburgh Neuroscience, 1 George Square, Edinburgh EH8 9JZ, UK
| | - P Kind
- Simons Initiative for the Developing Brain, Edinburgh Neuroscience, 1 George Square, Edinburgh EH8 9JZ, UK
- The Patrick Wild Centre, Edinburgh Neuroscience, 1 George Square, Edinburgh EH8 9JZ, UK
- Centre for Discovery Brain Sciences, Edinburgh Neuroscience, 1 George Square, Edinburgh EH8 9JZ, UK
- Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, 560065, India
| | - S Chattarji
- Simons Initiative for the Developing Brain, Edinburgh Neuroscience, 1 George Square, Edinburgh EH8 9JZ, UK
- The Patrick Wild Centre, Edinburgh Neuroscience, 1 George Square, Edinburgh EH8 9JZ, UK
- Centre for Discovery Brain Sciences, Edinburgh Neuroscience, 1 George Square, Edinburgh EH8 9JZ, UK
- Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, 560065, India
- National Centre for Biological Sciences, Bangalore, 560065, India
| | - R G M Morris
- Simons Initiative for the Developing Brain, Edinburgh Neuroscience, 1 George Square, Edinburgh EH8 9JZ, UK
- The Patrick Wild Centre, Edinburgh Neuroscience, 1 George Square, Edinburgh EH8 9JZ, UK
- Centre for Discovery Brain Sciences, Edinburgh Neuroscience, 1 George Square, Edinburgh EH8 9JZ, UK
- Centre for Cognitive and Neural Systems, Edinburgh Neuroscience, 1 George Square, Edinburgh EH8 9JZ, UK
- Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, 560065, India
| |
Collapse
|
27
|
Zeidler S, Pop AS, Jaafar IA, de Boer H, Buijsen RAM, de Esch CEF, Nieuwenhuizen‐Bakker I, Hukema RK, Willemsen R. Paradoxical effect of baclofen on social behavior in the fragile X syndrome mouse model. Brain Behav 2018; 8:e00991. [PMID: 29785777 PMCID: PMC5991574 DOI: 10.1002/brb3.991] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/29/2018] [Accepted: 03/31/2018] [Indexed: 11/09/2022] Open
Abstract
INTRODUCTION Fragile X syndrome (FXS) is a common monogenetic cause of intellectual disability, autism spectrum features, and a broad range of other psychiatric and medical problems. FXS is caused by the lack of the fragile X mental retardation protein (FMRP), a translational regulator of specific mRNAs at the postsynaptic compartment. The absence of FMRP leads to aberrant synaptic plasticity, which is believed to be caused by an imbalance in excitatory and inhibitory network functioning of the synapse. Evidence from studies in mice demonstrates that GABA, the major inhibitory neurotransmitter in the brain, and its receptors, is involved in the pathogenesis of FXS. Moreover, several FXS phenotypes, including social behavior deficits, could be corrected in Fmr1 KO mice after acute treatment with GABAB agonists. METHODS As FXS would probably require a lifelong treatment, we investigated the effect of chronic treatment with the GABAB agonist baclofen on social behavior in Fmr1 KO mice on two behavioral paradigms for social behavior: the automated tube test and the three-chamber sociability test. RESULTS Unexpectedly, chronic baclofen treatment resulted in worsening of the FXS phenotypes in these behavior tests. Strikingly, baclofen treatment also affected wild-type animals in both behavioral tests, inducing a phenotype similar to that of untreated Fmr1 KO mice. CONCLUSION Altogether, the disappointing results of recent clinical trials with the R-baclofen enantiomer arbaclofen and our current results indicate that baclofen should be reconsidered and further evaluated before its application in targeted treatment for FXS.
Collapse
Affiliation(s)
- Shimriet Zeidler
- Department of Clinical GeneticsErasmus University Medical CenterRotterdamThe Netherlands
| | - Andreea S. Pop
- Department of Clinical GeneticsErasmus University Medical CenterRotterdamThe Netherlands
| | - Israa A. Jaafar
- Department of Clinical GeneticsErasmus University Medical CenterRotterdamThe Netherlands
| | - Helen de Boer
- Department of Clinical GeneticsErasmus University Medical CenterRotterdamThe Netherlands
| | - Ronald A. M. Buijsen
- Department of Clinical GeneticsErasmus University Medical CenterRotterdamThe Netherlands
| | - Celine E. F. de Esch
- Department of Clinical GeneticsErasmus University Medical CenterRotterdamThe Netherlands
| | | | - Renate K. Hukema
- Department of Clinical GeneticsErasmus University Medical CenterRotterdamThe Netherlands
| | - Rob Willemsen
- Department of Clinical GeneticsErasmus University Medical CenterRotterdamThe Netherlands
| |
Collapse
|
28
|
Dahlhaus R. Of Men and Mice: Modeling the Fragile X Syndrome. Front Mol Neurosci 2018; 11:41. [PMID: 29599705 PMCID: PMC5862809 DOI: 10.3389/fnmol.2018.00041] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 01/31/2018] [Indexed: 12/26/2022] Open
Abstract
The Fragile X Syndrome (FXS) is one of the most common forms of inherited intellectual disability in all human societies. Caused by the transcriptional silencing of a single gene, the fragile x mental retardation gene FMR1, FXS is characterized by a variety of symptoms, which range from mental disabilities to autism and epilepsy. More than 20 years ago, a first animal model was described, the Fmr1 knock-out mouse. Several other models have been developed since then, including conditional knock-out mice, knock-out rats, a zebrafish and a drosophila model. Using these model systems, various targets for potential pharmaceutical treatments have been identified and many treatments have been shown to be efficient in preclinical studies. However, all attempts to turn these findings into a therapy for patients have failed thus far. In this review, I will discuss underlying difficulties and address potential alternatives for our future research.
Collapse
Affiliation(s)
- Regina Dahlhaus
- Institute for Biochemistry, Emil-Fischer Centre, University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
29
|
McGraw CM, Ward CS, Samaco RC. Genetic rodent models of brain disorders: Perspectives on experimental approaches and therapeutic strategies. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2018; 175:368-379. [PMID: 28910526 PMCID: PMC5659732 DOI: 10.1002/ajmg.c.31570] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 06/28/2017] [Indexed: 12/14/2022]
Abstract
Neurobehavioral disorders comprised of neurodegenerative, neurodevelopmental, and psychiatric disorders together represent leading causes of morbidity and mortality. Despite significant academic research and industry efforts to elucidate the disease mechanisms operative in these disorders and to develop mechanism‐based therapies, our understanding remains incomplete and our access to tractable therapeutic interventions severely limited. The magnitude of these short‐comings can be measured by the growing list of disappointing clinical trials based on initially promising compounds identified in genetic animal models. This review and commentary will explore why this may be so, focusing on the central role that genetic models of neurobehavioral disorders have come to occupy in current efforts to identify disease mechanisms and therapies. In particular, we will highlight the unique pitfalls and challenges that have hampered success in these models as compared to genetic models of non‐neurological diseases as well as to symptom‐based models of the early 20th century that led to the discovery of all major classes of psychoactive pharmaceutical compounds still used today. Using examples from specific genetic rodent models of human neurobehavioral disorders, we will highlight issues of reproducibility, construct validity, and translational relevance in the hopes that these examples will be instructive toward greater success in future endeavors. Lastly, we will champion a two‐pronged approach toward identifying novel therapies for neurobehavioral disorders that makes greater use of the historically more successful symptom‐based approaches in addition to more mechanism‐based approaches.
Collapse
Affiliation(s)
- Christopher M McGraw
- Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Christopher S Ward
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Rodney C Samaco
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.,Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
30
|
Drug development for neurodevelopmental disorders: lessons learned from fragile X syndrome. Nat Rev Drug Discov 2017; 17:280-299. [PMID: 29217836 DOI: 10.1038/nrd.2017.221] [Citation(s) in RCA: 230] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Neurodevelopmental disorders such as fragile X syndrome (FXS) result in lifelong cognitive and behavioural deficits and represent a major public health burden. FXS is the most frequent monogenic form of intellectual disability and autism, and the underlying pathophysiology linked to its causal gene, FMR1, has been the focus of intense research. Key alterations in synaptic function thought to underlie this neurodevelopmental disorder have been characterized and rescued in animal models of FXS using genetic and pharmacological approaches. These robust preclinical findings have led to the implementation of the most comprehensive drug development programme undertaken thus far for a genetically defined neurodevelopmental disorder, including phase IIb trials of metabotropic glutamate receptor 5 (mGluR5) antagonists and a phase III trial of a GABAB receptor agonist. However, none of the trials has been able to unambiguously demonstrate efficacy, and they have also highlighted the extent of the knowledge gaps in drug development for FXS and other neurodevelopmental disorders. In this Review, we examine potential issues in the previous studies and future directions for preclinical and clinical trials. FXS is at the forefront of efforts to develop drugs for neurodevelopmental disorders, and lessons learned in the process will also be important for such disorders.
Collapse
|
31
|
Zeidler S, de Boer H, Hukema RK, Willemsen R. Combination Therapy in Fragile X Syndrome; Possibilities and Pitfalls Illustrated by Targeting the mGluR5 and GABA Pathway Simultaneously. Front Mol Neurosci 2017; 10:368. [PMID: 29163043 PMCID: PMC5681991 DOI: 10.3389/fnmol.2017.00368] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 10/24/2017] [Indexed: 11/13/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common monogenetic cause of intellectual disability and autism. The disorder is characterized by altered synaptic plasticity in the brain. Synaptic plasticity is tightly regulated by a complex balance of different synaptic pathways. In FXS, various synaptic pathways are disrupted, including the excitatory metabotropic glutamate receptor 5 (mGluR5) and the inhibitory γ-aminobutyric acid (GABA) pathways. Targeting each of these pathways individually, has demonstrated beneficial effects in animal models, but not in patients with FXS. This lack of translation might be due to oversimplification of the disease mechanisms when targeting only one affected pathway, in spite of the complexity of the many pathways implicated in FXS. In this report we outline the hypothesis that targeting more than one pathway simultaneously, a combination therapy, might improve treatment effects in FXS. In addition, we present a glance of the first results of chronic combination therapy on social behavior in Fmr1 KO mice. In contrast to what we expected, targeting both the mGluR5 and the GABAergic pathways simultaneously did not result in a synergistic effect, but in a slight worsening of the social behavior phenotype. This does implicate that both pathways are interconnected and important for social behavior. Our results underline the tremendous fine-tuning that is needed to reach the excitatory-inhibitory balance in the synapse in relation to social behavior. We believe that alternative strategies focused on combination therapy should be further explored, including targeting pathways in different cellular compartments or cell-types.
Collapse
Affiliation(s)
- Shimriet Zeidler
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Helen de Boer
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Renate K Hukema
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Rob Willemsen
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
32
|
Ablation of TFR1 in Purkinje Cells Inhibits mGlu1 Trafficking and Impairs Motor Coordination, But Not Autistic-Like Behaviors. J Neurosci 2017; 37:11335-11352. [PMID: 29054881 DOI: 10.1523/jneurosci.1223-17.2017] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 09/28/2017] [Accepted: 10/06/2017] [Indexed: 11/21/2022] Open
Abstract
Group 1 metabotropic glutamate receptors (mGlu1/5s) are critical to synapse formation and participate in synaptic LTP and LTD in the brain. mGlu1/5 signaling alterations have been documented in cognitive impairment, neurodegenerative disorders, and psychiatric diseases, but underlying mechanisms for its modulation are not clear. Here, we report that transferrin receptor 1 (TFR1), a transmembrane protein of the clathrin complex, modulates the trafficking of mGlu1 in cerebellar Purkinje cells (PCs) from male mice. We show that conditional knock-out of TFR1 in PCs does not affect the cytoarchitecture of PCs, but reduces mGlu1 expression at synapses. This regulation by TFR1 acts in concert with that by Rab8 and Rab11, which modulate the internalization and recycling of mGlu1, respectively. TFR1 can bind to Rab proteins and facilitate their expression at synapses. PC ablation of TFR1 inhibits parallel fiber-PC LTD, whereas parallel fiber-LTP and PC intrinsic excitability are not affected. Finally, we demonstrate that PC ablation of TFR1 impairs motor coordination, but does not affect social behaviors in mice. Together, these findings underscore the importance of TFR1 in regulating mGlu1 trafficking and suggest that mGlu1- and mGlu1-dependent parallel fiber-LTD are associated with regulation of motor coordination, but not autistic behaviors.SIGNIFICANCE STATEMENT Group 1 metabotropic glutamate receptor (mGlu1/5) signaling alterations have been documented in cognitive impairment, neurodegenerative disorders, and psychiatric diseases. Recent work suggests that altered mGlu1 signaling in Purkinje cells (PCs) may be involved in not only motor learning, but also autistic-like behaviors. We find that conditional knock-out of transferrin receptor 1 (TFR1) in PCs reduces synaptic mGlu1 by tethering Rab8 and Rab11 in the cytosol. PC ablation of TFR1 inhibits parallel fiber-PC LTD, whereas parallel fiber-PC LTP and PC intrinsic excitability are intact. Motor coordination is impaired, but social behaviors are normal in TFR1flox/flox;pCP2-cre mice. Our data reveal a new regulator for trafficking and synaptic expression of mGlu1 and suggest that mGlu1-dependent LTD is associated with motor coordination, but not autistic-like behaviors.
Collapse
|
33
|
Longitudinal identification of clinically distinct neurophenotypes in young children with fragile X syndrome. Proc Natl Acad Sci U S A 2017; 114:10767-10772. [PMID: 28923933 DOI: 10.1073/pnas.1620994114] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Fragile X syndrome (FXS), due to mutations of the FMR1 gene, is the most common known inherited cause of developmental disability. The cognitive, behavioral, and neurological phenotypes observed in affected individuals can vary considerably, making it difficult to predict outcomes and determine the need for interventions. We sought to examine early structural brain growth as a potential marker for identification of clinically meaningful subgroups. Participants included 42 very young boys with FXS who completed a T1-weighted anatomical MRI and cognitive/behavioral assessment at two longitudinal time points, with mean ages of 2.89 y and 4.91 y. Topological data analysis (TDA), an unsupervised approach to multivariate pattern analysis, was applied to the longitudinal anatomical data to identify coherent but heretofore unknown subgroups. TDA revealed two large subgroups within the study population based solely on longitudinal MRI data. Post hoc comparisons of cognition, adaptive functioning, and autism severity scores between these groups demonstrated that one group was consistently higher functioning on all measures at both time points, with pronounced and significant unidirectional differences (P < 0.05 for time point 1 and/or time point 2 for each measure). These results support the existence of two longitudinally defined, neuroanatomically distinct, and clinically relevant phenotypes among boys with FXS. If confirmed by additional analyses, such information may be used to predict outcomes and guide design of targeted therapies. Furthermore, TDA of longitudinal anatomical MRI data may represent a useful method for reliably and objectively defining subtypes within other neuropsychiatric disorders.
Collapse
|
34
|
Borrie SC, Brems H, Legius E, Bagni C. Cognitive Dysfunctions in Intellectual Disabilities: The Contributions of the Ras-MAPK and PI3K-AKT-mTOR Pathways. Annu Rev Genomics Hum Genet 2017; 18:115-142. [DOI: 10.1146/annurev-genom-091416-035332] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Sarah C. Borrie
- Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Hilde Brems
- Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Eric Legius
- Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Claudia Bagni
- Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
- Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00173 Rome, Italy
| |
Collapse
|
35
|
Harkins CM, Dominick KC, Wink LK, Pedapati EV, Shaffer RC, Fitzpatrick SE, Davenport MH, Sweeney JA, Erickson CA. Challenges in Conducting Clinical Trials for Pharmacotherapies in Fragile X Syndrome: Lessons Learned. Pharmaceut Med 2017. [DOI: 10.1007/s40290-017-0199-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
36
|
Abstract
Human genetic studies have been the driving force in bringing to light the underlying biology of psychiatric conditions. As these studies fill in the gaps in our knowledge of the mechanisms at play, we will be better equipped to design therapies in rational and targeted ways, or repurpose existing therapies in previously unanticipated ways. This review is intended for those unfamiliar with psychiatric genetics as a field and provides a primer on different modes of genetic variation, the technologies currently used to probe them, and concepts that provide context for interpreting the gene-phenotype relationship. Like other subfields in human genetics, psychiatric genetics is moving from microarray technology to sequencing-based approaches as barriers of cost and expertise are removed, and the ramifications of this transition are discussed here. A summary is then given of recent genetic discoveries in a number of neuropsychiatric conditions, with particular emphasis on neurodevelopmental conditions. The general impact of genetics on drug development has been to underscore the extensive etiological heterogeneity in seemingly cohesive diagnostic categories. Consequently, the path forward is not in therapies hoping to reach large swaths of patients sharing a clinically defined diagnosis, but rather in targeting patients belonging to specific "biotypes" defined through a combination of objective, quantifiable data, including genotype.
Collapse
Affiliation(s)
- Jacob J Michaelson
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
- Department of Biomedical Engineering, University of Iowa College of Engineering, Iowa City, IA, USA.
- Department of Communication Sciences and Disorders, University of Iowa College of Liberal Arts and Sciences, Iowa City, IA, USA.
- Iowa Institute of Human Genetics, University of Iowa, Iowa City, IA, USA.
- Genetics Cluster Initiative, University of Iowa, Iowa City, IA, USA.
- The DeLTA Center, University of Iowa, Iowa City, IA, USA.
- University of Iowa Informatics Initiative, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
37
|
Pellerin D, Lortie A, Corbin F. Platelets as a surrogate disease model of neurodevelopmental disorders: Insights from Fragile X Syndrome. Platelets 2017; 29:113-124. [PMID: 28660769 DOI: 10.1080/09537104.2017.1317733] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Fragile X Syndrome (FXS) is the most common inherited form of intellectual disability and the leading monogenic cause of autism spectrum disorders (ASD). Despite a large number of therapeutics developed in past years, there is currently no targeted treatment approved for FXS. In fact, translation of the positive and very promising preclinical findings from animal models to human subjects has so far fallen short owing in part to the low predictive validity of the Fmr1 ko mouse, an overly simplistic model of the complex human disease. This issue stresses the critical need to identify new surrogate human peripheral cell models of FXS, which may in fact allow for the identification of novel and more efficient therapies. Of all described models, blood platelets appear to be one of the most promising and appropriate disease models of FXS, in part owing to their close biochemical similarities with neurons. Noteworthy, they also recapitulate some of FXS neuron's core molecular dysregulations, such as hyperactivity of the MAPK/ERK and PI3K/Akt/mTOR pathways, elevated enzymatic activity of MMP9 and decreased production of cAMP. Platelets might therefore help furthering our understanding of FXS pathophysiology and might also lead to the identification of disease-specific biomarkers, as was shown in several psychiatric disorders such as schizophrenia and Alzheimer's disease. Moreover, there is additional evidence suggesting that platelet signaling may assist with prediction of cognitive phenotype and could represent a potent readout of drug efficacy in clinical trials. Globally, given the neurobiological overlap between different forms of intellectual disability, platelets may be a valuable window to access the molecular underpinnings of ASD and other neurodevelopmental disorders (NDD) sharing similar synaptic plasticity defects with FXS. Platelets are indeed an attractive model for unraveling pathophysiological mechanisms involved in NDD as well as to search for diagnostic and therapeutic biomarkers.
Collapse
Affiliation(s)
- David Pellerin
- a Department of Biochemistry, Faculty of Medicine and Health Sciences , Université de Sherbrooke , Sherbrooke , QC , Canada.,b Department of Neurology and Neurosurgery, Faculty of Medicine , McGill University , Montreal , QC , Canada
| | - Audrey Lortie
- a Department of Biochemistry, Faculty of Medicine and Health Sciences , Université de Sherbrooke , Sherbrooke , QC , Canada
| | - François Corbin
- a Department of Biochemistry, Faculty of Medicine and Health Sciences , Université de Sherbrooke , Sherbrooke , QC , Canada
| |
Collapse
|
38
|
Rogers TD, Anacker AMJ, Kerr TM, Forsberg CG, Wang J, Zhang B, Veenstra-VanderWeele J. Effects of a social stimulus on gene expression in a mouse model of fragile X syndrome. Mol Autism 2017. [PMID: 28649315 PMCID: PMC5481916 DOI: 10.1186/s13229-017-0148-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND People with fragile X syndrome (FXS) often have deficits in social behavior, and a substantial portion meet criteria for autism spectrum disorder. Though the genetic cause of FXS is known to be due to the silencing of FMR1, and the Fmr1 null mouse model representing this lesion has been extensively studied, the contributions of this gene and its protein product, FMRP, to social behavior are not well understood. METHODS Fmr1 null mice and wildtype littermates were exposed to a social or non-social stimulus. In one experiment, subjects were assessed for expression of the inducible transcription factor c-Fos in response to the stimulus, to detect brain regions with social-specific activity. In a separate experiment, tissue was taken from those brain regions showing differential activity, and RNA sequencing was performed. RESULTS Immunohistochemistry revealed a significantly greater number of c-Fos-positive cells in the lateral amygdala and medial amygdala in the brains of mice exposed to a social stimulus, compared to a non-social stimulus. In the prelimbic cortex, there was no significant effect of social stimulus; although the number of c-Fos-positive cells was lower in the social condition compared to the non-social condition, and negatively correlated with c-Fos in the amygdala. RNA sequencing revealed differentially expressed genes enriched for molecules known to interact with FMRP and also for autism-related genes identified in the Simons Foundation Autism Research Initiative gene database. Ingenuity Pathway Analysis detected enrichment of differentially expressed genes in networks and pathways related to neuronal development, intracellular signaling, and inflammatory response. CONCLUSIONS Using the Fmr1 null mouse model of fragile X syndrome, we have identified brain regions, gene networks, and molecular pathways responsive to a social stimulus. These findings, and future experiments following up on the role of specific gene networks, may shed light on the neural mechanisms underlying dysregulated social behaviors in fragile X syndrome and more broadly.
Collapse
Affiliation(s)
- Tiffany D Rogers
- Department of Psychiatry, Vanderbilt University, 7158 MRBIII, 465 21st Avenue South, Nashville, TN 37232 USA.,Department of Psychology, Middle Tennessee State University, 355 Jones Hall, 624 Old Main Circle, Murfreesboro, TN 37132 USA
| | - Allison M J Anacker
- Department of Psychiatry, Columbia University; New York State Psychiatric Institute, 1051 Riverside Dr, Unit 78, New York, NY 10032 USA
| | - Travis M Kerr
- The University of Tennessee Health Science Center College of Medicine, 910 Madison Ave, Suite 1002, Memphis, TN 38163 USA
| | - C Gunnar Forsberg
- College of Medicine, Medical University of South Carolina, Charleston, SC 29425 USA
| | - Jing Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030 USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030 USA
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030 USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030 USA
| | - Jeremy Veenstra-VanderWeele
- Department of Psychiatry, Columbia University; New York State Psychiatric Institute, 1051 Riverside Dr, Unit 78, New York, NY 10032 USA
| |
Collapse
|
39
|
Niu M, Han Y, Dy ABC, Du J, Jin H, Qin J, Zhang J, Li Q, Hagerman RJ. Fragile X Syndrome: Prevalence, Treatment, and Prevention in China. Front Neurol 2017. [PMID: 28634468 PMCID: PMC5459883 DOI: 10.3389/fneur.2017.00254] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common inherited cause of intellectual disability and the leading monogenic cause of autism spectrum disorder. Although FXS has been studied for several decades, there is relatively little basic science or clinical research being performed on FXS in China. Indeed, there is a large gap between China and Western countries in the FXS field. China has a potentially large number of FXS patients. However, many of them are underdiagnosed or even misdiagnosed, and treatments are not always administered in the Chinese population. This review discusses the prevalence, treatment, and prevention of FXS in China to facilitate an understanding of this disease in the Chinese population.
Collapse
Affiliation(s)
- Manman Niu
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Ying Han
- Department of Pediatrics, Peking University First Hospital, Beijing, China.,MIND Institute, University of California-Davis Medical Center, Sacramento, CA, United States
| | - Angel Belle C Dy
- MIND Institute, University of California-Davis Medical Center, Sacramento, CA, United States.,Ateneo de Manila University - School of Medicine and Public Health, Quezon, Philippines
| | - Junbao Du
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Hongfang Jin
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Jiong Qin
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | - Jing Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Qinrui Li
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Randi J Hagerman
- MIND Institute, University of California-Davis Medical Center, Sacramento, CA, United States.,Department of Pediatrics, University of California-Davis Medical Center, Sacramento, CA, United States
| |
Collapse
|
40
|
Duy PQ, Budimirovic DB. Fragile X Syndrome: Lessons Learned from the Most Translated Neurodevelopmental Disorder in Clinical Trials. Transl Neurosci 2017; 8:7-8. [PMID: 28400977 PMCID: PMC5382936 DOI: 10.1515/tnsci-2017-0002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 02/23/2017] [Indexed: 11/25/2022] Open
Abstract
Fragile X syndrome (FXS) is the leading genetic cause of autism spectrum disorder (ASD) and inherited intellectual disability (ID) worldwide. Preclinical successes in understanding the biology of FXS have led to numerous translational attempts in human clinical trials of therapeutics that target the excitatory/inhibitory neural signaling imbalances thought to underlie FXS. Despite the preclinical success story, the negative results of the human clinical trials have been deemed to be at least in part disappointing by the field. In this commentary, we contend that such negative studies results in clinical trials may actually propel the FXS field forward by serving as important lessons for designing and implementing improved future clinical trials such that can objectively assess the full range of responses to new therapeutics.
Collapse
Affiliation(s)
- Phan Q. Duy
- Department of Biology Johns Hopkins University Baltimore, MD 21218, USA
- E-mail:
| | - Dejan B. Budimirovic
- Clinical Research Center, Clinical Trials Unit & Fragile X Clinic, Kennedy Krieger Institute, Johns Hopkins Medical Institutions Baltimore, MD 21205, USA
| |
Collapse
|
41
|
Sinclair D, Featherstone R, Naschek M, Nam J, Du A, Wright S, Pance K, Melnychenko O, Weger R, Akuzawa S, Matsumoto M, Siegel SJ. GABA-B Agonist Baclofen Normalizes Auditory-Evoked Neural Oscillations and Behavioral Deficits in the Fmr1 Knockout Mouse Model of Fragile X Syndrome. eNeuro 2017; 4:ENEURO.0380-16.2017. [PMID: 28451631 PMCID: PMC5394929 DOI: 10.1523/eneuro.0380-16.2017] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 02/09/2017] [Accepted: 02/13/2017] [Indexed: 12/15/2022] Open
Abstract
Fragile X syndrome is a genetic condition resulting from FMR1 gene mutation that leads to intellectual disability, autism-like symptoms, and sensory hypersensitivity. Arbaclofen, a GABA-B agonist, has shown efficacy in some individuals with FXS but has become unavailable after unsuccessful clinical trials, prompting interest in publicly available, racemic baclofen. The present study investigated whether racemic baclofen can remediate abnormalities of neural circuit function, sensory processing, and behavior in Fmr1 knockout mice, a rodent model of fragile X syndrome. Fmr1 knockout mice showed increased baseline and auditory-evoked high-frequency gamma (30-80 Hz) power relative to C57BL/6 controls, as measured by electroencephalography. These deficits were accompanied by decreased T maze spontaneous alternation, decreased social interactions, and increased open field center time, suggestive of diminished working memory, sociability, and anxiety-like behavior, respectively. Abnormal auditory-evoked gamma oscillations, working memory, and anxiety-related behavior were normalized by treatment with baclofen, but impaired sociability was not. Improvements in working memory were evident predominantly in mice whose auditory-evoked gamma oscillations were dampened by baclofen. These findings suggest that racemic baclofen may be useful for targeting sensory and cognitive disturbances in fragile X syndrome.
Collapse
Affiliation(s)
- D Sinclair
- Translational Neuroscience Program Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - R Featherstone
- Translational Neuroscience Program Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - M Naschek
- Translational Neuroscience Program Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - J Nam
- Translational Neuroscience Program Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - A Du
- Translational Neuroscience Program Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - S Wright
- Translational Neuroscience Program Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - K Pance
- Translational Neuroscience Program Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - O Melnychenko
- Translational Neuroscience Program Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - R Weger
- Translational Neuroscience Program Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - S Akuzawa
- Neuroscience Research Unit, DDR, Astellas Pharma Inc., Tsukuba-Shi, Ibaraki 305-8585, Japan
| | - M Matsumoto
- Neuroscience Research Unit, DDR, Astellas Pharma Inc., Tsukuba-Shi, Ibaraki 305-8585, Japan
| | - S J Siegel
- Translational Neuroscience Program Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
42
|
Genetic and Pharmacological Reversibility of Phenotypes in Mouse Models of Autism Spectrum Disorder. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2017; 224:189-211. [PMID: 28551757 DOI: 10.1007/978-3-319-52498-6_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
As autism spectrum disorder (ASD) is largely regarded as a neurodevelopmental condition, long-time consensus was that its hallmark features are irreversible. However, several studies from recent years using defined mouse models of ASD have provided clear evidence that in mice neurobiological and behavioural alterations can be ameliorated or even reversed by genetic restoration or pharmacological treatment either before or after symptom onset. Here, we review findings on genetic and pharmacological reversibility of phenotypes in mouse models of ASD. Our review should give a comprehensive overview on both aspects and encourage future studies to better understand the underlying molecular mechanisms that might be translatable from animals to humans.
Collapse
|
43
|
Berry-Kravis E, Des Portes V, Hagerman R, Jacquemont S, Charles P, Visootsak J, Brinkman M, Rerat K, Koumaras B, Zhu L, Barth GM, Jaecklin T, Apostol G, von Raison F. Mavoglurant in fragile X syndrome: Results of two randomized, double-blind, placebo-controlled trials. Sci Transl Med 2016; 8:321ra5. [PMID: 26764156 DOI: 10.1126/scitranslmed.aab4109] [Citation(s) in RCA: 179] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Fragile X syndrome (FXS), the most common cause of inherited intellectual disability and autistic spectrum disorder, is typically caused by transcriptional silencing of the X-linked FMR1 gene. Work in animal models has described altered synaptic plasticity, a result of the up-regulation of metabotropic glutamate receptor 5 (mGluR5)-mediated signaling, as a putative downstream effect. Post hoc analysis of a randomized, placebo-controlled, crossover phase 2 trial suggested that the selective mGluR5 antagonist mavoglurant improved behavioral symptoms in FXS patients with completely methylated FMR1 genes. We present the results of two phase 2b, multicenter, randomized, double-blind, placebo-controlled, parallel-group studies of mavoglurant in FXS, designed to confirm this result in adults (n = 175, aged 18 to 45 years) and adolescents (n = 139, aged 12 to 17 years). In both trials, participants were stratified by methylation status and randomized to receive mavoglurant (25, 50, or 100 mg twice daily) or placebo over 12 weeks. Neither of the studies achieved the primary efficacy end point of improvement on behavioral symptoms measured by the Aberrant Behavior Checklist-Community Edition using the FXS-specific algorithm (ABC-C(FX)) after 12 weeks of treatment with mavoglurant. The safety and tolerability profile of mavoglurant was as previously described, with few adverse events. Therefore, under the conditions of our study, we could not confirm the mGluR theory of FXS nor the ability of the methylation state of the FMR1 promoter to predict mavoglurant efficacy. Preclinical results suggest that future clinical trials might profitably explore initiating treatment in a younger population with longer treatment duration and longer placebo run-ins and identifying new markers to better assess behavioral and cognitive benefits.
Collapse
Affiliation(s)
- Elizabeth Berry-Kravis
- Departments of Pediatrics, Neurological Sciences, and Biochemistry, Rush University Medical Center, Chicago, IL 60612, USA
| | - Vincent Des Portes
- National Reference Center for Fragile X and Other X-Linked Intellectual Disabilities, CIC 1407 INSERM, Hospices Civils de Lyon, Université de Lyon, 69002 Lyon, France. CNRS UMR 5304 (Laboratoire sur le Langage, le Cerveau et la Cognition), 69500 Bron, France
| | - Randi Hagerman
- Medical Investigation of Neurodevelopmental Disorders Institute and Department of Pediatrics, UC Davis Medical Center, Sacramento, CA 95817, USA
| | - Sébastien Jacquemont
- Centre Hospitalier Universitaire Vaudois, CH-1011 Lausanne, Switzerland. CHU Sainte-Justine Research Centre, Montreal, Quebec H3T 1C5, Canada
| | - Perrine Charles
- Département de Génétique et Cytogénétique, Unité Fonctionnelle de Génétique Médicale, Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Pitié-Salpêtrière, 75013 Paris, France
| | - Jeannie Visootsak
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | - Karin Rerat
- Novartis Pharmaceuticals SAS, 92500 Rueil-Malmaison, France
| | - Barbara Koumaras
- Neurodegeneration Global Development, Novartis Pharmaceuticals Corporation, East Hanover, NJ 07936-1080, USA
| | - Liansheng Zhu
- Integrated Quantitative Science, Global Development, Novartis Pharmaceuticals Corporation, East Hanover, NJ 07936-1080, USA
| | - Gottfried Maria Barth
- Department of Child and Adolescent Psychiatry, University Hospital of Tübingen, 72076 Tübingen, Baden-Württemberg, Germany
| | - Thomas Jaecklin
- Neuroscience Development, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - George Apostol
- Neuroscience Development, Novartis Pharma AG, CH-4056 Basel, Switzerland
| | - Florian von Raison
- Neuroscience Development, Novartis Pharma AG, CH-4056 Basel, Switzerland.
| |
Collapse
|
44
|
Jeste SS, Geschwind DH. Clinical trials for neurodevelopmental disorders: At a therapeutic frontier. Sci Transl Med 2016; 8:321fs1. [PMID: 26764154 DOI: 10.1126/scitranslmed.aad9874] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
A well-powered clinical trial that failed to replicate promising results in animal models of fragile X syndrome yields important lessons for clinical trial design (Berry-Kravis et al., this issue).
Collapse
Affiliation(s)
- Shafali S Jeste
- Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California at Los Angeles (UCLA), Los Angeles, CA 90095-17610, USA
| | - Daniel H Geschwind
- Center for Autism Research and Treatment, Semel Institute, and Neurogenetics Program, Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095-1761, USA.
| |
Collapse
|
45
|
Abstract
The aims of this study were to investigate behaviour relevant to human autism spectrum disorder (ASD) and the fragile X syndrome in adolescent Fmr1 knockout (KO) mice and to evaluate the tissue levels of striatal monoamines. Fmr1 KO mice were evaluated in the open field, marble burying and three-chamber test for the presence of hyperactivity, anxiety, repetitive behaviour, sociability and observation of social novelty compared with wild-type (WT) mice. The Fmr1 KO mice expressed anxiety and hyperactivity in the open field compared with WT mice. This increased level of hyperactivity was confirmed in the three-chamber test. Fmr1 KO mice spent more time with stranger mice compared with the WT. However, after a correction for hyperactivity, their apparent increase in sociability became identical to that of the WT. Furthermore, the Fmr1 KO mice could not differentiate between a familiar or a novel mouse. Monoamines were measured by HPLC: Fmr1 KO mice showed an increase in the striatal dopamine level. We conclude that the fragile X syndrome model seems to be useful for understanding certain aspects of ASD and may have translational interest for studies of social behaviour when hyperactivity coexists in ASD patients.
Collapse
|
46
|
Fung LK, Reiss AL. Moving Toward Integrative, Multidimensional Research in Modern Psychiatry: Lessons Learned From Fragile X Syndrome. Biol Psychiatry 2016; 80:100-111. [PMID: 26868443 PMCID: PMC4912939 DOI: 10.1016/j.biopsych.2015.12.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Revised: 12/14/2015] [Accepted: 12/15/2015] [Indexed: 10/22/2022]
Abstract
The field of psychiatry is approaching a major inflection point. The basic science behind cognition, emotion, behavior, and social processes has been advancing rapidly in the past 20 years. However, clinical research supporting the classification system in psychiatry has not kept up with these scientific advances. To begin organizing the basic science of psychiatry in a comprehensive manner, we begin by selecting fragile X syndrome, a neurogenetic disease with cognitive-behavioral manifestations, to illustrate key concepts in an integrative, multidimensional model. Specifically, we describe key genetic and molecular mechanisms (e.g., gamma-aminobutyric acidergic dysfunction and metabotropic glutamate receptor 5-associated long-term depression) relevant to the pathophysiology of fragile X syndrome as well as neural correlates of cognitive-behavioral symptoms. We then describe what we have learned from fragile X syndrome that may be applicable to other psychiatric disorders. We conclude this review by discussing current and future opportunities in diagnosing and treating psychiatric diseases.
Collapse
Affiliation(s)
- Lawrence K. Fung
- Division of Child & Adolescent Psychiatry, Department of Psychiatry & Behavioral Sciences, Stanford University, Stanford, CA
| | - Allan L. Reiss
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry & Behavioral Sciences, Stanford University, Stanford, CA
| |
Collapse
|
47
|
Synaptic Plasticity, a Prominent Contributor to the Anxiety in Fragile X Syndrome. Neural Plast 2016; 2016:9353929. [PMID: 27239350 PMCID: PMC4864533 DOI: 10.1155/2016/9353929] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 04/04/2016] [Indexed: 01/03/2023] Open
Abstract
Fragile X syndrome (FXS) is an inheritable neuropsychological disease caused by expansion of the CGG trinucleotide repeat affecting the fmr1 gene on X chromosome, resulting in silence of the fmr1 gene and failed expression of FMRP. Patients with FXS suffer from cognitive impairment, sensory integration deficits, learning disability, anxiety, autistic traits, and so forth. Specifically, the morbidity of anxiety in FXS individuals remains high from childhood to adulthood. By and large, it is common that the change of brain plasticity plays a key role in the progression of disease. But for now, most studies excessively emphasized the one-sided factor on the change of synaptic plasticity participating in the generation of anxiety during the development of FXS. Here we proposed an integrated concept to acquire better recognition about the details of this process.
Collapse
|
48
|
Kazdoba TM, Leach PT, Yang M, Silverman JL, Solomon M, Crawley JN. Translational Mouse Models of Autism: Advancing Toward Pharmacological Therapeutics. Curr Top Behav Neurosci 2016; 28:1-52. [PMID: 27305922 PMCID: PMC5116923 DOI: 10.1007/7854_2015_5003] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Animal models provide preclinical tools to investigate the causal role of genetic mutations and environmental factors in the etiology of autism spectrum disorder (ASD). Knockout and humanized knock-in mice, and more recently knockout rats, have been generated for many of the de novo single gene mutations and copy number variants (CNVs) detected in ASD and comorbid neurodevelopmental disorders. Mouse models incorporating genetic and environmental manipulations have been employed for preclinical testing of hypothesis-driven pharmacological targets, to begin to develop treatments for the diagnostic and associated symptoms of autism. In this review, we summarize rodent behavioral assays relevant to the core features of autism, preclinical and clinical evaluations of pharmacological interventions, and strategies to improve the translational value of rodent models of autism.
Collapse
Affiliation(s)
- Tatiana M Kazdoba
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA
| | - Prescott T Leach
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA
| | - Mu Yang
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA
| | - Jill L Silverman
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA
| | - Marjorie Solomon
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA
| | - Jacqueline N Crawley
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA.
| |
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW This review discusses the ways that rodent models of autism spectrum disorders (ASDs) have been used to gain critical information about convergent molecular pathways, the mechanisms underlying altered microcircuit structure and function, and as a screen for potential cutting edge-treatments for ASDs. RECENT FINDINGS There is convergent evidence that impaired developmental pruning of connections may be a common finding among several mouse models of ASDs. Recent studies have uncovered impaired autophagy by pathological mTOR activation as a potential contributor to microcircuit dysfunction and behavior. ASD-related disinhibition and exaggerated synaptic plasticity in multiple distinct circuits in cortex and reward circuits in striatum also contribute to social dysfunction and repetitive behaviors. New exciting molecular therapeutic techniques have reversed cognitive deficits in models of ASD, indicating that mouse models could be used for preclinical translational studies of new treatments. SUMMARY Rodent models of ASDs coupled to new emerging technologies for genome editing, cell-specific functional and structural imaging, and neuronal activity manipulation will yield critical insights into ASD pathogenesis and fuel the emergence of new treatments.
Collapse
|
50
|
Sanders SJ. First glimpses of the neurobiology of autism spectrum disorder. Curr Opin Genet Dev 2015; 33:80-92. [PMID: 26547130 DOI: 10.1016/j.gde.2015.10.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 10/05/2015] [Accepted: 10/07/2015] [Indexed: 12/22/2022]
Abstract
Rapid progress in identifying the genes underlying autism spectrum disorder (ASD) has provided the substrate for a first wave of analyses into the underlying neurobiology. This review describes the consensus across these diverse analyses, highlighting two distinct sets of genes: 1) Genes that regulate chromatin and transcription, especially in cortical projection neurons and striatal medium spiny neurons during mid-fetal development; and 2) Genes involved in synapse development and function, especially during infancy and early childhood, and differentially expressed in the post mortem ASD brain. Both gene sets are also regulatory targets of the ASD genes CHD8 and FMRP. It remains to be seen whether these represent two independent paths to the ASD phenotype or two components of a common path.
Collapse
Affiliation(s)
- Stephan J Sanders
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|