1
|
Kaiser J, Risteska A, Muller AG, Sun H, Lei B, Nay K, Means AR, Cousin MA, Drewry DH, Oakhill JS, Kemp BE, Hannan AJ, Berk M, Febbraio MA, Gundlach AL, Hill-Yardin EL, Scott JW. Convergence on CaMK4: A Key Modulator of Autism-Associated Signaling Pathways in Neurons. Biol Psychiatry 2025; 97:439-449. [PMID: 39442785 DOI: 10.1016/j.biopsych.2024.10.012] [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: 07/09/2024] [Revised: 09/03/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
Although the precise underlying cause(s) of autism spectrum disorder remain unclear, more than 1000 rare genetic variations are associated with the condition. For many people living with profound autism, this genetic heterogeneity has impeded the identification of common biological targets for therapy development for core and comorbid traits that include significant impairments in social communication and repetitive and restricted behaviors. A substantial number of genes associated with autism encode proteins involved in signal transduction and synaptic transmission that are critical for brain development and function. CAMK4 is an emerging risk gene for autism spectrum disorder that encodes the CaMK4 (calcium/calmodulin-dependent protein kinase 4) enzyme. CaMK4 is a key component of a Ca2+-activated signaling pathway that regulates neurodevelopment and synaptic plasticity. In this review, we discuss 3 genetic variants of CAMK4 found in individuals with hyperkinetic movement disorder and comorbid neurological symptoms including autism spectrum disorder that are likely pathogenic with monogenic effect. We also comment on 4 other genetic variations in CAMK4 that show associations with autism spectrum disorder, as well as 12 examples of autism-associated variations in other genes that impact CaMK4 signaling pathways. Finally, we highlight 3 environmental risk factors that impact CaMK4 signaling based on studies of preclinical models of autism and/or clinical cohorts. Overall, we review molecular, genetic, physiological, and environmental evidence that suggest that defects in the CaMK4 signaling pathway may play an important role in a common autism pathogenesis network across numerous patient groups, and we propose CaMK4 as a potential therapeutic target.
Collapse
Affiliation(s)
- Jacqueline Kaiser
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia; Mary McKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, Australia
| | - Alana Risteska
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Abbey G Muller
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia; Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Haoxiong Sun
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Bethany Lei
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Kevin Nay
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Anthony R Means
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Margot A Cousin
- Center for Individualized Medicine, College of Medicine, Mayo Clinic, Rochester, Minnesota
| | - David H Drewry
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, the University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jonathan S Oakhill
- St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia; Mary McKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, Australia
| | - Bruce E Kemp
- St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia; Department of Anatomy and Physiology, the University of Melbourne, Melbourne, Victoria, Australia
| | - Michael Berk
- Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia; The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, Melbourne, Australia
| | - Mark A Febbraio
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia
| | - Andrew L Gundlach
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia; Department of Anatomy and Physiology, the University of Melbourne, Melbourne, Victoria, Australia
| | - Elisa L Hill-Yardin
- Department of Anatomy and Physiology, the University of Melbourne, Melbourne, Victoria, Australia; School of Health and Biomedical Sciences, STEM College, RMIT University, Melbourne, Victoria, Australia.
| | - John W Scott
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, Victoria, Australia; St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia; Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia.
| |
Collapse
|
2
|
Tuma J, Rana AN, Philip T, Park JB, Lee HY. Altered olfactory responses in Fmr1 KO mice. Sci Rep 2025; 15:2952. [PMID: 39848954 PMCID: PMC11758012 DOI: 10.1038/s41598-024-80000-5] [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: 04/16/2023] [Accepted: 11/14/2024] [Indexed: 01/25/2025] Open
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder oftentimes associated with abnormal social behaviors and altered sensory responsiveness. It is hypothesized that the inappropriate filtering of sensory stimuli, including olfaction, can lead to aberrant social behavior in FXS. However, previous studies investigating olfaction in animal models of FXS have shown inconsistent results. Here, we found that Fmr1 knock-out (KO) mice, a mouse model of FXS, showed increased sniffing duration for non-social odors during their first exposure. Additionally, while wild-type (WT) males demonstrated differences in behavioral patterns between non-social odors while Fmr1 KO males did not show such distinction. We also showed that Fmr1 KO males spent significantly less time sniffing female urine odor compared to WT males. Moreover, we found an increased volume of the olfactory bulb in Fmr1 KO males. Overall, our findings suggest that the Fmr1 KO mice demonstrate atypical olfactory behaviors as well as structural changes in the olfactory bulb.
Collapse
Affiliation(s)
- Jan Tuma
- The Department of Cellular and Integrative Physiology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Pathophysiology, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, 323 00, Plzen, Czech Republic
| | - Amtul-Noor Rana
- The Department of Cellular and Integrative Physiology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Teena Philip
- The Department of Cellular and Integrative Physiology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jeong Ben Park
- The Department of Cellular and Integrative Physiology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Hye Young Lee
- The Department of Cellular and Integrative Physiology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
3
|
Galineau L, Arlicot N, Dupont AC, Briend F, Houy-Durand E, Tauber C, Gomot M, Gissot V, Barantin L, Lefevre A, Vercouillie J, Roussel C, Roux S, Nadal L, Mavel S, Laumonnier F, Belzung C, Chalon S, Emond P, Santiago-Ribeiro MJ, Bonnet-Brilhault F. Glutamatergic synapse in autism: a complex story for a complex disorder. Mol Psychiatry 2023; 28:801-809. [PMID: 36434055 DOI: 10.1038/s41380-022-01860-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/12/2022] [Accepted: 10/28/2022] [Indexed: 11/27/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder whose pathophysiological mechanisms are still unclear. Hypotheses suggest a role for glutamate dysfunctions in ASD development, but clinical studies investigating brain and peripheral glutamate levels showed heterogenous results leading to hypo- and hyper-glutamatergic hypotheses of ASD. Recently, studies proposed the implication of elevated mGluR5 densities in brain areas in the pathophysiology of ASD. Thus, our objective was to characterize glutamate dysfunctions in adult subjects with ASD by quantifying (1) glutamate levels in the cingulate cortex and periphery using proton magnetic resonance spectroscopy and metabolomics, and (2) mGluR5 brain density in this population and in a validated animal model of ASD (prenatal exposure to valproate) at developmental stages corresponding to childhood and adolescence in humans using positron emission tomography. No modifications in cingulate Glu levels were observed between individuals with ASD and controls further supporting the difficulty to evaluate modifications in excitatory transmission using spectroscopy in this population, and the complexity of its glutamate-related changes. Our imaging results showed an overall increased density in mGluR5 in adults with ASD, that was only observed mostly subcortically in adolescent male rats prenatally exposed to valproic acid, and not detected in the stage corresponding to childhood in the same animals. This suggest that clinical changes in mGluR5 density could reflect the adaptation of the glutamatergic dysfunctions occurring earlier rather than being key to the pathophysiology of ASD.
Collapse
Affiliation(s)
| | - Nicolas Arlicot
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Unité de Radiopharmacie, CHRU de Tours, Tours, France
| | - Anne-Claire Dupont
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Unité de Radiopharmacie, CHRU de Tours, Tours, France.,Service de Médecine Nucléaire, CHRU de Tours, Tours, France
| | - Frederic Briend
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Excellence Center for Autism and Neurodevelopmental Disorders, CHRU de Tours, Tours, France
| | - Emmanuelle Houy-Durand
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Excellence Center for Autism and Neurodevelopmental Disorders, CHRU de Tours, Tours, France
| | - Clovis Tauber
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Marie Gomot
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Excellence Center for Autism and Neurodevelopmental Disorders, CHRU de Tours, Tours, France
| | | | | | - Antoine Lefevre
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | | | | | - Sylvie Roux
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Excellence Center for Autism and Neurodevelopmental Disorders, CHRU de Tours, Tours, France
| | - Lydie Nadal
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Sylvie Mavel
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | | | | | - Sylvie Chalon
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Patrick Emond
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Maria-Joao Santiago-Ribeiro
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Service de Médecine Nucléaire, CHRU de Tours, Tours, France
| | - Frédérique Bonnet-Brilhault
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France. .,Excellence Center for Autism and Neurodevelopmental Disorders, CHRU de Tours, Tours, France.
| |
Collapse
|
4
|
Kwan C, Kang W, Kim E, Belliveau S, Frouni I, Huot P. Metabotropic glutamate receptors in Parkinson's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:1-31. [PMID: 36868628 DOI: 10.1016/bs.irn.2022.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Parkinson's disease (PD) is a complex disorder that leads to alterations in multiple neurotransmitter systems, notably glutamate. As such, several drugs acting at glutamatergic receptors have been assessed to alleviate the manifestation of PD and treatment-related complications, culminating with the approval of the N-methyl-d-aspartate (NMDA) antagonist amantadine for l-3,4-dihydroxyphenylalanine (l-DOPA)-induced dyskinesia. Glutamate elicits its actions through several ionotropic and metabotropic (mGlu) receptors. There are 8 sub-types of mGlu receptors, with sub-types 4 (mGlu4) and 5 (mGlu5) modulators having been tested in the clinic for endpoints pertaining to PD, while sub-types 2 (mGlu2) and 3 (mGlu3) have been investigated in pre-clinical settings. In this book chapter, we provide an overview of mGlu receptors in PD, with a focus on mGlu5, mGlu4, mGlu2 and mGlu3 receptors. For each sub-type, we review, when applicable, their anatomical localization and possible mechanisms underlying their efficacy for specific disease manifestation or treatment-induced complications. We then summarize the findings of pre-clinical studies and clinical trials with pharmacological agents and discuss the potential strengths and limitations of each target. We conclude by offering some perspectives on the potential use of mGlu modulators in the treatment of PD.
Collapse
Affiliation(s)
- Cynthia Kwan
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada
| | - Woojin Kang
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada
| | - Esther Kim
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada
| | - Sébastien Belliveau
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada
| | - Imane Frouni
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada; Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
| | - Philippe Huot
- Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada; Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada; Movement Disorder Clinic, Division of Neurology, Department of Neurosciences, McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
5
|
Kampen S, Rodríguez D, Jørgensen M, Kruszyk-Kujawa M, Huang X, Collins M, Boyle N, Maurel D, Rudling A, Lebon G, Carlsson J. Structure-Based Discovery of Negative Allosteric Modulators of the Metabotropic Glutamate Receptor 5. ACS Chem Biol 2022; 17:2744-2752. [PMID: 36149353 PMCID: PMC9594040 DOI: 10.1021/acschembio.2c00234] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Recently determined structures of class C G protein-coupled receptors (GPCRs) revealed the location of allosteric binding sites and opened new opportunities for the discovery of novel modulators. In this work, molecular docking screens for allosteric modulators targeting the metabotropic glutamate receptor 5 (mGlu5) were performed. The mGlu5 receptor is activated by the main excitatory neurotransmitter of the nervous central system, L-glutamate, and mGlu5 receptor activity can be allosterically modulated by negative or positive allosteric modulators. The mGlu5 receptor is a promising target for the treatment of psychiatric and neurodegenerative diseases, and several allosteric modulators of this GPCR have been evaluated in clinical trials. Chemical libraries containing fragment- (1.6 million molecules) and lead-like (4.6 million molecules) compounds were docked to an allosteric binding site of mGlu5 identified in X-ray crystal structures. Among the top-ranked compounds, 59 fragments and 59 lead-like compounds were selected for experimental evaluation. Of these, four fragment- and seven lead-like compounds were confirmed to bind to the allosteric site with affinities ranging from 0.43 to 8.6 μM, corresponding to a hit rate of 9%. The four compounds with the highest affinities were demonstrated to be negative allosteric modulators of mGlu5 signaling in functional assays. The results demonstrate that virtual screens of fragment- and lead-like chemical libraries have complementary advantages and illustrate how access to high-resolution structures of GPCRs in complex with allosteric modulators can accelerate lead discovery.
Collapse
Affiliation(s)
- Stefanie Kampen
- Science
for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, SE-751 24 Uppsala, Sweden
| | - David Rodríguez
- Science
for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, SE-171 21 Solna, Sweden,H.
Lundbeck A/S, Ottiliavej
9, DK-2500 Valby, Denmark
| | | | | | - Xinyan Huang
- Lundbeck
Research USA, 215 College Road, Paramus, New Jersey 07652 - 1431, United States
| | - Michael Collins
- Lundbeck
Research USA, 215 College Road, Paramus, New Jersey 07652 - 1431, United States
| | - Noel Boyle
- Lundbeck
Research USA, 215 College Road, Paramus, New Jersey 07652 - 1431, United States
| | - Damien Maurel
- IGF,
Université de Montpellier, CNRS, INSERM, 34094 Montpellier, France
| | - Axel Rudling
- Science
for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, SE-171 21 Solna, Sweden
| | - Guillaume Lebon
- IGF,
Université de Montpellier, CNRS, INSERM, 34094 Montpellier, France
| | - Jens Carlsson
- Science
for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, SE-751 24 Uppsala, Sweden,
| |
Collapse
|
6
|
From bench to bedside: The mGluR5 system in people with and without Autism Spectrum Disorder and animal model systems. Transl Psychiatry 2022; 12:395. [PMID: 36127322 PMCID: PMC9489881 DOI: 10.1038/s41398-022-02143-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 08/19/2022] [Accepted: 09/01/2022] [Indexed: 11/08/2022] Open
Abstract
The metabotropic glutamate receptor 5 (mGluR5) is a key regulator of excitatory (E) glutamate and inhibitory (I) γ-amino butyric acid (GABA) signalling in the brain. Despite the close functional ties between mGluR5 and E/I signalling, no-one has directly examined the relationship between mGluR5 and glutamate or GABA in vivo in the human brain of autistic individuals. We measured [18F] FPEB (18F-3-fluoro-5-[(pyridin-3-yl)ethynyl]benzonitrile) binding in 15 adults (6 with Autism Spectrum Disorder) using two regions of interest, the left dorsomedial prefrontal cortex and a region primarily composed of left striatum and thalamus. These two regions were mapped out using MEGA-PRESS voxels and then superimposed on reconstructed PET images. This allowed for direct comparison between mGluR5, GABA + and Glx. To better understand the molecular underpinnings of our results we used an autoradiography study of mGluR5 in three mouse models associated with ASD: Cntnap2 knockout, Shank3 knockout, and 16p11.2 deletion. Autistic individuals had significantly higher [18F] FPEB binding (t (13) = -2.86, p = 0.047) in the left striatum/thalamus region of interest as compared to controls. Within this region, there was a strong negative correlation between GABA + and mGluR5 density across the entire cohort (Pearson's correlation: r (14) = -0.763, p = 0.002). Cntnap2 KO mice had significantly higher mGlu5 receptor binding in the striatum (caudate-putamen) as compared to wild-type (WT) mice (n = 15, p = 0.03). There were no differences in mGluR5 binding for mice with the Shank3 knockout or 16p11.2 deletion. Given that Cntnap2 is associated with a specific striatal deficit of parvalbumin positive GABA interneurons and 'autistic' features, our findings suggest that an increase in mGluR5 in ASD may relate to GABAergic interneuron abnormalities.
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
|
Budgett RF, Bakker G, Sergeev E, Bennett KA, Bradley SJ. Targeting the Type 5 Metabotropic Glutamate Receptor: A Potential Therapeutic Strategy for Neurodegenerative Diseases? Front Pharmacol 2022; 13:893422. [PMID: 35645791 PMCID: PMC9130574 DOI: 10.3389/fphar.2022.893422] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/18/2022] [Indexed: 01/13/2023] Open
Abstract
The type 5 metabotropic glutamate receptor, mGlu5, has been proposed as a potential therapeutic target for the treatment of several neurodegenerative diseases. In preclinical neurodegenerative disease models, novel allosteric modulators have been shown to improve cognitive performance and reduce disease-related pathology. A common pathological hallmark of neurodegenerative diseases is a chronic neuroinflammatory response, involving glial cells such as astrocytes and microglia. Since mGlu5 is expressed in astrocytes, targeting this receptor could provide a potential mechanism by which neuroinflammatory processes in neurodegenerative disease may be modulated. This review will discuss current evidence that highlights the potential of mGlu5 allosteric modulators to treat neurodegenerative diseases, including Alzheimer’s disease, Huntington’s disease, Parkinson’s disease, and amyotrophic lateral sclerosis. Furthermore, this review will explore the role of mGlu5 in neuroinflammatory responses, and the potential for this G protein-coupled receptor to modulate neuroinflammation.
Collapse
Affiliation(s)
- Rebecca F Budgett
- The Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | | | | | | | - Sophie J Bradley
- The Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom.,Sosei Heptares, Cambridge, United Kingdom
| |
Collapse
|
9
|
Altimiras F, Garcia JA, Palacios-García I, Hurley MJ, Deacon R, González B, Cogram P. Altered Gut Microbiota in a Fragile X Syndrome Mouse Model. Front Neurosci 2021; 15:653120. [PMID: 34121987 PMCID: PMC8190892 DOI: 10.3389/fnins.2021.653120] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/06/2021] [Indexed: 01/09/2023] Open
Abstract
The human gut microbiome is the ecosystem of microorganisms that live in the human digestive system. Several studies have related gut microbiome variants to metabolic, immune and nervous system disorders. Fragile X syndrome (FXS) is a neurodevelopmental disorder considered the most common cause of inherited intellectual disability and the leading monogenetic cause of autism. The role of the gut microbiome in FXS remains largely unexplored. Here, we report the results of a gut microbiome analysis using a FXS mouse model and 16S ribosomal RNA gene sequencing. We identified alterations in the fmr1 KO2 gut microbiome associated with different bacterial species, including those in the genera Akkermansia, Sutterella, Allobaculum, Bifidobacterium, Odoribacter, Turicibacter, Flexispira, Bacteroides, and Oscillospira. Several gut bacterial metabolic pathways were significantly altered in fmr1 KO2 mice, including menaquinone degradation, catechol degradation, vitamin B6 biosynthesis, fatty acid biosynthesis, and nucleotide metabolism. Several of these metabolic pathways, including catechol degradation, nucleotide metabolism and fatty acid biosynthesis, were previously reported to be altered in children and adults with autism. The present study reports a potential association of the gut microbiome with FXS, thereby opening new possibilities for exploring reliable treatments and non-invasive biomarkers.
Collapse
Affiliation(s)
- Francisco Altimiras
- Faculty of Engineering, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile.,Faculty of Engineering and Business, Universidad de las Américas, Santiago, Chile
| | - José Antonio Garcia
- Faculty of Engineering, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Ismael Palacios-García
- School of Psychology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Estudios en Neurociencia Humana y Neuropsicología, Facultad de Psicología, Universidad Diego Portales, Santiago, Chile
| | - Michael J Hurley
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Robert Deacon
- Department of Genetics, Institute of Ecology and Biodiversity (IEB), Faculty of Sciences, Universidad de Chile, Santiago, Chile.,FRAXA-DVI, FRAXA Research Foundation, Santiago, Chile
| | - Bernardo González
- Faculty of Engineering and Sciences, Universidad Adolfo Ibáñez, Santiago, Chile.,Center of Applied Ecology and Sustainability (CAPES), Santiago, Chile
| | - Patricia Cogram
- Department of Genetics, Institute of Ecology and Biodiversity (IEB), Faculty of Sciences, Universidad de Chile, Santiago, Chile.,FRAXA-DVI, FRAXA Research Foundation, Santiago, Chile
| |
Collapse
|
10
|
Feuge J, Scharkowski F, Michaelsen-Preusse K, Korte M. FMRP Modulates Activity-Dependent Spine Plasticity by Binding Cofilin1 mRNA and Regulating Localization and Local Translation. Cereb Cortex 2020; 29:5204-5216. [PMID: 30953439 DOI: 10.1093/cercor/bhz059] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/14/2019] [Accepted: 02/27/2019] [Indexed: 11/13/2022] Open
Abstract
Multiple variants of intellectual disability, e.g., the Fragile X Syndrome are associated with alterations in dendritic spine morphology, thereby pointing to dysregulated actin dynamics during development and processes of synaptic plasticity. Surprisingly, although the necessity of spine actin remodeling was demonstrated repeatedly, the importance and precise role of actin regulators is often undervalued. Here, we provide evidence that structural and functional plasticity are severely impaired after NMDAR-dependent LTP in the hippocampus of Fmr1 KO mice. We can link these defects to an aberrant activity-dependent regulation of Cofilin 1 (cof1) as activity-dependent modulations of local cof1 mRNA availability, local cof1 translation as well as total cof1 expression are impaired in the absence of FMRP. Finally, we can rescue activity-dependent structural plasticity in KO neurons by mimicking the regulation of cof1 observed in WT cells, thereby illustrating the potential of actin modulators to provide novel treatment strategies for the Fragile X Syndrome.
Collapse
Affiliation(s)
- Jonas Feuge
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Germany
| | | | | | - Martin Korte
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig, Germany.,Helmholtz Center for Infection Research, Research group Neuroinflammation and Neurodegeneration, Braunschweig, Germany
| |
Collapse
|
11
|
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 PMCID: PMC7705108 DOI: 10.1124/mol.119.119032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [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
|
12
|
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] [MESH Headings] [Grants] [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
|
13
|
Bonifacino T, Rebosio C, Provenzano F, Torazza C, Balbi M, Milanese M, Raiteri L, Usai C, Fedele E, Bonanno G. Enhanced Function and Overexpression of Metabotropic Glutamate Receptors 1 and 5 in the Spinal Cord of the SOD1 G93A Mouse Model of Amyotrophic Lateral Sclerosis during Disease Progression. Int J Mol Sci 2019; 20:ijms20184552. [PMID: 31540330 PMCID: PMC6774337 DOI: 10.3390/ijms20184552] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/26/2019] [Accepted: 09/12/2019] [Indexed: 12/11/2022] Open
Abstract
Glutamate (Glu)-mediated excitotoxicity is a major cause of amyotrophic lateral sclerosis (ALS) and our previous work highlighted that abnormal Glu release may represent a leading mechanism for excessive synaptic Glu. We demonstrated that group I metabotropic Glu receptors (mGluR1, mGluR5) produced abnormal Glu release in SOD1G93A mouse spinal cord at a late disease stage (120 days). Here, we studied this phenomenon in pre-symptomatic (30 and 60 days) and early-symptomatic (90 days) SOD1G93A mice. The mGluR1/5 agonist (S)-3,5-Dihydroxyphenylglycine (3,5-DHPG) concentration dependently stimulated the release of [3H]d-Aspartate ([3H]d-Asp), which was comparable in 30- and 60-day-old wild type mice and SOD1G93A mice. At variance, [3H]d-Asp release was significantly augmented in 90-day-old SOD1G93A mice and both mGluR1 and mGluR5 were involved. The 3,5-DHPG-induced [3H]d-Asp release was exocytotic, being of vesicular origin and mediated by intra-terminal Ca2+ release. mGluR1 and mGluR5 expression was increased in Glu spinal cord axon terminals of 90-day-old SOD1G93A mice, but not in the whole axon terminal population. Interestingly, mGluR1 and mGluR5 were significantly augmented in total spinal cord tissue already at 60 days. Thus, function and expression of group I mGluRs are enhanced in the early-symptomatic SOD1G93A mouse spinal cord, possibly participating in excessive Glu transmission and supporting their implication in ALS. Please define all abbreviations the first time they appear in the abstract, the main text, and the first figure or table caption.
Collapse
Affiliation(s)
- Tiziana Bonifacino
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, 16148 Genova, Italy.
| | - Claudia Rebosio
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, 16148 Genova, Italy.
| | - Francesca Provenzano
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, 16148 Genova, Italy.
| | - Carola Torazza
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, 16148 Genova, Italy.
| | - Matilde Balbi
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, 16148 Genova, Italy.
| | - Marco Milanese
- Department of Pharmacy, Unit of Pharmacology and Toxicology and Center of Excellence for Biomedical Research (CEBR), University of Genoa, 16132 Genova, Italy.
| | - Luca Raiteri
- Department of Pharmacy, Unit of Pharmacology and Toxicology and Center of Excellence for Biomedical Research (CEBR), University of Genoa, 16132 Genova, Italy.
| | - Cesare Usai
- Institute of Biophysics, National Research Council (CNR), 16149 Genova, Italy.
| | - Ernesto Fedele
- Department of Pharmacy, Unit of Pharmacology and Toxicology and Center of Excellence for Biomedical Research (CEBR), University of Genoa, 16132 Genova, Italy.
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, 16132 Genova, Italy.
| | - Giambattista Bonanno
- Department of Pharmacy, Unit of Pharmacology and Toxicology and Center of Excellence for Biomedical Research (CEBR), University of Genoa, 16132 Genova, Italy.
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Policlinico San Martino, 16132 Genova, Italy.
| |
Collapse
|
14
|
Masilamoni GJ, Smith Y. Group I metabotropic glutamate receptors in the primate motor thalamus: subsynaptic association with cortical and sub-cortical glutamatergic afferents. Brain Struct Funct 2019; 224:2787-2804. [PMID: 31422483 DOI: 10.1007/s00429-019-01937-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/07/2019] [Indexed: 12/21/2022]
Abstract
Preclinical evidence indicates that mGluR5 is a potential therapeutic target for Parkinson's disease and L-DOPA-induced dyskinesia. However, the mechanisms through which these therapeutic benefits are mediated remain poorly understood. Although the regulatory role of mGluR5 on glutamatergic transmission has been examined in various basal ganglia nuclei, very little is known about the localization and function of mGluR5 in the ventral motor and intralaminar thalamic nuclei, the main targets of basal ganglia output in mammals. Thus, we used immuno-electron microscopy to map the cellular and subcellular localization of group I mGluRs (mGluR1a and mGluR5) in the ventral motor and caudal intralaminar thalamic nuclei in rhesus monkeys. Furthermore, using double immuno-electron microscopy, we examined the subsynaptic localization of mGluR5 in relation to cortical and sub-cortical glutamatergic afferents. Four major conclusions can be drawn from these data. First, mGluR1a and mGluR5 are expressed postsynaptically on the plasma membrane of dendrites of projection neurons and GABAergic interneurons in the basal ganglia- and cerebellar-receiving regions of the ventral motor thalamus and in CM. Second, the plasma membrane-bound mGluR5 immunoreactivity is preferentially expressed perisynaptically at the edges of cortical and sub-cortical glutamatergic afferents. Third, the mGluR5 immunoreactivity is more strongly expressed in the lateral than the medial tiers of CM, suggesting a preferential association with thalamocortical over thalamostriatal neurons in the primate CM. Overall, mGluR5 is located to subserve powerful modulatory role of cortical and subcortical glutamatergic transmission in the primate ventral motor thalamus and CM.
Collapse
Affiliation(s)
- Gunasingh Jeyaraj Masilamoni
- Yerkes National Primate Research Center, Emory University, 954, Gatewood Rd NE, Atlanta, GA, 30329, USA. .,Udall Center of Excellence for Parkinson's Disease, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Yoland Smith
- Yerkes National Primate Research Center, Emory University, 954, Gatewood Rd NE, Atlanta, GA, 30329, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Udall Center of Excellence for Parkinson's Disease, Emory University School of Medicine, Atlanta, GA, 30322, USA
| |
Collapse
|
15
|
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
|
16
|
Biased agonism and allosteric modulation of metabotropic glutamate receptor 5. Clin Sci (Lond) 2018; 132:2323-2338. [PMID: 30389826 DOI: 10.1042/cs20180374] [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: 08/16/2018] [Revised: 10/03/2018] [Accepted: 10/05/2018] [Indexed: 12/18/2022]
Abstract
Metabotropic glutamate receptors belong to class C G-protein-coupled receptors and consist of eight subtypes that are ubiquitously expressed throughout the central nervous system. In recent years, the metabotropic glutamate receptor subtype 5 (mGlu5) has emerged as a promising target for a broad range of psychiatric and neurological disorders. Drug discovery programs targetting mGlu5 are primarily focused on development of allosteric modulators that interact with sites distinct from the endogenous agonist glutamate. Significant efforts have seen mGlu5 allosteric modulators progress into clinical trials; however, recent failures due to lack of efficacy or adverse effects indicate a need for a better understanding of the functional consequences of mGlu5 allosteric modulation. Biased agonism is an interrelated phenomenon to allosterism, describing how different ligands acting through the same receptor can differentially influence signaling to distinct transducers and pathways. Emerging evidence demonstrates that allosteric modulators can induce biased pharmacology at the level of intrinsic agonism as well as through differential modulation of orthosteric agonist-signaling pathways. Here, we present key considerations in the discovery and development of mGlu5 allosteric modulators and the opportunities and pitfalls offered by biased agonism and modulation.
Collapse
|
17
|
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
|
18
|
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: 87] [Impact Index Per Article: 12.4] [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
|
19
|
Lu S, Zhang J. Small Molecule Allosteric Modulators of G-Protein-Coupled Receptors: Drug–Target Interactions. J Med Chem 2018; 62:24-45. [DOI: 10.1021/acs.jmedchem.7b01844] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Shaoyong Lu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Jian Zhang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| |
Collapse
|
20
|
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
|
21
|
Goniotaki D, Lakkaraju AKK, Shrivastava AN, Bakirci P, Sorce S, Senatore A, Marpakwar R, Hornemann S, Gasparini F, Triller A, Aguzzi A. Inhibition of group-I metabotropic glutamate receptors protects against prion toxicity. PLoS Pathog 2017; 13:e1006733. [PMID: 29176838 PMCID: PMC5720820 DOI: 10.1371/journal.ppat.1006733] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/07/2017] [Accepted: 11/04/2017] [Indexed: 12/29/2022] Open
Abstract
Prion infections cause inexorable, progressive neurological dysfunction and neurodegeneration. Expression of the cellular prion protein PrPC is required for toxicity, suggesting the existence of deleterious PrPC-dependent signaling cascades. Because group-I metabotropic glutamate receptors (mGluR1 and mGluR5) can form complexes with the cellular prion protein (PrPC), we investigated the impact of mGluR1 and mGluR5 inhibition on prion toxicity ex vivo and in vivo. We found that pharmacological inhibition of mGluR1 and mGluR5 antagonized dose-dependently the neurotoxicity triggered by prion infection and by prion-mimetic anti-PrPC antibodies in organotypic brain slices. Prion-mimetic antibodies increased mGluR5 clustering around dendritic spines, mimicking the toxicity of Aβ oligomers. Oral treatment with the mGluR5 inhibitor, MPEP, delayed the onset of motor deficits and moderately prolonged survival of prion-infected mice. Although group-I mGluR inhibition was not curative, these results suggest that it may alleviate the neurological dysfunctions induced by prion diseases.
Collapse
Affiliation(s)
| | | | - Amulya N. Shrivastava
- École Normale Supérieure, Institut de Biologie de l'ENS (IBENS) INSERM CNRS PSL Research University, Paris, France
- Paris-Saclay Institute of Neuroscience, CNRS, Gif-sur-Yvette, France
| | - Pamela Bakirci
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Silvia Sorce
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Assunta Senatore
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | | | - Simone Hornemann
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | | | - Antoine Triller
- École Normale Supérieure, Institut de Biologie de l'ENS (IBENS) INSERM CNRS PSL Research University, Paris, France
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
22
|
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
|
23
|
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
|
24
|
Bian Y, Feng Z, Yang P, Xie XQ. Integrated In Silico Fragment-Based Drug Design: Case Study with Allosteric Modulators on Metabotropic Glutamate Receptor 5. AAPS JOURNAL 2017; 19:1235-1248. [PMID: 28560482 DOI: 10.1208/s12248-017-0093-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 04/26/2017] [Indexed: 12/22/2022]
Abstract
GPCR allosteric modulators target at the allosteric binding pockets of G protein-coupled receptors (GPCRs) with indirect influence on the effects of an orthosteric ligand. Such modulators exhibit significant advantages compared to the corresponding orthosteric ligands, including better chemical tractability or physicochemical properties, improved selectivity, and reduced risk of oversensitization towards their receptors. Metabotropic glutamate receptor 5 (mGlu5), a member of class C GPCRs, is a promising therapeutic target for treating many central nervous system diseases. The crystal structure of mGlu5 in the complex with the negative allosteric modulator mavoglurant was recently reported, providing a fundamental model for designing new allosteric modulators. Computational fragment-based drug discovery represents a powerful scaffold-hopping and lead structure-optimization tool for drug design. In the present work, a set of integrated computational methodologies was first used, such as fragment library generation and retrosynthetic combinatorial analysis procedure (RECAP) for novel compound generation. Then, the compounds generated were assessed by benchmark dataset verification, docking studies, and QSAR model simulation. Subsequently, structurally diverse compounds, with reported or unreported scaffolds, can be observed from top 20 in silico synthesized compounds, which were predicted to be potential mGlu5 modulators. In silico compounds with reported scaffolds may fill SAR holes in known, patented series of mGlu5 modulators. And the generation of compounds without reported tests on mGluR indicates that our approach is doable for exploring and designing novel compounds. Our case study of designing allosteric modulators on mGlu5 demonstrated that the established computational fragment-based approach is a useful methodology for facilitating new compound design in the future.
Collapse
Affiliation(s)
- Yuemin Bian
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA.,NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA.,Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA
| | - Zhiwei Feng
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA.,NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA.,Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA
| | - Peng Yang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA.,NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA.,Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA. .,NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA. .,Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA. .,Department of Computational Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA. .,Department of Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, USA.
| |
Collapse
|
25
|
Emmitte KA. mGlu5negative allosteric modulators: a patent review (2013 - 2016). Expert Opin Ther Pat 2017; 27:691-706. [DOI: 10.1080/13543776.2017.1280466] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Kyle A. Emmitte
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Lin YC, Frei JA, Kilander MBC, Shen W, Blatt GJ. A Subset of Autism-Associated Genes Regulate the Structural Stability of Neurons. Front Cell Neurosci 2016; 10:263. [PMID: 27909399 PMCID: PMC5112273 DOI: 10.3389/fncel.2016.00263] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 10/28/2016] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorder (ASD) comprises a range of neurological conditions that affect individuals’ ability to communicate and interact with others. People with ASD often exhibit marked qualitative difficulties in social interaction, communication, and behavior. Alterations in neurite arborization and dendritic spine morphology, including size, shape, and number, are hallmarks of almost all neurological conditions, including ASD. As experimental evidence emerges in recent years, it becomes clear that although there is broad heterogeneity of identified autism risk genes, many of them converge into similar cellular pathways, including those regulating neurite outgrowth, synapse formation and spine stability, and synaptic plasticity. These mechanisms together regulate the structural stability of neurons and are vulnerable targets in ASD. In this review, we discuss the current understanding of those autism risk genes that affect the structural connectivity of neurons. We sub-categorize them into (1) cytoskeletal regulators, e.g., motors and small RhoGTPase regulators; (2) adhesion molecules, e.g., cadherins, NCAM, and neurexin superfamily; (3) cell surface receptors, e.g., glutamatergic receptors and receptor tyrosine kinases; (4) signaling molecules, e.g., protein kinases and phosphatases; and (5) synaptic proteins, e.g., vesicle and scaffolding proteins. Although the roles of some of these genes in maintaining neuronal structural stability are well studied, how mutations contribute to the autism phenotype is still largely unknown. Investigating whether and how the neuronal structure and function are affected when these genes are mutated will provide insights toward developing effective interventions aimed at improving the lives of people with autism and their families.
Collapse
Affiliation(s)
- Yu-Chih Lin
- Laboratory of Neuronal Connectivity, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| | - Jeannine A Frei
- Laboratory of Neuronal Connectivity, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| | - Michaela B C Kilander
- Laboratory of Neuronal Connectivity, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| | - Wenjuan Shen
- Laboratory of Neuronal Connectivity, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| | - Gene J Blatt
- Laboratory of Autism Neurocircuitry, Program in Neuroscience, Hussman Institute for Autism, Baltimore MD, USA
| |
Collapse
|
29
|
Dean DD, Muthuswamy S, Agarwal S. Fragile X syndrome: Current insight. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2016. [DOI: 10.1016/j.ejmhg.2016.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
30
|
Characterization, treatment patterns, and patient-related outcomes of patients with Fragile X syndrome in Germany: final results of the observational EXPLAIN-FXS study. BMC Psychiatry 2016; 16:318. [PMID: 27612457 PMCID: PMC5018176 DOI: 10.1186/s12888-016-1020-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 08/29/2016] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND As data on the phenotype, characteristics and management of patients with Fragile X Syndrome (FXS) are limited, we aimed to collect such data in Germany in experienced centres involved in the treatment of such patients. METHODS EXPLAIN-FXS is a prospective observational (non-interventional) study (registry) performed between April 2013 and January 2016 at 18 sites in Germany. Requirements for patient participation included confirmed diagnosis of FXS by genetic testing (>200 CGG repeats) and written informed consent. Patients were followed for up to 2 years. RESULTS Seventy-five patients (84.0 % males, mean age 16.7 ± 14.5 years, ranging from 2 - 82 years) were analysed. The mean 6-item score, determined according to Giangreco (J Pediatr 129:611-614, 1996), was 6.9 ± 2.5 points. At least one neurological finding each was noted in 53 patients (69.7 %). Specifically, ataxia was noted in 5 patients (6.6 %), lack of fine motor skills in 40 patients, (52.6 %), muscle tonus disorder in 4 patients (5.3 %), and other neurological disorders in 39 patients (51.3 %). Spasticity was not noted in any patient. Seizures were reported in 6 patients (8.1 %), anxiety disorders in 22 patients (30.1 %), depression in 7 patients (9.6 %), ADHD/ADD in 36 patients (49.3 %), impairment of social behavior in 39 patients (53.4 %), and other comorbidities in 23 patients (31.5 %). The mean Aberrant Behaviour Checklist Community Edition (ABC-C) score on behavioral symptoms, obtained in 71 patients at first documentation, was 48.4 ± 27.8 (median 45.0, range 5-115). The mean visual analogue scale (VAS) score, obtained in 59 patients at first documentation, was 84.9 ± 14.6 points (median 90; range 50 - 100). CONCLUSIONS This report describes the largest cohort of patients with FXS in Europe. The reported observations indicate a substantial burden of disease for patients and their caregivers. Based on these observations, an early expert psychiatric diagnosis is recommended for suspected FXS patients. Further recommendations include multimodal and multi-professional management that is tailored to the individual patient's needs. TRIAL REGISTRATION The ClinTrials.gov identifier is NCT01711606 . Registered on 18 October 2012.
Collapse
|
31
|
Cheng C, Lau SKM, Doering LC. Astrocyte-secreted thrombospondin-1 modulates synapse and spine defects in the fragile X mouse model. Mol Brain 2016; 9:74. [PMID: 27485117 PMCID: PMC4971702 DOI: 10.1186/s13041-016-0256-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 07/15/2016] [Indexed: 01/24/2023] Open
Abstract
Astrocytes are key participants in various aspects of brain development and function, many of which are executed via secreted proteins. Defects in astrocyte signaling are implicated in neurodevelopmental disorders characterized by abnormal neural circuitry such as Fragile X syndrome (FXS). In animal models of FXS, the loss in expression of the Fragile X mental retardation 1 protein (FMRP) from astrocytes is associated with delayed dendrite maturation and improper synapse formation; however, the effect of astrocyte-derived factors on the development of neurons is not known. Thrombospondin-1 (TSP-1) is an important astrocyte-secreted protein that is involved in the regulation of spine development and synaptogenesis. In this study, we found that cultured astrocytes isolated from an Fmr1 knockout (Fmr1 KO) mouse model of FXS displayed a significant decrease in TSP-1 protein expression compared to the wildtype (WT) astrocytes. Correspondingly, Fmr1 KO hippocampal neurons exhibited morphological deficits in dendritic spines and alterations in excitatory synapse formation following long-term culture. All spine and synaptic abnormalities were prevented in the presence of either astrocyte-conditioned media or a feeder layer derived from FMRP-expressing astrocytes, or following the application of exogenous TSP-1. Importantly, this work demonstrates the integral role of astrocyte-secreted signals in the establishment of neuronal communication and identifies soluble TSP-1 as a potential therapeutic target for Fragile X syndrome.
Collapse
Affiliation(s)
- Connie Cheng
- McMaster Integrative Neuroscience Discovery and Study Program (MINDS), McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L8, Canada.,Department of Pathology and Molecular Medicine, McMaster University, 1200 Main Street West, HSC 1R15A, Hamilton, Ontario, L8N 3Z5, Canada
| | - Sally K M Lau
- Department of Pathology and Molecular Medicine, McMaster University, 1200 Main Street West, HSC 1R15A, Hamilton, Ontario, L8N 3Z5, Canada
| | - Laurie C Doering
- McMaster Integrative Neuroscience Discovery and Study Program (MINDS), McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L8, Canada. .,Department of Pathology and Molecular Medicine, McMaster University, 1200 Main Street West, HSC 1R15A, Hamilton, Ontario, L8N 3Z5, Canada.
| |
Collapse
|
32
|
Selective Disruption of Metabotropic Glutamate Receptor 5-Homer Interactions Mimics Phenotypes of Fragile X Syndrome in Mice. J Neurosci 2016; 36:2131-47. [PMID: 26888925 DOI: 10.1523/jneurosci.2921-15.2016] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
UNLABELLED Altered function of the Gq-coupled, Group 1 metabotropic glutamate receptors, specifically mGlu5, is implicated in multiple mouse models of autism and intellectual disability. mGlu5 dysfunction has been most well characterized in the fragile X syndrome mouse model, the Fmr1 knock-out (KO) mouse, where pharmacological and genetic reduction of mGlu5 reverses many phenotypes. mGlu5 is less associated with its scaffolding protein Homer in Fmr1 KO mice, and restoration of mGlu5-Homer interactions by genetic deletion of a short, dominant negative of Homer, H1a, rescues many phenotypes of Fmr1 KO mice. These results suggested that disruption of mGlu5-Homer leads to phenotypes of FXS. To test this idea, we examined mice with a knockin mutation of mGlu5 (F1128R; mGlu5(R/R)) that abrogates binding to Homer. Although FMRP levels were normal, mGlu5(R/R) mice mimicked multiple phenotypes of Fmr1 KO mice, including reduced mGlu5 association with the postsynaptic density, enhanced constitutive mGlu5 signaling to protein synthesis, deficits in agonist-induced translational control, protein synthesis-independent LTD, neocortical hyperexcitability, audiogenic seizures, and altered behaviors, including anxiety and sensorimotor gating. These results reveal new roles for the Homer scaffolds in regulation of mGlu5 function and implicate a specific molecular mechanism in a complex brain disease. SIGNIFICANCE STATEMENT Abnormal function of the metabotropic, or Gq-coupled, glutamate receptor 5 (mGlu5) has been implicated in neurodevelopmental disorders, including a genetic cause of intellectual disability and autism called fragile X syndrome. In brains of a mouse model of fragile X, mGlu5 is less associated with its binding partner Homer, a scaffolding protein that regulates mGlu5 localization to synapses and its ability to activate biochemical signaling pathways. Here we show that a mouse expressing a mutant mGlu5 that cannot bind to Homer is sufficient to mimic many of the biochemical, neurophysiological, and behavioral symptoms observed in the fragile X mouse. This work provides strong evidence that Homer-mGlu5 binding contributes to symptoms associated with neurodevelopmental disorders.
Collapse
|
33
|
LaCrosse AL, Taylor SB, Nemirovsky NE, Gass JT, Olive MF. mGluR5 Positive and Negative Allosteric Modulators Differentially Affect Dendritic Spine Density and Morphology in the Prefrontal Cortex. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2016; 14:476-85. [PMID: 25921744 DOI: 10.2174/1871527314666150429112849] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 01/17/2015] [Accepted: 01/19/2015] [Indexed: 12/31/2022]
Abstract
Positive and negative allosteric modulators (PAMs and NAMs, respectively) of type 5 metabotropic glutamate receptors (mGluR5) are currently being investigated as novel treatments for neuropsychiatric diseases including drug addiction, schizophrenia, and Fragile X syndrome. However, only a handful of studies have examined the effects of mGluR5 PAMs or NAMs on the structural plasticity of dendritic spines in otherwise naïve animals, particularly in brain regions mediating executive function. In the present study, we assessed dendritic spine density and morphology in pyramidal cells of the medial prefrontal cortex (mPFC) after repeated administration of either the prototypical mGluR5 PAM 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5- yl)benzamide (CDPPB, 20 mg/kg), the clinically utilized mGluR5 NAM 1-(3-chlorophenyl)-3-(3-methyl-5-oxo-4Himidazol- 2-yl)urea (fenobam, 20 mg/kg), or vehicle in male Sprague-Dawley rats. Following once daily treatment for 10 consecutive days, coronal brain sections containing the mPFC underwent diolistic labeling and 3D image analysis of dendritic spines. Compared to vehicle treated animals, rats administered fenobam exhibited significant increases in dendritic spine density and the overall frequency of spines with small (<0.2 μm) head diameters, decreases in frequency of spines with medium (0.2-0.4 μm) head diameters, and had no changes in frequency of spines with large head diameters (>0.4 μm). Administration of CDPPB had no discernable effects on dendritic spine density or morphology, and neither CDPPB nor fenobam had any effect on spine length or volume. We conclude that mGluR5 PAMs and NAMs differentially affect mPFC dendritic spine structural plasticity in otherwise naïve animals, and additional studies assessing their effects in combination with cognitive or behavioral tasks are needed.
Collapse
Affiliation(s)
| | | | | | | | - Michael F Olive
- Department of Psychology, Arizona State University, PO Box 871104, Tempe, AZ 85287, USA.
| |
Collapse
|
34
|
Galambos J, Domány G, Nógrádi K, Wágner G, Keserű GM, Bobok A, Kolok S, Mikó-Bakk ML, Vastag M, Sághy K, Kóti J, Szakács Z, Béni Z, Gál K, Szombathelyi Z, Greiner I. 4-Aryl-3-arylsulfonyl-quinolines as negative allosteric modulators of metabotropic GluR5 receptors: From HTS hit to development candidate. Bioorg Med Chem Lett 2016; 26:1249-52. [DOI: 10.1016/j.bmcl.2016.01.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 01/08/2016] [Accepted: 01/09/2016] [Indexed: 11/16/2022]
|
35
|
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] [MESH Headings] [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
|
36
|
Sengmany K, Gregory KJ. Metabotropic glutamate receptor subtype 5: molecular pharmacology, allosteric modulation and stimulus bias. Br J Pharmacol 2015; 173:3001-17. [PMID: 26276909 DOI: 10.1111/bph.13281] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 06/30/2015] [Accepted: 07/26/2015] [Indexed: 12/12/2022] Open
Abstract
The metabotropic glutamate receptor subtype 5 (mGlu5 ) is a family C GPCR that has been implicated in various neuronal processes and, consequently, in several CNS disorders. Over the past few decades, GPCR-based drug discovery, including that for mGlu5 receptors, has turned considerable attention to targeting allosteric binding sites. Modulation of endogenous agonists by allosteric ligands offers the advantages of spatial and temporal fine-tuning of receptor activity, increased selectivity and reduced adverse effects with the potential to elicit improved clinical outcomes. Further, with greater appreciation of the multifaceted nature of the transduction of mGlu5 receptor signalling, it is increasingly apparent that drug discovery must take into consideration unique receptor conformations and the potential for stimulus-bias. This novel paradigm proposes that different ligands may differentially modulate distinct signalling pathways arising from the same receptor. We review our current understanding of the complexities of mGlu5 receptor signalling and regulation, and how these relate to allosteric ligands. Ultimately, a deeper appreciation of these relationships will provide the foundation for targeted drug design of compounds with increased selectivity, not only for the desired receptor but also for the desired signalling outcome from the receptor. Linked Articles This article is part of a themed section on Molecular Pharmacology of G Protein-Coupled Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v173.20/issuetoc.
Collapse
Affiliation(s)
- K Sengmany
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, VIC, Australia
| | - K J Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, VIC, Australia.
| |
Collapse
|
37
|
Inhibition of Group I Metabotropic Glutamate Receptors Reverses Autistic-Like Phenotypes Caused by Deficiency of the Translation Repressor eIF4E Binding Protein 2. J Neurosci 2015; 35:11125-32. [PMID: 26245973 DOI: 10.1523/jneurosci.4615-14.2015] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Exacerbated mRNA translation during brain development has been linked to autism spectrum disorders (ASDs). Deletion of the eukaryotic initiation factor 4E (eIF4E)-binding protein 2 gene (Eif4ebp2), encoding the suppressor of mRNA translation initiation 4E-BP2, leads to an imbalance in excitatory-to-inhibitory neurotransmission and ASD-like behaviors. Inhibition of group I metabotropic glutamate receptors (mGluRs) mGluR1 and mGluR5 reverses the autistic phenotypes in several ASD mouse models. Importantly, these receptors control synaptic physiology via activation of mRNA translation. We investigated the potential reversal of autistic-like phenotypes in Eif4ebp2(-/-) mice by using antagonists of mGluR1 (JNJ16259685) or mGluR5 (fenobam). Augmented hippocampal mGluR-induced long-term depression (LTD; or chemically induced mGluR-LTD) in Eif4ebp2(-/-) mice was rescued by mGluR1 or mGluR5 antagonists. While rescue by mGluR5 inhibition occurs through the blockade of a protein synthesis-dependent component of LTD, normalization by mGluR1 antagonists requires the activation of protein synthesis. Synaptically induced LTD was deficient in Eif4ebp2(-/-) mice, and this deficit was not rescued by group I mGluR antagonists. Furthermore, a single dose of mGluR1 (0.3 mg/kg) or mGluR5 (3 mg/kg) antagonists in vivo reversed the deficits in social interaction and repetitive behaviors (marble burying) in Eif4ebp2(-/-) mice. Our results demonstrate that Eif4ebp2(-/-) mice serve as a relevant model to test potential therapies for ASD symptoms. In addition, we provide substantive evidence that the inhibition of mGluR1/mGluR5 is an effective treatment for physiological and behavioral alterations caused by exacerbated mRNA translation initiation. SIGNIFICANCE STATEMENT Exacerbated mRNA translation during brain development is associated with several autism spectrum disorders (ASDs). We recently demonstrated that the deletion of a negative regulator of mRNA translation initiation, the eukaryotic initiation factor 4E-binding protein 2, leads to ASD-like behaviors and increased excitatory synaptic activity. Here we demonstrated that autistic behavioral and electrophysiological phenotypes can be treated in adult mice with antagonists of group I metabotropic glutamate receptors (mGluRs), which have been previously used in other ASD models (i.e., fragile X syndrome). These findings support the use of group I mGluR antagonists as a potential therapy that extends to autism models involving exacerbated mRNA translation initiation.
Collapse
|
38
|
Koyama Y. Functional alterations of astrocytes in mental disorders: pharmacological significance as a drug target. Front Cell Neurosci 2015. [PMID: 26217185 PMCID: PMC4491615 DOI: 10.3389/fncel.2015.00261] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Astrocytes play an essential role in supporting brain functions in physiological and pathological states. Modulation of their pathophysiological responses have beneficial actions on nerve tissue injured by brain insults and neurodegenerative diseases, therefore astrocytes are recognized as promising targets for neuroprotective drugs. Recent investigations have identified several astrocytic mechanisms for modulating synaptic transmission and neural plasticity. These include altered expression of transporters for neurotransmitters, release of gliotransmitters and neurotrophic factors, and intercellular communication through gap junctions. Investigation of patients with mental disorders shows morphological and functional alterations in astrocytes. According to these observations, manipulation of astrocytic function by gene mutation and pharmacological tools reproduce mental disorder-like behavior in experimental animals. Some drugs clinically used for mental disorders affect astrocyte function. As experimental evidence shows their role in the pathogenesis of mental disorders, astrocytes have gained much attention as drug targets for mental disorders. In this paper, I review functional alterations of astrocytes in several mental disorders including schizophrenia, mood disorder, drug dependence, and neurodevelopmental disorders. The pharmacological significance of astrocytes in mental disorders is also discussed.
Collapse
Affiliation(s)
- Yutaka Koyama
- Laboratory of Pharmacology, Faculty of Pharmacy, Osaka Ohtani University Tondabayashi, Osaka, Japan
| |
Collapse
|
39
|
GPCR crystal structures: Medicinal chemistry in the pocket. Bioorg Med Chem 2015; 23:3880-906. [DOI: 10.1016/j.bmc.2014.12.034] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 12/12/2014] [Accepted: 12/16/2014] [Indexed: 12/20/2022]
|
40
|
Gross C, Hoffmann A, Bassell GJ, Berry-Kravis EM. Therapeutic Strategies in Fragile X Syndrome: From Bench to Bedside and Back. Neurotherapeutics 2015; 12:584-608. [PMID: 25986746 PMCID: PMC4489963 DOI: 10.1007/s13311-015-0355-9] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Fragile X syndrome (FXS), an inherited intellectual disability often associated with autism, is caused by the loss of expression of the fragile X mental retardation protein. Tremendous progress in basic, preclinical, and translational clinical research has elucidated a variety of molecular-, cellular-, and system-level defects in FXS. This has led to the development of several promising therapeutic strategies, some of which have been tested in larger-scale controlled clinical trials. Here, we will summarize recent advances in understanding molecular functions of fragile X mental retardation protein beyond the well-known role as an mRNA-binding protein, and will describe current developments and emerging limitations in the use of the FXS mouse model as a preclinical tool to identify therapeutic targets. We will review the results of recent clinical trials conducted in FXS that were based on some of the preclinical findings, and discuss how the observed outcomes and obstacles will inform future therapy development in FXS and other autism spectrum disorders.
Collapse
Affiliation(s)
- Christina Gross
- />Division of Neurology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229 USA
| | - Anne Hoffmann
- />Department of Pediatrics, Rush University Medical Center, Chicago, IL 60612 USA
| | - Gary J. Bassell
- />Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Elizabeth M. Berry-Kravis
- />Departments of Pediatrics, Neurological Sciences, Biochemistry, Rush University Medical Center, Chicago, IL 60612 USA
| |
Collapse
|
41
|
Schaefer TL, Davenport MH, Erickson CA. Emerging pharmacologic treatment options for fragile X syndrome. APPLICATION OF CLINICAL GENETICS 2015; 8:75-93. [PMID: 25897255 PMCID: PMC4396424 DOI: 10.2147/tacg.s35673] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Fragile X syndrome (FXS) is the most common single gene cause of intellectual disability and autism spectrum disorder. Caused by a silenced fragile X mental retardation 1 gene and the subsequent deficiency in fragile X mental retardation protein, patients with FXS experience a range of physical, behavioral, and intellectual debilitations. The FXS field, as a whole, has recently met with some challenges, as several targeted clinical trials with high expectations of success have failed to elucidate significant improvements in a variety of symptom domains. As new clinical trials in FXS are planned, there has been much discussion about the use of the commonly used clinical outcome measures, as well as study design considerations, patient stratification, and optimal age range for treatment. The evidence that modification of these drug targets and use of these failed compounds would prove to be efficacious in human clinical study were rooted in years of basic and translational research. There are questions arising as to the use of the mouse models for studying FXS treatment development. This issue is twofold: many of the symptom domains and molecular and biochemical changes assessed and indicative of efficacy in mouse model study are not easily amenable to clinical trials in people with FXS because of the intolerability of the testing paradigm or a lack of noninvasive techniques (prepulse inhibition, sensory hypersensitivity, startle reactivity, or electrophysiologic, biochemical, or structural changes in the brain); and capturing subtle yet meaningful changes in symptom domains such as sociability, anxiety, and hyperactivity in human FXS clinical trials is challenging with the currently used measures (typically parent/caregiver rating scales). Clinicians, researchers, and the pharmaceutical industry have all had to take a step back and critically evaluate the way we think about how to best optimize future investigations into pharmacologic FXS treatments. As new clinical trials are coming down the drug discovery pipeline, it is clear that the field is moving in a direction that values the development of molecular biomarkers, less subjective quantitative measures of symptom improvement, and rating scales developed specifically for use in FXS in conjunction with drug safety. While summarizing preclinical evidence, where applicable, and discussing challenges in FXS treatment development, this review details both completed clinical trials for the targeted and symptomatic treatment of FXS and introduces novel projects on the cusp of clinical trial investigation.
Collapse
Affiliation(s)
- Tori L Schaefer
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew H Davenport
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Craig A Erickson
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
42
|
Wang H, Pati S, Pozzo-Miller L, Doering LC. Targeted pharmacological treatment of autism spectrum disorders: fragile X and Rett syndromes. Front Cell Neurosci 2015; 9:55. [PMID: 25767435 PMCID: PMC4341567 DOI: 10.3389/fncel.2015.00055] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 02/05/2015] [Indexed: 12/27/2022] Open
Abstract
Autism spectrum disorders (ASDs) are genetically and clinically heterogeneous and lack effective medications to treat their core symptoms. Studies of syndromic ASDs caused by single gene mutations have provided insights into the pathophysiology of autism. Fragile X and Rett syndromes belong to the syndromic ASDs in which preclinical studies have identified rational targets for drug therapies focused on correcting underlying neural dysfunction. These preclinical discoveries are increasingly translating into exciting human clinical trials. Since there are significant molecular and neurobiological overlaps among ASDs, targeted treatments developed for fragile X and Rett syndromes may be helpful for autism of different etiologies. Here, we review the targeted pharmacological treatment of fragile X and Rett syndromes and discuss related issues in both preclinical studies and clinical trials of potential therapies for the diseases.
Collapse
Affiliation(s)
- Hansen Wang
- Faculty of Medicine, University of Toronto, 1 King's College Circle Toronto, ON, Canada
| | - Sandipan Pati
- Department of Neurology, Epilepsy Division, The University of Alabama at Birmingham Birmingham, AL, USA
| | - Lucas Pozzo-Miller
- Department of Neurobiology, Civitan International Research Center, The University of Alabama at Birmingham Birmingham, AL, USA
| | - Laurie C Doering
- Faculty of Health Sciences, Department of Pathology and Molecular Medicine, McMaster University Hamilton, ON, Canada
| |
Collapse
|
43
|
Deacon RMJ, Glass L, Snape M, Hurley MJ, Altimiras FJ, Biekofsky RR, Cogram P. NNZ-2566, a Novel Analog of (1–3) IGF-1, as a Potential Therapeutic Agent for Fragile X Syndrome. Neuromolecular Med 2015; 17:71-82. [DOI: 10.1007/s12017-015-8341-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 01/06/2015] [Indexed: 10/24/2022]
|
44
|
Mathews WB, Kuwabara H, Stansfield K, Valentine H, Alexander M, Kumar A, Hilton J, Dannals RF, Wong DF, Gasparini F. Dose-dependent, saturable occupancy of the metabotropic glutamate subtype 5 receptor by fenobam as measured with [ 11 C]ABP688 PET imaging. Synapse 2014; 68:565-573. [PMID: 25098663 PMCID: PMC4320023 DOI: 10.1002/syn.21775] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 06/25/2014] [Accepted: 07/06/2014] [Indexed: 12/24/2022]
Abstract
Fenobam is a negative allosteric modulator of the metabotropic glutamate receptor subtype 5 (mGluR5) with inverse agonist activity and is expected to contribute to the treatment of neuropsychiatric disorders involving dysfunction of mGluR5 including Fragile X syndrome. This study examined whether [11 C]ABP688, an antagonist PET radioligand, competes with fenobam for the same binding site in the nonhuman primate brain and would allow examination of occupancy-plasma concentration relationships in the evaluation of the drug for target disorders in the human brain. Four paired PET studies with [11 C]ABP688 were performed in baboons at a baseline condition and after intravenous treatment with fenobam at different dose levels (0.3-1.33 mg/kg). Total distribution volume (VT ) and binding potential (BPND ) using the cerebellum as a reference region were obtained by the plasma reference graphical method. Then it was examined whether occupancy follows a dose-dependent, saturating pattern that was predicted by a modified first-order Hill equation in individual regions. Baseline regional VT and BPND values agreed with previously published data. Occupancy showed dose-dependent and saturating patterns in individual regions, reaching >90% occupancy at 1.33 mg/kg dose of fenobam in the majority of regions. To our knowledge, this is the first use of PET to characterize the mGluR5 therapeutic drug fenobam. This study demonstrates a proof of principle for determining the in vivo occupancy of fenobam in primates. The results indicate that [11 C]ABP688 and PET may be useful for examination of occupancy of mGluR5 by fenobam, which should prove to be useful for designing future studies and treatment of human disease states. Synapse 68:565-573, 2014. © 2014 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- William B Mathews
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland
| | - Hiroto Kuwabara
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland
| | | | - Heather Valentine
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland
| | - Mohab Alexander
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland
| | - Anil Kumar
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland
| | - John Hilton
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland
| | - Robert F Dannals
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland
| | - Dean F Wong
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland
- Department of Psychiatry, Johns Hopkins University, Baltimore, Maryland
- Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland
- Department of Environmental Health Sciences, Johns Hopkins University, Baltimore, Maryland
| | | |
Collapse
|
45
|
Wendling T, Ogungbenro K, Pigeolet E, Dumitras S, Woessner R, Aarons L. Model-based evaluation of the impact of formulation and food intake on the complex oral absorption of mavoglurant in healthy subjects. Pharm Res 2014; 32:1764-78. [PMID: 25425054 DOI: 10.1007/s11095-014-1574-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 11/10/2014] [Indexed: 11/26/2022]
Abstract
PURPOSE To compare the pharmacokinetics of intravenous (IV), oral immediate-release (IR) and oral modified-release (MR) formulations of mavoglurant in healthy subjects, and to assess the food effect on the MR formulation's input characteristics. METHODS Plasma concentration-time data from two clinical studies in healthy volunteers were pooled and analysed using NONMEM®. Drug entry into the systemic circulation was modelled using a sum of inverse Gaussian (IG) functions as an input rate function, which was estimated specifically for each formulation and food state. RESULTS Mavoglurant pharmacokinetics was best described by a two-compartment model with a sum of either two or three IG functions as input function. The mean absolute bioavailability from the MR formulation (0.387) was less than from the IR formulation (0.436). The MR formulation pharmacokinetics were significantly impacted by food: bioavailability was higher (0.508) and the input process was shorter (complete in approximately 36 versus 12 h for the fasted and fed states, respectively). CONCLUSIONS Modelling and simulation of mavoglurant pharmacokinetics indicate that the MR formulation might provide a slightly lower steady-state concentration range with lower peaks (possibly better drug tolerance) than the IR formulation, and that the MR formulation's input properties strongly depend on the food conditions at drug administration.
Collapse
Affiliation(s)
- Thierry Wendling
- Manchester Pharmacy School, The University of Manchester, Manchester, UK
| | | | | | | | | | | |
Collapse
|
46
|
de Esch CE, Zeidler S, Willemsen R. Translational endpoints in fragile X syndrome. Neurosci Biobehav Rev 2014; 46 Pt 2:256-69. [DOI: 10.1016/j.neubiorev.2013.10.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 10/07/2013] [Accepted: 10/09/2013] [Indexed: 01/01/2023]
|
47
|
Structure of class C GPCR metabotropic glutamate receptor 5 transmembrane domain. Nature 2014; 511:557-62. [DOI: 10.1038/nature13396] [Citation(s) in RCA: 337] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 04/22/2014] [Indexed: 12/19/2022]
|
48
|
Sidorov MS, Krueger DD, Taylor M, Gisin E, Osterweil EK, Bear MF. Extinction of an instrumental response: a cognitive behavioral assay in Fmr1 knockout mice. GENES BRAIN AND BEHAVIOR 2014; 13:451-8. [PMID: 24684608 DOI: 10.1111/gbb.12137] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 03/14/2014] [Accepted: 03/28/2014] [Indexed: 01/26/2023]
Abstract
Fragile X (FX) is the most common genetic cause of intellectual disability and autism. Previous studies have shown that partial inhibition of metabotropic glutamate receptor signaling is sufficient to correct behavioral phenotypes in a mouse model of FX, including audiogenic seizures, open-field hyperactivity and social behavior. These phenotypes model well the epilepsy (15%), hyperactivity (20%) and autism (30%) that are comorbid with FX in human patients. Identifying reliable and robust mouse phenotypes to model cognitive impairments is critical considering the 90% comorbidity of FX and intellectual disability. Recent work characterized a five-choice visuospatial discrimination assay testing cognitive flexibility, in which FX model mice show impairments associated with decreases in synaptic proteins in prefrontal cortex (PFC). In this study, we sought to determine whether instrumental extinction, another process requiring PFC, is altered in FX model mice, and whether downregulation of metabotropic glutamate receptor signaling pathways is sufficient to correct both visuospatial discrimination and extinction phenotypes. We report that instrumental extinction is consistently exaggerated in FX model mice. However, neither the extinction phenotype nor the visuospatial discrimination phenotype is corrected by approaches targeting metabotropic glutamate receptor signaling. This work describes a novel behavioral extinction assay to model impaired cognition in mouse models of neurodevelopmental disorders, provides evidence that extinction is exaggerated in the FX mouse model and suggests possible limitations of metabotropic glutamate receptor-based pharmacotherapy.
Collapse
Affiliation(s)
- M S Sidorov
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - D D Krueger
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA.,Present address: Department of Molecular Neurobiology, Max Planck Institute for Experimental Medicine, Goettingen, Germany
| | - M Taylor
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - E Gisin
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - E K Osterweil
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA.,Present address: Centre for Integrative Physiology, University of Edinburgh, Edinburgh, UK
| | - M F Bear
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
49
|
|
50
|
Milanese M, Giribaldi F, Melone M, Bonifacino T, Musante I, Carminati E, Rossi PI, Vergani L, Voci A, Conti F, Puliti A, Bonanno G. Knocking down metabotropic glutamate receptor 1 improves survival and disease progression in the SOD1G93A mouse model of amyotrophic lateral sclerosis. Neurobiol Dis 2014; 64:48-59. [DOI: 10.1016/j.nbd.2013.11.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 10/17/2013] [Accepted: 11/12/2013] [Indexed: 11/26/2022] Open
|