1
|
van der Lei MB, Kooy RF. From Discovery to Innovative Translational Approaches in 80 Years of Fragile X Syndrome Research. Biomedicines 2025; 13:805. [PMID: 40299377 PMCID: PMC12024745 DOI: 10.3390/biomedicines13040805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Fragile X syndrome (FXS) is the most common inherited cause of intellectual disability and a major genetic contributor to autism spectrum disorder. It is caused by a CGG trinucleotide repeat expansion in the FMR1 gene, resulting in gene silencing and the loss of FMRP, an RNA-binding protein essential for synaptic plasticity. This review covers over 80 years of FXS research, highlighting key milestones, clinical features, genetic and molecular mechanisms, the FXS mouse model, disrupted molecular pathways, and current therapeutic strategies. Additionally, we discuss recent advances including AI-driven combination therapies, CRISPR-based gene editing, and antisense oligonucleotides (ASOs) therapies. Despite these scientific breakthroughs, translating preclinical findings into effective clinical treatments remains challenging. Clinical trials have faced several difficulties, including patient heterogeneity, inconsistent outcome measures, and variable therapeutic responses. Standardized preclinical testing protocols and refined clinical trial designs are required to overcome these challenges. The development of FXS-specific biomarkers could also improve the precision of treatment assessments. Ultimately, future therapies will need to combine pharmacological and behavioral interventions tailored to individual needs. While significant challenges remain, ongoing research continues to offer hope for transformative breakthroughs that could significantly improve the quality of life for individuals with FXS and their families.
Collapse
Affiliation(s)
| | - R. Frank Kooy
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, 2650 Edegem, Belgium;
| |
Collapse
|
2
|
Hakon J, Quattromani MJ, Sjölund C, Talhada D, Kim B, Moyanova S, Mastroiacovo F, Di Menna L, Olsson R, Englund E, Nicoletti F, Ruscher K, Bauer AQ, Wieloch T. Inhibiting metabotropic glutamate receptor 5 after stroke restores brain function and connectivity. Brain 2024; 147:186-200. [PMID: 37656990 PMCID: PMC10766240 DOI: 10.1093/brain/awad293] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 06/12/2023] [Accepted: 08/04/2023] [Indexed: 09/03/2023] Open
Abstract
Stroke results in local neural disconnection and brain-wide neuronal network dysfunction leading to neurological deficits. Beyond the hyper-acute phase of ischaemic stroke, there is no clinically-approved pharmacological treatment that alleviates sensorimotor impairments. Functional recovery after stroke involves the formation of new or alternative neuronal circuits including existing neural connections. The type-5 metabotropic glutamate receptor (mGluR5) has been shown to modulate brain plasticity and function and is a therapeutic target in neurological diseases outside of stroke. We investigated whether mGluR5 influences functional recovery and network reorganization rodent models of focal ischaemia. Using multiple behavioural tests, we observed that treatment with negative allosteric modulators (NAMs) of mGluR5 (MTEP, fenobam and AFQ056) for 12 days, starting 2 or 10 days after stroke, restored lost sensorimotor functions, without diminishing infarct size. Recovery was evident within hours after initiation of treatment and progressed over the subsequent 12 days. Recovery was prevented by activation of mGluR5 with the positive allosteric modulator VU0360172 and accelerated in mGluR5 knock-out mice compared with wild-type mice. After stroke, multisensory stimulation by enriched environments enhanced recovery, a result prevented by VU0360172, implying a role of mGluR5 in enriched environment-mediated recovery. Additionally, MTEP treatment in conjunction with enriched environment housing provided an additive recovery enhancement compared to either MTEP or enriched environment alone. Using optical intrinsic signal imaging, we observed brain-wide disruptions in resting-state functional connectivity after stroke that were prevented by mGluR5 inhibition in distinct areas of contralesional sensorimotor and bilateral visual cortices. The levels of mGluR5 protein in mice and in tissue samples of stroke patients were unchanged after stroke. We conclude that neuronal circuitry subserving sensorimotor function after stroke is depressed by a mGluR5-dependent maladaptive plasticity mechanism that can be restored by mGluR5 inhibition. Post-acute stroke treatment with mGluR5 NAMs combined with rehabilitative training may represent a novel post-acute stroke therapy.
Collapse
Affiliation(s)
- Jakob Hakon
- Division of Neurosurgery, Department of Clinical Sciences, Laboratory for Experimental Brain Research, Lund University, Lund 221 84, Sweden
| | - Miriana J Quattromani
- Division of Neurosurgery, Department of Clinical Sciences, Laboratory for Experimental Brain Research, Lund University, Lund 221 84, Sweden
| | - Carin Sjölund
- Division of Neurosurgery, Department of Clinical Sciences, Laboratory for Experimental Brain Research, Lund University, Lund 221 84, Sweden
| | - Daniela Talhada
- Division of Neurosurgery, Department of Clinical Sciences, Laboratory for Experimental Brain Research, Lund University, Lund 221 84, Sweden
| | - Byungchan Kim
- Department of Radiology, Washington University, Saint Louis, MO 63110, USA
| | - Slavianka Moyanova
- Department of Molecular Pathology, IRCCS Neuromed, 86077 Pozzilli, Italy
| | | | - Luisa Di Menna
- Department of Molecular Pathology, IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Roger Olsson
- Department of Experimental Medical Sciences, Chemical Biology & Therapeutics, Lund University, Lund 221 84, Sweden
| | - Elisabet Englund
- Division of Pathology, Department of Clinical Sciences, Lund University, Lund 221 84, Sweden
| | - Ferdinando Nicoletti
- Department of Molecular Pathology, IRCCS Neuromed, 86077 Pozzilli, Italy
- Department of Physiology and Pharmacology, University of Rome La Sapienza, 00185 Rome, Italy
| | - Karsten Ruscher
- Division of Neurosurgery, Department of Clinical Sciences, Laboratory for Experimental Brain Research, Lund University, Lund 221 84, Sweden
| | - Adam Q Bauer
- Department of Radiology, Washington University, Saint Louis, MO 63110, USA
| | - Tadeusz Wieloch
- Division of Neurosurgery, Department of Clinical Sciences, Laboratory for Experimental Brain Research, Lund University, Lund 221 84, Sweden
| |
Collapse
|
3
|
Kuchling J, Jurek B, Kents M, Kreye J, Geis C, Wickel J, Mueller S, Koch SP, Boehm-Sturm P, Prüss H, Finke C. Impaired functional connectivity of the hippocampus in translational murine models of NMDA-receptor antibody associated neuropsychiatric pathology. Mol Psychiatry 2024; 29:85-96. [PMID: 37875549 PMCID: PMC11078734 DOI: 10.1038/s41380-023-02303-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 09/28/2023] [Accepted: 10/11/2023] [Indexed: 10/26/2023]
Abstract
Decreased hippocampal connectivity and disruption of functional networks are established resting-state functional MRI (rs-fMRI) features that are associated with neuropsychiatric symptom severity in human anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis. However, the underlying pathophysiology of NMDAR encephalitis remains poorly understood. Application of patient-derived monoclonal antibodies against the NR1 (GluN1) subunit of the NMDAR now allows for the translational investigation of functional connectivity in experimental murine NMDAR antibody disease models with neurodevelopmental disorders. Using rs-fMRI, we studied functional connectivity alterations in (1) adult C57BL/6 J mice that were intrathecally injected with a recombinant human NR1 antibody over 14 days (n = 10) and in (2) a newly established mouse model with in utero exposure to a human recombinant NR1 antibody (NR1-offspring) at the age of (2a) 8 weeks (n = 15) and (2b) 10 months (n = 14). Adult NR1-antibody injected mice showed impaired functional connectivity within the left hippocampus compared to controls, resembling impaired connectivity patterns observed in human NMDAR encephalitis patients. Similarly, NR1-offspring showed significantly reduced functional connectivity in the hippocampus after 8 weeks, and impaired connectivity in the hippocampus was likewise observed in NR1-offspring at the age of 10 months. We successfully reproduced functional connectivity changes within the hippocampus in different experimental murine systems that were previously observed in human NMDAR encephalitis patients. Translational application of this method within a combined imaging and histopathological framework will allow future experimental studies to identify the underlying biological mechanisms and may eventually facilitate non-invasive monitoring of disease activity and treatment responses in autoimmune encephalitis.
Collapse
Grants
- J.Ku is participant in the BIH-Charité Junior Clinician Scientist Program
- J.Kr is participant in the BIH-Charité Clinician Scientist Program funded by the Charité – Universitätsmedizin Berlin and the Berlin Institute of Health.
- C.G. is funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation: grant numbers GE2519/8-1, GE2519/9-1, FOR3004 and GE2519/11-1), by the German Ministry of Education and Research (BMBF: grant numbers 01EW1901, 01GM1908B), and receives funding from Hermann und Lilly Schilling Foundation.
- H.P. is funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation: grant numbers PR 1274/2-1, PR 1274/3-1, FOR3004 and PR 1274/5-1), by the German Ministry of Education and Research (BMBF: grant numbers 01GM1908D, CONNECT-GENERATE), and by the Helmholtz Association (HIL-A03).
- C.F. is funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation: grant numbers FI 2309/1-1 and FI 2309/2-1), and by the German Ministry of Education and Research (BMBF; grant numbers 01GM1908D, CONNECT-GENERATE)
Collapse
Affiliation(s)
- Joseph Kuchling
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Neurocure Cluster of Excellence, NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Betty Jurek
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
| | - Mariya Kents
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
| | - Jakob Kreye
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Christian Geis
- Section of Translational Neuroimmunology, Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Jonathan Wickel
- Section of Translational Neuroimmunology, Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Susanne Mueller
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Neurocure Cluster of Excellence, Core Facility 7 T Experimental MRIs, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Center for Stroke Research, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Paul Koch
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Neurocure Cluster of Excellence, Core Facility 7 T Experimental MRIs, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Center for Stroke Research, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Philipp Boehm-Sturm
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Neurocure Cluster of Excellence, Core Facility 7 T Experimental MRIs, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Center for Stroke Research, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Harald Prüss
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany.
| | - Carsten Finke
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Humboldt-Universität zu Berlin, Berlin School of Mind and Brain, Berlin, Germany.
| |
Collapse
|
4
|
Abstract
The fragile X-related disorders are an important group of hereditary disorders that are caused by expanded CGG repeats in the 5' untranslated region of the FMR1 gene or by mutations in the coding sequence of this gene. Two categories of pathological CGG repeats are associated with these disorders, full mutation alleles and shorter premutation alleles. Individuals with full mutation alleles develop fragile X syndrome, which causes autism and intellectual disability, whereas those with premutation alleles, which have shorter CGG expansions, can develop fragile X-associated tremor/ataxia syndrome, a progressive neurodegenerative disease. Thus, fragile X-related disorders can manifest as neurodegenerative or neurodevelopmental disorders, depending on the size of the repeat expansion. Here, we review mouse models of fragile X-related disorders and discuss how they have informed our understanding of neurodegenerative and neurodevelopmental disorders. We also assess the translational value of these models for developing rational targeted therapies for intellectual disability and autism disorders.
Collapse
Affiliation(s)
- Rob Willemsen
- Department of Clinical Genetics, Erasmus University Medical Center, 3015 CN Rotterdam, the Netherlands. Department of Medical Genetics, University of Antwerp, 2000 Antwerp, Belgium
| | - R. Frank Kooy
- Department of Clinical Genetics, Erasmus University Medical Center, 3015 CN Rotterdam, the Netherlands. Department of Medical Genetics, University of Antwerp, 2000 Antwerp, Belgium
| |
Collapse
|
5
|
Carmichael O. The Role of fMRI in Drug Development: An Update. ADVANCES IN NEUROBIOLOGY 2023; 30:299-333. [PMID: 36928856 DOI: 10.1007/978-3-031-21054-9_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Functional magnetic resonance imaging (fMRI) of the brain is a technology that holds great potential for increasing the efficiency of drug development for the central nervous system (CNS). In preclinical studies and both early- and late-phase human trials, fMRI has the potential to improve cross-species translation of drug effects, help to de-risk compounds early in development, and contribute to the portfolio of evidence for a compound's efficacy and mechanism of action. However, to date, the utilization of fMRI in the CNS drug development process has been limited. The purpose of this chapter is to explore this mismatch between potential and utilization. This chapter provides introductory material related to fMRI and drug development, describes what is required of fMRI measurements for them to be useful in a drug development setting, lists current capabilities of fMRI in this setting and challenges faced in its utilization, and ends with directions for future development of capabilities in this arena. This chapter is the 5-year update of material from a previously published workshop summary (Carmichael et al., Drug DiscovToday 23(2):333-348, 2018).
Collapse
Affiliation(s)
- Owen Carmichael
- Pennington Biomedical Research Center, Baton Rouge, LA, USA.
| |
Collapse
|
6
|
Yang D, Zhao Y, Nie B, An L, Wan X, Wang Y, Wang W, Cai G, Wu S. Progress in magnetic resonance imaging of autism model mice brain. WILEY INTERDISCIPLINARY REVIEWS. COGNITIVE SCIENCE 2022; 13:e1616. [PMID: 35930672 DOI: 10.1002/wcs.1616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/11/2022] [Accepted: 06/29/2022] [Indexed: 06/15/2023]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disease characterized by social disorder and stereotypical behaviors with an increasing incidence. ASD patients are suffering from varying degrees of mental retardation and language development abnormalities. Magnetic resonance imaging (MRI) is a noninvasive imaging technology to detect brain structural and functional dysfunction in vivo, playing an important role in the early diagnosisbasic research of ASD. High-field, small-animal MRI in basic research of autism model mice has provided a new approach to research the pathogenesis, characteristics, and intervention efficacy in autism. This article reviews MRI studies of mouse models of autism over the past 20 years. Reduced gray matter, abnormal connections of brain networks, and abnormal development of white matter fibers have been demonstrated in these studies, which are present in different proportions in the various mouse models. This provides a more macroscopic view for subsequent research on autism model mice. This article is categorized under: Cognitive Biology > Genes and Environment Neuroscience > Computation Neuroscience > Genes, Molecules, and Cells Neuroscience > Development.
Collapse
Affiliation(s)
- Dingding Yang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Yan Zhao
- Department of Gastroenterology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Binbin Nie
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, China
| | - Leiting An
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Xiangdong Wan
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Yazhou Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Wenting Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Guohong Cai
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Shengxi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
7
|
An optimized protocol for assessing changes in mouse whole-brain activity using opto-fMRI. STAR Protoc 2022; 3:101761. [PMID: 36240060 PMCID: PMC9568887 DOI: 10.1016/j.xpro.2022.101761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/10/2022] [Accepted: 09/15/2022] [Indexed: 11/07/2022] Open
Abstract
Functional magnetic resonance imaging (fMRI) in mouse brain, paired with spatially and temporally defined manipulations, offers a powerful tool to causally explain the effect of specific neuronal activity on brain network dynamics. Here, we present an optimized protocol to measure cell-type-specific contributions to changes in whole-brain dynamics in mice using optogenetics (opto)-fMRI. This protocol details the injection of ChR2-expressing AAV, the implantation of optical fiber, the steps to perform opto-BOLD (blood-oxygenation-level-dependent) fMRI recording, and data analysis. For complete details on the use and execution of this protocol, please refer to Grimm et al. (2021).
Collapse
|
8
|
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
|
9
|
Afshar S, Lule S, Yuan G, Qu X, Pan C, Whalen M, Brownell AL, Mody M. Longitudinal PET studies of mGluR5 in FXS using an FMR1 knockout mouse model. Transl Neurosci 2022; 13:80-92. [PMID: 35582646 PMCID: PMC9055256 DOI: 10.1515/tnsci-2022-0217] [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] [Received: 02/04/2022] [Revised: 03/28/2022] [Accepted: 04/04/2022] [Indexed: 11/15/2022] Open
Abstract
Abstract
Fragile X syndrome (FXS) is a monogenic disorder characterized by intellectual disability and behavioral challenges. It is caused by aberrant methylation of the fragile X mental retardation 1 (FMR1) gene. Given the failure of clinical trials in FXS and growing evidence of a role of metabotropic glutamate subtype 5 receptors (mGluR5) in the pathophysiology of the disorder, we investigated mGluR5 function in FMR1 Knockout (FMR1-KO) mice and age- and sex-matched control mice using longitudinal positron emission tomography (PET) imaging to better understand the disorder. The studies were repeated at four time points to examine age- and disease-induced changes in mGluR5 availability using 3-fluoro-[18F]5-(2-pyridinylethynyl)benzonitrile ([18F]FPEB). We found that the binding potential (BP) of [18F]FPEB was significantly lower in the KO mice in mGluR5-implicated brain areas including striatum, cortex, hippocampus, thalamus, and olfactory bulb. The BP also changed with age, regardless of disorder status, increasing in early adulthood in male but not in female mice before decreasing later in both sexes. The difference in mGluR5 availability between the FMR1-KO and control mice and the change in BP in the KO mice as a function of age and sex illustrate the nature of the disorder and its progression, providing mechanistic insights for treatment design.
Collapse
Affiliation(s)
- Sepideh Afshar
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , United States of America
| | - Sevda Lule
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , United States of America
| | - Gengyang Yuan
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , United States of America
| | - Xiying Qu
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , United States of America
| | - Chuzhi Pan
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , United States of America
| | - Michael Whalen
- Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , United States of America
| | - Anna-Liisa Brownell
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , United States of America
| | - Maria Mody
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School , Charlestown , 02129 MA , USA
| |
Collapse
|
10
|
Brašić JR, Goodman JA, Nandi A, Russell DS, Jennings D, Barret O, Martin SD, Slifer K, Sedlak T, Mathur AK, Seibyl JP, Berry-Kravis EM, Wong DF, Budimirovic DB. Fragile X Mental Retardation Protein and Cerebral Expression of Metabotropic Glutamate Receptor Subtype 5 in Men with Fragile X Syndrome: A Pilot Study. Brain Sci 2022; 12:314. [PMID: 35326270 PMCID: PMC8946825 DOI: 10.3390/brainsci12030314] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/26/2022] [Accepted: 02/09/2022] [Indexed: 02/04/2023] Open
Abstract
Multiple lines of evidence suggest that a deficiency of Fragile X Mental Retardation Protein (FMRP) mediates dysfunction of the metabotropic glutamate receptor subtype 5 (mGluR5) in the pathogenesis of fragile X syndrome (FXS), the most commonly known single-gene cause of inherited intellectual disability (ID) and autism spectrum disorder (ASD). Nevertheless, animal and human studies regarding the link between FMRP and mGluR5 expression provide inconsistent or conflicting findings about the nature of those relationships. Since multiple clinical trials of glutamatergic agents in humans with FXS did not demonstrate the amelioration of the behavioral phenotype observed in animal models of FXS, we sought measure if mGluR5 expression is increased in men with FXS to form the basis for improved clinical trials. Unexpectedly marked reductions in mGluR5 expression were observed in cortical and subcortical regions in men with FXS. Reduced mGluR5 expression throughout the living brains of men with FXS provides a clue to examine FMRP and mGluR5 expression in FXS. In order to develop the findings of our previous study and to strengthen the objective tools for future clinical trials of glutamatergic agents in FXS, we sought to assess the possible value of measuring both FMRP levels and mGluR5 expression in men with FXS. We aimed to show the value of measurement of FMRP levels and mGluR5 expression for the diagnosis and treatment of individuals with FXS and related conditions. We administered 3-[18F]fluoro-5-(2-pyridinylethynyl)benzonitrile ([18F]FPEB), a specific mGluR5 radioligand for quantitative measurements of the density and the distribution of mGluR5s, to six men with the full mutation (FM) of FXS and to one man with allele size mosaicism for FXS (FXS-M). Utilizing the seven cortical and subcortical regions affected in neurodegenerative disorders as indicator variables, adjusted linear regression of mGluR5 expression and FMRP showed that mGluR5 expression was significantly reduced in the occipital cortex and the thalamus relative to baseline (anterior cingulate cortex) if FMRP levels are held constant (F(7,47) = 6.84, p < 0.001).These findings indicate the usefulness of cerebral mGluR5 expression measured by PET with [18F]FPEB and FMRP values in men with FXS and related conditions for assessments in community facilities within a hundred-mile radius of a production center with a cyclotron. These initial results of this pilot study advance our previous study regarding the measurement of mGluR5 expression by combining both FMRP levels and mGluR5 expression as tools for meaningful clinical trials of glutamatergic agents for men with FXS. We confirm the feasibility of this protocol as a valuable tool to measure FMRP levels and mGluR5 expression in clinical trials of individuals with FXS and related conditions and to provide the foundations to apply precision medicine to tailor treatment plans to the specific needs of individuals with FXS and related conditions.
Collapse
Affiliation(s)
- James Robert Brašić
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (S.D.M.); (T.S.); (A.K.M.); (D.F.W.)
| | - Jack Alexander Goodman
- Frank H. Netter MD School of Medicine, Quinnipiac University, North Haven, CT 06473, USA;
| | - Ayon Nandi
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (S.D.M.); (T.S.); (A.K.M.); (D.F.W.)
| | - David S. Russell
- Institute for Neurodegenerative Disorders, New Haven, CT 06510, USA; (D.S.R.); (D.J.); (O.B.); (J.P.S.)
- Invicro, New Haven, CT 06510, USA
| | - Danna Jennings
- Institute for Neurodegenerative Disorders, New Haven, CT 06510, USA; (D.S.R.); (D.J.); (O.B.); (J.P.S.)
- Invicro, New Haven, CT 06510, USA
- Denali Therapeutics, Inc., South San Francisco, CA 94080, USA
| | - Olivier Barret
- Institute for Neurodegenerative Disorders, New Haven, CT 06510, USA; (D.S.R.); (D.J.); (O.B.); (J.P.S.)
- Invicro, New Haven, CT 06510, USA
- Laboratoire des Maladies Neurodégénératives, Molecular Imaging Research Center (MIRCen), Institut de Biologie François Jacob, Centre National de la Recherche Scientifique (CNRS), Commissariat à l’Énergie Atomique et aux Énergies Alternatives (CEA), Université Paris-Saclay, CEDEX, 92265 Fontenay-aux-Roses, France
| | - Samuel D. Martin
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (S.D.M.); (T.S.); (A.K.M.); (D.F.W.)
- Department of Neuroscience, Zanvyl Krieger School of Arts and Sciences, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Keith Slifer
- Department of Psychiatry and Behavioral Sciences-Child Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
- Department of Behavioral Psychology, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Thomas Sedlak
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (S.D.M.); (T.S.); (A.K.M.); (D.F.W.)
- Department of Psychiatry and Behavioral Sciences-General Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Anil Kumar Mathur
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (S.D.M.); (T.S.); (A.K.M.); (D.F.W.)
| | - John P. Seibyl
- Institute for Neurodegenerative Disorders, New Haven, CT 06510, USA; (D.S.R.); (D.J.); (O.B.); (J.P.S.)
- Invicro, New Haven, CT 06510, USA
| | - Elizabeth M. Berry-Kravis
- Departments of Pediatrics, Neurological Sciences, and Biochemistry, Rush University Medical Center, Chicago, IL 60612, USA;
| | - Dean F. Wong
- Section of High Resolution Brain Positron Emission Tomography Imaging, Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.N.); (S.D.M.); (T.S.); (A.K.M.); (D.F.W.)
- Laboratory of Central Nervous System (CNS) Neuropsychopharmacology and Multimodal, Imaging (CNAMI), Mallinckrodt Institute of Radiology, Washington University, Saint Louis, MO 63110, USA
| | - Dejan B. Budimirovic
- Department of Psychiatry and Behavioral Sciences-Child Psychiatry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
- Department of Psychiatry, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| |
Collapse
|
11
|
Mirabella F, Desiato G, Mancinelli S, Fossati G, Rasile M, Morini R, Markicevic M, Grimm C, Amegandjin C, Termanini A, Peano C, Kunderfranco P, di Cristo G, Zerbi V, Menna E, Lodato S, Matteoli M, Pozzi D. Prenatal interleukin 6 elevation increases glutamatergic synapse density and disrupts hippocampal connectivity in offspring. Immunity 2021; 54:2611-2631.e8. [PMID: 34758338 PMCID: PMC8585508 DOI: 10.1016/j.immuni.2021.10.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/24/2021] [Accepted: 10/07/2021] [Indexed: 02/07/2023]
Abstract
Early prenatal inflammatory conditions are thought to be a risk factor for different neurodevelopmental disorders. Maternal interleukin-6 (IL-6) elevation during pregnancy causes abnormal behavior in offspring, but whether these defects result from altered synaptic developmental trajectories remains unclear. Here we showed that transient IL-6 elevation via injection into pregnant mice or developing embryos enhanced glutamatergic synapses and led to overall brain hyperconnectivity in offspring into adulthood. IL-6 activated synaptogenesis gene programs in glutamatergic neurons and required the transcription factor STAT3 and expression of the RGS4 gene. The STAT3-RGS4 pathway was also activated in neonatal brains during poly(I:C)-induced maternal immune activation, which mimics viral infection during pregnancy. These findings indicate that IL-6 elevation at early developmental stages is sufficient to exert a long-lasting effect on glutamatergic synaptogenesis and brain connectivity, providing a mechanistic framework for the association between prenatal inflammatory events and brain neurodevelopmental disorders.
Collapse
Affiliation(s)
- Filippo Mirabella
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Genni Desiato
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy
| | - Sara Mancinelli
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Giuliana Fossati
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Marco Rasile
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
| | - Raffaella Morini
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Marija Markicevic
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland
| | - Christina Grimm
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland
| | - Clara Amegandjin
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada; CHU Sainte-Justine Research Center, Montréal, QC, Canada
| | - Alberto Termanini
- Bioinformatic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Clelia Peano
- Institute of Genetic and Biomedical Research, UoS Milan, National Research Council, 20089 Rozzano, Milan, Italy; Genomic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Paolo Kunderfranco
- Bioinformatic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Graziella di Cristo
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada; CHU Sainte-Justine Research Center, Montréal, QC, Canada
| | - Valerio Zerbi
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland; Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zürich, Zürich 8057, Switzerland
| | - Elisabetta Menna
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy
| | - Simona Lodato
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Michela Matteoli
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy.
| | - Davide Pozzi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy.
| |
Collapse
|
12
|
Adil KJ, Gonzales EL, Remonde CG, Boo KJ, Jeon SJ, Shin CY. Autism-Like Behavioral Phenotypes in Mice Treated with Systemic N-Methyl-D-Aspartate. Biomol Ther (Seoul) 2021; 30:232-237. [PMID: 34702791 PMCID: PMC9047488 DOI: 10.4062/biomolther.2021.133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/15/2021] [Accepted: 09/17/2021] [Indexed: 11/05/2022] Open
Abstract
Autism spectrum disorder (ASD) having core characteristics of social interaction problems and repetitive behaviors and interests affects individuals at varying degrees and comorbidities, making it difficult to determine the precise etiology underlying the symptoms. Given its heterogeneity, ASD is difficult to treat and the development of therapeutics is slow due to the scarcity of animal models that are easy to produce and screen with. Based on the theory of excitation/inhibition imbalance in the brain with ASD which involves glutamatergic and/or GABAergic neurotransmission, a pharmacologic agent to modulate these receptors might be a good starting point for modeling. N-methyl-D-aspartic acid (NMDA) is an amino acid derivative acting as a specific agonist at the NMDA receptor and therefore imitates the action of the neurotransmitter glutamate on that receptor. In contrast to glutamate, NMDA selectively binds to and regulates the NMDA receptor, but not other glutamate receptors such as AMPA and kainite receptors. Given this role, we aimed to determine whether NMDA administration could result in autistic-like behavior in adolescent mice. Both male and female mice were treated with saline or NMDA (50 and 75 mg/kg) and were tested on various behavior experiments. Interestingly, acute NMDA-treated mice showed social deficits and repetitive behavior similar to ASD phenotypes. These results support the excitation/inhibition imbalance theory of ASD and that NMDA injection can be used as a pharmacologic model of ASD-like behaviors.
Collapse
Affiliation(s)
- Keremkleroo Jym Adil
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea
| | - Edson Luck Gonzales
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea
| | - Chilly Gay Remonde
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea
| | - Kyung-Jun Boo
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea
| | - Se Jin Jeon
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea
| | - Chan Young Shin
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
13
|
Markicevic M, Savvateev I, Grimm C, Zerbi V. Emerging imaging methods to study whole-brain function in rodent models. Transl Psychiatry 2021; 11:457. [PMID: 34482367 PMCID: PMC8418612 DOI: 10.1038/s41398-021-01575-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 08/05/2021] [Accepted: 08/23/2021] [Indexed: 02/07/2023] Open
Abstract
In the past decade, the idea that single populations of neurons support cognition and behavior has gradually given way to the realization that connectivity matters and that complex behavior results from interactions between remote yet anatomically connected areas that form specialized networks. In parallel, innovation in brain imaging techniques has led to the availability of a broad set of imaging tools to characterize the functional organization of complex networks. However, each of these tools poses significant technical challenges and faces limitations, which require careful consideration of their underlying anatomical, physiological, and physical specificity. In this review, we focus on emerging methods for measuring spontaneous or evoked activity in the brain. We discuss methods that can measure large-scale brain activity (directly or indirectly) with a relatively high temporal resolution, from milliseconds to seconds. We further focus on methods designed for studying the mammalian brain in preclinical models, specifically in mice and rats. This field has seen a great deal of innovation in recent years, facilitated by concomitant innovation in gene-editing techniques and the possibility of more invasive recordings. This review aims to give an overview of currently available preclinical imaging methods and an outlook on future developments. This information is suitable for educational purposes and for assisting scientists in choosing the appropriate method for their own research question.
Collapse
Affiliation(s)
- Marija Markicevic
- Neural Control of Movement Lab, HEST, ETH Zürich, Zürich, Switzerland
- Neuroscience Center Zurich, University and ETH Zürich, Zürich, Switzerland
| | - Iurii Savvateev
- Neural Control of Movement Lab, HEST, ETH Zürich, Zürich, Switzerland
- Neuroscience Center Zurich, University and ETH Zürich, Zürich, Switzerland
- Decision Neuroscience Lab, HEST, ETH Zürich, Zürich, Switzerland
| | - Christina Grimm
- Neural Control of Movement Lab, HEST, ETH Zürich, Zürich, Switzerland
- Neuroscience Center Zurich, University and ETH Zürich, Zürich, Switzerland
| | - Valerio Zerbi
- Neural Control of Movement Lab, HEST, ETH Zürich, Zürich, Switzerland.
- Neuroscience Center Zurich, University and ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
14
|
Siegel-Ramsay JE, Romaniuk L, Whalley HC, Roberts N, Branigan H, Stanfield AC, Lawrie SM, Dauvermann MR. Glutamate and functional connectivity - support for the excitatory-inhibitory imbalance hypothesis in autism spectrum disorders. Psychiatry Res Neuroimaging 2021; 313:111302. [PMID: 34030047 DOI: 10.1016/j.pscychresns.2021.111302] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/30/2021] [Accepted: 05/07/2021] [Indexed: 12/24/2022]
Abstract
It has been proposed that the Glutamate (Glu) system is implicated in autism spectrum disorders (ASD) via an imbalance between excitatory and inhibitory brain circuits, which impacts on brain function. Here, we investigated the excitatory-inhibitory imbalance theory by measuring Glu-concentrations and the relationship with resting-state function. Nineteen adult males with ASD and 19 age and sex-matched healthy controls (HC) (23 - 58 years) underwent Proton Magnetic Resonance Spectroscopy of the dorsal anterior cingulate cortex (dACC) and resting-state functional Magnetic Resonance Imaging (fMRI). Glu and Glx concentrations were compared between groups. Seed-based functional connectivity was analyzed with a priori seeds of the right and left dACC. Finally, metabolite concentrations were related to functional connectivity coefficients and compared between both groups. Individuals with ASD showed significantly negative associations between increased Glx concentrations and reduced functional connectivity between the dACC and insular, limbic and parietal regions. In contrast, HC displayed a positive relationship between the same metabolite and connectivity measures. We provided new evidence to support the excitatory-inhibitory imbalance theory, where excitatory Glx concentrations were related to functional dysconnectivity in ASD. Future research is needed to investigate large-scale functional networks in association with both excitatory and inhibitory metabolites in subpopulations of ASD.
Collapse
Affiliation(s)
- Jennifer E Siegel-Ramsay
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom; Department of Psychiatry and Behavioral Science, University of Texas, Austin, United States
| | - Liana Romaniuk
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - Heather C Whalley
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - Neil Roberts
- Centre for Reproductive Health (CRH), School of Clinical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Holly Branigan
- School of Psychology, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew C Stanfield
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom; Patrick Wild Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Stephen M Lawrie
- Division of Psychiatry, University of Edinburgh, Edinburgh, United Kingdom
| | - Maria R Dauvermann
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom; McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, United States.
| |
Collapse
|
15
|
Eed A, Cerdán Cerdá A, Lerma J, De Santis S. Diffusion-weighted MRI in neurodegenerative and psychiatric animal models: Experimental strategies and main outcomes. J Neurosci Methods 2020; 343:108814. [PMID: 32569785 DOI: 10.1016/j.jneumeth.2020.108814] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 12/31/2022]
Abstract
Preclinical MRI approaches constitute a key tool to study a wide variety of neurological and psychiatric illnesses, allowing a more direct investigation of the disorder substrate and, at the same time, the possibility of back-translating such findings to human subjects. However, the lack of consensus on the optimal experimental scheme used to acquire the data has led to relatively high heterogeneity in the choice of protocols, which can potentially impact the comparison between results obtained by different groups, even using the same animal model. This is especially true for diffusion-weighted MRI data, where certain experimental choices can impact not only on the accuracy and precision of the extracted biomarkers, but also on their biological meaning. With this in mind, we extensively examined preclinical imaging studies that used diffusion-weighted MRI to investigate neurodegenerative, neurodevelopmental and psychiatric disorders in rodent models. In this review, we discuss the main findings for each preclinical model, with a special focus on the analysis and comparison of the different acquisition strategies used across studies and their impact on the heterogeneity of the findings.
Collapse
Affiliation(s)
- Amr Eed
- Instituto de Neurociencias, CSIC, UMH, San Juan de Alicante, Alicante, Spain
| | | | - Juan Lerma
- Instituto de Neurociencias, CSIC, UMH, San Juan de Alicante, Alicante, Spain
| | - Silvia De Santis
- Instituto de Neurociencias, CSIC, UMH, San Juan de Alicante, Alicante, Spain; CUBRIC, School of Psychology, Cardiff University, Cardiff, UK.
| |
Collapse
|
16
|
Reduced axonal caliber and structural changes in a rat model of Fragile X syndrome with a deletion of a K-Homology domain of Fmr1. Transl Psychiatry 2020; 10:280. [PMID: 32788572 PMCID: PMC7423986 DOI: 10.1038/s41398-020-00943-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 04/15/2020] [Accepted: 04/21/2020] [Indexed: 12/30/2022] Open
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder that is caused by mutations in the FMR1 gene. Neuroanatomical alterations have been reported in both male and female individuals with FXS, yet the morphological underpinnings of these alterations have not been elucidated. In the current study, we found structural changes in both male and female rats that model FXS, some of which are similarly impaired in both sexes, including the superior colliculus and periaqueductal gray, and others that show sex-specific changes. The splenium of the corpus callosum, for example, was only impaired in males. We also found reduced axonal caliber in the splenium, offering a mechanism for its structural changes. Furthermore, we found that overall, male rats have higher brain-wide diffusion than female rats. Our results provide insight into which brain regions are vulnerable to a loss of Fmr1 expression and reveal an impairment at the level of the axon that could cause structural changes in white matter regions.
Collapse
|
17
|
Telias M. Pharmacological Treatments for Fragile X Syndrome Based on Synaptic Dysfunction. Curr Pharm Des 2020; 25:4394-4404. [PMID: 31682210 DOI: 10.2174/1381612825666191102165206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 10/31/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND Fragile X syndrome (FXS) is the most common form of monogenic hereditary cognitive impairment, including intellectual disability, autism, hyperactivity, and epilepsy. METHODS This article reviews the literature pertaining to the role of synaptic dysfunction in FXS. RESULTS In FXS, synaptic dysfunction alters the excitation-inhibition ratio, dysregulating molecular and cellular processes underlying cognition, learning, memory, and social behavior. Decades of research have yielded important hypotheses that could explain, at least in part, the development of these neurological disorders in FXS patients. However, the main goal of translating lab research in animal models to pharmacological treatments in the clinic has been so far largely unsuccessful, leaving FXS a still incurable disease. CONCLUSION In this concise review, we summarize and analyze the main hypotheses proposed to explain synaptic dysregulation in FXS, by reviewing the scientific evidence that led to pharmaceutical clinical trials and their outcome.
Collapse
Affiliation(s)
- Michael Telias
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, United States
| |
Collapse
|
18
|
Markicevic M, Fulcher BD, Lewis C, Helmchen F, Rudin M, Zerbi V, Wenderoth N. Cortical Excitation:Inhibition Imbalance Causes Abnormal Brain Network Dynamics as Observed in Neurodevelopmental Disorders. Cereb Cortex 2020; 30:4922-4937. [PMID: 32313923 PMCID: PMC7391279 DOI: 10.1093/cercor/bhaa084] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Abnormal brain development manifests itself at different spatial scales. However, whether abnormalities at the cellular level can be diagnosed from network activity measured with functional magnetic resonance imaging (fMRI) is largely unknown, yet of high clinical relevance. Here a putative mechanism reported in neurodevelopmental disorders, that is, excitation-to-inhibition ratio (E:I), was chemogenetically increased within cortical microcircuits of the mouse brain and measured via fMRI. Increased E:I caused a significant "reduction" of long-range connectivity, irrespective of whether excitatory neurons were facilitated or inhibitory Parvalbumin (PV) interneurons were suppressed. Training a classifier on fMRI signals, we were able to accurately classify cortical areas exhibiting increased E:I. This classifier was validated in an independent cohort of Fmr1y/- knockout mice, a model for autism with well-documented loss of parvalbumin neurons and chronic alterations of E:I. Our findings demonstrate a promising novel approach towards inferring microcircuit abnormalities from macroscopic fMRI measurements.
Collapse
Affiliation(s)
- Marija Markicevic
- Neural Control of Movement Lab, HEST, ETH Zürich, 8093 Zurich, Switzerland.,Neuroscience Center Zurich, University and ETH Zurich, 8057 Zurich, Switzerland
| | - Ben D Fulcher
- School of Physics, The University of Sydney, NSW 2006, Australia
| | - Christopher Lewis
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Fritjof Helmchen
- Neuroscience Center Zurich, University and ETH Zurich, 8057 Zurich, Switzerland.,Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Markus Rudin
- Neuroscience Center Zurich, University and ETH Zurich, 8057 Zurich, Switzerland.,Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland.,Institute for Biomedical Engineering, University and ETH Zurich, 8093 Zurich, Switzerland
| | - Valerio Zerbi
- Neural Control of Movement Lab, HEST, ETH Zürich, 8093 Zurich, Switzerland.,Neuroscience Center Zurich, University and ETH Zurich, 8057 Zurich, Switzerland
| | - Nicole Wenderoth
- Neural Control of Movement Lab, HEST, ETH Zürich, 8093 Zurich, Switzerland.,Neuroscience Center Zurich, University and ETH Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
19
|
Shitik EM, Velmiskina AA, Dolskiy AA, Yudkin DV. Reactivation of FMR1 gene expression is a promising strategy for fragile X syndrome therapy. Gene Ther 2020; 27:247-253. [PMID: 32203197 DOI: 10.1038/s41434-020-0141-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 02/06/2023]
Abstract
Fragile X syndrome (FXS) is the most common form of intellectual disability and autism spectrum disorder and is caused by CGG repeat expansion in the promoter region of the FMR1 gene, which encodes fragile X mental retardation protein. This event leads to gene silencing and the loss of gene products through DNA methylation and chromatin remodeling. Due to the pathogenesis of FXS, targeted, symptomatic, and etiological approaches have been developed for its treatment. Despite their rapid development, symptomatic and targeted treatment approaches have numerous limitations; etiological approaches have the greatest potential because they affect the main causes of transcriptional silencing. In this review, we consider three potential etiological therapeutic methods that affect the reactivation of FMR1 gene expression: treatment with inhibitors of chromatin-modifying enzymes, the use of noncoding RNAs and the application of gene therapy. Inhibitors of chromatin-modifying enzymes are not clinically applicable because of their low reactivity and high cytotoxicity, and noncoding RNAs are currently only under study. Thus, we discuss gene therapy as the most promising approach for treating FXS in the near future.
Collapse
Affiliation(s)
- Ekaterina M Shitik
- State Research Center of Virology and Biotechnology "Vector", Federal Service for Surveillance on Consumer Rights Protection and Human Well-being (FBRI SRC VB "Vector", Rospotrebnadzor), Koltsovo, Novosibirsk Region, Russia
| | - Anastasia A Velmiskina
- State Research Center of Virology and Biotechnology "Vector", Federal Service for Surveillance on Consumer Rights Protection and Human Well-being (FBRI SRC VB "Vector", Rospotrebnadzor), Koltsovo, Novosibirsk Region, Russia
| | - Alexander A Dolskiy
- State Research Center of Virology and Biotechnology "Vector", Federal Service for Surveillance on Consumer Rights Protection and Human Well-being (FBRI SRC VB "Vector", Rospotrebnadzor), Koltsovo, Novosibirsk Region, Russia
| | - Dmitry V Yudkin
- State Research Center of Virology and Biotechnology "Vector", Federal Service for Surveillance on Consumer Rights Protection and Human Well-being (FBRI SRC VB "Vector", Rospotrebnadzor), Koltsovo, Novosibirsk Region, Russia.
| |
Collapse
|