1
|
Yang X, Huang YWA, Marshall J. Targeting TrkB-PSD-95 coupling to mitigate neurological disorders. Neural Regen Res 2025; 20:715-724. [PMID: 38886937 PMCID: PMC11433911 DOI: 10.4103/nrr.nrr-d-23-02000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 03/15/2024] [Accepted: 03/30/2024] [Indexed: 06/20/2024] Open
Abstract
Tropomyosin receptor kinase B (TrkB) signaling plays a pivotal role in dendritic growth and dendritic spine formation to promote learning and memory. The activity-dependent release of brain-derived neurotrophic factor at synapses binds to pre- or postsynaptic TrkB resulting in the strengthening of synapses, reflected by long-term potentiation. Postsynaptically, the association of postsynaptic density protein-95 with TrkB enhances phospholipase Cγ-Ca2+/calmodulin-dependent protein kinase II and phosphatidylinositol 3-kinase-mechanistic target of rapamycin signaling required for long-term potentiation. In this review, we discuss TrkB-postsynaptic density protein-95 coupling as a promising strategy to magnify brain-derived neurotrophic factor signaling towards the development of novel therapeutics for specific neurological disorders. A reduction of TrkB signaling has been observed in neurodegenerative disorders, such as Alzheimer's disease and Huntington's disease, and enhancement of postsynaptic density protein-95 association with TrkB signaling could mitigate the observed deficiency of neuronal connectivity in schizophrenia and depression. Treatment with brain-derived neurotrophic factor is problematic, due to poor pharmacokinetics, low brain penetration, and side effects resulting from activation of the p75 neurotrophin receptor or the truncated TrkB.T1 isoform. Although TrkB agonists and antibodies that activate TrkB are being intensively investigated, they cannot distinguish the multiple human TrkB splicing isoforms or cell type-specific functions. Targeting TrkB-postsynaptic density protein-95 coupling provides an alternative approach to specifically boost TrkB signaling at localized synaptic sites versus global stimulation that risks many adverse side effects.
Collapse
Affiliation(s)
- Xin Yang
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Yu-Wen Alvin Huang
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
- Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, RI, USA
| | - John Marshall
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| |
Collapse
|
2
|
Antonijevic M, Dallemagne P, Rochais C. Indirect influence on the BDNF/TrkB receptor signaling pathway via GPCRs, an emerging strategy in the treatment of neurodegenerative disorders. Med Res Rev 2025; 45:274-310. [PMID: 39180386 DOI: 10.1002/med.22075] [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: 11/16/2021] [Revised: 12/06/2022] [Accepted: 08/04/2024] [Indexed: 08/26/2024]
Abstract
Neuronal survival depends on neurotrophins and their receptors. There are two types of neurotrophin receptors: a nonenzymatic, trans-membrane protein of the tumor necrosis factor receptor (TNFR) family-p75 receptor and the tyrosine kinase receptors (TrkR) A, B, and C. Activation of the TrkBR by brain-derived neurotrophic factor (BDNF) or neurotrophin 4/5 (NT-4/5) promotes neuronal survival, differentiation, and synaptic function. It is shown that in the pathogenesis of several neurodegenerative conditions (Alzheimer's disease, Parkinson's disease, Huntington's disease) the BDNF/TrkBR signaling pathway is impaired. Since it is known that GPCRs and TrkR are regulating several cell functions by interacting with each other and generating a cross-communication in this review we have focused on the interaction between different GPCRs and their ligands on BDNF/TrkBR signaling pathway.
Collapse
|
3
|
Dooley JC, van der Heijden ME. More Than a Small Brain: The Importance of Studying Neural Function during Development. J Neurosci 2024; 44:e1367242024. [PMID: 39603806 PMCID: PMC11604142 DOI: 10.1523/jneurosci.1367-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/17/2024] [Accepted: 10/17/2024] [Indexed: 11/29/2024] Open
Abstract
The nervous system contains complex circuits comprising thousands of cell types and trillions of connections. Here, we discuss how the field of "developmental systems neuroscience" combines the molecular and genetic perspectives of developmental neuroscience with the (typically adult-focused) functional perspective of systems neuroscience. This combination of approaches is critical to understanding how a handful of cells eventually produce the wide range of behaviors necessary for survival. Functional circuit development typically lags behind neural connectivity, leading to intermediate stages of neural activity that are either not seen in adults or, if present, are considered pathophysiological. Developmental systems neuroscience examines these intermediate stages of neural activity, mapping out the critical phases and inflection points of neural circuit function to understand how neural activity and behavior emerge across development. Beyond understanding typical development, this approach provides invaluable insight into the pathophysiology of neurodevelopmental disorders by identifying when and how functional development diverges between health and disease. We argue that developmental systems neuroscience will identify important periods of neural development, reveal novel therapeutic windows for treatment, and set the stage to answer fundamental questions about the brain in health and disease.
Collapse
Affiliation(s)
- James C Dooley
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana 47907
| | - Meike E van der Heijden
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia 24016
- Center for Neurobiology Research, Roanoke, Virginia 24016
- School of Neuroscience, Virginia Tech, Blacksburg, Virginia 24016
| |
Collapse
|
4
|
Faulkner IE, Pajak RZ, Harte MK, Glazier JD, Hager R. Voltage-gated potassium channels as a potential therapeutic target for the treatment of neurological and psychiatric disorders. Front Cell Neurosci 2024; 18:1449151. [PMID: 39411003 PMCID: PMC11473391 DOI: 10.3389/fncel.2024.1449151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024] Open
Abstract
Voltage-gated potassium channels are a widely distributed subgroup of potassium channels responsible for the efflux of potassium in the repolarisation of the cell membrane, and hence contribute to the latency and propagation of action potentials. As they are causal to synaptic transmission, alterations to the structure of these channels can lead to a variety of neurological and psychiatric diseases. The Kv3 subfamily of voltage-gated potassium channels are found on many neurons in the brain, including inhibitory interneurons where they contribute to fast-frequency firing. Changes to the firing ability of these interneurons can lead to an imbalance of inhibitory and excitatory neurotransmission. To date, we have little understanding of the mechanism by which excitatory and inhibitory inputs become imbalanced. This imbalance is associated with cognitive deficits seen across neurological and neuropsychiatric disorders, which are currently difficult to treat. In this review, we collate evidence supporting the hypothesis that voltage-gated potassium channels, specifically the Kv3 subfamily, are central to many neurological and psychiatric disorders, and may thus be considered as an effective drug target. The collective evidence provided by the studies reviewed here demonstrates that Kv3 channels may be amenable to novel treatments that modulate the activity of these channels, with the prospect of improved patient outcome.
Collapse
Affiliation(s)
- Isabel E. Faulkner
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Rachael Z. Pajak
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Michael K. Harte
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Jocelyn D. Glazier
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Reinmar Hager
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
5
|
Dumontier D, Liebman SA, Le VH, George S, Valdemar D, Van Aelst L, Pouchelon G. Restoring transient connectivity during development improves dysfunctions in fragile X mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.08.611918. [PMID: 39314327 PMCID: PMC11419037 DOI: 10.1101/2024.09.08.611918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Early-generated circuits are critical for the maturation of cortical network activity and the formation of excitation/inhibition (E/I) balance. This process involves the maturation of specific populations of inhibitory neurons. While parvalbumin (PV)-expressing neurons have been associated with E/I impairments observed in neurodevelopmental disorders, somatostatin-expressing (SST) neurons have recently been shown to regulate PV neuron maturation by controlling neural dynamics in the developing cortex. SST neurons receive transient connections from the sensory thalamus, yet the implications of transient connectivity in neurodevelopmental disorders remain unknown. Here, we show that thalamocortical connectivity to SST neurons is persistent rather than transient in a mouse model of Fragile X syndrome. We were able to restore the transient dynamics using chemogenetics, which led to the recovery of fragile X-associated dysfunctions in circuit maturation and sensory-dependent behavior. Overall, our findings unveil the role of early transient dynamics in controlling downstream maturation of sensory functions.
Collapse
Affiliation(s)
| | | | - Viet-Hang Le
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Shanu George
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | | | | |
Collapse
|
6
|
Kourdougli N, Nomura T, Wu MW, Heuvelmans A, Dobler Z, Contractor A, Portera-Cailliau C. The NKCC1 Inhibitor Bumetanide Restores Cortical Feedforward Inhibition and Lessens Sensory Hypersensitivity in Early Postnatal Fragile X Mice. Biol Psychiatry 2024:S0006-3223(24)01427-6. [PMID: 38950809 DOI: 10.1016/j.biopsych.2024.06.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 06/12/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024]
Abstract
BACKGROUND Exaggerated responses to sensory stimuli, a hallmark of fragile X syndrome, contribute to anxiety and learning challenges. Sensory hypersensitivity is recapitulated in the Fmr1 knockout (KO) mouse model of fragile X syndrome. Recent studies in Fmr1 KO mice have demonstrated differences in the activity of cortical interneurons and a delayed switch in the polarity of GABA (gamma-aminobutyric acid) signaling during development. Previously, we reported that blocking the chloride transporter NKCC1 with the diuretic bumetanide could rescue synaptic circuit phenotypes in the primary somatosensory cortex (S1) of Fmr1 KO mice. However, it remains unknown whether bumetanide can rescue earlier circuit phenotypes or sensory hypersensitivity in Fmr1 KO mice. METHODS We used acute and chronic systemic administration of bumetanide in Fmr1 KO mice and performed in vivo 2-photon calcium imaging to record neuronal activity, while tracking mouse behavior with high-resolution videos. RESULTS We demonstrated that layer 2/3 pyramidal neurons in the S1 of Fmr1 KO mice showed a higher frequency of synchronous events on postnatal day 6 than wild-type controls. This was reversed by acute administration of bumetanide. Furthermore, chronic bumetanide treatment (postnatal days 5-14) restored S1 circuit differences in Fmr1 KO mice, including reduced neuronal adaptation to repetitive whisker stimulation, and ameliorated tactile defensiveness. Bumetanide treatment also rectified the reduced feedforward inhibition of layer 2/3 neurons in the S1 and boosted the circuit participation of parvalbumin interneurons. CONCLUSIONS This further supports the notion that synaptic, circuit, and sensory behavioral phenotypes in Fmr1 KO can be mitigated by inhibitors of NKCC1, such as the Food and Drug Administration-approved diuretic bumetanide.
Collapse
Affiliation(s)
- Nazim Kourdougli
- Department of Neurology, University of California, Los Angeles, Los Angeles, California
| | - Toshihiro Nomura
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Michelle W Wu
- Department of Neurology, University of California, Los Angeles, Los Angeles, California; Neuroscience Interdepartmental Graduate Program, University of California, Los Angeles, Los Angeles, California; UCLA-Caltech Medical Scientist Training Program, University of California, Los Angeles, Los Angeles, California
| | - Anouk Heuvelmans
- Department of Neurology, University of California, Los Angeles, Los Angeles, California
| | - Zoë Dobler
- Department of Neurology, University of California, Los Angeles, Los Angeles, California; Neuroscience Interdepartmental Graduate Program, University of California, Los Angeles, Los Angeles, California
| | - Anis Contractor
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Carlos Portera-Cailliau
- Department of Neurology, University of California, Los Angeles, Los Angeles, California; Department of Neurobiology, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
7
|
Marshall AH, Hanson MA, Boyle DJ, Nagarajan D, Bibi N, Fitzgerald J, Gaitten E, Kokiko-Cochran ON, Gu B, Wester JC. Arid1b haploinsufficiency in pyramidal neurons causes cellular and circuit changes in neocortex but is not sufficient to produce behavioral or seizure phenotypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.597344. [PMID: 38895205 PMCID: PMC11185765 DOI: 10.1101/2024.06.04.597344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Arid1b is a high confidence risk gene for autism spectrum disorder that encodes a subunit of a chromatin remodeling complex expressed in neuronal progenitors. Haploinsufficiency causes a broad range of social, behavioral, and intellectual disability phenotypes, including Coffin-Siris syndrome. Recent work using transgenic mouse models suggests pathology is due to deficits in proliferation, survival, and synaptic development of cortical neurons. However, there is conflicting evidence regarding the relative roles of excitatory projection neurons and inhibitory interneurons in generating abnormal cognitive and behavioral phenotypes. Here, we conditionally knocked out either one or both copies of Arid1b from excitatory projection neuron progenitors and systematically investigated the effects on intrinsic membrane properties, synaptic physiology, social behavior, and seizure susceptibility. We found that disrupting Arid1b expression in excitatory neurons alters their membrane properties, including hyperpolarizing action potential threshold; however, these changes depend on neuronal subtype. Using paired whole-cell recordings, we found increased synaptic connectivity rate between projection neurons. Furthermore, we found reduced strength of excitatory synapses to parvalbumin (PV)-expression inhibitory interneurons. These data suggest an increase in the ratio of excitation to inhibition. However, the strength of inhibitory synapses from PV interneurons to excitatory neurons was enhanced, which may rebalance this ratio. Indeed, Arid1b haploinsufficiency in projection neurons was insufficient to cause social deficits and seizure phenotypes observed in a preclinical germline haploinsufficient mouse model. Our data suggest that while excitatory projection neurons likely contribute to autistic phenotypes, pathology in these cells is not the primary cause.
Collapse
|
8
|
Bhandari K, Kanodia H, Donato F, Caroni P. Selective vulnerability of the ventral hippocampus-prelimbic cortex axis parvalbumin interneuron network underlies learning deficits of fragile X mice. Cell Rep 2024; 43:114124. [PMID: 38630591 DOI: 10.1016/j.celrep.2024.114124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 03/07/2024] [Accepted: 04/02/2024] [Indexed: 04/19/2024] Open
Abstract
High-penetrance mutations affecting mental health can involve genes ubiquitously expressed in the brain. Whether the specific patterns of dysfunctions result from ubiquitous circuit deficits or might reflect selective vulnerabilities of targetable subnetworks has remained unclear. Here, we determine how loss of ubiquitously expressed fragile X mental retardation protein (FMRP), the cause of fragile X syndrome, affects brain networks in Fmr1y/- mice. We find that in wild-type mice, area-specific knockout of FMRP in the adult mimics behavioral consequences of area-specific silencing. By contrast, the functional axis linking the ventral hippocampus (vH) to the prelimbic cortex (PreL) is selectively affected in constitutive Fmr1y/- mice. A chronic alteration in late-born parvalbumin interneuron networks across the vH-PreL axis rescued by VIP signaling specifically accounts for deficits in vH-PreL theta-band network coherence, ensemble assembly, and learning functions of Fmr1y/- mice. Therefore, vH-PreL axis function exhibits a selective vulnerability to loss of FMRP in the vH or PreL, leading to learning and memory dysfunctions in fragile X mice.
Collapse
Affiliation(s)
- Komal Bhandari
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Harsh Kanodia
- Biozentrum, University of Basel, 4058 Basel, Switzerland
| | - Flavio Donato
- Biozentrum, University of Basel, 4058 Basel, Switzerland
| | - Pico Caroni
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland.
| |
Collapse
|
9
|
Croom K, Rumschlag JA, Erickson MA, Binder D, Razak KA. Sex differences during development in cortical temporal processing and event related potentials in wild-type and fragile X syndrome model mice. J Neurodev Disord 2024; 16:24. [PMID: 38720271 PMCID: PMC11077726 DOI: 10.1186/s11689-024-09539-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/17/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is currently diagnosed in approximately 1 in 44 children in the United States, based on a wide array of symptoms, including sensory dysfunction and abnormal language development. Boys are diagnosed ~ 3.8 times more frequently than girls. Auditory temporal processing is crucial for speech recognition and language development. Abnormal development of temporal processing may account for ASD language impairments. Sex differences in the development of temporal processing may underlie the differences in language outcomes in male and female children with ASD. To understand mechanisms of potential sex differences in temporal processing requires a preclinical model. However, there are no studies that have addressed sex differences in temporal processing across development in any animal model of ASD. METHODS To fill this major gap, we compared the development of auditory temporal processing in male and female wildtype (WT) and Fmr1 knock-out (KO) mice, a model of Fragile X Syndrome (FXS), a leading genetic cause of ASD-associated behaviors. Using epidural screw electrodes, we recorded auditory event related potentials (ERP) and auditory temporal processing with a gap-in-noise auditory steady state response (ASSR) paradigm at young (postnatal (p)21 and p30) and adult (p60) ages from both auditory and frontal cortices of awake, freely moving mice. RESULTS The results show that ERP amplitudes were enhanced in both sexes of Fmr1 KO mice across development compared to WT counterparts, with greater enhancement in adult female than adult male KO mice. Gap-ASSR deficits were seen in the frontal, but not auditory, cortex in early development (p21) in female KO mice. Unlike male KO mice, female KO mice show WT-like temporal processing at p30. There were no temporal processing deficits in the adult mice of both sexes. CONCLUSIONS These results show a sex difference in the developmental trajectories of temporal processing and hypersensitive responses in Fmr1 KO mice. Male KO mice show slower maturation of temporal processing than females. Female KO mice show stronger hypersensitive responses than males later in development. The differences in maturation rates of temporal processing and hypersensitive responses during various critical periods of development may lead to sex differences in language function, arousal and anxiety in FXS.
Collapse
Affiliation(s)
- Katilynne Croom
- Graduate Neuroscience Program, University of California, Riverside, USA
| | - Jeffrey A Rumschlag
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, USA
| | - Michael A Erickson
- Department of Psychology, University of California, 900 University Avenue, Riverside, USA
| | - Devin Binder
- Graduate Neuroscience Program, University of California, Riverside, USA
- Biomedical Sciences, School of Medicine, University of California, Riverside, USA
| | - Khaleel A Razak
- Graduate Neuroscience Program, University of California, Riverside, USA.
- Department of Psychology, University of California, 900 University Avenue, Riverside, USA.
| |
Collapse
|
10
|
Nomura T. In Vitro Patch-Clamp. Methods Mol Biol 2024; 2794:221-244. [PMID: 38630233 DOI: 10.1007/978-1-0716-3810-1_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
The patch-clamp technique is one of the most useful tools to analyze the function of electrically active cells such as neurons. This technique allows for the analysis of proteins (ion channels and receptors), cells (neurons), and synapses that are the building blocks of neuronal networks. Cortical development involves coordinated changes in functional measures at each of these levels of analysis that reflect both cellular and circuit maturation. This chapter explains the technical and theoretical basis of patch-clamp methodology and introduces several examples of how this technique can be applied in the context of cortical development.
Collapse
Affiliation(s)
- Toshihiro Nomura
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
11
|
Nomura T, Taniguchi S, Wang YZ, Yeh NH, Wilen AP, Castillon CCM, Foote KM, Xu J, Armstrong JN, Savas JN, Swanson GT, Contractor A. A Pathogenic Missense Mutation in Kainate Receptors Elevates Dendritic Excitability and Synaptic Integration through Dysregulation of SK Channels. J Neurosci 2023; 43:7913-7928. [PMID: 37802657 PMCID: PMC10669804 DOI: 10.1523/jneurosci.1259-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/12/2023] [Accepted: 09/16/2023] [Indexed: 10/08/2023] Open
Abstract
Numerous rare variants that cause neurodevelopmental disorders (NDDs) occur within genes encoding synaptic proteins, including ionotropic glutamate receptors. However, in many cases, it remains unclear how damaging missense variants affect brain function. We determined the physiological consequences of an NDD causing missense mutation in the GRIK2 kainate receptor (KAR) gene, that results in a single amino acid change p.Ala657Thr in the GluK2 receptor subunit. We engineered this mutation in the mouse Grik2 gene, yielding a GluK2(A657T) mouse, and studied mice of both sexes to determine how hippocampal neuronal function is disrupted. Synaptic KAR currents in hippocampal CA3 pyramidal neurons from heterozygous A657T mice exhibited slow decay kinetics, consistent with incorporation of the mutant subunit into functional receptors. Unexpectedly, CA3 neurons demonstrated elevated action potential spiking because of downregulation of the small-conductance Ca2+ activated K+ channel (SK), which mediates the post-spike afterhyperpolarization. The reduction in SK activity resulted in increased CA3 dendritic excitability, increased EPSP-spike coupling, and lowered the threshold for the induction of LTP of the associational-commissural synapses in CA3 neurons. Pharmacological inhibition of SK channels in WT mice increased dendritic excitability and EPSP-spike coupling, mimicking the phenotype in A657T mice and suggesting a causative role for attenuated SK activity in aberrant excitability observed in the mutant mice. These findings demonstrate that a disease-associated missense mutation in GRIK2 leads to altered signaling through neuronal KARs, pleiotropic effects on neuronal and dendritic excitability, and implicate these processes in neuropathology in patients with genetic NDDs.SIGNIFICANCE STATEMENT Damaging mutations in genes encoding synaptic proteins have been identified in various neurodevelopmental disorders, but the functional consequences at the cellular and circuit level remain elusive. By generating a novel knock-in mutant mouse, this study examined the role of a pathogenic mutation in the GluK2 kainate receptor (KAR) subunit, a subclass of ionotropic glutamate receptors. Analyses of hippocampal CA3 pyramidal neurons determined elevated action potential firing because of an increase in dendritic excitability. Increased dendritic excitability was attributable to reduced activity of a Ca2+ activated K+ channel. These results indicate that a pathogenic KAR mutation results in dysregulation of dendritic K+ channels, which leads to an increase in synaptic integration and backpropagation of action potentials into distal dendrites.
Collapse
Affiliation(s)
- Toshihiro Nomura
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Sakiko Taniguchi
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Yi-Zhi Wang
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Nai-Hsing Yeh
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Anika P Wilen
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Charlotte C M Castillon
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Kendall M Foote
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Jian Xu
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - John N Armstrong
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Jeffrey N Savas
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Geoffrey T Swanson
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
- Department of Neurobiology, Weinberg College of Arts and Sciences Northwestern University, Chicago, Illinois 60611
| | - Anis Contractor
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
- Department of Psychiatry and Behavioral Sciences Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
- Department of Neurobiology, Weinberg College of Arts and Sciences Northwestern University, Chicago, Illinois 60611
| |
Collapse
|
12
|
Kourdougli N, Suresh A, Liu B, Juarez P, Lin A, Chung DT, Graven Sams A, Gandal MJ, Martínez-Cerdeño V, Buonomano DV, Hall BJ, Mombereau C, Portera-Cailliau C. Improvement of sensory deficits in fragile X mice by increasing cortical interneuron activity after the critical period. Neuron 2023; 111:2863-2880.e6. [PMID: 37451263 PMCID: PMC10529373 DOI: 10.1016/j.neuron.2023.06.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 04/14/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023]
Abstract
Changes in the function of inhibitory interneurons (INs) during cortical development could contribute to the pathophysiology of neurodevelopmental disorders. Using all-optical in vivo approaches, we find that parvalbumin (PV) INs and their immature precursors are hypoactive and transiently decoupled from excitatory neurons in postnatal mouse somatosensory cortex (S1) of Fmr1 KO mice, a model of fragile X syndrome (FXS). This leads to a loss of parvalbumin INs (PV-INs) in both mice and humans with FXS. Increasing the activity of future PV-INs in neonatal Fmr1 KO mice restores PV-IN density and ameliorates transcriptional dysregulation in S1, but not circuit dysfunction. Critically, administering an allosteric modulator of Kv3.1 channels after the S1 critical period does rescue circuit dynamics and tactile defensiveness. Symptoms in FXS and related disorders could be mitigated by targeting PV-INs.
Collapse
Affiliation(s)
| | - Anand Suresh
- Department of Neurology, UCLA, Los Angeles, CA, USA
| | - Benjamin Liu
- Department of Neurology, UCLA, Los Angeles, CA, USA
| | - Pablo Juarez
- Department of Pathology, UC Davis, Davis, CA, USA
| | - Ashley Lin
- Department of Neurology, UCLA, Los Angeles, CA, USA
| | | | | | | | | | - Dean V Buonomano
- Department of Neurology, UCLA, Los Angeles, CA, USA; Department of Psychology, UCLA, Los Angeles, CA, USA
| | | | | | - Carlos Portera-Cailliau
- Department of Neurology, UCLA, Los Angeles, CA, USA; Department of Neurobiology, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
13
|
Monday HR, Wang HC, Feldman DE. Circuit-level theories for sensory dysfunction in autism: convergence across mouse models. Front Neurol 2023; 14:1254297. [PMID: 37745660 PMCID: PMC10513044 DOI: 10.3389/fneur.2023.1254297] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/14/2023] [Indexed: 09/26/2023] Open
Abstract
Individuals with autism spectrum disorder (ASD) exhibit a diverse range of behavioral features and genetic backgrounds, but whether different genetic forms of autism involve convergent pathophysiology of brain function is unknown. Here, we analyze evidence for convergent deficits in neural circuit function across multiple transgenic mouse models of ASD. We focus on sensory areas of neocortex, where circuit differences may underlie atypical sensory processing, a central feature of autism. Many distinct circuit-level theories for ASD have been proposed, including increased excitation-inhibition (E-I) ratio and hyperexcitability, hypofunction of parvalbumin (PV) interneuron circuits, impaired homeostatic plasticity, degraded sensory coding, and others. We review these theories and assess the degree of convergence across ASD mouse models for each. Behaviorally, our analysis reveals that innate sensory detection behavior is heightened and sensory discrimination behavior is impaired across many ASD models. Neurophysiologically, PV hypofunction and increased E-I ratio are prevalent but only rarely generate hyperexcitability and excess spiking. Instead, sensory tuning and other aspects of neural coding are commonly degraded and may explain impaired discrimination behavior. Two distinct phenotypic clusters with opposing neural circuit signatures are evident across mouse models. Such clustering could suggest physiological subtypes of autism, which may facilitate the development of tailored therapeutic approaches.
Collapse
Affiliation(s)
- Hannah R. Monday
- Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
| | | | - Daniel E. Feldman
- Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
| |
Collapse
|
14
|
Mohapatra AN, Wagner S. The role of the prefrontal cortex in social interactions of animal models and the implications for autism spectrum disorder. Front Psychiatry 2023; 14:1205199. [PMID: 37409155 PMCID: PMC10318347 DOI: 10.3389/fpsyt.2023.1205199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 05/26/2023] [Indexed: 07/07/2023] Open
Abstract
Social interaction is a complex behavior which requires the individual to integrate various internal processes, such as social motivation, social recognition, salience, reward, and emotional state, as well as external cues informing the individual of others' behavior, emotional state and social rank. This complex phenotype is susceptible to disruption in humans affected by neurodevelopmental and psychiatric disorders, including autism spectrum disorder (ASD). Multiple pieces of convergent evidence collected from studies of humans and rodents suggest that the prefrontal cortex (PFC) plays a pivotal role in social interactions, serving as a hub for motivation, affiliation, empathy, and social hierarchy. Indeed, disruption of the PFC circuitry results in social behavior deficits symptomatic of ASD. Here, we review this evidence and describe various ethologically relevant social behavior tasks which could be employed with rodent models to study the role of the PFC in social interactions. We also discuss the evidence linking the PFC to pathologies associated with ASD. Finally, we address specific questions regarding mechanisms employed by the PFC circuitry that may result in atypical social interactions in rodent models, which future studies should address.
Collapse
Affiliation(s)
- Alok Nath Mohapatra
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | | |
Collapse
|
15
|
Tempio A, Boulksibat A, Bardoni B, Delhaye S. Fragile X Syndrome as an interneuronopathy: a lesson for future studies and treatments. Front Neurosci 2023; 17:1171895. [PMID: 37188005 PMCID: PMC10176609 DOI: 10.3389/fnins.2023.1171895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
Fragile X Syndrome (FXS) is the most common form of inherited intellectual disability (ID) and a primary genetic cause of autism spectrum disorder (ASD). FXS arises from the silencing of the FMR1 gene causing the lack of translation of its encoded protein, the Fragile X Messenger RibonucleoProtein (FMRP), an RNA-binding protein involved in translational control and in RNA transport along dendrites. Although a large effort during the last 20 years has been made to investigate the cellular roles of FMRP, no effective and specific therapeutic intervention is available to treat FXS. Many studies revealed a role for FMRP in shaping sensory circuits during developmental critical periods to affect proper neurodevelopment. Dendritic spine stability, branching and density abnormalities are part of the developmental delay observed in various FXS brain areas. In particular, cortical neuronal networks in FXS are hyper-responsive and hyperexcitable, making these circuits highly synchronous. Overall, these data suggest that the excitatory/inhibitory (E/I) balance in FXS neuronal circuitry is altered. However, not much is known about how interneuron populations contribute to the unbalanced E/I ratio in FXS even if their abnormal functioning has an impact on the behavioral deficits of patients and animal models affected by neurodevelopmental disorders. We revise here the key literature concerning the role of interneurons in FXS not only with the purpose to better understand the pathophysiology of this disorder, but also to explore new possible therapeutic applications to treat FXS and other forms of ASD or ID. Indeed, for instance, the re-introduction of functional interneurons in the diseased brains has been proposed as a promising therapeutic approach for neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Alessandra Tempio
- Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
- Alessandra Tempio,
| | - Asma Boulksibat
- Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Barbara Bardoni
- Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
- Inserm, Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
- *Correspondence: Barbara Bardoni,
| | - Sébastien Delhaye
- Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| |
Collapse
|
16
|
Kalinowska M, van der Lei MB, Kitiashvili M, Mamcarz M, Oliveira MM, Longo F, Klann E. Deletion of Fmr1 in parvalbumin-expressing neurons results in dysregulated translation and selective behavioral deficits associated with fragile X syndrome. Mol Autism 2022; 13:29. [PMID: 35768828 PMCID: PMC9245312 DOI: 10.1186/s13229-022-00509-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 06/10/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Fragile X syndrome (FXS), the most common genetic cause of autism spectrum disorder and intellectual disability, is caused by the lack of fragile X mental retardation protein (FMRP) expression. FMRP is an mRNA binding protein with functions in mRNA transport, localization, and translational control. In Fmr1 knockout mice, dysregulated translation has been linked to pathophysiology, including abnormal synaptic function and dendritic morphology, and autistic-like behavioral phenotypes. The role of FMRP in morphology and function of excitatory neurons has been well studied in mice lacking Fmr1, but the impact of Fmr1 deletion on inhibitory neurons remains less characterized. Moreover, the contribution of FMRP in different cell types to FXS pathophysiology is not well defined. We sought to characterize whether FMRP loss in parvalbumin or somatostatin-expressing neurons results in FXS-like deficits in mice. METHODS We used Cre-lox recombinase technology to generate two lines of conditional knockout mice lacking FMRP in either parvalbumin or somatostatin-expressing cells and carried out a battery of behavioral tests to assess motor function, anxiety, repetitive, stereotypic, social behaviors, and learning and memory. In addition, we used fluorescent non-canonical amino acid tagging along with immunostaining to determine whether de novo protein synthesis is dysregulated in parvalbumin or somatostatin-expressing neurons. RESULTS De novo protein synthesis was elevated in hippocampal parvalbumin and somatostatin-expressing inhibitory neurons in Fmr1 knockout mice. Cell type-specific deletion of Fmr1 in parvalbumin-expressing neurons resulted in anxiety-like behavior, impaired social behavior, and dysregulated de novo protein synthesis. In contrast, deletion of Fmr1 in somatostatin-expressing neurons did not result in behavioral abnormalities and did not significantly impact de novo protein synthesis. This is the first report of how loss of FMRP in two specific subtypes of inhibitory neurons is associated with distinct FXS-like abnormalities. LIMITATIONS The mouse models we generated are limited by whole body knockout of FMRP in parvalbumin or somatostatin-expressing cells and further studies are needed to establish a causal relationship between cellular deficits and FXS-like behaviors. CONCLUSIONS Our findings indicate a cell type-specific role for FMRP in parvalbumin-expressing neurons in regulating distinct behavioral features associated with FXS.
Collapse
Affiliation(s)
- Magdalena Kalinowska
- grid.137628.90000 0004 1936 8753Center for Neural Science, New York University, New York, NY USA
| | - Mathijs B. van der Lei
- grid.5284.b0000 0001 0790 3681Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Michael Kitiashvili
- grid.137628.90000 0004 1936 8753Center for Neural Science, New York University, New York, NY USA
| | - Maggie Mamcarz
- grid.137628.90000 0004 1936 8753Center for Neural Science, New York University, New York, NY USA
| | - Mauricio M. Oliveira
- grid.137628.90000 0004 1936 8753Center for Neural Science, New York University, New York, NY USA
| | - Francesco Longo
- grid.8761.80000 0000 9919 9582Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden ,grid.8761.80000 0000 9919 9582Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Eric Klann
- Center for Neural Science, New York University, New York, NY, USA. .,NYU Neuroscience Institute, New York University Langone Medical Center, New York, NY, USA.
| |
Collapse
|
17
|
Donnard E, Shu H, Garber M. Single cell transcriptomics reveals dysregulated cellular and molecular networks in a fragile X syndrome model. PLoS Genet 2022; 18:e1010221. [PMID: 35675353 PMCID: PMC9212148 DOI: 10.1371/journal.pgen.1010221] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 06/21/2022] [Accepted: 04/27/2022] [Indexed: 02/07/2023] Open
Abstract
Despite advances in understanding the pathophysiology of Fragile X syndrome (FXS), its molecular basis is still poorly understood. Whole brain tissue expression profiles have proved surprisingly uninformative, therefore we applied single cell RNA sequencing to profile an FMRP deficient mouse model with higher resolution. We found that the absence of FMRP results in highly cell type specific gene expression changes that are strongest among specific neuronal types, where FMRP-bound mRNAs were prominently downregulated. Metabolic pathways including translation and respiration are significantly upregulated across most cell types with the notable exception of excitatory neurons. These effects point to a potential difference in the activity of mTOR pathways, and together with other dysregulated pathways, suggest an excitatory-inhibitory imbalance in the Fmr1-knock out cortex that is exacerbated by astrocytes. Our data demonstrate that FMRP loss affects abundance of key cellular communication genes that potentially affect neuronal synapses and provide a resource for interrogating the biological basis of this disorder. Fragile X syndrome is a leading genetic cause of inherited intellectual disability and autism spectrum disorder. It results from the inactivation of a single gene, FMR1 and hence the loss of its encoded protein FMRP. Despite decades of intensive research, we still lack an overview of the molecular and biological consequences of the disease. Using single cell RNA sequencing, we profiled cells from the brain of healthy mice and of knock-out mice lacking the FMRP protein, a common model for this disease, to identify molecular changes that happen across different cell types. We find neurons are the most impacted cell type, where genes in multiple pathways are similarly impacted. This includes transcripts known to be bound by FMRP, which are collectively decreased only in neurons but not in other cell types. Our results show how the loss of FMRP affects the intricate interactions between different brain cell types, which could provide new perspectives to the development of therapeutic interventions.
Collapse
Affiliation(s)
- Elisa Donnard
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail: (ED); (HS); (MG)
| | - Huan Shu
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail: (ED); (HS); (MG)
| | - Manuel Garber
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Department of Dermatology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail: (ED); (HS); (MG)
| |
Collapse
|
18
|
D’Incal C, Broos J, Torfs T, Kooy RF, Vanden Berghe W. Towards Kinase Inhibitor Therapies for Fragile X Syndrome: Tweaking Twists in the Autism Spectrum Kinase Signaling Network. Cells 2022; 11:cells11081325. [PMID: 35456004 PMCID: PMC9029738 DOI: 10.3390/cells11081325] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/01/2022] [Accepted: 04/03/2022] [Indexed: 12/12/2022] Open
Abstract
Absence of the Fragile X Mental Retardation Protein (FMRP) causes autism spectrum disorders and intellectual disability, commonly referred to as the Fragile X syndrome. FMRP is a negative regulator of protein translation and is essential for neuronal development and synapse formation. FMRP is a target for several post-translational modifications (PTMs) such as phosphorylation and methylation, which tightly regulate its cellular functions. Studies have indicated the involvement of FMRP in a multitude of cellular pathways, and an absence of FMRP was shown to affect several neurotransmitter receptors, for example, the GABA receptor and intracellular signaling molecules such as Akt, ERK, mTOR, and GSK3. Interestingly, many of these molecules function as protein kinases or phosphatases and thus are potentially amendable by pharmacological treatment. Several treatments acting on these kinase-phosphatase systems have been shown to be successful in preclinical models; however, they have failed to convincingly show any improvements in clinical trials. In this review, we highlight the different protein kinase and phosphatase studies that have been performed in the Fragile X syndrome. In our opinion, some of the paradoxical study conclusions are potentially due to the lack of insight into integrative kinase signaling networks in the disease. Quantitative proteome analyses have been performed in several models for the FXS to determine global molecular processes in FXS. However, only one phosphoproteomics study has been carried out in Fmr1 knock-out mouse embryonic fibroblasts, and it showed dysfunctional protein kinase and phosphatase signaling hubs in the brain. This suggests that the further use of phosphoproteomics approaches in Fragile X syndrome holds promise for identifying novel targets for kinase inhibitor therapies.
Collapse
Affiliation(s)
- Claudio D’Incal
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium; (C.D.); (J.B.); (T.T.)
- Department of Medical Genetics, University of Antwerp, 2000 Antwerp, Belgium;
| | - Jitse Broos
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium; (C.D.); (J.B.); (T.T.)
| | - Thierry Torfs
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium; (C.D.); (J.B.); (T.T.)
| | - R. Frank Kooy
- Department of Medical Genetics, University of Antwerp, 2000 Antwerp, Belgium;
| | - Wim Vanden Berghe
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium; (C.D.); (J.B.); (T.T.)
- Correspondence: ; Tel.: +0032-(0)-32-652-657
| |
Collapse
|
19
|
Deng PY, Avraham O, Cavalli V, Klyachko VA. Hyperexcitability of Sensory Neurons in Fragile X Mouse Model. Front Mol Neurosci 2022; 14:796053. [PMID: 35002623 PMCID: PMC8727524 DOI: 10.3389/fnmol.2021.796053] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 11/17/2021] [Indexed: 01/18/2023] Open
Abstract
Sensory hypersensitivity and somatosensory deficits represent the core symptoms of Fragile X syndrome (FXS). These alterations are believed to arise from changes in cortical sensory processing, while potential deficits in the function of peripheral sensory neurons residing in dorsal root ganglia remain unexplored. We found that peripheral sensory neurons exhibit pronounced hyperexcitability in Fmr1 KO mice, manifested by markedly increased action potential (AP) firing rate and decreased threshold. Unlike excitability changes found in many central neurons, no significant changes were observed in AP rising and falling time, peak potential, amplitude, or duration. Sensory neuron hyperexcitability was caused primarily by increased input resistance, without changes in cell capacitance or resting membrane potential. Analyses of the underlying mechanisms revealed reduced activity of HCN channels and reduced expression of HCN1 and HCN4 in Fmr1 KO compared to WT. A selective HCN channel blocker abolished differences in all measures of sensory neuron excitability between WT and Fmr1 KO neurons. These results reveal a hyperexcitable state of peripheral sensory neurons in Fmr1 KO mice caused by dysfunction of HCN channels. In addition to the intrinsic neuronal dysfunction, the accompanying paper examines deficits in sensory neuron association/communication with their enveloping satellite glial cells, suggesting contributions from both neuronal intrinsic and extrinsic mechanisms to sensory dysfunction in the FXS mouse model.
Collapse
Affiliation(s)
- Pan-Yue Deng
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Oshri Avraham
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, United States
| | - Valeria Cavalli
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, United States.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States.,Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | - Vitaly A Klyachko
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, United States.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
20
|
Rais M, Lovelace JW, Shuai XS, Woodard W, Bishay S, Estrada L, Sharma AR, Nguy A, Kulinich A, Pirbhoy PS, Palacios AR, Nelson DL, Razak KA, Ethell IM. Functional consequences of postnatal interventions in a mouse model of Fragile X syndrome. Neurobiol Dis 2022; 162:105577. [PMID: 34871737 DOI: 10.1016/j.nbd.2021.105577] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 10/22/2021] [Accepted: 12/02/2021] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is a leading genetic cause of autism and intellectual disability with cortical hyperexcitability and sensory hypersensitivity attributed to loss and hypofunction of inhibitory parvalbumin-expressing (PV) cells. Our studies provide novel insights into the role of excitatory neurons in abnormal development of PV cells during a postnatal period of inhibitory circuit refinement. METHODS To achieve Fragile X mental retardation gene (Fmr1) deletion and re-expression in excitatory neurons during the postnatal day (P)14-P21 period, we generated CreCaMKIIa/Fmr1Flox/y (cOFF) and CreCaMKIIa/Fmr1FloxNeo/y (cON) mice, respectively. Cortical phenotypes were evaluated in adult mice using biochemical, cellular, clinically relevant electroencephalogram (EEG) and behavioral tests. RESULTS We found that similar to global Fmr1 KO mice, the density of PV-expressing cells, their activation, and sound-evoked gamma synchronization were impaired in cOFF mice, but the phenotypes were improved in cON mice. cOFF mice also showed enhanced cortical gelatinase activity and baseline EEG gamma power, which were reduced in cON mice. In addition, TrkB phosphorylation and PV levels were lower in cOFF mice, which also showed increased locomotor activity and anxiety-like behaviors. Remarkably, when FMRP levels were restored in only excitatory neurons during the P14-P21 period, TrkB phosphorylation and mouse behaviors were also improved. CONCLUSIONS These results indicate that postnatal deletion or re-expression of FMRP in excitatory neurons is sufficient to elicit or ameliorate structural and functional cortical deficits, and abnormal behaviors in mice, informing future studies about appropriate treatment windows and providing fundamental insights into the cellular mechanisms of cortical circuit dysfunction in FXS.
Collapse
Affiliation(s)
- Maham Rais
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Jonathan W Lovelace
- Department of Psychology, University of California Riverside, Riverside, CA 92521, USA
| | - Xinghao S Shuai
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Walker Woodard
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Steven Bishay
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Leo Estrada
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Ashwin R Sharma
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Austin Nguy
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Anna Kulinich
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Patricia S Pirbhoy
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Arnold R Palacios
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | | | - Khaleel A Razak
- Department of Psychology, University of California Riverside, Riverside, CA 92521, USA; Neuroscience Graduate Program, University of California Riverside, Riverside, CA 92521, USA
| | - Iryna M Ethell
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, CA 92521, USA; Neuroscience Graduate Program, University of California Riverside, Riverside, CA 92521, USA.
| |
Collapse
|
21
|
Contractor A, Ethell IM, Portera-Cailliau C. Cortical interneurons in autism. Nat Neurosci 2021; 24:1648-1659. [PMID: 34848882 PMCID: PMC9798607 DOI: 10.1038/s41593-021-00967-6] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 09/21/2021] [Indexed: 01/01/2023]
Abstract
The mechanistic underpinnings of autism remain a subject of debate and controversy. Why do individuals with autism share an overlapping set of atypical behaviors and symptoms, despite having different genetic and environmental risk factors? A major challenge in developing new therapies for autism has been the inability to identify convergent neural phenotypes that could explain the common set of symptoms that result in the diagnosis. Although no striking macroscopic neuropathological changes have been identified in autism, there is growing evidence that inhibitory interneurons (INs) play an important role in its neural basis. In this Review, we evaluate and interpret this evidence, focusing on recent findings showing reduced density and activity of the parvalbumin class of INs. We discuss the need for additional studies that investigate how genes and the environment interact to change the developmental trajectory of INs, permanently altering their numbers, connectivity and circuit engagement.
Collapse
Affiliation(s)
- Anis Contractor
- Department of Neuroscience Feinberg School of Medicine, Weinberg College of Arts and Sciences, Northwestern University, Chicago, IL, USA
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Chicago, IL, USA
| | - Iryna M Ethell
- Division of Biomedical Sciences, UC Riverside School of Medicine, Riverside, CA, USA
| | - Carlos Portera-Cailliau
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
22
|
Razak KA, Binder DK, Ethell IM. Neural Correlates of Auditory Hypersensitivity in Fragile X Syndrome. Front Psychiatry 2021; 12:720752. [PMID: 34690832 PMCID: PMC8529206 DOI: 10.3389/fpsyt.2021.720752] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/16/2021] [Indexed: 01/20/2023] Open
Abstract
The mechanisms underlying the common association between autism spectrum disorders (ASD) and sensory processing disorders (SPD) are unclear, and treatment options to reduce atypical sensory processing are limited. Fragile X Syndrome (FXS) is a leading genetic cause of intellectual disability and ASD behaviors. As in most children with ASD, atypical sensory processing is a common symptom in FXS, frequently manifesting as sensory hypersensitivity. Auditory hypersensitivity is a highly debilitating condition in FXS that may lead to language delays, social anxiety and ritualized repetitive behaviors. Animal models of FXS, including Fmr1 knock out (KO) mouse, also show auditory hypersensitivity, providing a translation relevant platform to study underlying pathophysiological mechanisms. The focus of this review is to summarize recent studies in the Fmr1 KO mouse that identified neural correlates of auditory hypersensitivity. We review results of electroencephalography (EEG) recordings in the Fmr1 KO mice and highlight EEG phenotypes that are remarkably similar to EEG findings in humans with FXS. The EEG phenotypes associated with the loss of FMRP include enhanced resting EEG gamma band power, reduced cross frequency coupling, reduced sound-evoked synchrony of neural responses at gamma band frequencies, increased event-related potential amplitudes, reduced habituation of neural responses and increased non-phase locked power. In addition, we highlight the postnatal period when the EEG phenotypes develop and show a strong association of the phenotypes with enhanced matrix-metalloproteinase-9 (MMP-9) activity, abnormal development of parvalbumin (PV)-expressing inhibitory interneurons and reduced formation of specialized extracellular matrix structures called perineuronal nets (PNNs). Finally, we discuss how dysfunctions of inhibitory PV interneurons may contribute to cortical hyperexcitability and EEG abnormalities observed in FXS. Taken together, the studies reviewed here indicate that EEG recordings can be utilized in both pre-clinical studies and clinical trials, while at the same time, used to identify cellular and circuit mechanisms of dysfunction in FXS. New therapeutic approaches that reduce MMP-9 activity and restore functions of PV interneurons may succeed in reducing FXS sensory symptoms. Future studies should examine long-lasting benefits of developmental vs. adult interventions on sensory phenotypes.
Collapse
Affiliation(s)
- Khaleel A. Razak
- Department of Psychology, University of California, Riverside, Riverside, CA, United States
- Graduate Neuroscience Program, University of California, Riverside, Riverside, CA, United States
| | - Devin K. Binder
- Graduate Neuroscience Program, University of California, Riverside, Riverside, CA, United States
- Division of Biomedical Sciences and Graduate Biomedical Sciences Program, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Iryna M. Ethell
- Graduate Neuroscience Program, University of California, Riverside, Riverside, CA, United States
- Division of Biomedical Sciences and Graduate Biomedical Sciences Program, School of Medicine, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
23
|
Nomura T. Interneuron Dysfunction and Inhibitory Deficits in Autism and Fragile X Syndrome. Cells 2021; 10:cells10102610. [PMID: 34685590 PMCID: PMC8534049 DOI: 10.3390/cells10102610] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 01/18/2023] Open
Abstract
The alteration of excitatory–inhibitory (E–I) balance has been implicated in various neurological and psychiatric diseases, including autism spectrum disorder (ASD). Fragile X syndrome (FXS) is a single-gene disorder that is the most common known cause of ASD. Understanding the molecular and physiological features of FXS is thought to enhance our knowledge of the pathophysiology of ASD. Accumulated evidence implicates deficits in the inhibitory circuits in FXS that tips E–I balance toward excitation. Deficits in interneurons, the main source of an inhibitory neurotransmitter, gamma-aminobutyric acid (GABA), have been reported in FXS, including a reduced number of cells, reduction in intrinsic cellular excitability, or weaker synaptic connectivity. Manipulating the interneuron activity ameliorated the symptoms in the FXS mouse model, which makes it reasonable to conceptualize FXS as an interneuronopathy. While it is still poorly understood how the developmental profiles of the inhibitory circuit go awry in FXS, recent works have uncovered several developmental alterations in the functional properties of interneurons. Correcting disrupted E–I balance by potentiating the inhibitory circuit by targeting interneurons may have a therapeutic potential in FXS. I will review the recent evidence about the inhibitory alterations and interneuron dysfunction in ASD and FXS and will discuss the future directions of this field.
Collapse
Affiliation(s)
- Toshihiro Nomura
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
24
|
Delhaye S, Bardoni B. Role of phosphodiesterases in the pathophysiology of neurodevelopmental disorders. Mol Psychiatry 2021; 26:4570-4582. [PMID: 33414502 PMCID: PMC8589663 DOI: 10.1038/s41380-020-00997-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 12/03/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022]
Abstract
Phosphodiesterases (PDEs) are enzymes involved in the homeostasis of both cAMP and cGMP. They are members of a family of proteins that includes 11 subfamilies with different substrate specificities. Their main function is to catalyze the hydrolysis of cAMP, cGMP, or both. cAMP and cGMP are two key second messengers that modulate a wide array of intracellular processes and neurobehavioral functions, including memory and cognition. Even if these enzymes are present in all tissues, we focused on those PDEs that are expressed in the brain. We took into consideration genetic variants in patients affected by neurodevelopmental disorders, phenotypes of animal models, and pharmacological effects of PDE inhibitors, a class of drugs in rapid evolution and increasing application to brain disorders. Collectively, these data indicate the potential of PDE modulators to treat neurodevelopmental diseases characterized by learning and memory impairment, alteration of behaviors associated with depression, and deficits in social interaction. Indeed, clinical trials are in progress to treat patients with Alzheimer's disease, schizophrenia, depression, and autism spectrum disorders. Among the most recent results, the application of some PDE inhibitors (PDE2A, PDE3, PDE4/4D, and PDE10A) to treat neurodevelopmental diseases, including autism spectrum disorders and intellectual disability, is a significant advance, since no specific therapies are available for these disorders that have a large prevalence. In addition, to highlight the role of several PDEs in normal and pathological neurodevelopment, we focused here on the deregulation of cAMP and/or cGMP in Down Syndrome, Fragile X Syndrome, Rett Syndrome, and intellectual disability associated with the CC2D1A gene.
Collapse
Affiliation(s)
- Sébastien Delhaye
- grid.429194.30000 0004 0638 0649Université Côte d’Azur, CNRS UMR7275, Institute of Molecular and Cellular Pharmacology, 06560 Valbonne, France
| | - Barbara Bardoni
- Université Côte d'Azur, Inserm, CNRS UMR7275, Institute of Molecular and Cellular Pharmacology, 06560, Valbonne, France.
| |
Collapse
|
25
|
Cheffer A, Flitsch LJ, Krutenko T, Röderer P, Sokhranyaeva L, Iefremova V, Hajo M, Peitz M, Schwarz MK, Brüstle O. Human stem cell-based models for studying autism spectrum disorder-related neuronal dysfunction. Mol Autism 2020; 11:99. [PMID: 33308283 PMCID: PMC7733257 DOI: 10.1186/s13229-020-00383-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
The controlled differentiation of pluripotent stem cells (PSCs) into neurons and glia offers a unique opportunity to study early stages of human central nervous system development under controlled conditions in vitro. With the advent of cell reprogramming and the possibility to generate induced pluripotent stem cells (iPSCs) from any individual in a scalable manner, these studies can be extended to a disease- and patient-specific level. Autism spectrum disorder (ASD) is considered a neurodevelopmental disorder, with substantial evidence pointing to early alterations in neurogenesis and network formation as key pathogenic drivers. For that reason, ASD represents an ideal candidate for stem cell-based disease modeling. Here, we provide a concise review on recent advances in the field of human iPSC-based modeling of syndromic and non-syndromic forms of ASD, with a particular focus on studies addressing neuronal dysfunction and altered connectivity. We further discuss recent efforts to translate stem cell-based disease modeling to 3D via brain organoid and cell transplantation approaches, which enable the investigation of disease mechanisms in a tissue-like context. Finally, we describe advanced tools facilitating the assessment of altered neuronal function, comment on the relevance of iPSC-based models for the assessment of pharmaceutical therapies and outline potential future routes in stem cell-based ASD research.
Collapse
Affiliation(s)
- Arquimedes Cheffer
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Lea Jessica Flitsch
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Tamara Krutenko
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Pascal Röderer
- Life & Brain GmbH, Platform Cellomics, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Liubov Sokhranyaeva
- Institute of Experimental Epileptology and Cognition Research, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Vira Iefremova
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Mohamad Hajo
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Michael Peitz
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
- Life & Brain GmbH, Platform Cellomics, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
- Cell Programming Core Facility, University of Bonn Medical Faculty, Bonn, Germany
| | - Martin Karl Schwarz
- Life & Brain GmbH, Platform Cellomics, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
- Institute of Experimental Epileptology and Cognition Research, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty & University Hospital Bonn, Venusberg-Campus 1, Building 76, 53127, Bonn, Germany.
| |
Collapse
|
26
|
Adams I, Yang T, Longo FM, Katz DM. Restoration of motor learning in a mouse model of Rett syndrome following long-term treatment with a novel small-molecule activator of TrkB. Dis Model Mech 2020; 13:13/11/dmm044685. [PMID: 33361117 PMCID: PMC7710018 DOI: 10.1242/dmm.044685] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 09/23/2020] [Indexed: 12/23/2022] Open
Abstract
Reduced expression of brain-derived neurotrophic factor (BDNF) and impaired activation of the BDNF receptor, tropomyosin receptor kinase B (TrkB; also known as Ntrk2), are thought to contribute significantly to the pathophysiology of Rett syndrome (RTT), a severe neurodevelopmental disorder caused by loss-of-function mutations in the X-linked gene encoding methyl-CpG-binding protein 2 (MeCP2). Previous studies from this and other laboratories have shown that enhancing BDNF expression and/or TrkB activation in Mecp2-deficient mouse models of RTT can ameliorate or reverse abnormal neurological phenotypes that mimic human RTT symptoms. The present study reports on the preclinical efficacy of a novel, small-molecule, non-peptide TrkB partial agonist, PTX-BD4-3, in heterozygous female Mecp2 mutant mice, a well-established RTT model that recapitulates the genetic mosaicism of the human disease. PTX-BD4-3 exhibited specificity for TrkB in cell-based assays of neurotrophin receptor activation and neuronal cell survival and in in vitro receptor binding assays. PTX-BD4-3 also activated TrkB following systemic administration to wild-type and Mecp2 mutant mice and was rapidly cleared from the brain and plasma with a half-life of ∼2 h. Chronic intermittent treatment of Mecp2 mutants with a low dose of PTX-BD4-3 (5 mg/kg, intraperitoneally, once every 3 days for 8 weeks) reversed deficits in two core RTT symptom domains – respiration and motor control – and symptom rescue was maintained for at least 24 h after the last dose. Together, these data indicate that significant clinically relevant benefit can be achieved in a mouse model of RTT with a chronic intermittent, low-dose treatment paradigm targeting the neurotrophin receptor TrkB. Editor's choice: Long-term intermittent treatment with a newly developed partial agonist of the TrkB neurotrophin receptor reverses deficits in motor learning and respiration in a mouse model of Rett syndrome.
Collapse
Affiliation(s)
- Ian Adams
- Department of Neurosciences, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106-4975, USA
| | - Tao Yang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Frank M Longo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David M Katz
- Department of Neurosciences, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106-4975, USA
| |
Collapse
|
27
|
Cogram P, Alkon DL, Crockford D, Deacon RMJ, Hurley MJ, Altimiras F, Sun MK, Tranfaglia M. Chronic bryostatin-1 rescues autistic and cognitive phenotypes in the fragile X mice. Sci Rep 2020; 10:18058. [PMID: 33093534 PMCID: PMC7581799 DOI: 10.1038/s41598-020-74848-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 10/05/2020] [Indexed: 12/20/2022] Open
Abstract
Fragile X syndrome (FXS), an X-chromosome linked intellectual disability, is the leading monogenetic cause of autism spectrum disorder (ASD), a neurodevelopmental condition that currently has no specific drug treatment. Building upon the demonstrated therapeutic effects on spatial memory of bryostatin-1, a relatively specific activator of protein kinase C (PKC)ε, (also of PKCα) on impaired synaptic plasticity/maturation and spatial learning and memory in FXS mice, we investigated whether bryostatin-1 might affect the autistic phenotypes and other behaviors, including open field activity, activities of daily living (nesting and marble burying), at the effective therapeutic dose for spatial memory deficits. Further evaluation included other non-spatial learning and memory tasks. Interestingly, a short period of treatment (5 weeks) only produced very limited or no therapeutic effects on the autistic and cognitive phenotypes in the Fmr1 KO2 mice, while a longer treatment (13 weeks) with the same dose of bryostatin-1 effectively rescued the autistic and non-spatial learning deficit cognitive phenotypes. It is possible that longer-term treatment would result in further improvement in these fragile X phenotypes. This effect is clearly different from other treatment strategies tested to date, in that the drug shows little acute effect, but strong long-term effects. It also shows no evidence of tolerance, which has been a problem with other drug classes (mGluR5 antagonists, GABA-A and -B agonists). The results strongly suggest that, at appropriate dosing and therapeutic period, chronic bryostatin-1 may have great therapeutic value for both ASD and FXS.
Collapse
Affiliation(s)
- Patricia Cogram
- FRAXA-DVI, FRAXA, Santiago, Chile. .,IEB, Faculty of Science, University of Chile, Santiago, Chile.
| | | | | | - Robert M J Deacon
- FRAXA-DVI, FRAXA, Santiago, Chile.,IEB, Faculty of Science, University of Chile, Santiago, Chile
| | - Michael J Hurley
- Neuroimmunology, Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | - Francisco Altimiras
- Faculty of Engineering, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile.,Faculty of Engineering and Business, Universidad de las Américas, Santiago, Chile
| | | | | |
Collapse
|
28
|
Gilbert ME, O'Shaughnessy KL, Axelstad M. Regulation of Thyroid-disrupting Chemicals to Protect the Developing Brain. Endocrinology 2020; 161:bqaa106. [PMID: 32615585 PMCID: PMC8650774 DOI: 10.1210/endocr/bqaa106] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/30/2020] [Indexed: 12/18/2022]
Abstract
Synthetic chemicals with endocrine disrupting properties are pervasive in the environment and are present in the bodies of humans and wildlife. As thyroid hormones (THs) control normal brain development, and maternal hypothyroxinemia is associated with neurological impairments in children, chemicals that interfere with TH signaling are of considerable concern for children's health. However, identifying thyroid-disrupting chemicals (TDCs) in vivo is largely based on measuring serum tetraiodothyronine in rats, which may be inadequate to assess TDCs with disparate mechanisms of action and insufficient to evaluate the potential neurotoxicity of TDCs. In this review 2 neurodevelopmental processes that are dependent on TH action are highlighted, neuronal migration and maturation of gamma amino butyric acid-ergic interneurons. We discuss how interruption of these processes by TDCs may contribute to abnormal brain circuitry following developmental TH insufficiency. Finally, we identify issues in evaluating the developmental neurotoxicity of TDCs and the strengths and limitations of current approaches designed to regulate them. It is clear that an enhanced understanding of how THs affect brain development will lead to refined toxicity testing, reducing uncertainty and improving our ability to protect children's health.
Collapse
Affiliation(s)
- Mary E Gilbert
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Research Triangle Park, North Carolina
| | - Katherine L O'Shaughnessy
- Center for Public Health and Environmental Assessment, US Environmental Protection Agency, Research Triangle Park, North Carolina
| | - Marta Axelstad
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Kgs. Lyngby, Denmark
| |
Collapse
|
29
|
Lovelace JW, Ethell IM, Binder DK, Razak KA. Minocycline Treatment Reverses Sound Evoked EEG Abnormalities in a Mouse Model of Fragile X Syndrome. Front Neurosci 2020; 14:771. [PMID: 32848552 PMCID: PMC7417521 DOI: 10.3389/fnins.2020.00771] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/30/2020] [Indexed: 01/19/2023] Open
Abstract
Fragile X Syndrome (FXS) is a leading known genetic cause of intellectual disability. Many symptoms of FXS overlap with those in autism including repetitive behaviors, language delays, anxiety, social impairments and sensory processing deficits. Electroencephalogram (EEG) recordings from humans with FXS and an animal model, the Fmr1 knockout (KO) mouse, show remarkably similar phenotypes suggesting that EEG phenotypes can serve as biomarkers for developing treatments. This includes enhanced resting gamma band power and sound evoked total power, and reduced fidelity of temporal processing and habituation of responses to repeated sounds. Given the therapeutic potential of the antibiotic minocycline in humans with FXS and animal models, it is important to determine sensitivity and selectivity of EEG responses to minocycline. Therefore, in this study, we examined if a 10-day treatment of adult Fmr1 KO mice with minocycline (oral gavage, 30 mg/kg per day) would reduce EEG abnormalities. We tested if minocycline treatment has specific effects based on the EEG measurement type (e.g., resting versus sound-evoked) from the frontal and auditory cortex of the Fmr1 KO mice. We show increased resting EEG gamma power and reduced phase locking to time varying stimuli as well as the 40 Hz auditory steady state response in the Fmr1 KO mice in the pre-drug condition. Minocycline treatment increased gamma band phase locking in response to auditory stimuli, and reduced sound-evoked power of auditory event related potentials (ERP) in Fmr1 KO mice compared to vehicle treatment. Minocycline reduced resting EEG gamma power in Fmr1 KO mice, but this effect was similar to vehicle treatment. We also report frequency band-specific effects on EEG responses. Taken together, these data indicate that sound-evoked EEG responses may serve as more sensitive measures, compared to resting EEG measures, to isolate minocycline effects from placebo in humans with FXS. Given the use of minocycline and EEG recordings in a number of neurodegenerative and neurodevelopmental conditions, these findings may be more broadly applicable in translational neuroscience.
Collapse
Affiliation(s)
- Jonathan W Lovelace
- Department of Psychology and Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States
| | - Iryna M Ethell
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States.,Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Devin K Binder
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States.,Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Khaleel A Razak
- Department of Psychology and Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States.,Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
30
|
Kipnis PA, Sullivan BJ, Carter BM, Kadam SD. TrkB agonists prevent postischemic emergence of refractory neonatal seizures in mice. JCI Insight 2020; 5:136007. [PMID: 32427585 DOI: 10.1172/jci.insight.136007] [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: 01/06/2020] [Accepted: 05/14/2020] [Indexed: 12/19/2022] Open
Abstract
Refractory neonatal seizures do not respond to first-line antiseizure medications like phenobarbital (PB), a positive allosteric modulator for GABAA receptors. GABAA receptor-mediated inhibition is dependent upon electroneutral cation-chloride transporter KCC2, which mediates neuronal chloride extrusion and its age-dependent increase and postnatally shifts GABAergic signaling from depolarizing to hyperpolarizing. Brain-derived neurotropic factor-tyrosine receptor kinase B activation (BDNF-TrkB activation) after excitotoxic injury recruits downstream targets like PLCγ1, leading to KCC2 hypofunction. Here, the antiseizure efficacy of TrkB agonists LM22A-4, HIOC, and deoxygedunin (DG) on PB-refractory seizures and postischemic TrkB pathway activation was investigated in a mouse model (CD-1, P7) of refractory neonatal seizures. LM, a BDNF loop II mimetic, rescued PB-refractory seizures in a sexually dimorphic manner. Efficacy was associated with a substantial reduction in the postischemic phosphorylation of TrkB at Y816, a site known to mediate postischemic KCC2 hypofunction via PLCγ1 activation. LM rescued ischemia-induced phospho-KCC2-S940 dephosphorylation, preserving its membrane stability. Full TrkB agonists HIOC and DG similarly rescued PB refractoriness. Chemogenetic inactivation of TrkB substantially reduced postischemic neonatal seizure burdens at P7. Sex differences identified in developmental expression profiles of TrkB and KCC2 may underlie the sexually dimorphic efficacy of LM. These results support a potentially novel role for the TrkB receptor in the emergence of age-dependent refractory neonatal seizures.
Collapse
Affiliation(s)
- Pavel A Kipnis
- Neuroscience Laboratory, Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Brennan J Sullivan
- Neuroscience Laboratory, Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Brandon M Carter
- Neuroscience Laboratory, Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Shilpa D Kadam
- Neuroscience Laboratory, Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, Maryland, USA.,Department of Neurology and Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
31
|
Pirbhoy PS, Rais M, Lovelace JW, Woodard W, Razak KA, Binder DK, Ethell IM. Acute pharmacological inhibition of matrix metalloproteinase-9 activity during development restores perineuronal net formation and normalizes auditory processing in Fmr1 KO mice. J Neurochem 2020; 155:538-558. [PMID: 32374912 DOI: 10.1111/jnc.15037] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/31/2020] [Accepted: 04/30/2020] [Indexed: 12/13/2022]
Abstract
Individuals with Fragile X Syndrome (FXS) and autism spectrum disorder (ASD) exhibit cognitive impairments, social deficits, increased anxiety, and sensory hyperexcitability. Previously, we showed that elevated levels of matrix metalloproteinase-9 (MMP-9) may contribute to abnormal development of parvalbumin (PV) interneurons and perineuronal nets (PNNs) in the developing auditory cortex (AC) of Fmr1 knock-out (KO) mice, which likely underlie auditory hypersensitivity. Thus, MMP-9 may serve as a potential target for treatment of auditory hypersensitivity in FXS. Here, we used the MMP-2/9 inhibitor, SB-3CT, to pharmacologically inhibit MMP-9 activity during a specific developmental period and to test whether inhibition of MMP-9 activity reverses neural oscillation deficits and behavioral impairments by enhancing PNN formation around PV cells in Fmr1 KO mice. Electroencephalography (EEG) was used to measure resting state and sound-evoked electrocortical activity in auditory and frontal cortices of postnatal day (P)22-23 male mice before and one-day after treatment with SB-3CT (25 mg/kg) or vehicle. At P27-28, animal behaviors were tested to measure the effects of the treatment on anxiety and hyperactivity. Results show that acute inhibition of MMP-9 activity improved evoked synchronization to auditory stimuli and ameliorated mouse behavioral deficits. MMP-9 inhibition enhanced PNN formation, increased PV levels and TrkB phosphorylation yet reduced Akt phosphorylation in the AC of Fmr1 KO mice. Our results show that MMP-9 inhibition during early postnatal development is beneficial in reducing some auditory processing deficits in the FXS mouse model and may serve as a candidate therapeutic for reversing sensory hypersensitivity in FXS and possibly other ASDs.
Collapse
Affiliation(s)
- Patricia S Pirbhoy
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Maham Rais
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Jonathan W Lovelace
- Department of Psychology, University of California Riverside, Riverside, CA, USA
| | - Walker Woodard
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Khaleel A Razak
- Department of Psychology, University of California Riverside, Riverside, CA, USA
| | - Devin K Binder
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Iryna M Ethell
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| |
Collapse
|
32
|
Razak KA, Dominick KC, Erickson CA. Developmental studies in fragile X syndrome. J Neurodev Disord 2020; 12:13. [PMID: 32359368 PMCID: PMC7196229 DOI: 10.1186/s11689-020-09310-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 02/13/2020] [Indexed: 01/27/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common single gene cause of autism and intellectual disabilities. Humans with FXS exhibit increased anxiety, sensory hypersensitivity, seizures, repetitive behaviors, cognitive inflexibility, and social behavioral impairments. The main purpose of this review is to summarize developmental studies of FXS in humans and in the mouse model, the Fmr1 knockout mouse. The literature presents considerable evidence that a number of early developmental deficits can be identified and that these early deficits chart a course of altered developmental experience leading to symptoms well characterized in adolescents and adults. Nevertheless, a number of critical issues remain unclear or untested regarding the development of symptomology and underlying mechanisms. First, what is the role of FMRP, the protein product of Fmr1 gene, during different developmental ages? Does the absence of FMRP during early development lead to irreversible changes, or could reintroduction of FMRP or therapeutics aimed at FMRP-interacting proteins/pathways hold promise when provided in adults? These questions have implications for clinical trial designs in terms of optimal treatment windows, but few studies have systematically addressed these issues in preclinical and clinical work. Published studies also point to complex trajectories of symptom development, leading to the conclusion that single developmental time point studies are unlikely to disambiguate effects of genetic mutation from effects of altered developmental experience and compensatory plasticity. We conclude by suggesting a number of experiments needed to address these major gaps in the field.
Collapse
Affiliation(s)
- Khaleel A Razak
- Department of Psychology and Graduate Neuroscience Program, University of California, Riverside, USA
| | - Kelli C Dominick
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA.,Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 4002, Cincinnati, OH, 45229, USA
| | - Craig A Erickson
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA. .,Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue MLC 4002, Cincinnati, OH, 45229, USA.
| |
Collapse
|
33
|
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
|
34
|
Kulinich AO, Reinhard SM, Rais M, Lovelace JW, Scott V, Binder DK, Razak KA, Ethell IM. Beneficial effects of sound exposure on auditory cortex development in a mouse model of Fragile X Syndrome. Neurobiol Dis 2020; 134:104622. [PMID: 31698054 DOI: 10.1016/j.nbd.2019.104622] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/22/2019] [Accepted: 09/23/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is the most common genetic cause of autism and intellectual disability. Fragile X mental retardation gene (Fmr1) knock-out (KO) mice display core deficits of FXS, including abnormally increased sound-evoked responses, and show a delayed development of parvalbumin (PV) cells. Here, we present the surprising result that sound exposure during early development reduces correlates of auditory hypersensitivity in Fmr1 KO mice. METHODS Fmr1 KO and wild-type (WT) mice were raised in a sound-attenuated environment (AE) or sound-exposed (SE) to 14 kHz tones (5 Hz repetition rate) from P9 until P21. At P21-P23, event-related potentials (ERPs), dendritic spine density, PV expression and phosphorylation of tropomyosin receptor kinase B (TrkB) were analyzed in the auditory cortex of AE and SE mice. RESULTS Enhanced N1 amplitude of ERPs, impaired PV cell development, and increased spine density in layers (L) 2/3 and L5/6 excitatory neurons were observed in AE Fmr1 KO compared to WT mice. In contrast, developmental sound exposure normalized ERP N1 amplitude, density of PV cells and dendritic spines in SE Fmr1 KO mice. Finally, TrkB phosphorylation was reduced in AE Fmr1 KO, but was enhanced in SE Fmr1 KO mice, suggesting that BDNF-TrkB signaling may be regulated by sound exposure to influence PV cell development. CONCLUSIONS Our results demonstrate that sound exposure, but not attenuation, during early developmental window restores molecular, cellular and functional properties in the auditory cortex of Fmr1 KO mice, and suggest this approach as a potential treatment for sensory phenotypes in FXS.
Collapse
Affiliation(s)
- Anna O Kulinich
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Sarah M Reinhard
- Psychology Department, University of California, Riverside, CA, USA
| | - Maham Rais
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA; Biomedical Sciences Graduate Program, University of California, Riverside, CA, USA
| | | | - Veronica Scott
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA
| | - Devin K Binder
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA; Neuroscience Graduate Program, University of California, Riverside, CA, USA; Biomedical Sciences Graduate Program, University of California, Riverside, CA, USA
| | - Khaleel A Razak
- Psychology Department, University of California, Riverside, CA, USA; Neuroscience Graduate Program, University of California, Riverside, CA, USA; Biomedical Sciences Graduate Program, University of California, Riverside, CA, USA.
| | - Iryna M Ethell
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA, USA; Neuroscience Graduate Program, University of California, Riverside, CA, USA; Biomedical Sciences Graduate Program, University of California, Riverside, CA, USA.
| |
Collapse
|
35
|
Sullivan BJ, Kadam SD. The involvement of neuronal chloride transporter deficiencies in epilepsy. NEURONAL CHLORIDE TRANSPORTERS IN HEALTH AND DISEASE 2020:329-366. [DOI: 10.1016/b978-0-12-815318-5.00014-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
36
|
He Q, Arroyo ED, Smukowski SN, Xu J, Piochon C, Savas JN, Portera-Cailliau C, Contractor A. Critical period inhibition of NKCC1 rectifies synapse plasticity in the somatosensory cortex and restores adult tactile response maps in fragile X mice. Mol Psychiatry 2019; 24:1732-1747. [PMID: 29703945 PMCID: PMC6204122 DOI: 10.1038/s41380-018-0048-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 01/10/2018] [Accepted: 02/19/2018] [Indexed: 12/15/2022]
Abstract
Sensory perturbations in visual, auditory and tactile perception are core problems in fragile X syndrome (FXS). In the Fmr1 knockout mouse model of FXS, the maturation of synapses and circuits during critical period (CP) development in the somatosensory cortex is delayed, but it is unclear how this contributes to altered tactile sensory processing in the mature CNS. Here we demonstrate that inhibiting the juvenile chloride co-transporter NKCC1, which contributes to altered chloride homeostasis in developing cortical neurons of FXS mice, rectifies the chloride imbalance in layer IV somatosensory cortex neurons and corrects the development of thalamocortical excitatory synapses during the CP. Comparison of protein abundances demonstrated that NKCC1 inhibition during early development caused a broad remodeling of the proteome in the barrel cortex. In addition, the abnormally large size of whisker-evoked cortical maps in adult Fmr1 knockout mice was corrected by rectifying the chloride imbalance during the early CP. These data demonstrate that correcting the disrupted driving force through GABAA receptors during the CP in cortical neurons restores their synaptic development, has an unexpectedly large effect on differentially expressed proteins, and produces a long-lasting correction of somatosensory circuit function in FXS mice.
Collapse
Affiliation(s)
- Qionger He
- Department of Physiology, Northwestern University Feinberg School of Medicine
| | - Erica D Arroyo
- Departments of Neurology and Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095
| | - Samuel N Smukowski
- Department of Neurology, Northwestern University Feinberg School of Medicine
| | - Jian Xu
- Department of Physiology, Northwestern University Feinberg School of Medicine
| | - Claire Piochon
- Department of Physiology, Northwestern University Feinberg School of Medicine
| | - Jeffrey N Savas
- Department of Neurology, Northwestern University Feinberg School of Medicine
| | - Carlos Portera-Cailliau
- Departments of Neurology and Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095
| | - Anis Contractor
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA. .,Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
37
|
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder that causes intellectual disability. It is a leading known genetic cause of autism. In addition to cognitive, social, and communication deficits, humans with FXS demonstrate abnormal sensory processing including sensory hypersensitivity. Sensory hypersensitivity commonly manifests as auditory, tactile, or visual defensiveness or avoidance. Clinical, behavioral, and electrophysiological studies consistently show auditory hypersensitivity, impaired habituation to repeated sounds, and reduced auditory attention in humans with FXS. Children with FXS also exhibit significant visuospatial impairments. Studies in infants and toddlers with FXS have documented impairments in processing texture-defined motion stimuli, temporal flicker, perceiving ordinal numerical sequence, and the ability to maintain the identity of dynamic object information during occlusion. Consistent with the observations in humans with FXS, fragile X mental retardation 1 ( Fmr1) gene knockout (KO) rodent models of FXS also show seizures, abnormal visual-evoked responses, auditory hypersensitivity, and abnormal processing at multiple levels of the auditory system, including altered acoustic startle responses. Among other sensory symptoms, individuals with FXS exhibit tactile defensiveness. Fmr1 KO mice also show impaired encoding of tactile stimulation frequency and larger size of receptive fields in the somatosensory cortex. Since sensory deficits are relatively more tractable from circuit mechanisms and developmental perspectives than more complex social behaviors, the focus of this review is on clinical, functional, and structural studies that outline the auditory, visual, and somatosensory processing deficits in FXS. The similarities in sensory phenotypes between humans with FXS and animal models suggest a likely conservation of basic sensory processing circuits across species and may provide a translational platform to not just develop biomarkers but also to understand underlying mechanisms. We argue that preclinical studies in animal models of FXS can facilitate the ongoing search for new therapeutic approaches in FXS by understanding mechanisms of basic sensory processing circuits and behaviors that are conserved across species.
Collapse
Affiliation(s)
- Maham Rais
- 1 Division of Biomedical Sciences, University of California Riverside School of Medicine, CA, USA.,2 Biomedical Sciences Graduate Program, University of California Riverside, CA, USA
| | - Devin K Binder
- 1 Division of Biomedical Sciences, University of California Riverside School of Medicine, CA, USA.,2 Biomedical Sciences Graduate Program, University of California Riverside, CA, USA.,3 Neuroscience Graduate Program, University of California Riverside, CA, USA
| | - Khaleel A Razak
- 2 Biomedical Sciences Graduate Program, University of California Riverside, CA, USA.,3 Neuroscience Graduate Program, University of California Riverside, CA, USA.,4 Psychology Department, University of California Riverside, CA, USA
| | - Iryna M Ethell
- 1 Division of Biomedical Sciences, University of California Riverside School of Medicine, CA, USA.,2 Biomedical Sciences Graduate Program, University of California Riverside, CA, USA.,3 Neuroscience Graduate Program, University of California Riverside, CA, USA
| |
Collapse
|
38
|
Bagni C, Zukin RS. A Synaptic Perspective of Fragile X Syndrome and Autism Spectrum Disorders. Neuron 2019; 101:1070-1088. [PMID: 30897358 PMCID: PMC9628679 DOI: 10.1016/j.neuron.2019.02.041] [Citation(s) in RCA: 216] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/25/2019] [Accepted: 02/27/2019] [Indexed: 12/28/2022]
Abstract
Altered synaptic structure and function is a major hallmark of fragile X syndrome (FXS), autism spectrum disorders (ASDs), and other intellectual disabilities (IDs), which are therefore classified as synaptopathies. FXS and ASDs, while clinically and genetically distinct, share significant comorbidity, suggesting that there may be a common molecular and/or cellular basis, presumably at the synapse. In this article, we review brain architecture and synaptic pathways that are dysregulated in FXS and ASDs, including spine architecture, signaling in synaptic plasticity, local protein synthesis, (m)RNA modifications, and degradation. mRNA repression is a powerful mechanism for the regulation of synaptic structure and efficacy. We infer that there is no single pathway that explains most of the etiology and discuss new findings and the implications for future work directed at improving our understanding of the pathogenesis of FXS and related ASDs and the design of therapeutic strategies to ameliorate these disorders.
Collapse
Affiliation(s)
- Claudia Bagni
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland; Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy.
| | - R Suzanne Zukin
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York City, NY, USA.
| |
Collapse
|
39
|
Telias M. Molecular Mechanisms of Synaptic Dysregulation in Fragile X Syndrome and Autism Spectrum Disorders. Front Mol Neurosci 2019; 12:51. [PMID: 30899214 PMCID: PMC6417395 DOI: 10.3389/fnmol.2019.00051] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 02/12/2019] [Indexed: 12/21/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common form of monogenic hereditary cognitive impairment. FXS patient exhibit a high comorbidity rate with autism spectrum disorders (ASDs). This makes FXS a model disease for understanding how synaptic dysregulation alters neuronal excitability, learning and memory, social behavior, and more. Since 1991, with the discovery of fragile X mental retardation 1 (FMR1) as the sole gene that is mutated in FXS, thousands of studies into the function of the gene and its encoded protein FMR1 protein (FMRP), have been conducted, yielding important information regarding the pathophysiology of the disease, as well as insight into basic synaptic mechanisms that control neuronal networking and circuitry. Among the most important, are molecular mechanisms directly involved in plasticity, including glutamate and γ-aminobutyric acid (GABA) receptors, which can control synaptic transmission and signal transduction, including short- and long-term plasticity. More recently, several novel mechanisms involving growth factors, enzymatic cascades and transcription factors (TFs), have been proposed to have the potential of explaining some of the synaptic dysregulation in FXS. In this review article, I summarize the main mechanisms proposed to underlie synaptic disruption in FXS and ASDs. I focus on studies conducted on the Fmr1 knock-out (KO) mouse model and on FXS-human pluripotent stem cells (hPSCs), emphasizing the differences and even contradictions between mouse and human, whenever possible. As FXS and ASDs are both neurodevelopmental disorders that follow a specific time-course of disease progression, I highlight those studies focusing on the differential developmental regulation of synaptic abnormalities in these diseases.
Collapse
Affiliation(s)
- Michael Telias
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| |
Collapse
|
40
|
Wen TH, Lovelace JW, Ethell IM, Binder DK, Razak KA. Developmental Changes in EEG Phenotypes in a Mouse Model of Fragile X Syndrome. Neuroscience 2018; 398:126-143. [PMID: 30528856 DOI: 10.1016/j.neuroscience.2018.11.047] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 11/27/2018] [Accepted: 11/29/2018] [Indexed: 01/29/2023]
Abstract
Fragile X Syndrome (FXS) is a leading genetic cause of autism and intellectual disabilities. Sensory-processing deficits are common in humans with FXS and an animal model, the Fmr1 knockout (KO) mouse, manifesting in the auditory system as debilitating hypersensitivity and abnormal electroencephalographic (EEG) and event-related potential (ERP) phenotypes. FXS is a neurodevelopmental disorder, but how EEG/ERP phenotypes change during development is unclear. Therefore, we characterized baseline and stimulus-evoked EEG in auditory and frontal cortex of developing (postnatal day (P) 21 and P30) and adult (P60) wildtype (WT) and Fmr1 KO mice with the FVB genetic background. We found that baseline gamma-band power and N1 amplitude of auditory ERP were increased in frontal cortex of Fmr1 KO mice during development and in adults. Baseline gamma power was increased in auditory cortex at P30. Genotype differences in stimulus-evoked gamma power were present in both cortical regions, but the direction and strength of the changes were age-dependent. These findings suggest that cortical deficits are present during early development and may contribute to sensory-processing deficits in FXS, which in turn may lead to anxiety and delayed language. Developmental changes in EEG measures indicate that observations at a single time-point during development are not reflective of FXS disease progression and highlight the need to identify developmental trajectories and optimal windows for treatment.
Collapse
Affiliation(s)
- Teresa H Wen
- Neuroscience Graduate Program, University of California Riverside, Riverside, CA 92521, USA
| | - Jonathan W Lovelace
- Psychology Department and Psychology Graduate Program, University of California Riverside, Riverside, CA 92521, USA
| | - Iryna M Ethell
- Neuroscience Graduate Program, University of California Riverside, Riverside, CA 92521, USA; Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Devin K Binder
- Neuroscience Graduate Program, University of California Riverside, Riverside, CA 92521, USA; Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA 92521, USA
| | - Khaleel A Razak
- Neuroscience Graduate Program, University of California Riverside, Riverside, CA 92521, USA; Psychology Department and Psychology Graduate Program, University of California Riverside, Riverside, CA 92521, USA.
| |
Collapse
|
41
|
Development of GABAergic Inputs Is Not Altered in Early Maturation of Adult Born Dentate Granule Neurons in Fragile X Mice. eNeuro 2018; 5:eN-NRS-0137-18. [PMID: 30627633 PMCID: PMC6325535 DOI: 10.1523/eneuro.0137-18.2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 09/27/2018] [Accepted: 10/17/2018] [Indexed: 02/08/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common form of inherited mental retardation and the most common known cause of autism. Loss of fragile X mental retardation protein (FMRP) in mice (Fmr1 KO) leads to altered synaptic and circuit maturation in the hippocampus that is correlated with alterations in hippocampal-dependent behaviors. Previous studies have demonstrated that loss of FMRP increased the rate of proliferation of progenitor cells and altered their fate specification in adult Fmr1 KO mice. While these studies clearly demonstrate a role for FMRP in adult neurogenesis in the hippocampus, it is not known whether the functional synaptic maturation and integration of adult-born dentate granule cells (abDGCs) into hippocampal circuits is affected in Fmr1 KO mice. Here, we used retroviral labeling to birthdate abDGCs in Fmr1 KO mice which allowed us to perform targeted patch clamp recording to measure the development of synaptic inputs to these neurons at precise time points after differentiation. The frequency and amplitude of spontaneous GABAergic events increased over the first three weeks after differentiation; however, this normal development of GABAergic synapses was not altered in Fmr1 KO mice. Furthermore, the relatively depolarized GABA reversal potential (EGABA) in immature abDGCs was unaffected by loss of FMRP as was the development of dendritic arbor of the adult generated neurons. These studies systematically characterized the functional development of abDGCs during the first four weeks after differentiation and demonstrate that the maturation of GABAergic synaptic inputs to these neurons is not grossly affected by the loss of FMRP.
Collapse
|
42
|
Ali Shariati M, Kumar V, Yang T, Chakraborty C, Barres BA, Longo FM, Liao YJ. A Small Molecule TrkB Neurotrophin Receptor Partial Agonist as Possible Treatment for Experimental Nonarteritic Anterior Ischemic Optic Neuropathy. Curr Eye Res 2018; 43:1489-1499. [PMID: 30273053 PMCID: PMC10710940 DOI: 10.1080/02713683.2018.1508726] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 07/25/2018] [Accepted: 07/30/2018] [Indexed: 10/28/2022]
Abstract
PURPOSE Brain-derived neurotrophic factor (BDNF) and activation of its high affinity receptor tropomyosin kinase (Trk) B promote retinal ganglion cells (RGCs) survival following injury. In this study, we tested the effects of LM22A-4, a small molecule TrkB receptor-specific partial agonist, on RGC survival in vitro and in experimental nonarteritic anterior ischemic optic neuropathy (AION), the most common acute optic neuropathy in those older than 50 years. METHODS We assessed drug effects on immunopanned, cultured RGCs and calculated RGC survival and assessed TrkB receptor activation by mitogen-activated protein (MAP) kinase translocation. To assess effects in vivo, we induced murine AION and treated the animals with one intravitreal injection and three-week systemic treatment. We measured drug effects using serial spectral-domain optical coherence tomography (OCT) and quantified retinal Brn3A+ RGC density three weeks after ischemia. RESULTS In vitro, LM22A-4 significantly increased the survival of cultured RGCs at day 2 (95% CI control: 8.4-13.6; LM22A-4: 23.7-30.3; BDNF: 24.3-29.9; P ≤ 0.0001), similar to the effect of the endogenous TrkB receptor ligand BDNF. There was also significant nuclear and cytoplasmic translocation of MAP kinase (95% CI control: 0.9-6.8; LM22A-4: 38.8-84.4; BDNF: 64.0-93.0; P = 0.0002), a known downstream event of TrkB receptor activation. Following AION, LM22A-4 treatment led to significant preservation of the ganglion cell complex (95% CI: AION-PBS: 66.8-70.7%; AION-LM22A-4: 70.0-73.1; P = 0.03) and total retinal thickness (95% CI: AION-PBS: 185-196%; AION-LM22A-4: 195-203; P = 0.002) as measured by OCT compared with non-treated eyes. There was also significant rescue of the Brn3A+ RGC density on morphometric analysis of whole mount retinae (95% CI control: 2360-2629; AION-PBS: 1647-2008 cells/mm2; AION-LM22A-4: 1958-2216 cells/mm2; P = 0.02). CONCLUSIONS TrkB receptor partial agonist LM22A-4 promoted survival of cultured RGCs in vitro by TrkB receptor activation, and treatment in vivo led to increased survival of RGCs after optic nerve ischemia, providing support that LM22A-4 may be effective therapy to treat ischemic optic neuropathy. ABBREVIATIONS AION: anterior ischemic optic neuropathy, BDNF: Brain-derived neurotrophic factor, GCC: ganglion cell complex, MAP: mitogen-activated protein, OCT: spectral-domain optical coherence tomography, OD: right eye, ON: optic nerve, ONH: optic nerve head, OS: left eye, RGC: retinal ganglion cell; Trk: tropomyosin kinase.
Collapse
Affiliation(s)
- Mohammad Ali Shariati
- a Departments of Ophthalmology , Stanford University School of Medicine , Stanford , CA , USA
| | - Varun Kumar
- a Departments of Ophthalmology , Stanford University School of Medicine , Stanford , CA , USA
| | - Tao Yang
- b Neurobiology , Stanford University School of Medicine , Stanford , CA , USA
| | | | - Ben Anthony Barres
- b Neurobiology , Stanford University School of Medicine , Stanford , CA , USA
| | - Frank Michael Longo
- b Neurobiology , Stanford University School of Medicine , Stanford , CA , USA
| | - Yaping Joyce Liao
- a Departments of Ophthalmology , Stanford University School of Medicine , Stanford , CA , USA
- b Neurobiology , Stanford University School of Medicine , Stanford , CA , USA
| |
Collapse
|
43
|
Impaired GABA Neural Circuits Are Critical for Fragile X Syndrome. Neural Plast 2018; 2018:8423420. [PMID: 30402088 PMCID: PMC6192167 DOI: 10.1155/2018/8423420] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/17/2018] [Indexed: 12/24/2022] Open
Abstract
Fragile X syndrome (FXS) is an inheritable neuropsychological disease caused by silence of the fmr1 gene and the deficiency of Fragile X mental retardation protein (FMRP). Patients present neuronal alterations that lead to severe intellectual disability and altered sleep rhythms. However, the neural circuit mechanisms underlying FXS remain unclear. Previous studies have suggested that metabolic glutamate and gamma-aminobutyric acid (GABA) receptors/circuits are two counter-balanced factors involved in FXS pathophysiology. More and more studies demonstrated that attenuated GABAergic circuits in the absence of FMRP are critical for abnormal progression of FXS. Here, we reviewed the changes of GABA neural circuits that were attributed to intellectual-deficient FXS, from several aspects including deregulated GABA metabolism, decreased expressions of GABA receptor subunits, and impaired GABAergic neural circuits. Furthermore, the activities of GABA neural circuits are modulated by circadian rhythm of FMRP metabolism and reviewed the abnormal condition of FXS mice or patients.
Collapse
|
44
|
Wen TH, Binder DK, Ethell IM, Razak KA. The Perineuronal 'Safety' Net? Perineuronal Net Abnormalities in Neurological Disorders. Front Mol Neurosci 2018; 11:270. [PMID: 30123106 PMCID: PMC6085424 DOI: 10.3389/fnmol.2018.00270] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 07/17/2018] [Indexed: 12/22/2022] Open
Abstract
Perineuronal nets (PNN) are extracellular matrix (ECM) assemblies that preferentially ensheath parvalbumin (PV) expressing interneurons. Converging evidence indicates that PV cells and PNN are impaired in a variety of neurological disorders. PNN development and maintenance is necessary for a number of processes within the CNS, including regulation of GABAergic cell function, protection of neurons from oxidative stress, and closure of developmental critical period plasticity windows. Understanding PNN functions may be essential for characterizing the mechanisms of altered cortical excitability observed in neurodegenerative and neurodevelopmental disorders. Indeed, PNN abnormalities have been observed in post-mortem brain tissues of patients with schizophrenia and Alzheimer’s disease. There is impaired development of PNNs and enhanced activity of its key regulator matrix metalloproteinase-9 (MMP-9) in Fragile X Syndrome, a common genetic cause of autism. MMP-9, a protease that cleaves ECM, is differentially regulated in a number of these disorders. Despite this, few studies have addressed the interactions between PNN expression, MMP-9 activity and neuronal excitability. In this review, we highlight the current evidence for PNN abnormalities in CNS disorders associated with altered network function and MMP-9 levels, emphasizing the need for future work targeting PNNs in pathophysiology and therapeutic treatment of neurological disorders.
Collapse
Affiliation(s)
- Teresa H Wen
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States
| | - Devin K Binder
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States.,Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Iryna M Ethell
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States.,Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Khaleel A Razak
- Neuroscience Graduate Program, University of California, Riverside, Riverside, CA, United States.,Psychology Graduate Program, Department of Psychology, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
45
|
Synaptic and circuit development of the primary sensory cortex. Exp Mol Med 2018; 50:1-9. [PMID: 29628505 PMCID: PMC5938038 DOI: 10.1038/s12276-018-0029-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 12/06/2017] [Indexed: 01/06/2023] Open
Abstract
Animals, including humans, optimize their primary sensory cortex through the use of input signals, which allow them to adapt to the external environment and survive. The time window at the beginning of life in which external input signals are connected sensitively and strongly to neural circuit optimization is called the critical period. The critical period has attracted the attention of many neuroscientists due to the rapid activity-/experience-dependent circuit development that occurs, which is clearly differentiated from other developmental time periods and brain areas. This process involves various types of GABAergic inhibitory neurons, the extracellular matrix, neuromodulators, transcription factors, and neurodevelopmental factors. In this review, I discuss recent progress regarding the biological nature of the critical period that contribute to a better understanding of brain development.
Collapse
|