1
|
Reis SL, Monteiro P. From synaptic dysfunction to atypical emotional processing in autism. FEBS Lett 2024; 598:269-282. [PMID: 38233224 DOI: 10.1002/1873-3468.14801] [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: 09/04/2023] [Revised: 12/21/2023] [Accepted: 12/27/2023] [Indexed: 01/19/2024]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition mainly characterized by social impairments and repetitive behaviors. Among these core symptoms, a notable aspect of ASD is the presence of emotional complexities, including high rates of anxiety disorders. The inherent heterogeneity of ASD poses a unique challenge in understanding its etiological origins, yet the utilization of diverse animal models replicating ASD traits has enabled researchers to dissect the intricate relationship between autism and atypical emotional processing. In this review, we delve into the general findings about the neural circuits underpinning one of the most extensively researched and evolutionarily conserved emotional states: fear and anxiety. Additionally, we explore how distinct ASD animal models exhibit various anxiety phenotypes, making them a crucial tool for dissecting ASD's multifaceted nature. Overall, to a proper display of fear response, it is crucial to properly process and integrate sensorial and visceral cues to the fear-induced stimuli. ASD individuals exhibit altered sensory processing, possibly contributing to the emergence of atypical phobias, a prevailing anxiety disorder manifested in this population. Moreover, these individuals display distinctive alterations in a pivotal fear and anxiety processing hub, the amygdala. By examining the neurobiological mechanisms underlying fear and anxiety regulation, we can gain insights into the factors contributing to the distinctive emotional profile observed in individuals with ASD. Such insights hold the potential to pave the way for more targeted interventions and therapies that address the emotional challenges faced by individuals within the autism spectrum.
Collapse
Affiliation(s)
- Sara L Reis
- Department of Biomedicine - Experimental Biology Unit, Faculty of Medicine of the University of Porto, Portugal
| | - Patricia Monteiro
- Department of Biomedicine - Experimental Biology Unit, Faculty of Medicine of the University of Porto, Portugal
| |
Collapse
|
2
|
Milla LA, Corral L, Rivera J, Zuñiga N, Pino G, Nunez-Parra A, Cea-Del Rio CA. Neurodevelopment and early pharmacological interventions in Fragile X Syndrome. Front Neurosci 2023; 17:1213410. [PMID: 37599992 PMCID: PMC10433175 DOI: 10.3389/fnins.2023.1213410] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Fragile X Syndrome (FXS) is a neurodevelopmental disorder and the leading monogenic cause of autism and intellectual disability. For years, several efforts have been made to develop an effective therapeutic approach to phenotypically rescue patients from the disorder, with some even advancing to late phases of clinical trials. Unfortunately, none of these attempts have completely succeeded, bringing urgency to further expand and refocus research on FXS therapeutics. FXS arises at early stages of postnatal development due to the mutation and transcriptional silencing of the Fragile X Messenger Ribonucleoprotein 1 gene (FMR1) and consequent loss of the Fragile X Messenger Ribonucleoprotein (FMRP) expression. Importantly, FMRP expression is critical for the normal adult nervous system function, particularly during specific windows of embryogenic and early postnatal development. Cellular proliferation, migration, morphology, axonal guidance, synapse formation, and in general, neuronal network establishment and maturation are abnormally regulated in FXS, underlying the cognitive and behavioral phenotypes of the disorder. In this review, we highlight the relevance of therapeutically intervening during critical time points of development, such as early postnatal periods in infants and young children and discuss past and current clinical trials in FXS and their potential to specifically target those periods. We also discuss potential benefits, limitations, and disadvantages of these pharmacological tools based on preclinical and clinical research.
Collapse
Affiliation(s)
- Luis A. Milla
- Centro de Investigacion Biomedica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Medicas, Universidad de Santiago de Chile, Santiago, Chile
| | - Lucia Corral
- Laboratorio de Neurofisiopatologia, Centro de Investigacion Biomedica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Medicas, Universidad de Santiago de Chile, Santiago, Chile
| | - Jhanpool Rivera
- Laboratorio de Neurofisiopatologia, Centro de Investigacion Biomedica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Medicas, Universidad de Santiago de Chile, Santiago, Chile
| | - Nolberto Zuñiga
- Laboratorio de Neurofisiopatologia, Centro de Investigacion Biomedica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Medicas, Universidad de Santiago de Chile, Santiago, Chile
| | - Gabriela Pino
- Laboratorio de Neurofisiopatologia, Centro de Investigacion Biomedica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Medicas, Universidad de Santiago de Chile, Santiago, Chile
| | - Alexia Nunez-Parra
- Physiology Laboratory, Department of Biology, Faculty of Science, Universidad de Chile, Santiago, Chile
- Cell Physiology Center, Universidad de Chile, Santiago, Chile
| | - Christian A. Cea-Del Rio
- Laboratorio de Neurofisiopatologia, Centro de Investigacion Biomedica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Medicas, Universidad de Santiago de Chile, Santiago, Chile
| |
Collapse
|
3
|
Svalina MN, Sullivan R, Restrepo D, Huntsman MM. From circuits to behavior: Amygdala dysfunction in fragile X syndrome. Front Integr Neurosci 2023; 17:1128529. [PMID: 36969493 PMCID: PMC10034113 DOI: 10.3389/fnint.2023.1128529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/23/2023] [Indexed: 03/12/2023] Open
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by a repeat expansion mutation in the promotor region of the FMR1 gene resulting in transcriptional silencing and loss of function of fragile X messenger ribonucleoprotein 1 protein (FMRP). FMRP has a well-defined role in the early development of the brain. Thus, loss of the FMRP has well-known consequences for normal cellular and synaptic development leading to a variety of neuropsychiatric disorders including an increased prevalence of amygdala-based disorders. Despite our detailed understanding of the pathophysiology of FXS, the precise cellular and circuit-level underpinnings of amygdala-based disorders is incompletely understood. In this review, we discuss the development of the amygdala, the role of neuromodulation in the critical period plasticity, and recent advances in our understanding of how synaptic and circuit-level changes in the basolateral amygdala contribute to the behavioral manifestations seen in FXS.
Collapse
Affiliation(s)
- Matthew N. Svalina
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Regina Sullivan
- Brain Institute, Nathan Kline Institute, Orangeburg, NY, United States
- Child and Adolescent Psychiatry, Child Study Center, New York University School of Medicine, New York, NY, United States
| | - Diego Restrepo
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Molly M. Huntsman
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- *Correspondence: Molly M. Huntsman,
| |
Collapse
|
4
|
Kim Y, Jeon SJ, Gonzales EL, Shin D, Remonde CG, Ahn T, Shin CY. Pirenperone relieves the symptoms of fragile X syndrome in Fmr1 knockout mice. Sci Rep 2022; 12:20966. [PMID: 36470953 PMCID: PMC9723111 DOI: 10.1038/s41598-022-25582-8] [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: 09/02/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder that is caused by the loss of Fragile X-linked mental retardation protein (FMRP), an RNA binding protein that can bind and recognize different RNA structures and regulate the target mRNAs' translation involved in neuronal synaptic plasticity. Perturbations of this gene expression network have been related to abnormal behavioral symptoms such as hyperactivity, and impulsivity. Considering the roles of FMRP in the modulation of mRNA translation, we investigated the differentially expressed genes which might be targeted to revert to normal and ameliorate behavioral symptoms. Gene expression data was analyzed and used the connectivity map (CMap) to understand the changes in gene expression in FXS and predict the effective drug candidates. We analyzed the GSE7329 dataset that had 15 control and 8 FXS patients' lymphoblastoid samples. Among 924 genes, 42 genes were selected as signatures for CMap analysis, and 24 associated drugs were found. Pirenperone was selected as a potential drug candidate for FXS for its possible antipsychotic effect. Treatment of pirenperone increased the expression level of Fmr1 gene. Moreover, pirenperone rescued the behavioral deficits in Fmr1 KO mice including hyperactivity, spatial memory, and impulsivity. These results suggest that pirenperone is a new drug candidate for FXS, which should be verified in future studies.
Collapse
Affiliation(s)
- Yujeong Kim
- grid.258676.80000 0004 0532 8339Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, Seoul, 05029 Republic of Korea
| | - Se Jin Jeon
- grid.412357.60000 0004 0533 2063Department of Integrative Biotechnology, College of Science and Technology, Sahmyook University, Seoul, 01795 Republic of Korea
| | - Edson Luck Gonzales
- grid.258676.80000 0004 0532 8339Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, Seoul, 05029 Republic of Korea
| | - Dongpil Shin
- grid.258676.80000 0004 0532 8339Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, Seoul, 05029 Republic of Korea
| | - Chilly Gay Remonde
- grid.258676.80000 0004 0532 8339Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, Seoul, 05029 Republic of Korea
| | - TaeJin Ahn
- grid.411957.f0000 0004 0647 2543Department of Life Science, Handong Global University, Nehemiah 36, Handong-ro 558, Pohang, 37554 Republic of Korea
| | - Chan Young Shin
- grid.258676.80000 0004 0532 8339Department of Pharmacology and Department of Advanced Translational Medicine, School of Medicine, Konkuk University, Seoul, 05029 Republic of Korea
| |
Collapse
|
5
|
Svalina MN, Rio CACD, Kushner JK, Levy A, Baca SM, Guthman EM, Opendak M, Sullivan RM, Restrepo D, Huntsman MM. Basolateral Amygdala Hyperexcitability Is Associated with Precocious Developmental Emergence of Fear-Learning in Fragile X Syndrome. J Neurosci 2022; 42:7294-7308. [PMID: 35970562 PMCID: PMC9512574 DOI: 10.1523/jneurosci.1776-21.2022] [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: 09/01/2021] [Revised: 08/02/2022] [Accepted: 08/05/2022] [Indexed: 12/24/2022] Open
Abstract
Fragile X Syndrome is a neurodevelopmental disorder and the most common monogenic cause of intellectual disability, autism spectrum disorders, and anxiety disorders. Loss of fragile x mental retardation protein results in disruptions of synaptic development during a critical period of circuit formation in the BLA. However, it is unknown how these alterations impact microcircuit development and function. Using a combination of electrophysiologic and behavioral approaches in both male (Fmr1-/y) and female (Fmr1-/-) mice, we demonstrate that principal neurons in the Fmr1KO BLA exhibit hyperexcitability during a sensitive period in amygdala development. This hyperexcitability contributes to increased excitatory gain in fear-learning circuits. Further, synaptic plasticity is enhanced in the BLA of Fmr1KO mice. Behavioral correlation demonstrates that fear-learning emerges precociously in the Fmr1KO mouse. Early life 4,5,6,7-tetrahydroisoxazolo [5,4-c]pyridin-3ol intervention ameliorates fear-learning in Fmr1KO mice. These results suggest that critical period plasticity in the amygdala of the Fmr1KO mouse may be shifted to earlier developmental time points.SIGNIFICANCE STATEMENT In these studies, we identify early developmental alterations in principal neurons in the Fragile X syndrome BLA. We show that, as early as P14, excitability and feedforward excitation, and synaptic plasticity are enhanced in Fmr1KO lateral amygdala. This correlates with precocious emergence of fear-learning in the Fmr1KO mouse. Early life 4,5,6,7-tetrahydroisoxazolo [5,4-c]pyridin-3ol intervention restores critical period plasticity in WT mice and ameliorates fear-learning in the Fmr1KO mouse.
Collapse
Affiliation(s)
- Matthew N Svalina
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Christian A Cea-Del Rio
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
- CIBAP, Escuela de Medicina, Facultad de Ciencias Medicas, Universidad de Santiago de Chile, Santiago, Chile 9170201
| | - J Keenan Kushner
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Abigail Levy
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Serapio M Baca
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - E Mae Guthman
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Maya Opendak
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, New York 10962
- Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, 10016
| | - Regina M Sullivan
- Emotional Brain Institute, Nathan Kline Institute, Orangeburg, New York 10962
- Child Study Center, Child & Adolescent Psychiatry, New York University School of Medicine, New York, 10016
| | - Diego Restrepo
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Molly M Huntsman
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| |
Collapse
|
6
|
Sun Y, Peng Z, Wei X, Zhang N, Huang CS, Wallner M, Mody I, Houser CR. Virally-induced expression of GABAA receptor δ subunits following their pathological loss reveals their role in regulating GABAA receptor assembly. Prog Neurobiol 2022; 218:102337. [PMID: 35934131 PMCID: PMC10091858 DOI: 10.1016/j.pneurobio.2022.102337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/26/2022] [Accepted: 08/03/2022] [Indexed: 10/31/2022]
Abstract
Decreased expression of the δ subunit of the GABAA receptor (GABAAR) has been found in the dentate gyrus in several animal models of epilepsy and other disorders with increased excitability and is associated with altered modulation of tonic inhibition in dentate granule cells (GCs). In contrast, other GABAAR subunits, including α4 and γ2 subunits, are increased, but the relationship between these changes is unclear. The goals of this study were to determine if viral transfection of δ subunits in dentate GCs could increase δ subunit expression, alter expression of potentially-related GABAAR subunits, and restore more normal network excitability in the dentate gyrus in a mouse model of epilepsy. Pilocarpine-induced seizures were elicited in DOCK10-Cre mice that express Cre selectively in dentate GCs, and two weeks later the mice were injected unilaterally with a Cre-dependent δ-GABAAR viral vector. At 4-6 weeks following transfection, δ subunit immunolabeling was substantially increased in dentate GCs on the transfected side compared to the nontransfected side. Importantly, α4 and γ2 subunit labeling was downregulated on the transfected side. Electrophysiological studies revealed enhanced tonic inhibition, decreased network excitability, and increased neurosteroid sensitivity in slices from the δ subunit-transfected side compared to those from the nontransfected side of the same pilocarpine-treated animal, consistent with the formation of δ subunit-containing GABAARs. No differences were observed between sides of eYFP-transfected animals. These findings are consistent with the idea that altering expression of key subunits, such as the δ subunit, regulates GABAAR subunit assemblies, resulting in substantial effects on network excitability.
Collapse
|
7
|
Deng PY, Kumar A, Cavalli V, Klyachko VA. FMRP regulates GABA A receptor channel activity to control signal integration in hippocampal granule cells. Cell Rep 2022; 39:110820. [PMID: 35584668 DOI: 10.1016/j.celrep.2022.110820] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/11/2022] [Accepted: 04/21/2022] [Indexed: 11/29/2022] Open
Abstract
Fragile X syndrome, the most common inherited form of intellectual disability, is caused by loss of fragile X mental retardation protein (FMRP). GABAergic system dysfunction is one of the hallmarks of FXS, yet the underlying mechanisms remain poorly understood. Here, we report that FMRP interacts with GABAA receptor (GABAAR) and modulates its single-channel activity. Specifically, FMRP regulates spontaneous GABAAR opening through modulating its single-channel conductance and open probability in dentate granule cells. FMRP loss reduces spontaneous GABAAR activity underlying tonic inhibition, while N-terminal FMRP fragment (aa 1-297) is sufficient to rapidly normalize tonic inhibition in Fmr1 knockout (KO) granule cells. FMRP-GABAAR interaction is supported by co-immunoprecipitation of FMRP with at least one GABAAR subunit, the α5. Functionally, FMRP-GABAAR interaction ensures accuracy of coincidence detection of granule cells, which is markedly reduced in Fmr1 KOs. Our study reveals a mechanism underlying FMRP regulation of the GABAergic system and information processing in the hippocampus.
Collapse
Affiliation(s)
- Pan-Yue Deng
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Ajeet Kumar
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Valeria Cavalli
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Vitaly A Klyachko
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, MO 63110, USA.
| |
Collapse
|
8
|
Chen SY, Yao J, Hu YD, Chen HY, Liu PC, Wang WF, Zeng YH, Zhuang CW, Zeng SX, Li YP, Yang LY, Huang ZX, Huang KQ, Lai ZT, Hu YH, Cai P, Chen L, Wu S. Control of Behavioral Arousal and Defense by a Glutamatergic Midbrain-Amygdala Pathway in Mice. Front Neurosci 2022; 16:850193. [PMID: 35527820 PMCID: PMC9070111 DOI: 10.3389/fnins.2022.850193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
In response to external threatening signals, animals evolve a series of defensive behaviors that depend on heightened arousal. It is believed that arousal and defensive behaviors are coordinately regulated by specific neurocircuits in the central nervous system. The ventral tegmental area (VTA) is a key structure located in the ventral midbrain of mice. The activity of VTA glutamatergic neurons has recently been shown to be closely related to sleep–wake behavior. However, the specific role of VTA glutamatergic neurons in sleep–wake regulation, associated physiological functions, and underlying neural circuits remain unclear. In the current study, using an optogenetic approach and synchronous polysomnographic recording, we demonstrated that selective activation of VTA glutamatergic neurons induced immediate transition from sleep to wakefulness and obviously increased the amount of wakefulness in mice. Furthermore, optogenetic activation of VTA glutamatergic neurons induced multiple defensive behaviors, including burrowing, fleeing, avoidance and hiding. Finally, viral-mediated anterograde activation revealed that projections from the VTA to the central nucleus of the amygdala (CeA) mediated the wake- and defense-promoting effects of VTA glutamatergic neurons. Collectively, our results illustrate that the glutamatergic VTA is a key neural substrate regulating wakefulness and defensive behaviors that controls these behaviors through its projection into the CeA. We further discuss the possibility that the glutamatergic VTA-CeA pathway may be involved in psychiatric diseases featuring with excessive defense.
Collapse
Affiliation(s)
- Shang-Yi Chen
- Department of Epidemiology and Health Statistics, The School of Public Health, Fujian Medical University, Fuzhou, China
| | - Jing Yao
- Fujian Province Key Laboratory of Environment and Health, The School of Public Health, Fujian Medical University, Fuzhou, China
| | - Yu-Duan Hu
- Department of Epidemiology and Health Statistics, The School of Public Health, Fujian Medical University, Fuzhou, China
| | - Hui-Yun Chen
- Fujian Province Key Laboratory of Environment and Health, The School of Public Health, Fujian Medical University, Fuzhou, China
| | - Pei-Chang Liu
- Department of Anesthesiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Wen-Feng Wang
- School of Basic Medicine, Fujian Medical University, Fuzhou, China
| | - Yu-Hang Zeng
- School of Basic Medicine, Fujian Medical University, Fuzhou, China
| | - Cong-Wen Zhuang
- School of Basic Medicine, Fujian Medical University, Fuzhou, China
| | - Shun-Xing Zeng
- School of Clinical Medicine, Fujian Medical University, Fuzhou, China
| | - Yue-Ping Li
- School of Clinical Medicine, Fujian Medical University, Fuzhou, China
| | - Liu-Yun Yang
- School of Clinical Medicine, Fujian Medical University, Fuzhou, China
| | - Zi-Xuan Huang
- School of Basic Medicine, Fujian Medical University, Fuzhou, China
| | - Kai-Qi Huang
- School of Basic Medicine, Fujian Medical University, Fuzhou, China
| | - Zhen-Ting Lai
- School of Basic Medicine, Fujian Medical University, Fuzhou, China
| | - Yong-Huai Hu
- School of Basic Medicine, Fujian Medical University, Fuzhou, China
| | - Ping Cai
- Fujian Province Key Laboratory of Environment and Health, The School of Public Health, Fujian Medical University, Fuzhou, China
- *Correspondence: Ping Cai,
| | - Li Chen
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China
- Li Chen,
| | - Siying Wu
- Department of Epidemiology and Health Statistics, The School of Public Health, Fujian Medical University, Fuzhou, China
- Siying Wu,
| |
Collapse
|
9
|
Liu X, Kumar V, Tsai NP, Auerbach BD. Hyperexcitability and Homeostasis in Fragile X Syndrome. Front Mol Neurosci 2022; 14:805929. [PMID: 35069112 PMCID: PMC8770333 DOI: 10.3389/fnmol.2021.805929] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/14/2021] [Indexed: 01/13/2023] Open
Abstract
Fragile X Syndrome (FXS) is a leading inherited cause of autism and intellectual disability, resulting from a mutation in the FMR1 gene and subsequent loss of its protein product FMRP. Despite this simple genetic origin, FXS is a phenotypically complex disorder with a range of physical and neurocognitive disruptions. While numerous molecular and cellular pathways are affected by FMRP loss, there is growing evidence that circuit hyperexcitability may be a common convergence point that can account for many of the wide-ranging phenotypes seen in FXS. The mechanisms for hyperexcitability in FXS include alterations to excitatory synaptic function and connectivity, reduced inhibitory neuron activity, as well as changes to ion channel expression and conductance. However, understanding the impact of FMR1 mutation on circuit function is complicated by the inherent plasticity in neural circuits, which display an array of homeostatic mechanisms to maintain activity near set levels. FMRP is also an important regulator of activity-dependent plasticity in the brain, meaning that dysregulated plasticity can be both a cause and consequence of hyperexcitable networks in FXS. This makes it difficult to separate the direct effects of FMR1 mutation from the myriad and pleiotropic compensatory changes associated with it, both of which are likely to contribute to FXS pathophysiology. Here we will: (1) review evidence for hyperexcitability and homeostatic plasticity phenotypes in FXS models, focusing on similarities/differences across brain regions, cell-types, and developmental time points; (2) examine how excitability and plasticity disruptions interact with each other to ultimately contribute to circuit dysfunction in FXS; and (3) discuss how these synaptic and circuit deficits contribute to disease-relevant behavioral phenotypes like epilepsy and sensory hypersensitivity. Through this discussion of where the current field stands, we aim to introduce perspectives moving forward in FXS research.
Collapse
Affiliation(s)
- Xiaopeng Liu
- Deparment of Molecular & Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Beckman Institute for Advanced Science & Technology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Vipendra Kumar
- Deparment of Molecular & Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Nien-Pei Tsai
- Deparment of Molecular & Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Benjamin D. Auerbach
- Deparment of Molecular & Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Beckman Institute for Advanced Science & Technology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- *Correspondence: Benjamin D. Auerbach
| |
Collapse
|
10
|
Bach S, Shovlin S, Moriarty M, Bardoni B, Tropea D. Rett Syndrome and Fragile X Syndrome: Different Etiology With Common Molecular Dysfunctions. Front Cell Neurosci 2021; 15:764761. [PMID: 34867203 PMCID: PMC8640214 DOI: 10.3389/fncel.2021.764761] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/27/2021] [Indexed: 01/04/2023] Open
Abstract
Rett syndrome (RTT) and Fragile X syndrome (FXS) are two monogenetic neurodevelopmental disorders with complex clinical presentations. RTT is caused by mutations in the Methyl-CpG binding protein 2 gene (MECP2) altering the function of its protein product MeCP2. MeCP2 modulates gene expression by binding methylated CpG dinucleotides, and by interacting with transcription factors. FXS is caused by the silencing of the FMR1 gene encoding the Fragile X Mental Retardation Protein (FMRP), a RNA binding protein involved in multiple steps of RNA metabolism, and modulating the translation of thousands of proteins including a large set of synaptic proteins. Despite differences in genetic etiology, there are overlapping features in RTT and FXS, possibly due to interactions between MeCP2 and FMRP, and to the regulation of pathways resulting in dysregulation of common molecular signaling. Furthermore, basic physiological mechanisms are regulated by these proteins and might concur to the pathophysiology of both syndromes. Considering that RTT and FXS are disorders affecting brain development, and that most of the common targets of MeCP2 and FMRP are involved in brain activity, we discuss the mechanisms of synaptic function and plasticity altered in RTT and FXS, and we consider the similarities and the differences between these two disorders.
Collapse
Affiliation(s)
- Snow Bach
- School of Mathematical Sciences, Dublin City University, Dublin, Ireland.,Neuropsychiatric Genetics, Department of Psychiatry, School of Medicine, Trinity College Dublin, Trinity Translational Medicine Institute, St James's Hospital, Dublin, Ireland
| | - Stephen Shovlin
- Neuropsychiatric Genetics, Department of Psychiatry, School of Medicine, Trinity College Dublin, Trinity Translational Medicine Institute, St James's Hospital, Dublin, Ireland
| | | | - Barbara Bardoni
- Inserm, CNRS UMR 7275, Institute of Molecular and Cellular Pharmacology, Université Côte d'Azur, Valbonne, France
| | - Daniela Tropea
- Neuropsychiatric Genetics, Department of Psychiatry, School of Medicine, Trinity College Dublin, Trinity Translational Medicine Institute, St James's Hospital, Dublin, Ireland.,Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland.,FutureNeuro, The SFI Research Centre for Chronic and Rare Neurological Diseases, Dublin, Ireland
| |
Collapse
|
11
|
Budimirovic DB, Dominick KC, Gabis LV, Adams M, Adera M, Huang L, Ventola P, Tartaglia NR, Berry-Kravis E. Gaboxadol in Fragile X Syndrome: A 12-Week Randomized, Double-Blind, Parallel-Group, Phase 2a Study. Front Pharmacol 2021; 12:757825. [PMID: 34690787 PMCID: PMC8531725 DOI: 10.3389/fphar.2021.757825] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/23/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Fragile X syndrome (FXS), the most common single-gene cause of intellectual disability and autism spectrum disorder (ASD), is caused by a >200-trinucleotide repeat expansion in the 5' untranslated region of the fragile X mental retardation 1 (FMR1) gene. Individuals with FXS can present with a range of neurobehavioral impairments including, but not limited to: cognitive, language, and adaptive deficits; ASD; anxiety; social withdrawal and avoidance; and aggression. Decreased expression of the γ-aminobutyric acid type A (GABAA) receptor δ subunit and deficient GABAergic tonic inhibition could be associated with symptoms of FXS. Gaboxadol (OV101) is a δ-subunit-selective, extrasynaptic GABAA receptor agonist that enhances GABAergic tonic inhibition, providing the rationale for assessment of OV101 as a potential targeted treatment of FXS. No drug is approved in the United States for the treatment of FXS. Methods: This 12-weeks, randomized (1:1:1), double-blind, parallel-group, phase 2a study was designed to assess the safety, tolerability, efficacy, and optimal daily dose of OV101 5 mg [once (QD), twice (BID), or three-times daily (TID)] when administered for 12 weeks to adolescent and adult men with FXS. Safety was the primary study objective, with key assessments including treatment-emergent adverse events (TEAEs), treatment-related adverse events leading to study discontinuation, and serious adverse events (SAEs). The secondary study objective was to evaluate the effect of OV101 on a variety of problem behaviors. Results: A total of 23 participants with FXS (13 adolescents, 10 adults) with moderate-to-severe neurobehavioral phenotypes (Full Scale Intelligence Quotient, 41.5 ± 3.29; ASD, 82.6%) were randomized to OV101 5 mg QD (n = 8), 5 mg BID (n = 8), or 5 mg TID (n = 7) for 12 weeks. OV101 was well tolerated across all 3 treatment regimens. The most common TEAEs were upper respiratory tract infection (n = 4), headache (n = 3), diarrhea (n = 2), and irritability (n = 2). No SAEs were reported. Improvements from baseline to end-of-treatment were observed on several efficacy endpoints, and 60% of participants were identified as treatment responders based on Clinical Global Impressions-Improvement. Conclusions: Overall, OV101 was safe and well tolerated. Efficacy results demonstrate an initial signal for OV101 in individuals with FXS. These results need to be confirmed in a larger, randomized, placebo-controlled study with optimal outcomes and in the most appropriate age group. Clinical Trial Registration: www.ClinicalTrials.gov, identifier: NCT03697161.
Collapse
Affiliation(s)
- Dejan B Budimirovic
- Department of Psychiatry, Kennedy Krieger Institute, Johns Hopkins University, Baltimore, MD, United States.,Department of Psychiatry and Behavioral Sciences-Child Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Kelli C Dominick
- Department of Psychiatry, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Lidia V Gabis
- Maccabi HMO, Tel Aviv-Yafo, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | | | | | - Linda Huang
- Ovid Therapeutics Inc., New York, NY, United States
| | - Pamela Ventola
- Child Study Center, Yale University, New Haven, CT, United States
| | - Nicole R Tartaglia
- University of Colorado School of Medicine, Children's Hospital Colorado, Aurora, CO, United States
| | - Elizabeth Berry-Kravis
- Department of Pediatrics, Neurological Sciences, Biochemistry, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|
12
|
Deemyad T, Puig S, Papale AE, Qi H, LaRocca GM, Aravind D, LaNoce E, Urban NN. Lateralized Decrease of Parvalbumin+ Cells in the Somatosensory Cortex of ASD Models Is Correlated with Unilateral Tactile Hypersensitivity. Cereb Cortex 2021; 32:554-568. [PMID: 34347040 DOI: 10.1093/cercor/bhab233] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 05/31/2021] [Accepted: 06/21/2021] [Indexed: 12/27/2022] Open
Abstract
Inhibitory control of excitatory networks contributes to cortical functions. Increasing evidence indicates that parvalbumin (PV+)-expressing basket cells (BCs) are a major player in maintaining the balance between excitation (E) and inhibition (I). Disruption of E/I balance in cortical networks is believed to be a hallmark of autism spectrum disorder (ASD). Here, we report a lateralized decrease in the number of PV+ BCs in L2/3 of the somatosensory cortex in the dominant hemisphere of Shank3-/- and Cntnap2-/- mouse models of ASD. The dominant hemisphere was identified during a reaching task to establish each animal's dominant forepaw. Double labeling with anti-PV antibody and a biotinylated lectin (Vicia villosa lectin [VVA]) showed that the number of BCs was not different but rather, some BCs did not express PV (PV-), resulting in an elevated number of PV- VVA+ BCs. Finally, we showed that dominant hindpaws had higher mechanical sensitivity when compared with the other hindpaws. This mechanical hypersensitivity in the dominant paw strongly correlated with the decrease in the number of PV+ interneurons and reduced PV expression in the corresponding cortex. Together, these results suggest that the hypersensitivity in ASD patients could be due to decreased inhibitory inputs to the dominant somatosensory cortex.
Collapse
Affiliation(s)
- Tara Deemyad
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Stephanie Puig
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Andrew E Papale
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Hang Qi
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Gregory M LaRocca
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Deepthi Aravind
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Emma LaNoce
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Nathaniel N Urban
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Center for Neuroscience at the University of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
13
|
Liu DC, Lee KY, Lizarazo S, Cook JK, Tsai NP. ER stress-induced modulation of neural activity and seizure susceptibility is impaired in a fragile X syndrome mouse model. Neurobiol Dis 2021; 158:105450. [PMID: 34303799 DOI: 10.1016/j.nbd.2021.105450] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/18/2021] [Indexed: 01/29/2023] Open
Abstract
Imbalanced neuronal excitability homeostasis is commonly observed in patients with fragile X syndrome (FXS) and the animal model of FXS, the Fmr1 KO. While alterations of neuronal intrinsic excitability and synaptic activity at the steady state in FXS have been suggested to contribute to such a deficit and ultimately the increased susceptibility to seizures in FXS, it remains largely unclear whether and how the homeostatic response of neuronal excitability following extrinsic challenges is disrupted in FXS. Our previous work has shown that the acute response following induction of endoplasmic reticulum (ER) stress can reduce neural activity and seizure susceptibility. Because many signaling pathways associated with ER stress response are mediated by Fmr1, we asked whether acute ER stress-induced reduction of neural activity and seizure susceptibility are altered in FXS. Our results first revealed that acute ER stress can trigger a protein synthesis-dependent prevention of neural network synchronization in vitro and a reduction of susceptibility to kainic acid-induced seizures in vivo in wild-type but not in Fmr1 KO mice. Mechanistically, we found that acute ER stress-induced activation of murine double minute-2 (Mdm2), ubiquitination of p53, and the subsequent transient protein synthesis are all impaired in Fmr1 KO neurons. Employing a p53 inhibitor, Pifithrin-α, to mimic p53 inactivation, we were able to blunt the increase in neural network synchronization and reduce the seizure susceptibility in Fmr1 KO mice following ER stress induction. In summary, our data revealed a novel cellular defect in Fmr1 KO mice and suggest that an impaired response to common extrinsic challenges may contribute to imbalanced neuronal excitability homeostasis in FXS.
Collapse
Affiliation(s)
- Dai-Chi Liu
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Kwan Young Lee
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Simon Lizarazo
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Jessie K Cook
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Nien-Pei Tsai
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
14
|
Rudolph S, Guo C, Pashkovski SL, Osorno T, Gillis WF, Krauss JM, Nyitrai H, Flaquer I, El-Rifai M, Datta SR, Regehr WG. Cerebellum-Specific Deletion of the GABA A Receptor δ Subunit Leads to Sex-Specific Disruption of Behavior. Cell Rep 2021; 33:108338. [PMID: 33147470 PMCID: PMC7700496 DOI: 10.1016/j.celrep.2020.108338] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 08/04/2020] [Accepted: 10/08/2020] [Indexed: 12/19/2022] Open
Abstract
Granule cells (GCs) of the cerebellar input layer express high-affinity δ GABAA subunit-containing GABAA receptors (δGABAARs) that respond to ambient GABA levels and context-dependent neuromodulators like steroids. We find that GC-specific deletion of δGABAA (cerebellar [cb] δ knockout [KO]) decreases tonic inhibition, makes GCs hyperexcitable, and in turn, leads to differential activation of cb output regions as well as many cortical and subcortical brain areas involved in cognition, anxiety-like behaviors, and the stress response. Cb δ KO mice display deficits in many behaviors, but motor function is normal. Strikingly, δGABAA deletion alters maternal behavior as well as spontaneous, stress-related, and social behaviors specifically in females. Our findings establish that δGABAARs enable the cerebellum to control diverse behaviors not previously associated with the cerebellum in a sex-dependent manner. These insights may contribute to a better understanding of the mechanisms that underlie behavioral abnormalities in psychiatric and neurodevelopmental disorders that display a gender bias. Rudolph et al. show that deletion of the neuromodulator and hormone-sensitive δGABAA receptor subunit from cerebellar granule cells results in anxiety-like behaviors and female-specific deficits in social behavior and maternal care. δGABAA deletion is associated with hyperexcitability of the cerebellar input layer and altered activation of many stress-related brain regions.
Collapse
Affiliation(s)
- Stephanie Rudolph
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Chong Guo
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Stan L Pashkovski
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Tomas Osorno
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Winthrop F Gillis
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Jeremy M Krauss
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Hajnalka Nyitrai
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Isabella Flaquer
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Mahmoud El-Rifai
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Wade G Regehr
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
15
|
Abstract
Fragile X syndrome (FXS) is the most common inherited form of intellectual disability and the leading monogenic cause of autism. The condition stems from loss of fragile X mental retardation protein (FMRP), which regulates a wide range of ion channels via translational control, protein-protein interactions and second messenger pathways. Rapidly increasing evidence demonstrates that loss of FMRP leads to numerous ion channel dysfunctions (that is, channelopathies), which in turn contribute significantly to FXS pathophysiology. Consistent with this, pharmacological or genetic interventions that target dysregulated ion channels effectively restore neuronal excitability, synaptic function and behavioural phenotypes in FXS animal models. Recent studies further support a role for direct and rapid FMRP-channel interactions in regulating ion channel function. This Review lays out the current state of knowledge in the field regarding channelopathies and the pathogenesis of FXS, including promising therapeutic implications.
Collapse
|
16
|
Svalina MN, Guthman EM, Cea-Del Rio CA, Kushner JK, Baca SM, Restrepo D, Huntsman MM. Hyperexcitability and Loss of Feedforward Inhibition Contribute to Aberrant Plasticity in the Fmr1KO Amygdala. eNeuro 2021; 8:ENEURO.0113-21.2021. [PMID: 33893168 PMCID: PMC8121259 DOI: 10.1523/eneuro.0113-21.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/05/2021] [Accepted: 04/09/2021] [Indexed: 12/15/2022] Open
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder (NDD) characterized by intellectual disability, autism spectrum disorders (ASDs), and anxiety disorders. The disruption in the function of the FMR1 gene results in a range of alterations in cellular and synaptic function. Previous studies have identified dynamic alterations in inhibitory neurotransmission in early postnatal development in the amygdala of the mouse model of FXS. However, little is known about how these changes alter microcircuit development and plasticity in the lateral amygdala (LA). Using whole-cell patch clamp electrophysiology, we demonstrate that principal neurons (PNs) in the LA exhibit hyperexcitability with a concomitant increase in the synaptic strength of excitatory synapses in the BLA. Further, reduced feed-forward inhibition appears to enhance synaptic plasticity in the FXS amygdala. These results demonstrate that plasticity is enhanced in the amygdala of the juvenile Fmr1 knock-out (KO) mouse and that E/I imbalance may underpin anxiety disorders commonly seen in FXS and ASDs.
Collapse
Affiliation(s)
- Matthew N Svalina
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - E Mae Guthman
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Christian A Cea-Del Rio
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Centro de Investigación Biomédica y Aplicada, Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Santiago 9160000, Chile
| | - J Keenan Kushner
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Serapio M Baca
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Diego Restrepo
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Molly M Huntsman
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
17
|
Schaefer TL, Ashworth AA, Tiwari D, Tomasek MP, Parkins EV, White AR, Snider A, Davenport MH, Grainger LM, Becker RA, Robinson CK, Mukherjee R, Williams MT, Gibson JR, Huber KM, Gross C, Erickson CA. GABA A Alpha 2,3 Modulation Improves Select Phenotypes in a Mouse Model of Fragile X Syndrome. Front Psychiatry 2021; 12:678090. [PMID: 34093287 PMCID: PMC8175776 DOI: 10.3389/fpsyt.2021.678090] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/26/2021] [Indexed: 11/22/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common cause of inherited intellectual disability. FXS is caused by functional loss of the Fragile X Protein (FXP), also known as Fragile X Mental Retardation Protein (FMRP). In humans and animal models, loss of FXP leads to sensory hypersensitivity, increased susceptibility to seizures and cortical hyperactivity. Several components of the GABAergic system, the major inhibitory system in the brain, are dysregulated in FXS, and thus modulation of GABAergic transmission was suggested and tested as a treatment strategy. However, so far, clinical trials using broad spectrum GABAA or GABAB receptor-specific agonists have not yielded broad improvement of FXS phenotypes in humans. Here, we tested a more selective strategy in Fmr1 knockout (KO) mice using the experimental drug BAER-101, which is a selective GABAA α2/α3 agonist. Our results suggest that BAER-101 reduces hyperexcitability of cortical circuits, partially corrects increased frequency-specific baseline cortical EEG power, reduces susceptibility to audiogenic seizures and improves novel object memory. Other Fmr1 KO-specific phenotypes were not improved by the drug, such as increased hippocampal dendritic spine density, open field activity and marble burying. Overall, this work shows that BAER-101 improves select phenotypes in Fmr1 KO mice and encourages further studies into the efficacy of GABAA-receptor subunit-selective agonists for the treatment of FXS.
Collapse
Affiliation(s)
- Tori L Schaefer
- Division of Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Amy A Ashworth
- Division of Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Durgesh Tiwari
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Madison P Tomasek
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Emma V Parkins
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Angela R White
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Andrew Snider
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Matthew H Davenport
- Division of Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Lindsay M Grainger
- Division of Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Robert A Becker
- Division of Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Chandler K Robinson
- Division of Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Rishav Mukherjee
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Michael T Williams
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Jay R Gibson
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Kimberly M Huber
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Christina Gross
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Craig A Erickson
- Division of Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
18
|
Telias M. Pharmacological Treatments for Fragile X Syndrome Based on Synaptic Dysfunction. Curr Pharm Des 2020; 25:4394-4404. [PMID: 31682210 DOI: 10.2174/1381612825666191102165206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 10/31/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND Fragile X syndrome (FXS) is the most common form of monogenic hereditary cognitive impairment, including intellectual disability, autism, hyperactivity, and epilepsy. METHODS This article reviews the literature pertaining to the role of synaptic dysfunction in FXS. RESULTS In FXS, synaptic dysfunction alters the excitation-inhibition ratio, dysregulating molecular and cellular processes underlying cognition, learning, memory, and social behavior. Decades of research have yielded important hypotheses that could explain, at least in part, the development of these neurological disorders in FXS patients. However, the main goal of translating lab research in animal models to pharmacological treatments in the clinic has been so far largely unsuccessful, leaving FXS a still incurable disease. CONCLUSION In this concise review, we summarize and analyze the main hypotheses proposed to explain synaptic dysregulation in FXS, by reviewing the scientific evidence that led to pharmaceutical clinical trials and their outcome.
Collapse
Affiliation(s)
- Michael Telias
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, United States
| |
Collapse
|
19
|
Disrupted inhibitory plasticity and homeostasis in Fragile X syndrome. Neurobiol Dis 2020; 142:104959. [PMID: 32512151 PMCID: PMC7959200 DOI: 10.1016/j.nbd.2020.104959] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/28/2020] [Accepted: 06/03/2020] [Indexed: 12/21/2022] Open
Abstract
Fragile X Syndrome (FXS) is a neurodevelopmental disorder instigated by the absence of a key translation regulating protein, Fragile X Mental Retardation Protein (FMRP). The loss of FMRP in the CNS leads to abnormal synaptic development, disruption of critical periods of plasticity, and an overall deficiency in proper sensory circuit coding leading to hyperexcitable sensory networks. However, little is known about how this hyperexcitable environment affects inhibitory synaptic plasticity. Here, we show that in vivo layer 2/3 of the primary somatosensory cortex of the Fmr1 KO mouse exhibits basal hyperexcitability and an increase in neuronal firing rate suppression during whisker activation. This aligns with our in vitro data that indicate an increase in GABAergic spontaneous activity, a faulty mGluR-mediated inhibitory input and impaired inhibitory plasticity processes. Specifically, we find that mGluR activation sensitivity is overall diminished in the Fmr1 KO mouse leading to both a decreased spontaneous inhibitory postsynaptic input to principal cells and a disrupted form of inhibitory long-term depression (I-LTD). These data suggest an adaptive mechanism that acts to homeostatically counterbalance the cortical hyperexcitability observed in FXS.
Collapse
|
20
|
Pan HQ, Zhang WH, Liao CZ, He Y, Xiao ZM, Qin X, Liu WZ, Wang N, Zou JX, Liu XX, Pan BX. Chronic Stress Oppositely Regulates Tonic Inhibition in Thy1-Expressing and Non-expressing Neurons in Amygdala. Front Neurosci 2020; 14:299. [PMID: 32362809 PMCID: PMC7180173 DOI: 10.3389/fnins.2020.00299] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 03/16/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic or prolonged exposure to stress ranks among the most important socioenvironmental factors contributing to the development of neuropsychiatric diseases, a process generally associated with loss of inhibitory tone in amygdala. Recent studies have identified distinct neuronal circuits within the basolateral amygdala (BLA) engaged in different emotional processes. However, the potential circuit involved in stress-induced dysregulation of inhibitory tones in BLA remains elusive. Here, a transgenic mouse model expressing yellow fluorescent protein under control of the Thy1 promoter was used to differentiate subpopulations of projection neurons (PNs) within the BLA. We observed that the tonic inhibition in amygdala neurons expressing and not expressing Thy1 (Thy1+/-) was oppositely regulated by chronic social defeat stress (CSDS). In unstressed control mice, the tonic inhibitory currents were significantly stronger in Thy1- PNs than their Thy1+ counterparts. CSDS markedly reduced the currents in Thy1- projection neurons (PNs), but increased that in Thy1+ ones. By contrast, CSDS failed to affect both the phasic A-type γ-aminobutyric acid receptor (GABAAR) currents and GABABR currents in these two PN populations. Moreover, chronic corticosterone administration was sufficient to mimic the effect of CSDS on the tonic inhibition of Thy1+ and Thy1- PNs. As a consequence, the suppression of tonic GABAAR currents on the excitability of Thy1- PNs was weakened by CSDS, but enhanced in Thy1+ PNs. The differential regulation of chronic stress on the tonic inhibition in Thy1+ and Thy1- neurons may orchestrate cell-specific adaptation of amygdala neurons to chronic stress.
Collapse
Affiliation(s)
- Han-Qing Pan
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang, China.,Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China
| | - Wen-Hua Zhang
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang, China.,Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China
| | - Cai-Zhi Liao
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang, China.,Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China
| | - Ye He
- Center for Medical Experiments, Nanchang University, Nanchang, China
| | - Zhi-Ming Xiao
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China
| | - Xia Qin
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang, China.,Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China
| | - Wei-Zhu Liu
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang, China.,Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China
| | - Na Wang
- Department of Physiology, Mudanjiang Medical University, Mudanjiang, China
| | - Jia-Xin Zou
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang, China.,Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China
| | - Xiao-Xuan Liu
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang, China.,Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China
| | - Bing-Xing Pan
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang, China.,Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China
| |
Collapse
|
21
|
McCullagh EA, Rotschafer SE, Auerbach BD, Klug A, Kaczmarek LK, Cramer KS, Kulesza RJ, Razak KA, Lovelace JW, Lu Y, Koch U, Wang Y. Mechanisms underlying auditory processing deficits in Fragile X syndrome. FASEB J 2020; 34:3501-3518. [PMID: 32039504 DOI: 10.1096/fj.201902435r] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/31/2019] [Accepted: 01/18/2020] [Indexed: 01/14/2023]
Abstract
Autism spectrum disorders (ASD) are strongly associated with auditory hypersensitivity or hyperacusis (difficulty tolerating sounds). Fragile X syndrome (FXS), the most common monogenetic cause of ASD, has emerged as a powerful gateway for exploring underlying mechanisms of hyperacusis and auditory dysfunction in ASD. This review discusses examples of disruption of the auditory pathways in FXS at molecular, synaptic, and circuit levels in animal models as well as in FXS individuals. These examples highlight the involvement of multiple mechanisms, from aberrant synaptic development and ion channel deregulation of auditory brainstem circuits, to impaired neuronal plasticity and network hyperexcitability in the auditory cortex. Though a relatively new area of research, recent discoveries have increased interest in auditory dysfunction and mechanisms underlying hyperacusis in this disorder. This rapidly growing body of data has yielded novel research directions addressing critical questions regarding the timing and possible outcomes of human therapies for auditory dysfunction in ASD.
Collapse
Affiliation(s)
- Elizabeth A McCullagh
- Department of Physiology and Biophysics, University of Colorado Anschutz, Aurora, CO, USA.,Department of Integrative Biology, Oklahoma State University, Stillwater, OK, USA
| | - Sarah E Rotschafer
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA.,Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA
| | - Benjamin D Auerbach
- Center for Hearing and Deafness, Department of Communicative Disorders & Sciences, SUNY at Buffalo, Buffalo, NY, USA
| | - Achim Klug
- Department of Physiology and Biophysics, University of Colorado Anschutz, Aurora, CO, USA
| | - Leonard K Kaczmarek
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
| | - Karina S Cramer
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Randy J Kulesza
- Department of Anatomy, Lake Erie College of Osteopathic Medicine, Erie, PA, USA
| | - Khaleel A Razak
- Department of Psychology, University of California, Riverside, CA, USA
| | | | - Yong Lu
- Department of Anatomy and Neurobiology, College of Medicine, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Ursula Koch
- Institute of Biology, Neurophysiology, Freie Universität Berlin, Berlin, Germany
| | - Yuan Wang
- Department of Biomedical Sciences, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| |
Collapse
|
22
|
Ciranna L, Costa L. Pituitary Adenylate Cyclase-Activating Polypeptide Modulates Hippocampal Synaptic Transmission and Plasticity: New Therapeutic Suggestions for Fragile X Syndrome. Front Cell Neurosci 2019; 13:524. [PMID: 31827422 PMCID: PMC6890831 DOI: 10.3389/fncel.2019.00524] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 11/08/2019] [Indexed: 12/13/2022] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) modulates glutamatergic synaptic transmission and plasticity in the hippocampus, a brain area with a key role in learning and memory. In agreement, several studies have demonstrated that PACAP modulates learning in physiological conditions. Recent publications show reduced PACAP levels and/or alterations in PACAP receptor expression in different conditions associated with cognitive disability. It is noteworthy that PACAP administration rescued impaired synaptic plasticity and learning in animal models of aging, Alzheimer's disease, Parkinson's disease, and Huntington's chorea. In this context, results from our laboratory demonstrate that PACAP rescued metabotropic glutamate receptor-mediated synaptic plasticity in the hippocampus of a mouse model of fragile X syndrome (FXS), a genetic form of intellectual disability. PACAP is actively transported through the blood-brain barrier and reaches the brain following intranasal or intravenous administration. Besides, new studies have identified synthetic PACAP analog peptides with improved selectivity and pharmacokinetic properties with respect to the native peptide. Our review supports the shared idea that pharmacological activation of PACAP receptors might be beneficial for brain pathologies with cognitive disability. In addition, we suggest that the effects of PACAP treatment might be further studied as a possible therapy in FXS.
Collapse
Affiliation(s)
- Lucia Ciranna
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Lara Costa
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| |
Collapse
|
23
|
Horder J, Andersson M, Mendez MA, Singh N, Tangen Ä, Lundberg J, Gee A, Halldin C, Veronese M, Bölte S, Farde L, Sementa T, Cash D, Higgins K, Spain D, Turkheimer F, Mick I, Selvaraj S, Nutt DJ, Lingford-Hughes A, Howes OD, Murphy DG, Borg J. GABA A receptor availability is not altered in adults with autism spectrum disorder or in mouse models. Sci Transl Med 2019; 10:10/461/eaam8434. [PMID: 30282698 DOI: 10.1126/scitranslmed.aam8434] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 08/15/2017] [Accepted: 12/15/2017] [Indexed: 01/16/2023]
Abstract
Preliminary studies have suggested that γ-aminobutyric acid type A (GABAA) receptors, and potentially the GABAA α5 subtype, are deficient in autism spectrum disorder (ASD). However, prior studies have been confounded by the effects of medications, and these studies did not compare findings across different species. We measured both total GABAA and GABAA α5 receptor availability in two positron emission tomography imaging studies. We used the tracer [11C]flumazenil in 15 adults with ASD and in 15 control individuals without ASD and the tracer [11C]Ro15-4513 in 12 adults with ASD and in 16 control individuals without ASD. All participants were free of medications. We also performed autoradiography, using the same tracers, in three mouse models of ASD: the Cntnap2 knockout mouse, the Shank3 knockout mouse, and mice carrying a 16p11.2 deletion. We found no differences in GABAA receptor or GABAA α5 subunit availability in any brain region of adults with ASD compared to those without ASD. There were no differences in GABAA receptor or GABAA α5 subunit availability in any of the three mouse models. However, adults with ASD did display altered performance on a GABA-sensitive perceptual task. Our data suggest that GABAA receptor availability may be normal in adults with ASD, although GABA signaling may be functionally impaired.
Collapse
Affiliation(s)
- Jamie Horder
- Department of Forensic and Neurodevelopmental Sciences and Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Max Andersson
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm County Council, SE-171 76 Stockholm, Sweden
| | - Maria A Mendez
- Department of Forensic and Neurodevelopmental Sciences and Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Nisha Singh
- Department of Neuroimaging, Institute of Psychiatry Psychology, and Neuroscience, King's College London, London, UK.,Division of Imaging Sciences and Biomedical Engineering, King's College London, London, UK
| | - Ämma Tangen
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm County Council, SE-171 76 Stockholm, Sweden
| | - Johan Lundberg
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm County Council, SE-171 76 Stockholm, Sweden
| | - Antony Gee
- Division of Imaging Sciences and Biomedical Engineering, King's College London, London, UK
| | - Christer Halldin
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm County Council, SE-171 76 Stockholm, Sweden
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry Psychology, and Neuroscience, King's College London, London, UK
| | - Sven Bölte
- Center of Neurodevelopmental Disorders at Karolinska Institutet (KIND), Pediatric Neuropsychiatry Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Child and Adolescent Psychiatry, Center for Psychiatry Research, Stockholm County Council, Stockholm, Sweden
| | - Lars Farde
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm County Council, SE-171 76 Stockholm, Sweden.,Personalised Healthcare and Biomarkers, AstraZeneca, PET Science Centre, Karolinska Institutet, Stockholm, Sweden
| | - Teresa Sementa
- Division of Imaging Sciences and Biomedical Engineering, King's College London, London, UK
| | - Diana Cash
- Department of Neuroimaging, Institute of Psychiatry Psychology, and Neuroscience, King's College London, London, UK
| | - Karen Higgins
- Department of Neuroimaging, Institute of Psychiatry Psychology, and Neuroscience, King's College London, London, UK
| | - Debbie Spain
- Department of Forensic and Neurodevelopmental Sciences and Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Federico Turkheimer
- Neuropsychopharmacology Unit, Centre for Psychiatry, Division of Brain Sciences, Imperial College London, London, UK
| | - Inge Mick
- Neuropsychopharmacology Unit, Centre for Psychiatry, Division of Brain Sciences, Imperial College London, London, UK
| | - Sudhakar Selvaraj
- Neuropsychopharmacology Unit, Centre for Psychiatry, Division of Brain Sciences, Imperial College London, London, UK
| | - David J Nutt
- Neuropsychopharmacology Unit, Centre for Psychiatry, Division of Brain Sciences, Imperial College London, London, UK
| | - Anne Lingford-Hughes
- Neuropsychopharmacology Unit, Centre for Psychiatry, Division of Brain Sciences, Imperial College London, London, UK
| | - Oliver D Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Declan G Murphy
- Department of Forensic and Neurodevelopmental Sciences and Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Jacqueline Borg
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm County Council, SE-171 76 Stockholm, Sweden.,Center of Neurodevelopmental Disorders at Karolinska Institutet (KIND), Pediatric Neuropsychiatry Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
24
|
Reinhard SM, Rais M, Afroz S, Hanania Y, Pendi K, Espinoza K, Rosenthal R, Binder DK, Ethell IM, Razak KA. Reduced perineuronal net expression in Fmr1 KO mice auditory cortex and amygdala is linked to impaired fear-associated memory. Neurobiol Learn Mem 2019; 164:107042. [PMID: 31326533 PMCID: PMC7519848 DOI: 10.1016/j.nlm.2019.107042] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 06/20/2019] [Accepted: 07/10/2019] [Indexed: 02/06/2023]
Abstract
Fragile X Syndrome (FXS) is a leading cause of heritable intellectual disability and autism. Humans with FXS show anxiety, sensory hypersensitivity and impaired learning. The mechanisms of learning impairments can be studied in the mouse model of FXS, the Fmr1 KO mouse, using tone-associated fear memory paradigms. Our previous study reported impaired development of parvalbumin (PV) positive interneurons and perineuronal nets (PNN) in the auditory cortex of Fmr1 KO mice. A recent study suggested PNN dynamics in the auditory cortex following tone-shock association is necessary for fear expression. Together these data suggest that abnormal PNN regulation may underlie tone-fear association learning deficits in Fmr1 KO mice. We tested this hypothesis by quantifying PV and PNN expression in the amygdala, hippocampus and auditory cortex of Fmr1 KO mice following fear conditioning. We found impaired tone-associated memory formation in Fmr1 KO mice. This was paralleled by impaired learning-associated regulation of PNNs in the superficial layers of auditory cortex in Fmr1 KO mice. PV cell density decreased in the auditory cortex in response to fear conditioning in both WT and Fmr1 KO mice. Learning-induced increase of PV expression in the CA3 hippocampus was only observed in WT mice. We also found reduced PNN density in the amygdala and auditory cortex of Fmr1 KO mice in all conditions, as well as reduced PNN intensity in CA2 hippocampus. There was a positive correlation between tone-associated memory and PNN density in the amygdala and auditory cortex, consistent with a tone-association deficit. Altogether our studies suggest a link between impaired PV and PNN regulation within specific regions of the fear conditioning circuit and impaired tone memory formation in Fmr1 KO mice.
Collapse
Affiliation(s)
- Sarah M Reinhard
- Psychology Department and Psychology Graduate Program, University of California, Riverside, CA 92521, USA
| | - Maham Rais
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Sonia Afroz
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Yasmien Hanania
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Kasim Pendi
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Katherine Espinoza
- Psychology Department and Psychology Graduate Program, University of California, Riverside, CA 92521, USA; Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Robert Rosenthal
- Psychology Department and Psychology Graduate Program, University of California, Riverside, CA 92521, USA
| | - Devin K Binder
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA; Neuroscience Graduate Program, University of California, Riverside, CA 92521, USA
| | - Iryna M Ethell
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA; Neuroscience Graduate Program, University of California, Riverside, CA 92521, USA.
| | - Khaleel A Razak
- Psychology Department and Psychology Graduate Program, University of California, Riverside, CA 92521, USA; Neuroscience Graduate Program, University of California, Riverside, CA 92521, USA.
| |
Collapse
|
25
|
Wang X, Tao J, Qiao Y, Luo S, Zhao Z, Gao Y, Guo J, Kong J, Chen C, Ge L, Zhang B, Guo P, Liu L, Song Y. Gastrodin Rescues Autistic-Like Phenotypes in Valproic Acid-Induced Animal Model. Front Neurol 2018; 9:1052. [PMID: 30581411 PMCID: PMC6293267 DOI: 10.3389/fneur.2018.01052] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/20/2018] [Indexed: 12/13/2022] Open
Abstract
Autism spectrum disorder (ASD) is an immensely challenging developmental disorder characterized by impaired social interaction, restricted/repetitive behavior, and anxiety. GABAergic dysfunction has been postulated to underlie these autistic symptoms. Gastrodin is widely used clinically in the treatment of neurological disorders and showed to modulate GABAergic signaling in the animal brain. The present study aimed to determine whether treatment with gastrodin can rescue valproic acid (VPA) induced autistic-like phenotypes, and to determine its possible mechanism of action. Our results showed that administration of gastrodin effectively alleviated the autistic-associated behavioral abnormalities as reflected by an increase in social interaction and decrement in repetitive/stereotyped behavior and anxiety in mice as compared to those in untreated animals. Remarkably, the amelioration in autistic-like phenotypes was accompanied by the restoration of inhibitory synaptic transmission, α5 GABAA receptor, and type 1 GABA transporter (GAT1) expression in the basolateral amygdala (BLA) of VPA-treated mice. These findings indicate that gastrodin may alleviate the autistic symptoms caused by VPA through regulating GABAergic synaptic transmission, suggesting that gastrodin may be a potential therapeutic target in autism.
Collapse
Affiliation(s)
- Xiaona Wang
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Jing Tao
- Department of Pathology, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Yidan Qiao
- Department of Pathology, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Shuying Luo
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Zhenqin Zhao
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Yinbo Gao
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Jisheng Guo
- Center for Translational Medicine, The Sixth People's Hospital of Zhengzhou, Zhengzhou, China
| | - Jinghui Kong
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Chongfen Chen
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Lili Ge
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Bo Zhang
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Pengbo Guo
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Lei Liu
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Yinsen Song
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Jewett KA, Lee KY, Eagleman DE, Soriano S, Tsai NP. Dysregulation and restoration of homeostatic network plasticity in fragile X syndrome mice. Neuropharmacology 2018; 138:182-192. [PMID: 29890190 DOI: 10.1016/j.neuropharm.2018.06.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 05/01/2018] [Accepted: 06/06/2018] [Indexed: 01/06/2023]
Abstract
Chronic activity perturbations in neurons induce homeostatic plasticity through modulation of synaptic strength or other intrinsic properties to maintain the correct physiological range of excitability. Although similar plasticity can also occur at the population level, what molecular mechanisms are involved remain unclear. In the current study, we utilized a multielectrode array (MEA) recording system to evaluate homeostatic neural network activity of primary mouse cortical neuron cultures. We demonstrated that chronic elevation of neuronal activity through the inhibition of GABA(A) receptors elicits synchronization of neural network activity and homeostatic reduction of the amplitude of spontaneous neural network spikes. We subsequently showed that this phenomenon is mediated by the ubiquitination of tumor suppressor p53, which is triggered by murine double minute-2 (Mdm2). Using a mouse model of fragile X syndrome, in which fragile X mental retardation protein (FMRP) is absent (Fmr1 knockout), we found that Mdm2-p53 signaling, network synchronization, and the reduction of network spike amplitude upon chronic activity stimulation were all impaired. Pharmacologically inhibiting p53 with Pifithrin-α or genetically employing p53 heterozygous mice to enforce the inactivation of p53 in Fmr1 knockout cultures restored the synchronization of neural network activity after chronic activity stimulation and partially corrects the homeostatic reduction of neural network spike amplitude. Together, our findings reveal the roles of both Fmr1 and Mdm2-p53 signaling in the homeostatic regulation of neural network activity and provide insight into the deficits of excitability homeostasis seen when Fmr1 is compromised, such as occurs with fragile X syndrome.
Collapse
Affiliation(s)
- Kathryn A Jewett
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Kwan Young Lee
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Daphne E Eagleman
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Stephanie Soriano
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Nien-Pei Tsai
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
29
|
Synaptic dysfunction in amygdala in intellectual disorder models. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:392-397. [PMID: 28774568 DOI: 10.1016/j.pnpbp.2017.07.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 07/31/2017] [Accepted: 07/31/2017] [Indexed: 11/24/2022]
Abstract
The amygdala is a part of the limbic circuit that has been extensively studied in terms of synaptic connectivity, plasticity and cellular organization since decades (Ehrlich et al., 2009; Ledoux, 2000; Maren, 2001). Amygdala sub-nuclei, including lateral, basolateral and central amygdala appear now as "hubs" providing in parallel and in series neuronal processing enabling the animal to elicit freezing or escaping behavior in response to external threats. In rodents, these behaviors are easily observed and quantified following associative fear conditioning. Thus, studies on amygdala circuit in association with threat/fear behavior became very popular in laboratories and are often used among other behavioral tests to evaluate learning abilities of mouse models for various neuropsychiatric conditions including genetically encoded intellectual disabilities (ID). Yet, more than 100 human X-linked genes - and several hundreds of autosomal genes - have been associated with ID in humans. These mutations introduced in mice can generate social deficits, anxiety dysregulations and fear learning impairments (McNaughton et al., 2008; Houbaert et al., 2013; Jayachandran et al., 2014; Zhang et al., 2015). Noteworthy, a significant proportion of the coded ID gene products are synaptic proteins. It is postulated that the loss of function of these proteins could destabilize neuronal circuits by global changes of the balance between inhibitory and excitatory drives onto neurons. However, whereas amygdala related behavioral deficits are commonly observed in ID models, the role of most of these ID-genes in synaptic function and plasticity in the amygdala are only sparsely studied. We will here discuss some of the concepts that emerged from amygdala-targeted studies examining the role of syndromic and non-syndromic ID genes in fear-related behaviors and/or synaptic function. Along describing these cases, we will discuss how synaptic deficits observed in amygdala circuits could impact memory formation and expression of conditioned fear.
Collapse
|
30
|
Ali Rodriguez R, Joya C, Hines RM. Common Ribs of Inhibitory Synaptic Dysfunction in the Umbrella of Neurodevelopmental Disorders. Front Mol Neurosci 2018; 11:132. [PMID: 29740280 PMCID: PMC5928253 DOI: 10.3389/fnmol.2018.00132] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/03/2018] [Indexed: 01/06/2023] Open
Abstract
The term neurodevelopmental disorder (NDD) is an umbrella term used to group together a heterogeneous class of disorders characterized by disruption in cognition, emotion, and behavior, early in the developmental timescale. These disorders are heterogeneous, yet they share common behavioral symptomatology as well as overlapping genetic contributors, including proteins involved in the formation, specialization, and function of synaptic connections. Advances may arise from bridging the current knowledge on synapse related factors indicated from both human studies in NDD populations, and in animal models. Mounting evidence has shown a link to inhibitory synapse formation, specialization, and function among Autism, Angelman, Rett and Dravet syndromes. Inhibitory signaling is diverse, with numerous subtypes of inhibitory interneurons, phasic and tonic modes of inhibition, and the molecular and subcellular diversity of GABAA receptors. We discuss common ribs of inhibitory synapse dysfunction in the umbrella of NDD, highlighting alterations in the developmental switch to inhibitory GABA, dysregulation of neuronal activity patterns by parvalbumin-positive interneurons, and impaired tonic inhibition. Increasing our basic understanding of inhibitory synapses, and their role in NDDs is likely to produce significant therapeutic advances in behavioral symptom alleviation for interrelated NDDs.
Collapse
Affiliation(s)
- Rachel Ali Rodriguez
- Neuroscience Emphasis, Department of Psychology, University of Nevada, Las Vegas, Las Vegas, NV, United States
| | - Christina Joya
- Neuroscience Emphasis, Department of Psychology, University of Nevada, Las Vegas, Las Vegas, NV, United States
| | - Rochelle M Hines
- Neuroscience Emphasis, Department of Psychology, University of Nevada, Las Vegas, Las Vegas, NV, United States
| |
Collapse
|
31
|
Xu NZ, Ernst M, Treven M, Cerne R, Wakulchik M, Li X, Jones TM, Gleason SD, Morrow D, Schkeryantz JM, Rahman MT, Li G, Poe MM, Cook JM, Witkin JM. Negative allosteric modulation of alpha 5-containing GABA A receptors engenders antidepressant-like effects and selectively prevents age-associated hyperactivity in tau-depositing mice. Psychopharmacology (Berl) 2018; 235:1151-1161. [PMID: 29374303 DOI: 10.1007/s00213-018-4832-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 01/07/2018] [Indexed: 02/08/2023]
Abstract
RATIONALE Associated with frank neuropathology, patients with Alzheimer's disease suffer from a host of neuropsychiatric symptoms that include depression, apathy, agitation, and aggression. Negative allosteric modulators (NAMs) of α5-containing GABAA receptors have been suggested to be a novel target for antidepressant action. We hypothesized that pharmacological modulation of this target would engender increased motivation in stressful environments. METHODS We utilized electrophysiological recordings from Xenopus oocytes and behavioral measures in mice to address this hypothesis. RESULTS In the forced-swim assay in mice that detects antidepressant drugs, the α5β3γ2 GABAΑ receptor NAM, RY-080 produced a marked antidepressant phenotype. Another compound, PWZ-029, was characterized as an α5β3γ2 receptor NAM of lower intrinsic efficacy in electrophysiological studies in Xenopus oocytes. In contrast to RY-080, PWZ-029 was only moderately active in the forced-swim assay and the α5β3γ2 receptor antagonist, Xli-093, was not active at all. The effects of RY-080 were prevented by the non-selective benzodiazepine receptor antagonist flumazenil as well as by the selective ligands, PWZ-029 and Xli-093. These findings demonstrate that this effect of RY-080 is driven by negative allosteric modulation of α5βγ2 GABAA receptors. RY-080 was not active in the tail-suspension test. We also demonstrated a reduction in the age-dependent hyperactivity exhibited by transgenic mice that accumulate pathological tau (rTg4510 mice) by RY-080. The decrease in hyperactivity by RY-080 was selective for the hyperactivity of the rTg4510 mice since the locomotion of control strains of mice were not significantly affected by RY-080. CONCLUSIONS α5βγ2 GABAA receptor NAMs might function as a pharmacological treatment for mood, amotivational syndromes, and psychomotor agitation in patients with Alzheimer's and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Nina Z Xu
- The Lilly Research Labs, Eli Lilly and Company, Indianapolis, IN, USA
| | - Margot Ernst
- Department of Molecular Neurosciences Center for Brain Research, Medical University of Vienna, Spitalgasse 4, Vienna, Austria
| | - Marco Treven
- Department of Molecular Neurosciences Center for Brain Research, Medical University of Vienna, Spitalgasse 4, Vienna, Austria
| | - Rok Cerne
- The Lilly Research Labs, Eli Lilly and Company, Indianapolis, IN, USA
| | - Mark Wakulchik
- The Lilly Research Labs, Eli Lilly and Company, Indianapolis, IN, USA
| | - Xia Li
- The Lilly Research Labs, Eli Lilly and Company, Indianapolis, IN, USA
| | - Timothy M Jones
- The Lilly Research Labs, Eli Lilly and Company, Indianapolis, IN, USA
| | - Scott D Gleason
- The Lilly Research Labs, Eli Lilly and Company, Indianapolis, IN, USA
| | - Denise Morrow
- The Lilly Research Labs, Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Md Toufiqur Rahman
- Department of Chemistry and Biochemistry, University of Wisconsin Milwaukee, Milwaukee, WI, USA
| | - Guanguan Li
- Department of Chemistry and Biochemistry, University of Wisconsin Milwaukee, Milwaukee, WI, USA
| | - Michael M Poe
- Department of Chemistry and Biochemistry, University of Wisconsin Milwaukee, Milwaukee, WI, USA
| | - James M Cook
- Department of Chemistry and Biochemistry, University of Wisconsin Milwaukee, Milwaukee, WI, USA
| | - Jeffrey M Witkin
- The Lilly Research Labs, Eli Lilly and Company, Indianapolis, IN, USA.
| |
Collapse
|
32
|
Meis S, Endres T, Munsch T, Lessmann V. Presynaptic Regulation of Tonic Inhibition by Neuromodulatory Transmitters in the Basal Amygdala. Mol Neurobiol 2018; 55:8509-8521. [PMID: 29560580 DOI: 10.1007/s12035-018-0984-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 02/28/2018] [Indexed: 01/23/2023]
Abstract
Tonic inhibition mediated by ambient levels of GABA that activate extrasynaptic GABAA receptors emerges as an essential factor that tunes neuronal network excitability in vitro and shapes behavioral responses in vivo. To address the role of neuromodulatory transmitter systems on this type of inhibition, we employed patch clamp recordings in mouse amygdala slice preparations. Our results show that the current amplitude of tonic inhibition (Itonic) in projection neurons of the basal amygdala (BA) is increased by preincubation with the neurosteroid THDOC, while the benzodiazepine diazepam is ineffective. This suggests involvement of THDOC sensitive δ subunit containing GABAA receptors in mediating tonic inhibition. Moreover, we provide evidence that the neuromodulatory transmitters NE, 5HT, and ACh strongly enhance spontaneous IPSCs as well as Itonic in the BA. As the increase in frequency, amplitude, and charge of sIPSCs by these neuromodulatory transmitters strongly correlated with the amplitude of Itonic, we conclude that spill-over of synaptic GABA leads to activation of Itonic and thereby to dampening of amygdala excitability. Since local injection of THDOC, as a positive modulator of tonic inhibition, into the BA interfered with the expression of contextual fear memory, our results point to a prominent role of Itonic in fear learning.
Collapse
Affiliation(s)
- S Meis
- Institut für Physiologie, Otto-von-Guericke-Universität, Leipziger Str. 44, D-39120, Magdeburg, Germany. .,Center for Behavioral Brain Sciences, Magdeburg, Germany.
| | - T Endres
- Institut für Physiologie, Otto-von-Guericke-Universität, Leipziger Str. 44, D-39120, Magdeburg, Germany
| | - T Munsch
- Institut für Physiologie, Otto-von-Guericke-Universität, Leipziger Str. 44, D-39120, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - V Lessmann
- Institut für Physiologie, Otto-von-Guericke-Universität, Leipziger Str. 44, D-39120, Magdeburg, Germany. .,Center for Behavioral Brain Sciences, Magdeburg, Germany.
| |
Collapse
|
33
|
Dahlhaus R. Of Men and Mice: Modeling the Fragile X Syndrome. Front Mol Neurosci 2018; 11:41. [PMID: 29599705 PMCID: PMC5862809 DOI: 10.3389/fnmol.2018.00041] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 01/31/2018] [Indexed: 12/26/2022] Open
Abstract
The Fragile X Syndrome (FXS) is one of the most common forms of inherited intellectual disability in all human societies. Caused by the transcriptional silencing of a single gene, the fragile x mental retardation gene FMR1, FXS is characterized by a variety of symptoms, which range from mental disabilities to autism and epilepsy. More than 20 years ago, a first animal model was described, the Fmr1 knock-out mouse. Several other models have been developed since then, including conditional knock-out mice, knock-out rats, a zebrafish and a drosophila model. Using these model systems, various targets for potential pharmaceutical treatments have been identified and many treatments have been shown to be efficient in preclinical studies. However, all attempts to turn these findings into a therapy for patients have failed thus far. In this review, I will discuss underlying difficulties and address potential alternatives for our future research.
Collapse
Affiliation(s)
- Regina Dahlhaus
- Institute for Biochemistry, Emil-Fischer Centre, University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
34
|
Developmental Disruption of GABA AR-Meditated Inhibition in Cntnap2 KO Mice. eNeuro 2017; 4:eN-NWR-0162-17. [PMID: 28966979 PMCID: PMC5617210 DOI: 10.1523/eneuro.0162-17.2017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 09/06/2017] [Accepted: 09/07/2017] [Indexed: 12/24/2022] Open
Abstract
GABA released from presynaptic sites induces short-lived phasic inhibition mediated by synaptic GABAA receptors (GABAARs) and longer-duration tonic inhibition mediated by extrasynaptic GABAA or GABAB receptors (GABABRs). A number of studies have found that contactin-associated protein 2 (Cntnap2) knockout (KO) mice, a well-established mouse model of autism, exhibit reduced interneuron numbers and aberrant phasic inhibition. However, little is known about whether tonic inhibition is disrupted in Cntnap2 KO mice and when the disruption of inhibition begins to occur during postnatal development. We examined tonic and phasic inhibition in layer 2/3 pyramidal cells of primary visual cortex of Cntnap2 KO at two different developmental stages, three to four and six to eight weeks of age. We found that both phasic inhibition and GABAAR but not GABABR-mediated tonic inhibition was reduced in pyramidal cells from six- to eight-week-old Cntnap2 KO mice, while in three- to four-week-old mice, no significant effects of genotype on tonic or phasic inhibition was observed. We further found that activation of tonic currents mediated by δ-subunit-containing GABAARs reduced neural excitability, an effect that was attenuated by loss of Cntnap2. While the relative contribution of tonic versus phasic inhibition to autism-related symptoms remains unclear, our data suggest that reduced tonic inhibition may play an important role, and δ-subunit-containing GABAARs may be a useful target for therapeutic intervention in autism.
Collapse
|
35
|
Decreased surface expression of the δ subunit of the GABA A receptor contributes to reduced tonic inhibition in dentate granule cells in a mouse model of fragile X syndrome. Exp Neurol 2017; 297:168-178. [PMID: 28822839 DOI: 10.1016/j.expneurol.2017.08.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 07/21/2017] [Accepted: 08/15/2017] [Indexed: 11/22/2022]
Abstract
While numerous changes in the GABA system have been identified in models of Fragile X Syndrome (FXS), alterations in subunits of the GABAA receptors (GABAARs) that mediate tonic inhibition are particularly intriguing. Considering the key role of tonic inhibition in controlling neuronal excitability, reduced tonic inhibition could contribute to FXS-associated disorders such as hyperactivity, hypersensitivity, and increased seizure susceptibility. The current study has focused on the expression and function of the δ subunit of the GABAAR, a major subunit involved in tonic inhibition, in granule cells of the dentate gyrus in the Fmr1 knockout (KO) mouse model of FXS. Electrophysiological studies of dentate granule cells revealed a marked, nearly four-fold, decrease in tonic inhibition in the Fmr1 KO mice, as well as reduced effects of two δ subunit-preferring pharmacological agents, THIP and DS2, supporting the suggestion that δ subunit-containing GABAARs are compromised in the Fmr1 KO mice. Immunohistochemistry demonstrated a small but statistically significant decrease in δ subunit labeling in the molecular layer of the dentate gyrus in Fmr1 KO mice compared to wildtype (WT) littermates. The discrepancy between the large deficits in GABA-mediated tonic inhibition in granule cells in the Fmr1 KO mice and only modest reductions in immunolabeling of the δ subunit led to studies of surface expression of the δ subunit. Cross-linking experiments followed by Western blot analysis demonstrated a small, non-significant decrease in total δ subunit protein in the hippocampus of Fmr1 KO mice, but a four-fold decrease in surface expression of the δ subunit in these mice. No significant changes were observed in total or surface expression of the α4 subunit protein, a major partner of the δ subunit in the forebrain. Postembedding immunogold labeling for the δ subunit demonstrated a large, three-fold, decrease in the number of symmetric synapses with immunolabeling at perisynaptic locations in Fmr1 KO mice. While α4 immunogold particles were also reduced at perisynaptic locations in the Fmr1 KO mice, the labeling was increased at synaptic sites. Together these findings suggest that, in the dentate gyrus, altered surface expression of the δ subunit, rather than a decrease in δ subunit expression alone, could be limiting δ subunit-mediated tonic inhibition in this model of FXS. Finding ways to increase surface expression of the δ subunit of the GABAAR could be a novel approach to treatment of hyperexcitability-related alterations in FXS.
Collapse
|
36
|
Ben-Ari Y. NKCC1 Chloride Importer Antagonists Attenuate Many Neurological and Psychiatric Disorders. Trends Neurosci 2017; 40:536-554. [PMID: 28818303 DOI: 10.1016/j.tins.2017.07.001] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/03/2017] [Accepted: 07/10/2017] [Indexed: 12/23/2022]
Abstract
In physiological conditions, adult neurons have low intracellular Cl- [(Cl-)I] levels underlying the γ-aminobutyric acid (GABA)ergic inhibitory drive. In contrast, neurons have high (Cl-)I levels and excitatory GABA actions in a wide range of pathological conditions including spinal cord lesions, chronic pain, brain trauma, cerebrovascular infarcts, autism, Rett and Down syndrome, various types of epilepsies, and other genetic or environmental insults. The diuretic highly specific NKCC1 chloride importer antagonist bumetanide (PubChem CID: 2461) efficiently restores low (Cl-)I levels and attenuates many disorders in experimental conditions and in some clinical trials. Here, I review the mechanisms of action, therapeutic effects, promises, and pitfalls of bumetanide.
Collapse
Affiliation(s)
- Yehezkel Ben-Ari
- New INMED, Aix-Marseille University, Campus Scientifique de Luminy, Marseilles, France.
| |
Collapse
|
37
|
Sharp BM. Basolateral amygdala and stress-induced hyperexcitability affect motivated behaviors and addiction. Transl Psychiatry 2017; 7:e1194. [PMID: 28786979 PMCID: PMC5611728 DOI: 10.1038/tp.2017.161] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 05/16/2017] [Accepted: 06/08/2017] [Indexed: 12/11/2022] Open
Abstract
The amygdala integrates and processes incoming information pertinent to reward and to emotions such as fear and anxiety that promote survival by warning of potential danger. Basolateral amygdala (BLA) communicates bi-directionally with brain regions affecting cognition, motivation and stress responses including prefrontal cortex, hippocampus, nucleus accumbens and hindbrain regions that trigger norepinephrine-mediated stress responses. Disruption of intrinsic amygdala and BLA regulatory neurocircuits is often caused by dysfunctional neuroplasticity frequently due to molecular alterations in local GABAergic circuits and principal glutamatergic output neurons. Changes in local regulation of BLA excitability underlie behavioral disturbances characteristic of disorders including post-traumatic stress syndrome (PTSD), autism, attention-deficit hyperactivity disorder (ADHD) and stress-induced relapse to drug use. In this Review, we discuss molecular mechanisms and neural circuits that regulate physiological and stress-induced dysfunction of BLA/amygdala and its principal output neurons. We consider effects of stress on motivated behaviors that depend on BLA; these include drug taking and drug seeking, with emphasis on nicotine-dependent behaviors. Throughout, we take a translational approach by integrating decades of addiction research on animal models and human trials. We show that changes in BLA function identified in animal addiction models illuminate human brain imaging and behavioral studies by more precisely delineating BLA mechanisms. In summary, BLA is required to promote responding for natural reward and respond to second-order drug-conditioned cues; reinstate cue-dependent drug seeking; express stress-enhanced reacquisition of nicotine intake; and drive anxiety and fear. Converging evidence indicates that chronic stress causes BLA principal output neurons to become hyperexcitable.
Collapse
Affiliation(s)
- B M Sharp
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
38
|
Wang L, Almeida LEF, Nettleton M, Khaibullina A, Albani S, Kamimura S, Nouraie M, Quezado ZMN. Altered nocifensive behavior in animal models of autism spectrum disorder: The role of the nicotinic cholinergic system. Neuropharmacology 2016; 111:323-334. [PMID: 27638450 PMCID: PMC5075237 DOI: 10.1016/j.neuropharm.2016.09.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 08/21/2016] [Accepted: 09/12/2016] [Indexed: 02/05/2023]
Abstract
Caretakers and clinicians alike have long recognized that individuals with autism spectrum disorder (ASD) can have altered sensory processing, which can contribute to its core symptoms. However, the pathobiology of sensory alterations in ASD is poorly understood. Here we examined nocifensive behavior in ASD mouse models, the BTBR T+Itpr3tf/J (BTBR) and the fragile-X mental retardation-1 knockout (Fmr1-KO) mice. We also examined the effects of nicotine on nocifensive behavior given that nicotine, a nicotinic cholinergic receptor (nAChR) agonist that has antinociceptive effects, was shown to improve social deficits and decrease repetitive behaviors in BTBR mice. Compared to respective controls, both BTBR and Fmr1-KO had hyporesponsiveness to noxious thermal stimuli and electrical stimulation of C-sensory fibers, normal responsiveness to electrical stimulation of Aβ- and Aδ-fiber, and hyperresponsiveness to visceral pain after acetic acid intraperitoneal injection. In BTBR, nicotine at lower doses increased, whereas at higher doses, it decreased hotplate latency compared to vehicle. In a significantly different effect pattern, in control mice, nicotine had antinociceptive effects to noxious heat only at the high dose. Interestingly, these nocifensive behavior alterations and differential responses to nicotine antinociceptive effects in BTBR mice were associated with significant downregulation of α3, α4, α5, α7, β2, β3, and β4 nAChR subunits in several cerebral regions both, during embryonic development and adulthood. Taken together, these findings further implicate nAChRs in behaviors alterations in the BTBR model and lend support to the hypothesis that nAChRs may be a target for treatment of behavior deficits and sensory dysfunction in ASD.
Collapse
Affiliation(s)
- Li Wang
- The Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's Research Institute, Division of Anesthesiology, Pain and Perioperative Medicine, Children's National Health System, School of Medicine and Health Sciences, George Washington University, Washington, DC, 20010, USA
| | - Luis E F Almeida
- The Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's Research Institute, Division of Anesthesiology, Pain and Perioperative Medicine, Children's National Health System, School of Medicine and Health Sciences, George Washington University, Washington, DC, 20010, USA
| | - Margaret Nettleton
- The Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's Research Institute, Division of Anesthesiology, Pain and Perioperative Medicine, Children's National Health System, School of Medicine and Health Sciences, George Washington University, Washington, DC, 20010, USA
| | - Alfia Khaibullina
- The Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's Research Institute, Division of Anesthesiology, Pain and Perioperative Medicine, Children's National Health System, School of Medicine and Health Sciences, George Washington University, Washington, DC, 20010, USA
| | - Sarah Albani
- The Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's Research Institute, Division of Anesthesiology, Pain and Perioperative Medicine, Children's National Health System, School of Medicine and Health Sciences, George Washington University, Washington, DC, 20010, USA
| | - Sayuri Kamimura
- The Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's Research Institute, Division of Anesthesiology, Pain and Perioperative Medicine, Children's National Health System, School of Medicine and Health Sciences, George Washington University, Washington, DC, 20010, USA
| | - Mehdi Nouraie
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Zenaide M N Quezado
- The Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's Research Institute, Division of Anesthesiology, Pain and Perioperative Medicine, Children's National Health System, School of Medicine and Health Sciences, George Washington University, Washington, DC, 20010, USA; Center for Neuroscience Research, Children's Research Institute, Children's National Health System, School of Medicine and Health Sciences, George Washington University, Washington, DC, 20010, USA.
| |
Collapse
|
39
|
Fragile X mental retardation protein knockdown in the developing Xenopus tadpole optic tectum results in enhanced feedforward inhibition and behavioral deficits. Neural Dev 2016; 11:14. [PMID: 27503008 PMCID: PMC4977860 DOI: 10.1186/s13064-016-0069-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 08/03/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Fragile X Syndrome is the leading monogenetic cause of autism and most common form of intellectual disability. Previous studies have implicated changes in dendritic spine architecture as the primary result of loss of Fragile X Mental Retardation Protein (FMRP), but recent work has shown that neural proliferation is decreased and cell death is increased with either loss of FMRP or overexpression of FMRP. The purpose of this study was to investigate the effects of loss of FMRP on behavior and cellular activity. METHODS We knocked down FMRP expression using morpholino oligos in the optic tectum of Xenopus laevis tadpoles and performed a series of behavioral and electrophysiological assays. We investigated visually guided collision avoidance, schooling, and seizure propensity. Using single cell electrophysiology, we assessed intrinsic excitability and synaptic connectivity of tectal neurons. RESULTS We found that FMRP knockdown results in decreased swimming speed, reduced schooling behavior and decreased seizure severity. In single cells, we found increased inhibition relative to excitation in response to sensory input. CONCLUSIONS Our results indicate that the electrophysiological development of single cells in the absence of FMRP is largely unaffected despite the large neural proliferation defect. The changes in behavior are consistent with an increase in inhibition, which could be due to either changes in cell number or altered inhibitory drive, and indicate that FMRP can play a significant role in neural development much earlier than previously thought.
Collapse
|
40
|
Deprez F, Vogt F, Floriou-Servou A, Lafourcade C, Rudolph U, Tyagarajan SK, Fritschy JM. Partial inactivation of GABAA receptors containing the α5 subunit affects the development of adult-born dentate gyrus granule cells. Eur J Neurosci 2016; 44:2258-71. [PMID: 27364953 DOI: 10.1111/ejn.13329] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 06/28/2016] [Indexed: 01/13/2023]
Abstract
Alterations of neuronal activity due to changes in GABAA receptors (GABAA R) mediating tonic inhibition influence different hippocampal functions. Gabra5-null mice and α5 subunit((H105R)) knock-in mice exhibit signs of hippocampal dysfunction, but are capable of improved performance in several learning and memory tasks. Accordingly, alleviating abnormal GABAergic tonic inhibition in the hippocampal formation by selective α5-GABAA R modulators represents a possible therapeutic approach for several intellectual deficit disorders. Adult neurogenesis in the dentate gyrus is an important facet of hippocampal plasticity; it is regulated by tonic GABAergic transmission, as shown by deficits in proliferation, migration and dendritic development of adult-born neurons in Gabra4-null mice. Here, we investigated the contribution of α5-GABAA Rs to granule cell development, using retroviral vectors expressing eGFP for labeling precursor cells in the subgranular zone. Global α5-GABAA R knockout (α5-KO) mice showed no alterations in migration and morphological development of eGFP-positive granule cells. However, upregulation of α1 subunit-immunoreactivity was observed in the hippocampal formation and cerebral cortex. In contrast, partial gene inactivation in α5-heterozygous (α5-het) mice, as well as single-cell deletion of Gabra5 in newborn granule cells from α5-floxed mice, caused severe alterations of migration and dendrite development. In α5-het mice, retrovirally mediated overexpression of Cdk5 resulted in normal migration and dendritic branching, suggesting that Cdk5 cooperates with α5-GABAA Rs to regulate neuronal development. These results show that minor imbalance of α5-GABAA R-mediated transmission may have major consequences for neuronal plasticity; and call for caution upon chronic therapeutic use of negative allosteric modulators acting at these receptors.
Collapse
Affiliation(s)
- Francine Deprez
- Institute of Pharmacology and Toxicology, University of Zurich, 8057, Zurich, Switzerland.,Neuroscience Center Zurich, ETH and University of Zurich, Zurich, Switzerland
| | - Fabia Vogt
- Institute of Pharmacology and Toxicology, University of Zurich, 8057, Zurich, Switzerland
| | - Amalia Floriou-Servou
- Institute of Pharmacology and Toxicology, University of Zurich, 8057, Zurich, Switzerland
| | - Carlos Lafourcade
- Institute of Pharmacology and Toxicology, University of Zurich, 8057, Zurich, Switzerland
| | - Uwe Rudolph
- Laboratory of Genetic Neuropharmacology, McLean Hospital, Belmont, MA, USA.,Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Shiva K Tyagarajan
- Institute of Pharmacology and Toxicology, University of Zurich, 8057, Zurich, Switzerland.,Neuroscience Center Zurich, ETH and University of Zurich, Zurich, Switzerland
| | - Jean-Marc Fritschy
- Institute of Pharmacology and Toxicology, University of Zurich, 8057, Zurich, Switzerland.,Neuroscience Center Zurich, ETH and University of Zurich, Zurich, Switzerland
| |
Collapse
|
41
|
Fuccillo MV. Striatal Circuits as a Common Node for Autism Pathophysiology. Front Neurosci 2016; 10:27. [PMID: 26903795 PMCID: PMC4746330 DOI: 10.3389/fnins.2016.00027] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 01/22/2016] [Indexed: 12/17/2022] Open
Abstract
Autism spectrum disorders (ASD) are characterized by two seemingly unrelated symptom domains-deficits in social interactions and restrictive, repetitive patterns of behavioral output. Whether the diverse nature of ASD symptomatology represents distributed dysfunction of brain networks or abnormalities within specific neural circuits is unclear. Striatal dysfunction is postulated to underlie the repetitive motor behaviors seen in ASD, and neurological and brain-imaging studies have supported this assumption. However, as our appreciation of striatal function expands to include regulation of behavioral flexibility, motivational state, goal-directed learning, and attention, we consider whether alterations in striatal physiology are a central node mediating a range of autism-associated behaviors, including social and cognitive deficits that are hallmarks of the disease. This review investigates multiple genetic mouse models of ASD to explore whether abnormalities in striatal circuits constitute a common pathophysiological mechanism in the development of autism-related behaviors. Despite the heterogeneity of genetic insult investigated, numerous genetic ASD models display alterations in the structure and function of striatal circuits, as well as abnormal behaviors including repetitive grooming, stereotypic motor routines, deficits in social interaction and decision-making. Comparative analysis in rodents provides a unique opportunity to leverage growing genetic association data to reveal canonical neural circuits whose dysfunction directly contributes to discrete aspects of ASD symptomatology. The description of such circuits could provide both organizing principles for understanding the complex genetic etiology of ASD as well as novel treatment routes. Furthermore, this focus on striatal mechanisms of behavioral regulation may also prove useful for exploring the pathogenesis of other neuropsychiatric diseases, which display overlapping behavioral deficits with ASD.
Collapse
Affiliation(s)
- Marc V. Fuccillo
- Department of Neuroscience, Perelman School of Medicine, University of PennsylvaniaPhiladelphia, PA, USA
| |
Collapse
|
42
|
Prager EM, Bergstrom HC, Wynn GH, Braga MFM. The basolateral amygdala γ-aminobutyric acidergic system in health and disease. J Neurosci Res 2015; 94:548-67. [PMID: 26586374 DOI: 10.1002/jnr.23690] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 10/01/2015] [Accepted: 10/18/2015] [Indexed: 01/13/2023]
Abstract
The brain comprises an excitatory/inhibitory neuronal network that maintains a finely tuned balance of activity critical for normal functioning. Excitatory activity in the basolateral amygdala (BLA), a brain region that plays a central role in emotion and motivational processing, is tightly regulated by a relatively small population of γ-aminobutyric acid (GABA) inhibitory neurons. Disruption in GABAergic inhibition in the BLA can occur when there is a loss of local GABAergic interneurons, an alteration in GABAA receptor activation, or a dysregulation of mechanisms that modulate BLA GABAergic inhibition. Disruptions in GABAergic control of the BLA emerge during development, in aging populations, or after trauma, ultimately resulting in hyperexcitability. BLA hyperexcitability manifests behaviorally as an increase in anxiety, emotional dysregulation, or development of seizure activity. This Review discusses the anatomy, development, and physiology of the GABAergic system in the BLA and circuits that modulate GABAergic inhibition, including the dopaminergic, serotonergic, noradrenergic, and cholinergic systems. We highlight how alterations in various neurotransmitter receptors, including the acid-sensing ion channel 1a, cannabinoid receptor 1, and glutamate receptor subtypes, expressed on BLA interneurons, modulate GABAergic transmission and how defects of these systems affect inhibitory tonus within the BLA. Finally, we discuss alterations in the BLA GABAergic system in neurodevelopmental (autism/fragile X syndrome) and neurodegenerative (Alzheimer's disease) diseases and after the development of epilepsy, anxiety, and traumatic brain injury. A more complete understanding of the intrinsic excitatory/inhibitory circuit balance of the amygdala and how imbalances in inhibitory control contribute to excessive BLA excitability will guide the development of novel therapeutic approaches in neuropsychiatric diseases.
Collapse
Affiliation(s)
- Eric M Prager
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services, University of the Health Sciences, Bethesda, Maryland
| | | | - Gary H Wynn
- Center for the Study of Traumatic Stress, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland.,Department of Psychiatry, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland.,Program in Neuroscience, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Maria F M Braga
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services, University of the Health Sciences, Bethesda, Maryland.,Center for the Study of Traumatic Stress, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland.,Department of Psychiatry, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland.,Program in Neuroscience, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
43
|
Abstract
Fragile X syndrome (FXS) results from a genetic mutation in a single gene yet produces a phenotypically complex disorder with a range of neurological and psychiatric problems. Efforts to decipher how perturbations in signaling pathways lead to the myriad alterations in synaptic and cellular functions have provided insights into the molecular underpinnings of this disorder. From this large body of data, the theme of circuit hyperexcitability has emerged as a potential explanation for many of the neurological and psychiatric symptoms in FXS. The mechanisms for hyperexcitability range from alterations in the expression or activity of ion channels to changes in neurotransmitters and receptors. Contributions of these processes are often brain region and cell type specific, resulting in complex effects on circuit function that manifest as altered excitability. Here, we review the current state of knowledge of the molecular, synaptic, and circuit-level mechanisms underlying hyperexcitability and their contributions to the FXS phenotypes.
Collapse
|
44
|
Compromising the phosphodependent regulation of the GABAAR β3 subunit reproduces the core phenotypes of autism spectrum disorders. Proc Natl Acad Sci U S A 2015; 112:14805-10. [PMID: 26627235 DOI: 10.1073/pnas.1514657112] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Alterations in the efficacy of neuronal inhibition mediated by GABAA receptors (GABAARs) containing β3 subunits are continually implicated in autism spectrum disorders (ASDs). In vitro, the plasma membrane stability of GABAARs is potentiated via phosphorylation of serine residues 408 and 409 (S408/9) in the β3 subunit, an effect that is mimicked by their mutation to alanines. To assess if modifications in β3 subunit expression contribute to ASDs, we have created a mouse in which S408/9 have been mutated to alanines (S408/9A). S408/9A homozygotes exhibited increased phasic, but decreased tonic, inhibition, events that correlated with alterations in the membrane stability and synaptic accumulation of the receptor subtypes that mediate these distinct forms of inhibition. S408/9A mice exhibited alterations in dendritic spine structure, increased repetitive behavior, and decreased social interaction, hallmarks of ASDs. ASDs are frequently comorbid with epilepsy, and consistent with this comorbidity, S408/9A mice exhibited a marked increase in sensitivity to seizures induced by the convulsant kainic acid. To assess the relevance of our studies using S408/9A mice for the pathophysiology of ASDs, we measured S408/9 phosphorylation in Fmr1 KO mice, a model of fragile X syndrome, the most common monogenetic cause of ASDs. Phosphorylation of S408/9 was selectively and significantly enhanced in Fmr1 KO mice. Collectively, our results suggest that alterations in phosphorylation and/or activity of β3-containing GABAARs may directly contribute to the pathophysiology of ASDs.
Collapse
|
45
|
Wahlstrom-Helgren S, Klyachko VA. GABAB receptor-mediated feed-forward circuit dysfunction in the mouse model of fragile X syndrome. J Physiol 2015; 593:5009-24. [PMID: 26282581 DOI: 10.1113/jp271190] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/12/2015] [Indexed: 12/18/2022] Open
Abstract
KEY POINTS Cortico-hippocampal feed-forward circuits formed by the temporoammonic (TA) pathway exhibit a marked increase in excitation/inhibition ratio and abnormal spike modulation functions in Fmr1 knock-out (KO) mice. Inhibitory, but not excitatory, synapse dysfunction underlies cortico-hippocampal feed-forward circuit abnormalities in Fmr1 KO mice. GABA release is reduced in TA-associated inhibitory synapses of Fmr1 KO mice in a GABAB receptor-dependent manner. Inhibitory synapse and feed-forward circuit defects are mediated predominately by presynaptic GABAB receptor signalling in the TA pathway of Fmr1 KO mice. GABAB receptor-mediated inhibitory synapse defects are circuit-specific and are not observed in the Schaffer collateral pathway-associated inhibitory synapses in stratum radiatum. ABSTRACT Circuit hyperexcitability has been implicated in neuropathology of fragile X syndrome, the most common inheritable cause of intellectual disability. Yet, how canonical unitary circuits are affected in this disorder remains poorly understood. Here, we examined this question in the context of the canonical feed-forward inhibitory circuit formed by the temporoammonic (TA) branch of the perforant path, the major cortical input to the hippocampus. TA feed-forward circuits exhibited a marked increase in excitation/inhibition ratio and major functional defects in spike modulation tasks in Fmr1 knock-out (KO) mice, a fragile X mouse model. Changes in feed-forward circuits were caused specifically by inhibitory, but not excitatory, synapse defects. TA-associated inhibitory synapses exhibited increase in paired-pulse ratio and in the coefficient of variation of IPSPs, consistent with decreased GABA release probability. TA-associated inhibitory synaptic transmission in Fmr1 KO mice was also more sensitive to inhibition of GABAB receptors, suggesting an increase in presynaptic GABAB receptor (GABAB R) signalling. Indeed, the differences in inhibitory synaptic transmission between Fmr1 KO and wild-type (WT) mice were eliminated by a GABAB R antagonist. Inhibition of GABAB Rs or selective activation of presynaptic GABAB Rs also abolished the differences in the TA feed-forward circuit properties between Fmr1 KO and WT mice. These GABAB R-mediated defects were circuit-specific and were not observed in the Schaffer collateral pathway-associated inhibitory synapses. Our results suggest that the inhibitory synapse dysfunction in the cortico-hippocampal pathway of Fmr1 KO mice causes hyperexcitability and feed-forward circuit defects, which are mediated in part by a presynaptic GABAB R-dependent reduction in GABA release.
Collapse
Affiliation(s)
- Sarah Wahlstrom-Helgren
- Departments of Cell Biology and Physiology, Biomedical Engineering, Center for the Investigation of Membrane Excitable Diseases, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Vitaly A Klyachko
- Departments of Cell Biology and Physiology, Biomedical Engineering, Center for the Investigation of Membrane Excitable Diseases, Washington University School of Medicine, St Louis, MO, 63110, USA
| |
Collapse
|
46
|
Fatemi SH, Folsom TD. GABA receptor subunit distribution and FMRP-mGluR5 signaling abnormalities in the cerebellum of subjects with schizophrenia, mood disorders, and autism. Schizophr Res 2015; 167:42-56. [PMID: 25432637 PMCID: PMC5301472 DOI: 10.1016/j.schres.2014.10.010] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 10/06/2014] [Accepted: 10/08/2014] [Indexed: 12/24/2022]
Abstract
Gamma-aminobutyric acid (GABA) is the main inhibitory neurotransmitter in the brain. GABAergic receptor abnormalities have been documented in several major psychiatric disorders including schizophrenia, mood disorders, and autism. Abnormal expression of mRNA and protein for multiple GABA receptors has also been observed in multiple brain regions leading to alterations in the balance between excitatory/inhibitory signaling in the brain with potential profound consequences for normal cognition and maintenance of mood and perception. Altered expression of GABAA receptor subunits has been documented in fragile X mental retardation 1 (FMR1) knockout mice, suggesting that loss of its protein product, fragile X mental retardation protein (FMRP), impacts GABAA subunit expression. Recent postmortem studies from our laboratory have shown reduced expression of FMRP in the brains of subjects with schizophrenia, bipolar disorder, major depression, and autism. FMRP acts as a translational repressor and, under normal conditions, inhibits metabotropic glutamate receptor 5 (mGluR5)-mediated signaling. In fragile X syndrome (FXS), the absence of FMRP is hypothesized to lead to unregulated mGluR5 signaling, ultimately resulting in the behavioral and intellectual impairments associated with this disorder. Our laboratory has identified changes in mGluR5 expression in autism, schizophrenia, and mood disorders. In the current review article, we discuss our postmortem data on GABA receptors, FMRP, and mGluR5 levels and compare our results with other laboratories. Finally, we discuss the interactions between these molecules and the potential for new therapeutic interventions that target these interconnected signaling systems.
Collapse
Affiliation(s)
- S Hossein Fatemi
- Department of Psychiatry, Division of Neuroscience Research, University of Minnesota Medical School, 420 Delaware St SE, MMC 392, Minneapolis, MN 55455, USA; Department of Neuroscience, University of Minnesota Medical School, 321 Church St. SE, Minneapolis, MN 55455, USA.
| | - Timothy D Folsom
- Department of Psychiatry, Division of Neuroscience Research, University of Minnesota Medical School, 420 Delaware St SE, MMC 392, Minneapolis, MN 55455, USA.
| |
Collapse
|
47
|
Martin BS, Martinez-Botella G, Loya CM, Salituro FG, Robichaud AJ, Huntsman MM, Ackley MA, Doherty JJ, Corbin JG. Rescue of deficient amygdala tonic γ-aminobutyric acidergic currents in the Fmr-/y mouse model of fragile X syndrome by a novel γ-aminobutyric acid type A receptor-positive allosteric modulator. J Neurosci Res 2015; 94:568-78. [PMID: 26308557 DOI: 10.1002/jnr.23632] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 08/01/2015] [Accepted: 08/04/2015] [Indexed: 11/07/2022]
Abstract
Alterations in the ratio of excitatory to inhibitory transmission are emerging as a common component of many nervous system disorders, including autism spectrum disorders (ASDs). Tonic γ-aminobutyric acidergic (GABAergic) transmission provided by peri- and extrasynaptic GABA type A (GABAA ) receptors powerfully controls neuronal excitability and plasticity and, therefore, provides a rational therapeutic target for normalizing hyperexcitable networks across a variety of disorders, including ASDs. Our previous studies revealed tonic GABAergic deficits in principal excitatory neurons in the basolateral amygdala (BLA) in the Fmr1(-/y) knockout (KO) mouse model fragile X syndrome. To correct amygdala deficits in tonic GABAergic neurotransmission in Fmr1(-/y) KO mice, we developed a novel positive allosteric modulator of GABAA receptors, SGE-872, based on endogenously active neurosteroids. This study shows that SGE-872 is nearly as potent and twice as efficacious for positively modulating GABAA receptors as its parent molecule, allopregnanolone. Furthermore, at submicromolar concentrations (≤1 μM), SGE-872 is selective for tonic, extrasynaptic α4β3δ-containing GABAA receptors over typical synaptic α1β2γ2 receptors. We further find that SGE-872 strikingly rescues the tonic GABAergic transmission deficit in principal excitatory neurons in the Fmr1(-/y) KO BLA, a structure heavily implicated in the neuropathology of ASDs. Therefore, the potent and selective action of SGE-872 on tonic GABAA receptors containing α4 subunits may represent a novel and highly useful therapeutic avenue for ASDs and related disorders involving hyperexcitability of neuronal networks.
Collapse
Affiliation(s)
- Brandon S Martin
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC
| | | | - Carlos M Loya
- SAGE Therapeutics, 215 First Street, Cambridge, MA 0214243
| | | | | | - Molly M Huntsman
- Pharmaceutical Sciences and Department of Pediatrics, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Mike A Ackley
- SAGE Therapeutics, 215 First Street, Cambridge, MA 0214243
| | | | - Joshua G Corbin
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC
| |
Collapse
|
48
|
GABAB Receptor Agonist R-Baclofen Reverses Social Deficits and Reduces Repetitive Behavior in Two Mouse Models of Autism. Neuropsychopharmacology 2015; 40:2228-39. [PMID: 25754761 PMCID: PMC4613612 DOI: 10.1038/npp.2015.66] [Citation(s) in RCA: 156] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 02/28/2015] [Accepted: 03/03/2015] [Indexed: 12/19/2022]
Abstract
Autism spectrum disorder (ASD) is diagnosed by two core behavioral criteria, unusual reciprocal social interactions and communication, and stereotyped, repetitive behaviors with restricted interests. Excitatory/inhibitory imbalance is a prominent hypothesis for the etiology of autism. The selective GABAB receptor agonist R-baclofen previously reversed social deficits and reduced repetitive behaviors in a mouse model of Fragile X syndrome, and Arbaclofen improved some clinical symptoms in some Fragile X and ASD patients. To evaluate R-baclofen in a broader range of mouse models of ASD, we tested both the R-baclofen enantiomer and the less potent S-baclofen enantiomer in two inbred strains of mice that display low sociability and/or high repetitive or stereotyped behaviors. R-baclofen treatment reversed social approach deficits in BTBR T+ Itpr3tf/J (BTBR), reduced repetitive self-grooming and high marble burying scores in BTBR, and reduced stereotyped jumping in C58/J (C58), at nonsedating doses. S-baclofen produced minimal effects at the same doses. These findings encourage investigations of R-baclofen in other preclinical model systems. Additional clinical studies may be warranted to further evaluate the hypothesis that the GABAB receptor represents a promising pharmacological target for treating appropriately stratified subsets of individuals with ASD.
Collapse
|
49
|
Schaefer TL, Davenport MH, Erickson CA. Emerging pharmacologic treatment options for fragile X syndrome. APPLICATION OF CLINICAL GENETICS 2015; 8:75-93. [PMID: 25897255 PMCID: PMC4396424 DOI: 10.2147/tacg.s35673] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Fragile X syndrome (FXS) is the most common single gene cause of intellectual disability and autism spectrum disorder. Caused by a silenced fragile X mental retardation 1 gene and the subsequent deficiency in fragile X mental retardation protein, patients with FXS experience a range of physical, behavioral, and intellectual debilitations. The FXS field, as a whole, has recently met with some challenges, as several targeted clinical trials with high expectations of success have failed to elucidate significant improvements in a variety of symptom domains. As new clinical trials in FXS are planned, there has been much discussion about the use of the commonly used clinical outcome measures, as well as study design considerations, patient stratification, and optimal age range for treatment. The evidence that modification of these drug targets and use of these failed compounds would prove to be efficacious in human clinical study were rooted in years of basic and translational research. There are questions arising as to the use of the mouse models for studying FXS treatment development. This issue is twofold: many of the symptom domains and molecular and biochemical changes assessed and indicative of efficacy in mouse model study are not easily amenable to clinical trials in people with FXS because of the intolerability of the testing paradigm or a lack of noninvasive techniques (prepulse inhibition, sensory hypersensitivity, startle reactivity, or electrophysiologic, biochemical, or structural changes in the brain); and capturing subtle yet meaningful changes in symptom domains such as sociability, anxiety, and hyperactivity in human FXS clinical trials is challenging with the currently used measures (typically parent/caregiver rating scales). Clinicians, researchers, and the pharmaceutical industry have all had to take a step back and critically evaluate the way we think about how to best optimize future investigations into pharmacologic FXS treatments. As new clinical trials are coming down the drug discovery pipeline, it is clear that the field is moving in a direction that values the development of molecular biomarkers, less subjective quantitative measures of symptom improvement, and rating scales developed specifically for use in FXS in conjunction with drug safety. While summarizing preclinical evidence, where applicable, and discussing challenges in FXS treatment development, this review details both completed clinical trials for the targeted and symptomatic treatment of FXS and introduces novel projects on the cusp of clinical trial investigation.
Collapse
Affiliation(s)
- Tori L Schaefer
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew H Davenport
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Craig A Erickson
- Division of Child and Adolescent Psychiatry, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
50
|
Romo-Parra H, Blaesse P, Sosulina L, Pape HC. Neurosteroids increase tonic GABAergic inhibition in the lateral section of the central amygdala in mice. J Neurophysiol 2015; 113:3421-31. [PMID: 25787948 DOI: 10.1152/jn.00045.2015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/12/2015] [Indexed: 01/12/2023] Open
Abstract
Neurosteroids are formed de novo in the brain and can modulate both inhibitory and excitatory neurotransmission. Recent evidence suggests that the anxiolytic effects of neurosteroids are mediated by the amygdala, a key structure for emotional and cognitive behaviors. Tonic inhibitory signaling via extrasynaptic type A γ-aminobutyric acid receptors (GABA(A)Rs) is known to be crucially involved in regulating network activity in various brain regions including subdivisions of the amygdala. Here we provide evidence for the existence of tonic GABAergic inhibition generated by the activation of δ-subunit-containing GABA(A)Rs in neurons of the lateral section of the mouse central amygdala (CeAl). Furthermore, we show that neurosteroids play an important role in the modulation of tonic GABAergic inhibition in the CeAl. Taken together, these findings provide new mechanistic insights into the effects of pharmacologically relevant neurosteroids in the amygdala and might be extrapolated to the regulation of anxiety.
Collapse
Affiliation(s)
- H Romo-Parra
- Institute of Physiology I, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - P Blaesse
- Institute of Physiology I, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - L Sosulina
- Institute of Physiology I, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - H-C Pape
- Institute of Physiology I, Westfälische Wilhelms-Universität Münster, Münster, Germany
| |
Collapse
|