1
|
Cesaroni CA, Pisanò G, Trimarchi G, Caraffi SG, Scandolo G, Gnazzo M, Frattini D, Spagnoli C, Rizzi S, Dittadi C, Sigona G, Garavelli L, Fusco C. Severe Neurodevelopmental Disorder in Autosomal Recessive Spinocerebellar Ataxia 13 (SCAR13) Caused by Two Novel Frameshift Variants in GRM1. CEREBELLUM (LONDON, ENGLAND) 2024; 23:1768-1771. [PMID: 37831383 DOI: 10.1007/s12311-023-01617-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/05/2023] [Indexed: 10/14/2023]
Abstract
Autosomal recessive spinocerebellar ataxia 13 (SCAR13) is a neurological disease characterized by psychomotor delay, mild to profound intellectual disability with poor or absent language, nystagmus, stance ataxia, and, if walking is acquired, gait ataxia. Epilepsy and polyneuropathy have also been documented in some patients. Cerebellar atrophy and/or ventriculomegaly may be present on brain MRI. SCAR13 is caused by pathogenic variants in the GRM1 gene encoding the metabotropic receptor of glutamate type 1 (mGlur1), which is highly expressed in Purkinje cerebellar cells, where it plays a fundamental role in cerebellar development. Here we discuss the case of an 8-year-old patient who presented with a severe neurodevelopmental disorder with balance disturbance, absence of independent walking, absence of language, diffuse hypotonia, mild nystagmus, and mild dysphagia. Whole-exome sequencing revealed a compound heterozygosity for two likely pathogenic variants in the GRM1 gene, responsible for the patient's phenotype, and made it possible to diagnose autosomal recessive spinocerebellar ataxia SCAR13. The detected (novel) variants appear to be causative of a particularly severe picture with regard to neurodevelopment, in the context of the typical neurological signs of spinocerebellar ataxia.
Collapse
Affiliation(s)
- Carlo Alberto Cesaroni
- Child Neurology and Psychiatry Unit, Pediatric Neurophysiology Laboratory, Mother-Child Department, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy.
| | - Giulia Pisanò
- Child Neurology and Psychiatry Unit, Pediatric Neurophysiology Laboratory, Mother-Child Department, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Gabriele Trimarchi
- Medical Genetics Unit, Mother-Child Department, Azienda USL-IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | - Stefano Giuseppe Caraffi
- Medical Genetics Unit, Mother-Child Department, Azienda USL-IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | - Giulia Scandolo
- Child Neurology and Psychiatry Unit, Pediatric Neurophysiology Laboratory, Mother-Child Department, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Martina Gnazzo
- Child Neurology and Psychiatry Unit, Pediatric Neurophysiology Laboratory, Mother-Child Department, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Daniele Frattini
- Child Neurology and Psychiatry Unit, Pediatric Neurophysiology Laboratory, Mother-Child Department, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Carlotta Spagnoli
- Child Neurology and Psychiatry Unit, Pediatric Neurophysiology Laboratory, Mother-Child Department, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Susanna Rizzi
- Child Neurology and Psychiatry Unit, Pediatric Neurophysiology Laboratory, Mother-Child Department, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Claudia Dittadi
- Child Neurology and Psychiatry Unit, Pediatric Neurophysiology Laboratory, Mother-Child Department, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Giulia Sigona
- Child Neurology and Psychiatry Unit, Pediatric Neurophysiology Laboratory, Mother-Child Department, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Livia Garavelli
- Medical Genetics Unit, Mother-Child Department, Azienda USL-IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | - Carlo Fusco
- Child Neurology and Psychiatry Unit, Pediatric Neurophysiology Laboratory, Mother-Child Department, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
2
|
Islam F, Roy S, Zehravi M, Paul S, Sutradhar H, Yaidikar L, Kumar BR, Dogiparthi LK, Prema S, Nainu F, Rab SO, Doukani K, Emran TB. Polyphenols Targeting MAP Kinase Signaling Pathway in Neurological Diseases: Understanding Molecular Mechanisms and Therapeutic Targets. Mol Neurobiol 2024; 61:2686-2706. [PMID: 37922063 DOI: 10.1007/s12035-023-03706-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/10/2023] [Indexed: 11/05/2023]
Abstract
Polyphenols are a class of secondary metabolic products found in plants that have been extensively studied for how well they regulate biological processes, such as the proliferation of cells, autophagy, and apoptosis. The mitogen-activated protein kinase (MAPK)-mediated signaling cascade is currently identified as a crucial pro-inflammatory pathway that plays a significant role in the development of neuroinflammation. This process has been shown to contribute to the pathogenesis of several neurological conditions, such as Alzheimer's disease (AD), Parkinson's disease (PD), CNS damage, and cerebral ischemia. Getting enough polyphenols through eating habits has resulted in mitigating the effects of oxidative stress (OS) and lowering the susceptibility to associated neurodegenerative disorders, including but not limited to multiple sclerosis (MS), AD, stroke, and PD. Polyphenols possess significant promise in dealing with the root cause of neurological conditions by modulating multiple therapeutic targets simultaneously, thereby attenuating their complicated physiology. Several polyphenolic substances have demonstrated beneficial results in various studies and are presently undergoing clinical investigation to treat neurological diseases (NDs). The objective of this review is to provide a comprehensive summary of the different aspects of the MAPK pathway involved in neurological conditions, along with an appraisal of the progress made in using polyphenols to regulate the MAPK signaling system to facilitate the management of NDs.
Collapse
Affiliation(s)
- Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Sumon Roy
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy, College of Dentistry & Pharmacy, Buraydah Private Colleges, Buraydah, 51418, Kingdom of Saudi Arabia.
| | - Shyamjit Paul
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Hriday Sutradhar
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Lavanya Yaidikar
- Department of Pharmacology, Seven Hills College of Pharmacy, Tirupati, India
| | - B Raj Kumar
- Department of Pharmaceutical Analysis, Moonray Institute of Pharmaceutical Sciences, Raikal (V), Farooq Nagar (Tlq), Shadnagar (M), R.R Dist., Telangana, 501512, India
| | - Lakshman Kumar Dogiparthi
- Department of Pharmacognosy, MB School of Pharmaceutical Sciences, MBU, Tirupati, Andhra Pradesh, India
| | - S Prema
- Crescent School of Pharmacy, BS Abdur Rahman Crescent Institute of Science and Technology, Vandalur, Chennai, 600048, India
| | - Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, 90245, Indonesia
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Koula Doukani
- Faculty of Nature and Life Sciences, University of Ibn Khaldoun-Tiaret, Tiaret, Algeria
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh.
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School & Legorreta Cancer Center, Brown University, Providence, RI, 02912, USA.
| |
Collapse
|
3
|
Jiang J, Wang D, Jiang Y, Yang X, Sun R, Chang J, Zhu W, Yao P, Song K, Chang S, Wang H, Zhou L, Zhang XS, Li H, Li N. The gut metabolite indole-3-propionic acid activates ERK1 to restore social function and hippocampal inhibitory synaptic transmission in a 16p11.2 microdeletion mouse model. MICROBIOME 2024; 12:66. [PMID: 38549163 PMCID: PMC10976717 DOI: 10.1186/s40168-024-01755-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 01/04/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Microdeletion of the human chromosomal region 16p11.2 (16p11.2+ / - ) is a prevalent genetic factor associated with autism spectrum disorder (ASD) and other neurodevelopmental disorders. However its pathogenic mechanism remains unclear, and effective treatments for 16p11.2+ / - syndrome are lacking. Emerging evidence suggests that the gut microbiota and its metabolites are inextricably linked to host behavior through the gut-brain axis and are therefore implicated in ASD development. Despite this, the functional roles of microbial metabolites in the context of 16p11.2+ / - are yet to be elucidated. This study aims to investigate the therapeutic potential of indole-3-propionic acid (IPA), a gut microbiota metabolite, in addressing behavioral and neural deficits associated with 16p11.2+ / - , as well as the underlying molecular mechanisms. RESULTS Mice with the 16p11.2+ / - showed dysbiosis of the gut microbiota and a significant decrease in IPA levels in feces and blood circulation. Further, these mice exhibited significant social and cognitive memory impairments, along with hyperactivation of hippocampal dentate gyrus neurons and reduced inhibitory synaptic transmission in this region. However, oral administration of IPA effectively mitigated the histological and electrophysiological alterations, thereby ameliorating the social and cognitive deficits of the mice. Remarkably, IPA treatment significantly increased the phosphorylation level of ERK1, a protein encoded by the Mapk3 gene in the 16p11.2 region, without affecting the transcription and translation of the Mapk3 gene. CONCLUSIONS Our study reveals that 16p11.2+ / - leads to a decline in gut metabolite IPA levels; however, IPA supplementation notably reverses the behavioral and neural phenotypes of 16p11.2+ / - mice. These findings provide new insights into the critical role of gut microbial metabolites in ASD pathogenesis and present a promising treatment strategy for social and cognitive memory deficit disorders, such as 16p11.2 microdeletion syndrome. Video Abstract.
Collapse
Affiliation(s)
- Jian Jiang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Dilong Wang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
- Department of Pediatrics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Youheng Jiang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Xiuyan Yang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Runfeng Sun
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jinlong Chang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Wenhui Zhu
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Peijia Yao
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Kun Song
- Brain Research Centre, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Shuwen Chang
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hong Wang
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen-Hong Kong Institute of Brain Science Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Lei Zhou
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Xue-Song Zhang
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, USA.
| | - Huiliang Li
- Wolfson Institute for Biomedical Research, Division of Medicine, Faculty of Medical Sciences, University College London, London, UK.
| | - Ningning Li
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China.
- China-UK Institute for Frontier Science, Shenzhen, China.
- Department of Anesthesiology, The Afliated Hospital of Youjiang Medical University for Nationalities, Baise, China.
| |
Collapse
|
4
|
Lee KY, Wang H, Yook Y, Rhodes JS, Christian-Hinman CA, Tsai NP. Tumor suppressor p53 modulates activity-dependent synapse strengthening, autism-like behavior and hippocampus-dependent learning. Mol Psychiatry 2023; 28:3782-3794. [PMID: 37759036 PMCID: PMC11392564 DOI: 10.1038/s41380-023-02268-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/30/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023]
Abstract
Synaptic potentiation underlies various forms of behavior and depends on modulation by multiple activity-dependent transcription factors to coordinate the expression of genes necessary for sustaining synaptic transmission. Our current study identified the tumor suppressor p53 as a novel transcription factor involved in this process. We first revealed that p53 could be elevated upon chemically induced long-term potentiation (cLTP) in cultured primary neurons. By knocking down p53 in neurons, we further showed that p53 is required for cLTP-induced elevation of surface GluA1 and GluA2 subunits of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR). Because LTP is one of the principal plasticity mechanisms underlying behaviors, we employed forebrain-specific knockdown of p53 to evaluate the role of p53 in behavior. Our results showed that, while knocking down p53 in mice does not alter locomotion or anxiety-like behavior, it significantly promotes repetitive behavior and reduces sociability in mice of both sexes. In addition, knocking down p53 also impairs hippocampal LTP and hippocampus-dependent learning and memory. Most importantly, these learning-associated defects are more pronounced in male mice than in female mice, suggesting a sex-specific role of p53 in these behaviors. Using RNA sequencing (RNAseq) to identify p53-associated genes in the hippocampus, we showed that knocking down p53 up- or down-regulates multiple genes with known functions in synaptic plasticity and neurodevelopment. Altogether, our study suggests p53 as an activity-dependent transcription factor that mediates the surface expression of AMPAR, permits hippocampal synaptic plasticity, represses autism-like behavior, and promotes hippocampus-dependent learning and memory.
Collapse
Affiliation(s)
- 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.
| | - Haohan Wang
- School of Information Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yeeun Yook
- Department of Molecular and Integrative Physiology, School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Justin S Rhodes
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Psychology, University of Illinois at Urbana-Champaign, Champaign, IL, 61820, USA
| | - Catherine A Christian-Hinman
- 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
- Beckman Institute for Advanced Science and Technology, 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.
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
5
|
Murari K, Abushaibah A, Rho JM, Turner RW, Cheng N. A clinically relevant selective ERK-pathway inhibitor reverses core deficits in a mouse model of autism. EBioMedicine 2023; 91:104565. [PMID: 37088035 PMCID: PMC10149189 DOI: 10.1016/j.ebiom.2023.104565] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 03/07/2023] [Accepted: 03/29/2023] [Indexed: 04/25/2023] Open
Abstract
BACKGROUND Extracellular signal-regulated kinase (ERK/MAPK) pathway in the brain is hypothesized to be a critical convergent node in the development of autism spectrum disorder. We reasoned that selectively targeting this pathway could reverse core autism-like phenotype in animal models. METHODS Here we tested a clinically relevant, selective inhibitor of ERK pathway, PD325901 (Mirdametinib), in a mouse model of idiopathic autism, the BTBR mice. FINDINGS We report that treating juvenile mice with PD325901 reduced ERK pathway activation, dose and duration-dependently reduced core disease-modeling deficits in sociability, vocalization and repetitive behavior, and reversed abnormal EEG signals. Further analysis revealed that subchronic treatment did not affect weight gain, locomotion, or neuronal density in the brain. Parallel treatment in the C57BL/6J mice did not alter their phenotype. INTERPRETATION Our data indicate that selectively inhibiting ERK pathway using PD325901 is beneficial in the BTBR model, thus further support the notion that ERK pathway is critically involved in the pathophysiology of autism. These results suggest that a similar approach could be applied to animal models of syndromic autism with dysregulated ERK signaling, to further test selectively targeting ERK pathway as a new approach for treating autism. FUNDING This has beenwork was supported by Alberta Children's Hospital Research Foundation (JMR & NC), University of Calgary Faculty of Veterinary Medicine (NC), Kids Brain Health Network (NC), and Natural Sciences and Engineering Research Council of Canada (NC).
Collapse
Affiliation(s)
- Kartikeya Murari
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Canada; Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Canada; Department of Electrical and Software Engineering, Schulich School of Engineering, University of Calgary, Canada
| | - Abdulrahman Abushaibah
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Canada; Bachelor of Health Sciences, Cumming School of Medicine, University of Calgary, Canada
| | - Jong M Rho
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Canada; Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Canada
| | - Ray W Turner
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Canada; Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Canada; Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Canada
| | - Ning Cheng
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Canada; Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Canada; Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Canada.
| |
Collapse
|
6
|
Singh A, Hadjinicolaou A, Peters JM, Salussolia CL. Treatment-Resistant Epilepsy and Tuberous Sclerosis Complex: Treatment, Maintenance, and Future Directions. Neuropsychiatr Dis Treat 2023; 19:733-748. [PMID: 37041855 PMCID: PMC10083014 DOI: 10.2147/ndt.s347327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/22/2023] [Indexed: 04/13/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is a neurogenetic disorder that affects multiple organ systems, including the heart, kidneys, eyes, skin, and central nervous system. The neurologic manifestations have the highest morbidity and mortality, in particular in children. Clinically, patients with TSC often present with new-onset seizures within the first year of life. TSC-associated epilepsy is often difficult to treat and refractory to multiple antiseizure medications. Refractory TSC-associated epilepsy is associated with increased risk of neurodevelopmental comorbidities, including developmental delay, intellectual disability, autism spectrum disorder, and attention hyperactivity disorder. An increasing body of research suggests that early, effective treatment of TSC-associated epilepsy during critical neurodevelopmental periods can potentially improve cognitive outcomes. Therefore, it is important to treat TSC-associated epilepsy aggressively, whether it be with pharmacological therapy, surgical intervention, and/or neuromodulation. This review discusses current and future pharmacological treatments for TSC-associated epilepsy, as well as the importance of early surgical evaluation for refractory epilepsy in children with TSC and consideration of neuromodulatory interventions in young adults.
Collapse
Affiliation(s)
- Avantika Singh
- Division of Epilepsy and Neurophysiology, Department of Neurology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Aristides Hadjinicolaou
- Division of Epilepsy and Neurophysiology, Department of Neurology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Jurriaan M Peters
- Division of Epilepsy and Neurophysiology, Department of Neurology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Catherine L Salussolia
- Division of Epilepsy and Neurophysiology, Department of Neurology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
- Correspondence: Catherine L Salussolia, 3 Blackfan Circle, Center for Life Sciences 14060, Boston, MA, 02115, USA, Tel +617-355-7970, Email
| |
Collapse
|
7
|
Matrisciano F, Locci V, Dong E, Nicoletti F, Guidotti A, Grayson DR. Altered Expression and In Vivo Activity of mGlu5 Variant a Receptors in the Striatum of BTBR Mice: Novel Insights Into the Pathophysiology of Adult Idiopathic Forms of Autism Spectrum Disorders. Curr Neuropharmacol 2022; 20:2354-2368. [PMID: 35139800 PMCID: PMC9890299 DOI: 10.2174/1567202619999220209112609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND mGlu5 metabotropic glutamate receptors are considered as candidate drug targets in the treatment of "monogenic" forms of autism spectrum disorders (ASD), such as Fragile- X syndrome (FXS). However, despite promising preclinical data, clinical trials using mGlu5 receptor antagonists to treat FXS showed no beneficial effects. OBJECTIVE Here, we studied the expression and function of mGlu5 receptors in the striatum of adult BTBR mice, which model idiopathic forms of ASD, and behavioral phenotype. METHODS Behavioral tests were associated with biochemistry analysis including qPCR and western blot for mRNA and protein expression. In vivo analysis of polyphosphoinositides hydrolysis was performed to study the mGlu5-mediated intracellular signaling in the striatum of adult BTBR mice under basal conditions and after MTEP exposure. RESULTS Expression of mGlu5 receptors and mGlu5 receptor-mediated polyphosphoinositides hydrolysis were considerably high in the striatum of BTBR mice, sensitive to MTEP treatment. Changes in the expression of genes encoding for proteins involved in excitatory and inhibitory neurotransmission and synaptic plasticity, including Fmr1, Dlg4, Shank3, Brd4, bdnf-exon IX, Mef2c, and Arc, GriA2, Glun1, Nr2A, and Grm1, Grm2, GriA1, and Gad1 were also found. Behaviorally, BTBR mice showed high repetitive stereotypical behaviors, including self-grooming and deficits in social interactions. Acute or repeated injections with MTEP reversed the stereotyped behavior and the social interaction deficit. Similar effects were observed with the NMDA receptor blockers MK-801 or ketamine. CONCLUSION These findings support a pivotal role of mGlu5 receptor abnormal expression and function in idiopathic ASD adult forms and unveil novel potential targets for therapy.
Collapse
Affiliation(s)
- Francesco Matrisciano
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Valentina Locci
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Erbo Dong
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Center for Alcohol Research in Epigenetics Department of Psychiatry College of Medicine University of Illinois Chicago, Chicago, IL 60612, USA
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Alessandro Guidotti
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Center for Alcohol Research in Epigenetics Department of Psychiatry College of Medicine University of Illinois Chicago, Chicago, IL 60612, USA
| | - Dennis R. Grayson
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Center for Alcohol Research in Epigenetics Department of Psychiatry College of Medicine University of Illinois Chicago, Chicago, IL 60612, USA
| |
Collapse
|
8
|
Atypical perineuronal nets in the CA2 region interfere with social memory in a mouse model of social dysfunction. Mol Psychiatry 2022; 27:3520-3531. [PMID: 34183768 PMCID: PMC8712624 DOI: 10.1038/s41380-021-01174-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 05/04/2021] [Accepted: 05/12/2021] [Indexed: 12/16/2022]
Abstract
Social memory dysfunction is an especially devastating symptom of many neuropsychiatric disorders, which makes understanding the cellular and molecular processes that contribute to such abnormalities important. Evidence suggests that the hippocampus, particularly the CA2 region, plays an important role in social memory. We sought to identify potential mechanisms of social memory dysfunction in the hippocampus by investigating features of neurons, glia, and the extracellular matrix (ECM) of BTBR mice, an inbred mouse strain with deficient social memory. The CA2 is known to receive inputs from dentate gyrus adult-born granule cells (abGCs), neurons known to participate in social memory, so we examined this cell population and found fewer abGCs, as well as fewer axons from abGCs in the CA2 of BTBR mice compared to controls. We also found that BTBR mice had fewer pyramidal cell dendritic spines, in addition to fewer microglia and astrocytes, in the CA2 compared to controls. Along with diminished neuronal and glial elements, we found atypical perineuronal nets (PNNs), specialized ECM structures that regulate plasticity, in the CA2 of BTBR mice. By diminishing PNNs in the CA2 of BTBR mice to control levels, we observed a partial restoration of social memory. Our findings suggest that the CA2 region of BTBR mice exhibits multiple cellular and extracellular abnormalities and identify atypical PNNs as one mechanism producing social memory dysfunction, although the contribution of reduced abGC afferents, pyramidal cell dendritic spine, and glial cell numbers remains unexplored.
Collapse
|
9
|
Taheri F, Esmaeilpour K, Sepehri G, Sheibani V, Ur Rehman N, Maneshian M. Histamine H3 receptor antagonist, ciproxifan, alleviates cognition and synaptic plasticity alterations in a valproic acid-induced animal model of autism. Psychopharmacology (Berl) 2022; 239:2673-2693. [PMID: 35538250 DOI: 10.1007/s00213-022-06155-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 04/21/2022] [Indexed: 11/27/2022]
Abstract
RATIONALE Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by deficits in social communication and cognitive behaviors. Histamine H3 receptor (H3R) antagonists are considered as therapeutic factors for treating cognitive impairments. OBJECTIVES The aim of the present study was to evaluate the effects of the H3R antagonist, ciproxifan (CPX), on cognition impairment especially, spatial learning memory, and synaptic plasticity in the CA1 region of the hippocampus in autistic rats. METHODS Pregnant rats were injected with either valproic acid (VPA) (600 mg/kg, i.p.) or saline on an embryonic day 12.5 (E12.5). The effects of the H3R antagonist, ciproxifan (CPX) (1, 3 mg/kg, i.p.), were investigated on learning and memory in VPA-exposed rat pups and saline-exposed rat pups using Morris water maze (MWM) and social interaction tasks. The H2R antagonist, famotidine (FAM) (10, 20, 40 mg/kg, i.p.), was used to determine whether brain histaminergic neurotransmission exerted its procognitive effects through the H2R. In addition, synaptic reinforcement was evaluated by in vivo field potential recording. RESULTS The results showed that VPA-exposed rat pups had significantly lower sociability and social memory performance compared to the saline rats. VPA-exposed rat pups exhibited learning and memory impairments in the MWM task. In addition, VPA caused suppression of long-term potentiation (LTP) in the CA1 area of the hippocampus. Our results demonstrated that CPX 3 mg/kg improved VPA-induced cognitive impairments and FAM 20 mg/kg attenuated cognitive behaviors as well as electrophysiological properties. CONCLUSIONS CPX 3 mg/kg improved VPA-induced impairments of LTP as well as learning and memory deficits through H2R.
Collapse
Affiliation(s)
- Farahnaz Taheri
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Khadijeh Esmaeilpour
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
- Physics and Astronomy Department, University of Waterloo, Waterloo, Ontario, Canada.
| | - Gholamreza Sepehri
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Vahid Sheibani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Naeem Ur Rehman
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, Islamabad, Pakistan
| | - Marzieh Maneshian
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
10
|
Caruso A, Ricceri L, Caruso A, Nicoletti F, Gaetano A, Scaccianoce S. Postweaning social isolation and autism-like phenotype: a biochemical and behavioral comparative analysis. Behav Brain Res 2022; 428:113891. [PMID: 35421428 DOI: 10.1016/j.bbr.2022.113891] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/15/2022] [Accepted: 04/07/2022] [Indexed: 12/16/2022]
Abstract
Adolescence is a critical period for brain development. In most mammalian species, disturbances experienced during adolescence constitute a risk factor for several neuropsychiatric disorders. In this study, we compared the biochemical and behavioral profile induced by postweaning social isolation (PWSI) in inbred C57BL/6N mice with that of BTBR mice, a rodent model of autism spectrum disorders. Male C57BL/6N mice were either housed in groups of four or isolated from weaning (postnatal day 21) for four weeks before experimental analyses. After weaning, male BTBR mice were housed four per cage and analyzed at 48 days of age. PWSI reduced hippocampal levels of type 2 metabotropic glutamate (mGlu2) receptors, and glucocorticoid and mineralocorticoid receptors. A similar reduction was seen in group-housed BTBR mice. Plasma corticosterone levels in basal conditions were not influenced by PWSI, but were increased in BTBR mice. Social investigation (total and head sniffing) and the number of ultrasonic vocalizations were reduced in both PWSI mice and age-matched group-housed BTBR mice, indicating a lower social responsiveness in both groups of mice. These results suggest that absence of social stimuli during adolescence induces an endophenotype with social deficit features, which mimics the phenotype of a mouse model of autism spectrum disorders.
Collapse
Affiliation(s)
- Alessandra Caruso
- Department of Physiology and Pharmacology "V. Erspamer" University Sapienza of Rome, Italy.
| | - Laura Ricceri
- Centre for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy.
| | - Angela Caruso
- Research Coordination and Support Service, Istituto Superiore di Sanità, Rome, Italy.
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology "V. Erspamer" University Sapienza of Rome, Italy; IRCCS Neuromed, Pozzilli, Italy.
| | - Alessandra Gaetano
- Department of Physiology and Pharmacology "V. Erspamer" University Sapienza of Rome, Italy.
| | - Sergio Scaccianoce
- Department of Physiology and Pharmacology "V. Erspamer" University Sapienza of Rome, Italy.
| |
Collapse
|
11
|
Chen J, Zhang C, Wang R, Jiang P, Cai H, Zhao W, Zhu J, Yu Y. Molecular basis underlying functional connectivity of fusiform gyrus subregions: A transcriptome-neuroimaging spatial correlation study. Cortex 2022; 152:59-73. [DOI: 10.1016/j.cortex.2022.03.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 02/13/2022] [Accepted: 03/30/2022] [Indexed: 01/07/2023]
|
12
|
Kanlayaprasit S, Thongkorn S, Panjabud P, Jindatip D, Hu VW, Kikkawa T, Osumi N, Sarachana T. Autism-Related Transcription Factors Underlying the Sex-Specific Effects of Prenatal Bisphenol A Exposure on Transcriptome-Interactome Profiles in the Offspring Prefrontal Cortex. Int J Mol Sci 2021; 22:13201. [PMID: 34947998 PMCID: PMC8708761 DOI: 10.3390/ijms222413201] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/03/2021] [Accepted: 12/05/2021] [Indexed: 11/16/2022] Open
Abstract
Bisphenol A (BPA) is an environmental risk factor for autism spectrum disorder (ASD). BPA exposure dysregulates ASD-related genes in the hippocampus and neurological functions of offspring. However, whether prenatal BPA exposure has an impact on genes in the prefrontal cortex, another brain region highly implicated in ASD, and through what mechanisms have not been investigated. Here, we demonstrated that prenatal BPA exposure disrupts the transcriptome-interactome profiles of the prefrontal cortex of neonatal rats. Interestingly, the list of BPA-responsive genes was significantly enriched with known ASD candidate genes, as well as genes that were dysregulated in the postmortem brain tissues of ASD cases from multiple independent studies. Moreover, several differentially expressed genes in the offspring's prefrontal cortex were the targets of ASD-related transcription factors, including AR, ESR1, and RORA. The hypergeometric distribution analysis revealed that BPA may regulate the expression of such genes through these transcription factors in a sex-dependent manner. The molecular docking analysis of BPA and ASD-related transcription factors revealed novel potential targets of BPA, including RORA, SOX5, TCF4, and YY1. Our findings indicated that prenatal BPA exposure disrupts ASD-related genes in the offspring's prefrontal cortex and may increase the risk of ASD through sex-dependent molecular mechanisms, which should be investigated further.
Collapse
Grants
- FRB65_hea(80)_175_37_05 Fundamental Fund, Chulalongkorn University
- AHS-CU 61004 Faculty of Allied Health Sciences Research Fund, Chulalongkorn University
- GRU 6300437001-1 Ratchadapisek Somphot Fund for Supporting Research Unit, Chulalongkorn University
- GRU_64_033_37_004 Ratchadapisek Somphot Fund for Supporting Research Unit, Chulalongkorn University
- The 100th Anniversary Chulalongkorn University Fund for Doctoral Scholarship, Graduate School, Chulalongkorn University
- The Overseas Research Experience Scholarship for Graduate Students from Graduate School, Chulalongkorn University
- PHD/0029/2561 The Royal Golden Jubilee Ph.D. Programme Scholarship, Thailand Research Fund and National Research Council of Thailand
- National Research Council of Thailand (NRCT)
- GCUGR1125623067D-67 The 90th Anniversary Chulalongkorn University Fund (Ratchadaphiseksomphot Endowment Fund), Graduate School, Chulalongkorn University
- GCUGR1125632108D-108 The 90th Anniversary Chulalongkorn University Fund (Ratchadaphiseksomphot Endowment Fund), Graduate School, Chulalongkorn University
- 2073011 Chulalongkorn University Laboratory Animal Center (CULAC) Grant
Collapse
Affiliation(s)
- Songphon Kanlayaprasit
- The Ph.D. Program in Clinical Biochemistry and Molecular Medicine, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (S.K.); (S.T.); (P.P.)
| | - Surangrat Thongkorn
- The Ph.D. Program in Clinical Biochemistry and Molecular Medicine, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (S.K.); (S.T.); (P.P.)
| | - Pawinee Panjabud
- The Ph.D. Program in Clinical Biochemistry and Molecular Medicine, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (S.K.); (S.T.); (P.P.)
| | - Depicha Jindatip
- Systems Neuroscience of Autism and PSychiatric Disorders (SYNAPS) Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
- Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Valerie W. Hu
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA;
| | - Takako Kikkawa
- Department of Developmental Neuroscience, United Centers for Advanced Research and Translational Medicine (ART), Tohoku University Graduate School of Medicine, Sendai 980-8577, Miyagi, Japan; (T.K.); (N.O.)
| | - Noriko Osumi
- Department of Developmental Neuroscience, United Centers for Advanced Research and Translational Medicine (ART), Tohoku University Graduate School of Medicine, Sendai 980-8577, Miyagi, Japan; (T.K.); (N.O.)
| | - Tewarit Sarachana
- Systems Neuroscience of Autism and PSychiatric Disorders (SYNAPS) Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
| |
Collapse
|
13
|
ERK/MAPK signalling in the developing brain: Perturbations and consequences. Neurosci Biobehav Rev 2021; 131:792-805. [PMID: 34634357 DOI: 10.1016/j.neubiorev.2021.10.009] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 09/26/2021] [Accepted: 10/05/2021] [Indexed: 12/18/2022]
Abstract
The extracellular regulated kinase/microtubule-associated protein kinase (ERK/MAPK) signalling pathway transduces signals that cause an alteration in the ongoing metabolic pathways and modifies gene expression patterns; thus, influencing cellular behaviour. ERK/MAPK signalling is essential for the proper development of the nervous system from neural progenitor cells derived from the embryonic mesoderm. Several signalling molecules that regulate the well-coordinated process of neurodevelopment transduce developmental information through the ERK/MAPK signalling pathway. The ERK/MAPK is a potential novel therapeutic target in several neurodevelopmental disorders, however, despite years of study, there is still significant uncertainty about the exact mechanism by which the ERK/MAPK signalling pathway elicits specific responses in neurodevelopment. Here, we will review the evidence highlighting the role of ERK/MAPK signalling in neurodevelopment. We will also discuss the structural implication and behavioural deficits associated with perturbed ERK/MAPK signalling pathway in cortical development, whilst examining its contribution to the neuropathology of several neurodevelopmental disorders, such as Autism Spectrum Disorder, Schizophrenia, Fragile X, and Attention Deficit Hyperactive Disorder.
Collapse
|
14
|
Ash RT, Buffington SA, Park J, Suter B, Costa-Mattioli M, Zoghbi HY, Smirnakis SM. Inhibition of Elevated Ras-MAPK Signaling Normalizes Enhanced Motor Learning and Excessive Clustered Dendritic Spine Stabilization in the MECP2-Duplication Syndrome Mouse Model of Autism. eNeuro 2021; 8:ENEURO.0056-21.2021. [PMID: 34021030 PMCID: PMC8260274 DOI: 10.1523/eneuro.0056-21.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/26/2022] Open
Abstract
The inflexible repetitive behaviors and "insistence on sameness" seen in autism imply a defect in neural processes controlling the balance between stability and plasticity of synaptic connections in the brain. It has been proposed that abnormalities in the Ras-ERK/MAPK pathway, a key plasticity-related cell signaling pathway known to drive consolidation of clustered synaptic connections, underlie altered learning phenotypes in autism. However, a link between altered Ras-ERK signaling and clustered dendritic spine plasticity has yet to be explored in an autism animal model in vivo The formation and stabilization of dendritic spine clusters is abnormally increased in the MECP2-duplication syndrome mouse model of syndromic autism, suggesting that ERK signaling may be increased. Here, we show that the Ras-ERK pathway is indeed hyperactive following motor training in MECP2-duplication mouse motor cortex. Pharmacological inhibition of ERK signaling normalizes the excessive clustered spine stabilization and enhanced motor learning behavior in MECP2-duplication mice. We conclude that hyperactive ERK signaling may contribute to abnormal clustered dendritic spine consolidation and motor learning in this model of syndromic autism.
Collapse
Affiliation(s)
- Ryan Thomas Ash
- Department of Psychiatry and Behavioral Sciences, Stanford University, CA 94305
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030
- Department of Neurology, Brigham and Women's Hospital and Jamaica Plain Veterans Administration Hospital, Harvard Medical School, Boston, MA 02115
| | - Shelly Alexandra Buffington
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX 77030
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, TX 77555
| | - Jiyoung Park
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Department of Neurology, Brigham and Women's Hospital and Jamaica Plain Veterans Administration Hospital, Harvard Medical School, Boston, MA 02115
| | - Bernhard Suter
- Department of Neurology, Brigham and Women's Hospital and Jamaica Plain Veterans Administration Hospital, Harvard Medical School, Boston, MA 02115
- Department of Pediatrics, Texas Children's Hospital and Baylor College of Medicine, Houston, TX 77030
| | - Mauro Costa-Mattioli
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX 77030
| | - Huda Yaya Zoghbi
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Department of Pediatrics, Texas Children's Hospital and Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030
| | - Stelios Manolis Smirnakis
- Department of Neurology, Brigham and Women's Hospital and Jamaica Plain Veterans Administration Hospital, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
15
|
Su LD, Wang N, Han J, Shen Y. Group 1 Metabotropic Glutamate Receptors in Neurological and Psychiatric Diseases: Mechanisms and Prospective. Neuroscientist 2021; 28:453-468. [PMID: 34088252 PMCID: PMC9449437 DOI: 10.1177/10738584211021018] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Metabotropic glutamate receptors (mGluRs) are G-protein coupled receptors
that are activated by glutamate in the central nervous system (CNS).
Basically, mGluRs contribute to fine-tuning of synaptic efficacy and
control the accuracy and sharpness of neurotransmission. Among eight
subtypes, mGluR1 and mGluR5 belong to group 1 (Gp1) family, and are
implicated in multiple CNS disorders, such as Alzheimer’s disease,
autism, Parkinson’s disease, and so on. In the present review, we
systematically discussed underlying mechanisms and prospective of Gp1
mGluRs in a group of neurological and psychiatric diseases, including
Alzheimer’s disease, Parkinson’s disease, autism spectrum disorder,
epilepsy, Huntington’s disease, intellectual disability, Down’s
syndrome, Rett syndrome, attention-deficit hyperactivity disorder,
addiction, anxiety, nociception, schizophrenia, and depression, in
order to provide more insights into the therapeutic potential of Gp1
mGluRs.
Collapse
Affiliation(s)
- Li-Da Su
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Na Wang
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Junhai Han
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Ying Shen
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
16
|
Chang H, Cai X, Li HJ, Liu WP, Zhao LJ, Zhang CY, Wang JY, Liu JW, Ma XL, Wang L, Yao YG, Luo XJ, Li M, Xiao X. Functional Genomics Identify a Regulatory Risk Variation rs4420550 in the 16p11.2 Schizophrenia-Associated Locus. Biol Psychiatry 2021; 89:246-255. [PMID: 33246552 DOI: 10.1016/j.biopsych.2020.09.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 09/17/2020] [Accepted: 09/17/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Genome-wide association studies (GWASs) have reported hundreds of genomic loci associated with schizophrenia, yet identifying the functional risk variations is a key step in elucidating the underlying mechanisms. METHODS We applied multiple bioinformatics and molecular approaches, including expression quantitative trait loci analyses, epigenome signature identification, luciferase reporter assay, chromatin conformation capture, homology-directed genome editing by CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/Cas9), RNA sequencing, and ATAC-Seq (assay for transposase-accessible chromatin using sequencing). RESULTS We found that the schizophrenia GWAS risk variations at 16p11.2 were significantly associated with messenger RNA levels of multiple genes in human brain, and one of the leading expression quantitative trait loci genes, MAPK3, is located ∼200 kb away from these risk variations in the genome. Further analyses based on the epigenome marks in human brain and cell lines suggested that a noncoding single nucleotide polymorphism, rs4420550 (p = 2.36 × 10-9 in schizophrenia GWAS), was within a DNA enhancer region, which was validated via in vitro luciferase reporter assays. The chromatin conformation capture experiment showed that the rs4420550 region physically interacted with the MAPK3 promoter and TAOK2 promoter. Precise CRISPR/Cas9 editing of a single base pair in cells followed by RNA sequencing further confirmed the regulatory effects of rs4420550 on the transcription of 16p11.2 genes, and ATAC-Seq demonstrated that rs4420550 affected chromatin accessibility at the 16p11.2 region. The rs4420550-[A/A] cells showed significantly higher proliferation rates compared with rs4420550-[G/G] cells. CONCLUSIONS These results together suggest that rs4420550 is a functional risk variation, and this study illustrates an example of comprehensive functional characterization of schizophrenia GWAS risk loci.
Collapse
Affiliation(s)
- Hong Chang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Shanghai, China
| | - Xin Cai
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Shanghai, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Shanghai, China
| | - Hui-Juan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Shanghai, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Shanghai, China
| | - Wei-Peng Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Shanghai, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Shanghai, China
| | - Li-Juan Zhao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Shanghai, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Shanghai, China
| | - Chu-Yi Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Shanghai, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Shanghai, China
| | - Jun-Yang Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Shanghai, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Shanghai, China
| | - Jie-Wei Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Shanghai, China
| | - Xiao-Lei Ma
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Shanghai, China
| | - Lu Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Shanghai, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Shanghai, China; KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Shanghai, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xiong-Jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Shanghai, China; KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Shanghai, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Shanghai, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Shanghai, China; KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Shanghai, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
17
|
Mahmood HM, Aldhalaan HM, Alshammari TK, Alqasem MA, Alshammari MA, Albekairi NA, AlSharari SD. The Role of Nicotinic Receptors in the Attenuation of Autism-Related Behaviors in a Murine BTBR T + tf/J Autistic Model. Autism Res 2020; 13:1311-1334. [PMID: 32691528 DOI: 10.1002/aur.2342] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 04/28/2020] [Accepted: 05/31/2020] [Indexed: 12/15/2022]
Abstract
Nicotinic receptors are distributed throughout the central and peripheral nervous system. Postmortem studies have reported that some nicotinic receptor subtypes are altered in the brains of autistic people. Recent studies have demonstrated the importance of nicotinic acetylcholine receptors (nAChRs) in the autistic behavior of BTBR T + tf/J mouse model of autism. This study was undertaken to examine the behavioral effects of targeted nAChRs using pharmacological ligands, including nicotine and mecamylamine in BTBR T + tf/J and C57BL/6J mice in a panel of behavioral tests relating to autism. These behavioral tests included the three-chamber social interaction, self-grooming, marble burying, locomotor activity, and rotarod test. We examined the effect of various oral doses of nicotine (50, 100, and 400 mcg/mL; po) over a period of 2 weeks in BTBR T + tf/J mouse model. The results indicated that the chronic administration of nicotine modulated sociability and repetitive behavior in BTBR T + tf/J mice while no effects observed in C57BL/6J mice. Furthermore, the nonselective nAChR antagonist, mecamylamine, reversed nicotine effects on sociability and increased repetitive behaviors in BTBR T + tf/J mice. Overall, the findings indicate that the pharmacological modulation of nicotinic receptors is involved in modulating core behavioral phenotypes in the BTBR T + tf/J mouse model. LAY SUMMARY: The involvement of brain nicotinic neurotransmission system plays a crucial role in regulating autism-related behavioral features. In addition, the brain of the autistic-like mouse model has a low acetylcholine level. Here, we report that nicotine, at certain doses, improved sociability and reduced repetitive behaviors in a mouse model of autism, implicating the potential therapeutic values of a pharmacological intervention targeting nicotinic receptors for autism therapy. Autism Res 2020, 13: 1311-1334. © 2020 International Society for Autism Research, Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Hafiz M Mahmood
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hesham M Aldhalaan
- Department of Neuroscience, Center for Autism Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Tahani K Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mashael A Alqasem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Musaad A Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Norah A Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Shakir D AlSharari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.,Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
18
|
Albert-Gascó H, Ros-Bernal F, Castillo-Gómez E, Olucha-Bordonau FE. MAP/ERK Signaling in Developing Cognitive and Emotional Function and Its Effect on Pathological and Neurodegenerative Processes. Int J Mol Sci 2020; 21:E4471. [PMID: 32586047 PMCID: PMC7352860 DOI: 10.3390/ijms21124471] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/14/2020] [Accepted: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
The signaling pathway of the microtubule-associated protein kinase or extracellular regulated kinase (MAPK/ERK) is a common mechanism of extracellular information transduction from extracellular stimuli to the intracellular space. The transduction of information leads to changes in the ongoing metabolic pathways and the modification of gene expression patterns. In the central nervous system, ERK is expressed ubiquitously, both temporally and spatially. As for the temporal ubiquity, this signaling system participates in three key moments: (i) Embryonic development; (ii) the early postnatal period; and iii) adulthood. During embryonic development, the system is partly responsible for the patterning of segmentation in the encephalic vesicle through the FGF8-ERK pathway. In addition, during this period, ERK directs neurogenesis migration and the final fate of neural progenitors. During the early postnatal period, ERK participates in the maturation process of dendritic trees and synaptogenesis. During adulthood, ERK participates in social and emotional behavior and memory processes, including long-term potentiation. Alterations in mechanisms related to ERK are associated with different pathological outcomes. Genetic alterations in any component of the ERK pathway result in pathologies associated with neural crest derivatives and mental dysfunctions associated with autism spectrum disorders. The MAP-ERK pathway is a key element of the neuroinflammatory pathway triggered by glial cells during the development of neurodegenerative diseases, such as Parkinson's and Alzheimer's disease, Huntington's disease, and amyotrophic lateral sclerosis, as well as prionic diseases. The process triggered by MAPK/ERK activation depends on the stage of development (mature or senescence), the type of cellular element in which the pathway is activated, and the anatomic neural structure. However, extensive gaps exist with regards to the targets of the phosphorylated ERK in many of these processes.
Collapse
Affiliation(s)
- Héctor Albert-Gascó
- UK Dementia Research Institute, Department of Clinical Neurosciences, University of Cambridge, Hills Road, Cambridge CB2 0AH, UK;
| | - Francisco Ros-Bernal
- U.P Medicina, Facultad de Ciencias de la Salud, Universitat Jaume I, Avda. de Vicent Sos Baynat s/n, 12071 Castelló de la Plana, Spain; (F.R.-B.); (E.C.-G.)
| | - Esther Castillo-Gómez
- U.P Medicina, Facultad de Ciencias de la Salud, Universitat Jaume I, Avda. de Vicent Sos Baynat s/n, 12071 Castelló de la Plana, Spain; (F.R.-B.); (E.C.-G.)
- Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Planta 0, 28029 Madrid, Spain
| | - Francisco E. Olucha-Bordonau
- U.P Medicina, Facultad de Ciencias de la Salud, Universitat Jaume I, Avda. de Vicent Sos Baynat s/n, 12071 Castelló de la Plana, Spain; (F.R.-B.); (E.C.-G.)
- Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Planta 0, 28029 Madrid, Spain
| |
Collapse
|
19
|
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
|
20
|
Morè L, Lauterborn JC, Papaleo F, Brambilla R. Enhancing cognition through pharmacological and environmental interventions: Examples from preclinical models of neurodevelopmental disorders. Neurosci Biobehav Rev 2020; 110:28-45. [PMID: 30981451 DOI: 10.1016/j.neubiorev.2019.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 02/04/2019] [Accepted: 02/05/2019] [Indexed: 12/29/2022]
Abstract
In this review we discuss the role of environmental and pharmacological treatments to enhance cognition with special regards to neurodevelopmental related disorders and aging. How the environment influences brain structure and function, and the interactions between rearing conditions and gene expression, are fundamental questions that are still poorly understood. We propose a model that can explain some of the discrepancies in findings for effects of environmental enrichment on outcome measures. Evidence of a direct causal correlation of nootropics and treatments that enhanced cognition also will be presented, and possible molecular mechanisms that include neurotrophin signaling and downstream pathways underlying these processes are discussed. Finally we review recent findings achieved with a wide set of behavioral and cognitive tasks that have translational validity to humans, and should be useful for future work on devising appropriate therapies. As will be discussed, the collective findings suggest that a combinational therapeutic approach of environmental enrichment and nootropics could be particularly successful for improving learning and memory in both developmental disorders and normal aging.
Collapse
Affiliation(s)
- Lorenzo Morè
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, PR1 2XT, Preston, UK.
| | - Julie C Lauterborn
- Department of Anatomy & Neurobiology, School of Medicine, University of California, Irvine, CA, 92617, USA.
| | - Francesco Papaleo
- Genetics of Cognition Laboratory, Istituto Italiano di Tecnologia, Via Morego, 30, 16163, Genova, Italy.
| | - Riccardo Brambilla
- Neuroscience and Mental Health Research Institute (NMHRI), Division of Neuroscience, School of Biosciences, Cardiff University, CF24 4HQ, Cardiff, UK.
| |
Collapse
|
21
|
Seese RR, Le AA, Wang K, Cox CD, Lynch G, Gall CM. A TrkB agonist and ampakine rescue synaptic plasticity and multiple forms of memory in a mouse model of intellectual disability. Neurobiol Dis 2020; 134:104604. [PMID: 31494285 PMCID: PMC7258745 DOI: 10.1016/j.nbd.2019.104604] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/26/2019] [Accepted: 09/04/2019] [Indexed: 12/20/2022] Open
Abstract
Fragile X syndrome (FXS) is associated with deficits in various types of learning, including those that require the hippocampus. Relatedly, hippocampal long-term potentiation (LTP) is impaired in the Fmr1 knockout (KO) mouse model of FXS. Prior research found that infusion of brain-derived neurotrophic factor (BDNF) rescues LTP in the KOs. Here, we tested if, in Fmr1 KO mice, up-regulating BDNF production or treatment with an agonist for BDNF's TrkB receptor restores synaptic plasticity and improves learning. In hippocampal slices, bath infusion of the TrkB agonist 7,8-dihydroxyflavone (7,8-DHF) completely restored otherwise impaired hippocampal field CA1 LTP of Fmr1 KOs without effect in wild types (WTs). Similarly, acute, semi-chronic, or chronic treatments with 7,8-DHF rescued a simple hippocampus-dependent form of spatial learning (object location memory: OLM) in Fmr1 KOs without effect in WTs. The agonist also restored object recognition memory, which depends on cortical regions. Semi-chronic, but not acute, treatment with the ampakine CX929, which up-regulates BDNF expression, lowered the training threshold for OLM in WT mice and rescued learning in the KOs. Positive results were also obtained in a test for social recognition. An mGluR5 antagonist did not improve learning. Quantification of synaptic immunolabeling demonstrated that 7,8-DHF and CX929 increase levels of activated TrkB at excitatory synapses. Moreover, CX929 induced a robust synaptic activation of the TrkB effector ERK1/2. These results suggest that enhanced synaptic BDNF signaling constitutes a plausible strategy for treating certain aspects of the cognitive disabilities associated with FXS.
Collapse
Affiliation(s)
- Ronald R Seese
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, United States of America
| | - Aliza A Le
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, United States of America
| | - Kathleen Wang
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, United States of America
| | - Conor D Cox
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, United States of America
| | - Gary Lynch
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, United States of America; Department of Psychiatry and Human Behavior, University of California, Irvine, CA, United States of America.
| | - Christine M Gall
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, United States of America; Department of Neurobiology and Behavior, University of California, Irvine, CA, United States of America.
| |
Collapse
|
22
|
Alhowail AH, Bloemer J, Majrashi M, Pinky PD, Bhattacharya S, Yongli Z, Bhattacharya D, Eggert M, Woodie L, Buabeid MA, Johnson N, Broadwater A, Smith B, Dhanasekaran M, Arnold RD, Suppiramaniam V. Doxorubicin-induced neurotoxicity is associated with acute alterations in synaptic plasticity, apoptosis, and lipid peroxidation. Toxicol Mech Methods 2019; 29:457-466. [PMID: 31010378 DOI: 10.1080/15376516.2019.1600086] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cognitive deficits are commonly reported by patients following treatment with chemotherapeutic agents. Anthracycline-containing chemotherapy regimens are associated with cognitive impairment and reductions in neuronal connectivity in cancer survivors, and doxorubicin (Dox) is a commonly used anthracycline. Although it has been reported that Dox distribution to the central nervous system (CNS) is limited, considerable Dox concentrations are observed in the brain with co-administration of certain medications. Additionally, pro-inflammatory cytokines, which are overproduced in cancer or in response to chemotherapy, can reduce the integrity of the blood-brain barrier (BBB). Therefore, the aim of this study was to evaluate the acute neurotoxic effects of Dox on hippocampal neurons. In this study, we utilized a hippocampal cell line (H19-7/IGF-IR) along with rodent hippocampal slices to evaluate the acute neurotoxic effects of Dox. Hippocampal slices were used to measure long-term potentiation (LTP), and expression of proteins was determined by immunoblotting. Cellular assays for mitochondrial complex activity and lipid peroxidation were also utilized. We observed reduction in LTP in hippocampal slices with Dox. In addition, lipid peroxidation was increased as measured by thiobarbituric acid reactive substances content indicating oxidative stress. Caspase-3 expression was increased indicating an increased propensity for cell death. Finally, the phosphorylation of signaling molecules which modulate LTP including extracellular signal-regulated kinase 1/2 (ERK1/2), p38 mitogen-activated protein kinase, and Akt were increased. This data indicates that acute Dox exposure dose-dependently impairs synaptic processes associated with hippocampal neurotransmission, induces apoptosis, and increases lipid peroxidation leading to neurotoxicity.
Collapse
Affiliation(s)
- Ahmad H Alhowail
- a Department of Drug Discovery and Development , Auburn University , Auburn , AL , USA
| | - Jenna Bloemer
- a Department of Drug Discovery and Development , Auburn University , Auburn , AL , USA
| | - Mohammed Majrashi
- a Department of Drug Discovery and Development , Auburn University , Auburn , AL , USA
| | - Priyanka D Pinky
- a Department of Drug Discovery and Development , Auburn University , Auburn , AL , USA
| | | | - Zhang Yongli
- a Department of Drug Discovery and Development , Auburn University , Auburn , AL , USA.,b Tianjin Huanhu Hospital , Tianjin , PR China
| | - Dwipayan Bhattacharya
- a Department of Drug Discovery and Development , Auburn University , Auburn , AL , USA
| | - Matthew Eggert
- a Department of Drug Discovery and Development , Auburn University , Auburn , AL , USA
| | - Lauren Woodie
- c Department of Nutrition, Dietetics and Hospitality Management , College of Human Sciences, Auburn University , Auburn , AL , USA
| | - Manal A Buabeid
- d College of Pharmacy and Health Sciences , Ajman University , Ajman , UAE
| | - Nathaniel Johnson
- a Department of Drug Discovery and Development , Auburn University , Auburn , AL , USA
| | - Alyssa Broadwater
- a Department of Drug Discovery and Development , Auburn University , Auburn , AL , USA
| | - Bruce Smith
- e Department of Anatomy, Physiology and Pharmacology , College of Veterinary Medicine, Auburn University , Auburn , AL , USA
| | | | - Robert D Arnold
- a Department of Drug Discovery and Development , Auburn University , Auburn , AL , USA
| | - Vishnu Suppiramaniam
- a Department of Drug Discovery and Development , Auburn University , Auburn , AL , USA
| |
Collapse
|
23
|
Lauterborn JC, Schultz MN, Le AA, Amani M, Friedman AE, Leach PT, Gall CM, Lynch GS, Crawley JN. Spaced training improves learning in Ts65Dn and Ube3a mouse models of intellectual disabilities. Transl Psychiatry 2019; 9:166. [PMID: 31182707 PMCID: PMC6557858 DOI: 10.1038/s41398-019-0495-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 03/14/2019] [Accepted: 03/23/2019] [Indexed: 12/29/2022] Open
Abstract
Benefits of distributed learning strategies have been extensively described in the human literature, but minimally investigated in intellectual disability syndromes. We tested the hypothesis that training trials spaced apart in time could improve learning in two distinct genetic mouse models of neurodevelopmental disorders characterized by intellectual impairments. As compared to training with massed trials, spaced training significantly improved learning in both the Ts65Dn trisomy mouse model of Down syndrome and the maternally inherited Ube3a mutant mouse model of Angelman syndrome. Spacing the training trials at 1 h intervals accelerated acquisition of three cognitive tasks by Ts65Dn mice: (1) object location memory, (2) novel object recognition, (3) water maze spatial learning. Further, (4) spaced training improved water maze spatial learning by Ube3a mice. In contrast, (5) cerebellar-mediated rotarod motor learning was not improved by spaced training. Corroborations in three assays, conducted in two model systems, replicated within and across two laboratories, confirm the strength of the findings. Our results indicate strong translational relevance of a behavioral intervention strategy for improving the standard of care in treating the learning difficulties that are characteristic and clinically intractable features of many neurodevelopmental disorders.
Collapse
Affiliation(s)
- J C Lauterborn
- Department of Anatomy & Neurobiology, School of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - M N Schultz
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - A A Le
- Department of Anatomy & Neurobiology, School of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - M Amani
- Department of Psychiatry and Human Behavior, School of Medicine, University of California Irvine, Irvine, CA, 92697, USA
- Department of Physiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - A E Friedman
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, USA
- Harvard University, Cambridge, MA, USA
| | - P T Leach
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, USA
- Biogen Inc., Cambridge, MA, USA
| | - C M Gall
- Department of Anatomy & Neurobiology, School of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - G S Lynch
- Department of Anatomy & Neurobiology, School of Medicine, University of California Irvine, Irvine, CA, 92697, USA
- Department of Psychiatry and Human Behavior, School of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - J N Crawley
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, USA.
| |
Collapse
|
24
|
Lee K, Vyas Y, Garner CC, Montgomery JM. Autism‐associated
Shank3
mutations alter mGluR expression and mGluR‐dependent but not NMDA receptor‐dependent long‐term depression. Synapse 2019; 73:e22097. [DOI: 10.1002/syn.22097] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 02/20/2019] [Accepted: 03/10/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Kevin Lee
- Department of Physiology, Centre for Brain Research University of Auckland Auckland New Zealand
| | - Yukti Vyas
- Department of Physiology, Centre for Brain Research University of Auckland Auckland New Zealand
| | - Craig C. Garner
- German Center for Neurodegenerative Disorders Charité – Universitätsmedizin Berlin Berlin Germany
| | - Johanna M. Montgomery
- Department of Physiology, Centre for Brain Research University of Auckland Auckland New Zealand
| |
Collapse
|
25
|
Telias M. Molecular Mechanisms of Synaptic Dysregulation in Fragile X Syndrome and Autism Spectrum Disorders. Front Mol Neurosci 2019; 12:51. [PMID: 30899214 PMCID: PMC6417395 DOI: 10.3389/fnmol.2019.00051] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 02/12/2019] [Indexed: 12/21/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common form of monogenic hereditary cognitive impairment. FXS patient exhibit a high comorbidity rate with autism spectrum disorders (ASDs). This makes FXS a model disease for understanding how synaptic dysregulation alters neuronal excitability, learning and memory, social behavior, and more. Since 1991, with the discovery of fragile X mental retardation 1 (FMR1) as the sole gene that is mutated in FXS, thousands of studies into the function of the gene and its encoded protein FMR1 protein (FMRP), have been conducted, yielding important information regarding the pathophysiology of the disease, as well as insight into basic synaptic mechanisms that control neuronal networking and circuitry. Among the most important, are molecular mechanisms directly involved in plasticity, including glutamate and γ-aminobutyric acid (GABA) receptors, which can control synaptic transmission and signal transduction, including short- and long-term plasticity. More recently, several novel mechanisms involving growth factors, enzymatic cascades and transcription factors (TFs), have been proposed to have the potential of explaining some of the synaptic dysregulation in FXS. In this review article, I summarize the main mechanisms proposed to underlie synaptic disruption in FXS and ASDs. I focus on studies conducted on the Fmr1 knock-out (KO) mouse model and on FXS-human pluripotent stem cells (hPSCs), emphasizing the differences and even contradictions between mouse and human, whenever possible. As FXS and ASDs are both neurodevelopmental disorders that follow a specific time-course of disease progression, I highlight those studies focusing on the differential developmental regulation of synaptic abnormalities in these diseases.
Collapse
Affiliation(s)
- Michael Telias
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| |
Collapse
|
26
|
Rhine MA, Parrott JM, Schultz MN, Kazdoba TM, Crawley JN. Hypothesis-driven investigations of diverse pharmacological targets in two mouse models of autism. Autism Res 2019; 12:401-421. [PMID: 30653853 PMCID: PMC6402976 DOI: 10.1002/aur.2066] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/13/2018] [Accepted: 12/06/2018] [Indexed: 12/19/2022]
Abstract
Autism spectrum disorder is a neurodevelopmental syndrome diagnosed primarily by persistent deficits in social interactions and communication, unusual sensory reactivity, motor stereotypies, repetitive behaviors, and restricted interests. No FDA‐approved medical treatments exist for the diagnostic symptoms of autism. Here we interrogate multiple pharmacological targets in two distinct mouse models that incorporate well‐replicated autism‐relevant behavioral phenotypes. Compounds that modify inhibitory or excitatory neurotransmission were selected to address hypotheses based on previously published biological abnormalities in each model. Shank3B is a genetic model of a mutation found in autism and Phelan‐McDermid syndrome, in which deficits in excitatory neurotransmission and synaptic plasticity have been reported. BTBR is an inbred strain model of forms of idiopathic autism in which reduced inhibitory neurotransmission and excessive mTOR signaling have been reported. The GABA‐A receptor agonist gaboxadol significantly reduced repetitive self‐grooming in three independent cohorts of BTBR. The TrkB receptor agonist 7,8‐DHF improved spatial learning in Shank3B mice, and reversed aspects of social deficits in BTBR. CX546, a positive allosteric modulator of the glutamatergic AMPA receptor, and d‐cycloserine, a partial agonist of the glycine site on the glutamatergic NMDA receptor, did not rescue aberrant behaviors in Shank3B mice. The mTOR inhibitor rapamycin did not ameliorate social deficits or repetitive behavior in BTBR mice. Comparison of positive and negative pharmacological outcomes, on multiple phenotypes, evaluated for replicability across independent cohorts, enhances the translational value of mouse models of autism for therapeutic discovery. GABA agonists present opportunities for personalized interventions to treat components of autism spectrum disorder. Autism Res 2019, 12: 401–421 © 2019 The Authors. Autism Research published by International Society for Autism Research published by Wiley Periodicals, Inc. Lay Summary Many of the risk genes for autism impair synapses, the connections between nerve cells in the brain. A drug that reverses the synaptic effects of a mutation could offer a precision therapy. Combining pharmacological and behavioral therapies could reduce symptoms and improve the quality of life for people with autism. Here we report reductions in repetitive behavior by a GABA‐A receptor agonist, gaboxadol, and improvements in social and cognitive behaviors by a TrkB receptor agonist, in mouse models of autism.
Collapse
Affiliation(s)
- Maya A Rhine
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, 95817
| | - Jennifer M Parrott
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, 95817
| | - Maria N Schultz
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, 95817
| | - Tatiana M Kazdoba
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, 95817
| | - Jacqueline N Crawley
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, 95817
| |
Collapse
|
27
|
Sun W, Li X, Tang C, An L. Acute Low Alcohol Disrupts Hippocampus-Striatum Neural Correlate of Learning Strategy by Inhibition of PKA/CREB Pathway in Rats. Front Pharmacol 2018; 9:1439. [PMID: 30574089 PMCID: PMC6291496 DOI: 10.3389/fphar.2018.01439] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 11/22/2018] [Indexed: 11/30/2022] Open
Abstract
The hippocampus and striatum guide place-strategy and response-strategy learning, respectively, and they have dissociable roles in memory systems, which could compensate in case of temporary or permanent damage. Although acute alcohol (AA) treatment had been shown to have adverse effects on hippocampal function, whether it causes the functional compensation and the underlying mechanisms is unknown. In this study, rats treated with a low dose of AA avoided a hippocampus-dependent spatial strategy, instead preferring a striatum-dependent response strategy. Consistently, the learning-induced increase in hippocampal, but not striatal, pCREB was rendered less pronounced due to diminished activity of pPKA, but not pERK or pCaMKII. As rats approached the turn-decision area, Sp-cAMP, a PKA activator, was found to mitigate the inhibitory effect of AA on intra- and cross-structure synchronized neuronal oscillations, and rescue response-strategy bias and spatial learning deficits. Our study provides strong evidence of the critical link between neural couplings and strategy selection. Moreover, the PKA/CREB-signaling pathway is involved in the suppressive effect of AA on neural correlates of place-learning strategy. The novel important evidence provided here shows the functional couplings between the hippocampus and striatum in spatial learning processing and suggests possible avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Wei Sun
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoliang Li
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chunzhi Tang
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lei An
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China.,College of Acupuncture-Moxibustion and Orthopedics, Guiyang University of Chinese Medicine, Guiyang, China.,Department of Physiology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
28
|
Abstract
The MAPK pathway is a prominent intracellular signaling pathway regulating various intracellular functions. Components of this pathway are mutated in a related collection of congenital syndromes collectively referred to as neuro-cardio-facio-cutaneous syndromes (NCFC) or Rasopathies. Recently, it has been appreciated that these disorders are associated with autism spectrum disorders (ASD). In addition, idiopathic ASD has also implicated the MAPK signaling cascade as a common pathway that is affected by many of the genetic variants that have been found to be linked to ASDs. This chapter describes the components of the MAPK pathway and how it is regulated. Furthermore, this chapter will highlight the various functions of the MAPK pathway during both embryonic development of the central nervous system (CNS) and its roles in neuronal physiology and ultimately, behavior. Finally, we will summarize the perturbations to MAPK signaling in various models of autism spectrum disorders and Rasopathies to highlight how dysregulation of this pivotal pathway may contribute to the pathogenesis of autism.
Collapse
|
29
|
Albert-Gasco H, Sanchez-Sarasua S, Ma S, García-Díaz C, Gundlach AL, Sanchez-Perez AM, Olucha-Bordonau FE. Central relaxin-3 receptor (RXFP3) activation impairs social recognition and modulates ERK-phosphorylation in specific GABAergic amygdala neurons. Brain Struct Funct 2018; 224:453-469. [PMID: 30368554 DOI: 10.1007/s00429-018-1763-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 09/27/2018] [Indexed: 11/25/2022]
Abstract
In mammals, the extended amygdala is a neural hub for social and emotional information processing. In the rat, the extended amygdala receives inhibitory GABAergic projections from the nucleus incertus (NI) in the pontine tegmentum. NI neurons produce the neuropeptide relaxin-3, which acts via the Gi/o-protein-coupled receptor, RXFP3. A putative role for RXFP3 signalling in regulating social interaction was investigated by assessing the effect of intracerebroventricular infusion of the RXFP3 agonist, RXFP3-A2, on performance in the 3-chamber social interaction paradigm. Central RXFP3-A2, but not vehicle, infusion, disrupted the capacity to discriminate between a familiar and novel conspecific subject, but did not alter differentiation between a conspecific and an inanimate object. Subsequent studies revealed that agonist-infused rats displayed increased phosphoERK(pERK)-immunoreactivity in specific amygdaloid nuclei at 20 min post-infusion, with levels similar to control again after 90 min. In parallel, we used immunoblotting to profile ERK phosphorylation dynamics in whole amygdala after RXFP3-A2 treatment; and multiplex histochemical labelling techniques to reveal that after RXFP3-A2 infusion and social interaction, pERK-immunopositive neurons in amygdala expressed vesicular GABA-transporter mRNA and displayed differential profiles of RXFP3 and oxytocin receptor mRNA. Overall, these findings demonstrate that central relaxin-3/RXFP3 signalling can modulate social recognition in rats via effects within the amygdala and likely interactions with GABA and oxytocin signalling.
Collapse
MESH Headings
- Amygdala/cytology
- Amygdala/drug effects
- Amygdala/enzymology
- Animals
- Behavior, Animal/drug effects
- Extracellular Signal-Regulated MAP Kinases/metabolism
- GABAergic Neurons/drug effects
- GABAergic Neurons/enzymology
- Infusions, Intraventricular
- Intercellular Signaling Peptides and Proteins
- Male
- Oxytocin/metabolism
- Peptides/administration & dosage
- Phosphorylation
- Rats, Wistar
- Receptors, G-Protein-Coupled/agonists
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Oxytocin/genetics
- Receptors, Oxytocin/metabolism
- Receptors, Peptide/agonists
- Receptors, Peptide/genetics
- Receptors, Peptide/metabolism
- Recognition, Psychology/drug effects
- Signal Transduction/drug effects
- Social Behavior
- Vesicular Inhibitory Amino Acid Transport Proteins/genetics
- Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
- gamma-Aminobutyric Acid/metabolism
Collapse
Affiliation(s)
- Hector Albert-Gasco
- Departamento de Medicina, Facultad de Ciencias de la Salud, Universitat Jaume I, Av de Vicent Sos Baynat, s/n, 12071, Castellón de la Plana, Castellón, Spain
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Sandra Sanchez-Sarasua
- Departamento de Medicina, Facultad de Ciencias de la Salud, Universitat Jaume I, Av de Vicent Sos Baynat, s/n, 12071, Castellón de la Plana, Castellón, Spain
| | - Sherie Ma
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Cristina García-Díaz
- Departamento de Medicina, Facultad de Ciencias de la Salud, Universitat Jaume I, Av de Vicent Sos Baynat, s/n, 12071, Castellón de la Plana, Castellón, Spain
| | - Andrew L Gundlach
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Ana M Sanchez-Perez
- Departamento de Medicina, Facultad de Ciencias de la Salud, Universitat Jaume I, Av de Vicent Sos Baynat, s/n, 12071, Castellón de la Plana, Castellón, Spain.
| | - Francisco E Olucha-Bordonau
- Departamento de Medicina, Facultad de Ciencias de la Salud, Universitat Jaume I, Av de Vicent Sos Baynat, s/n, 12071, Castellón de la Plana, Castellón, Spain.
| |
Collapse
|
30
|
Frazzini V, Granzotto A, Bomba M, Massetti N, Castelli V, d'Aurora M, Punzi M, Iorio M, Mosca A, Delli Pizzi S, Gatta V, Cimini A, Sensi SL. The pharmacological perturbation of brain zinc impairs BDNF-related signaling and the cognitive performances of young mice. Sci Rep 2018; 8:9768. [PMID: 29950603 PMCID: PMC6021411 DOI: 10.1038/s41598-018-28083-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 06/15/2018] [Indexed: 01/13/2023] Open
Abstract
Zinc (Zn2+) is a pleiotropic modulator of the neuronal and brain activity. The disruption of intraneuronal Zn2+ levels triggers neurotoxic processes and affects neuronal functioning. In this study, we investigated how the pharmacological modulation of brain Zn2+ affects synaptic plasticity and cognition in wild-type mice. To manipulate brain Zn2+ levels, we employed the Zn2+ (and copper) chelator 5-chloro-7-iodo-8-hydroxyquinoline (clioquinol, CQ). CQ was administered for two weeks to 2.5-month-old (m.o.) mice, and effects studied on BDNF-related signaling, metalloproteinase activity as well as learning and memory performances. CQ treatment was found to negatively affect short- and long-term memory performances. The CQ-driven perturbation of brain Zn2+ was found to reduce levels of BDNF, synaptic plasticity-related proteins and dendritic spine density in vivo. Our study highlights the importance of choosing "when", "where", and "how much" in the modulation of brain Zn2+ levels. Our findings confirm the importance of targeting Zn2+ as a therapeutic approach against neurodegenerative conditions but, at the same time, underscore the potential drawbacks of reducing brain Zn2+ availability upon the early stages of development.
Collapse
Affiliation(s)
- Valerio Frazzini
- Center of Excellence on Aging and Translational Medicine - CeSI-MeT, Chieti, Italy
- Institut du Cerveau et de la Moelle épinière, ICM, INSERM UMRS 1127, CNRS UMR 7225, Pitié-Salpêtrière Hospital, Paris, France
- AP-HP, GH Pitie-Salpêtrière-Charles Foix, Epilepsy Unit and Neurophysiology Department, Paris, France
| | - Alberto Granzotto
- Center of Excellence on Aging and Translational Medicine - CeSI-MeT, Chieti, Italy
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti-Pescara, Italy
| | - Manuela Bomba
- Center of Excellence on Aging and Translational Medicine - CeSI-MeT, Chieti, Italy
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti-Pescara, Italy
| | - Noemi Massetti
- Center of Excellence on Aging and Translational Medicine - CeSI-MeT, Chieti, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Marco d'Aurora
- Center of Excellence on Aging and Translational Medicine - CeSI-MeT, Chieti, Italy
- Department of Psychological Sciences, School of Medicine and Health Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti-Pescara, Italy
| | - Miriam Punzi
- Center of Excellence on Aging and Translational Medicine - CeSI-MeT, Chieti, Italy
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti-Pescara, Italy
| | - Mariangela Iorio
- Center of Excellence on Aging and Translational Medicine - CeSI-MeT, Chieti, Italy
| | - Alessandra Mosca
- Center of Excellence on Aging and Translational Medicine - CeSI-MeT, Chieti, Italy
| | - Stefano Delli Pizzi
- Center of Excellence on Aging and Translational Medicine - CeSI-MeT, Chieti, Italy
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti-Pescara, Italy
| | - Valentina Gatta
- Center of Excellence on Aging and Translational Medicine - CeSI-MeT, Chieti, Italy
- Department of Psychological Sciences, School of Medicine and Health Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti-Pescara, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, USA
- National Institute for Nuclear Physics (INFN), Gran Sasso National Laboratory (LNGS), Assergi, Italy
| | - Stefano L Sensi
- Center of Excellence on Aging and Translational Medicine - CeSI-MeT, Chieti, Italy.
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti-Pescara, Italy.
- Departments of Neurology and Pharmacology, Institute for Mind Impairments and Neurological Disorders, University of California - Irvine, Irvine, USA.
| |
Collapse
|
31
|
mGluR5 Modulation of Behavioral and Epileptic Phenotypes in a Mouse Model of Tuberous Sclerosis Complex. Neuropsychopharmacology 2018; 43:1457-1465. [PMID: 29206810 PMCID: PMC5916364 DOI: 10.1038/npp.2017.295] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 11/03/2017] [Accepted: 11/28/2017] [Indexed: 12/13/2022]
Abstract
Drugs targeting metabotropic glutamate receptor 5 (mGluR5) have therapeutic potential in autism spectrum disorders (ASD), including tuberous sclerosis complex (TSC). The question whether inhibition or potentiation of mGluR5 could be beneficial depends, among other factors, on the specific indication. To facilitate the development of mGluR5 treatment strategies, we tested the therapeutic utility of mGluR5 negative and positive allosteric modulators (an mGluR5 NAM and PAM) for TSC, using a mutant mouse model with neuronal loss of Tsc2 that demonstrates disease-related phenotypes, including behavioral symptoms of ASD and epilepsy. This model uniquely enables the in vivo characterization and rescue of the electrographic seizures associated with TSC. We demonstrate that inhibition of mGluR5 corrects hyperactivity, seizures, and elevated de novo synaptic protein synthesis. Conversely, positive allosteric modulation of mGluR5 results in the exacerbation of hyperactivity and epileptic phenotypes. The data suggest a meaningful therapeutic potential for mGluR5 NAMs in TSC, which warrants clinical exploration and the continued development of mGluR5 therapies.
Collapse
|
32
|
Nardecchia F, Orlando R, Iacovelli L, Colamartino M, Fiori E, Leuzzi V, Piccinin S, Nistico R, Puglisi-Allegra S, Di Menna L, Battaglia G, Nicoletti F, Pascucci T. Targeting mGlu5 Metabotropic Glutamate Receptors in the Treatment of Cognitive Dysfunction in a Mouse Model of Phenylketonuria. Front Neurosci 2018; 12:154. [PMID: 29615849 PMCID: PMC5864888 DOI: 10.3389/fnins.2018.00154] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 02/26/2018] [Indexed: 11/23/2022] Open
Abstract
We studied group-I metabotropic glutamate (mGlu) receptors in Pahenu2 (ENU2) mice, which mimic the genetics and neurobiology of human phenylketonuria (PKU), a metabolic disorder characterized, if untreated, by autism, and intellectual disability (ID). Male ENU2 mice showed increased mGlu5 receptor protein levels in the hippocampus and corpus striatum (but not in the prefrontal cortex) whereas the transcript of the mGlu5 receptor was unchanged. No changes in mGlu1 receptor mRNA and protein levels were found in any of the three brain regions of ENU2 mice. We extended the analysis to Homer proteins, which act as scaffolds by linking mGlu1 and mGlu5 receptors to effector proteins. Expression of the long isoforms of Homer was significantly reduced in the hippocampus of ENU2 mice, whereas levels of the short Homer isoform (Homer 1a) were unchanged. mGlu5 receptors were less associated to immunoprecipitated Homer in the hippocampus of ENU2 mice. The lack of mGlu5 receptor-mediated long-term depression (LTD) in wild-type mice (of BTBR strain) precluded the analysis of hippocampal synaptic plasticity in ENU2 mice. We therefore performed a behavioral analysis to examine whether pharmacological blockade of mGlu5 receptors could correct behavioral abnormalities in ENU2 mice. Using the same apparatus we sequentially assessed locomotor activity, object exploration, and spatial object recognition (spatial novelty test) after displacing some of the objects from their original position in the arena. Systemic treatment with the mGlu5 receptor antagonist, MPEP (20 mg/kg, i.p.), had a striking effect in the spatial novelty test by substantially increasing the time spent in exploring the displaced objects in ENU2 mice (but not in wild-type mice). These suggest a role for mGlu5 receptors in the pathophysiology of ID in PKU and suggest that, also in adult untreated animals, cognitive dysfunction may be improved by targeting these receptors with an appropriate therapy.
Collapse
Affiliation(s)
- Francesca Nardecchia
- Department of Physiology and Pharmacology, Sapienza Università di Roma, Rome, Italy.,Department of Pediatrics and Child Neuropsychiatry, Sapienza Università di Roma, Rome, Italy
| | - Rosamaria Orlando
- Department of Physiology and Pharmacology, Sapienza Università di Roma, Rome, Italy
| | - Luisa Iacovelli
- Department of Physiology and Pharmacology, Sapienza Università di Roma, Rome, Italy
| | - Marco Colamartino
- Daniel Bovet Department of Psychology, Neurobiology Research Center, Sapienza Università di Roma, Rome, Italy
| | - Elena Fiori
- Daniel Bovet Department of Psychology, Neurobiology Research Center, Sapienza Università di Roma, Rome, Italy
| | - Vincenzo Leuzzi
- Department of Pediatrics and Child Neuropsychiatry, Sapienza Università di Roma, Rome, Italy
| | - Sonia Piccinin
- Department of Physiology and Pharmacology, Sapienza Università di Roma, Rome, Italy.,Department of Biology, Università degli Studi di Roma Tor Vergata, Rome, Italy
| | - Robert Nistico
- Department of Biology, Università degli Studi di Roma Tor Vergata, Rome, Italy
| | - Stefano Puglisi-Allegra
- Daniel Bovet Department of Psychology, Neurobiology Research Center, Sapienza Università di Roma, Rome, Italy.,IRCCS Foundation Santa Lucia, Rome, Italy
| | | | | | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, Sapienza Università di Roma, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| | - Tiziana Pascucci
- Daniel Bovet Department of Psychology, Neurobiology Research Center, Sapienza Università di Roma, Rome, Italy.,IRCCS Foundation Santa Lucia, Rome, Italy
| |
Collapse
|
33
|
Wu H, Zhang Q, Gao J, Sun C, Wang J, Xia W, Cao Y, Hao Y, Wu L. Modulation of sphingosine 1-phosphate (S1P) attenuates spatial learning and memory impairments in the valproic acid rat model of autism. Psychopharmacology (Berl) 2018; 235:873-886. [PMID: 29218394 DOI: 10.1007/s00213-017-4805-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 11/27/2017] [Indexed: 12/26/2022]
Abstract
RATIONALE Autism spectrum disorders (ASD) are a set of pervasive neurodevelopmental disorders that manifest in early childhood, and it is growing up to be a major cause of disability in children. However, the etiology and treatment of ASD are not well understood. In our previous study, we found that serum levels of sphingosine 1-phosphate (S1P) were increased significantly in children with autism, indicating that S1P levels may be involved in ASD. OBJECTIVE The objective of this study was to identify a link between increased levels of S1P and neurobehavioral changes in autism. METHODS We utilized a valproic acid (VPA) -induced rat model of autism to evaluate the levels of S1P and the expression of sphingosine kinase (SphK), a key enzyme for S1P production, in serum and hippocampal tissue. Furthermore, we assessed cognitive functional changes and histopathological and neurochemical alterations in VPA-exposed rats after SphK blockade to explore the possible link between increased levels of S1P and neurobehavioral changes in autism. RESULTS We found that SphK2 and S1P are upregulated in hippocampal tissue from VPA-exposed rats, while pharmacological inhibition of SphK reduced S1P levels, attenuated spatial learning and memory impairments, increased the expression of phosphorylated CaMKII and CREB and autophagy-related proteins, inhibited cytochrome c release, decreased the expression of apoptosis related proteins, and protected against neuronal loss in the hippocampus. CONCLUSION We have demonstrated that an increased level of SphK2/S1P is involved in the spatial learning and memory impairments of autism, and this signaling pathway represents a novel therapeutic target and direction for future studies.
Collapse
Affiliation(s)
- Hongmei Wu
- Department of Nursing, Harbin Medical University in Daqing, Daqing, Heilongjiang, 163319, China
| | - Quanzhi Zhang
- Department of Nursing, Harbin Medical University in Daqing, Daqing, Heilongjiang, 163319, China.,Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Heilongjiang, 150081, China
| | - Jingquan Gao
- Department of Nursing, Harbin Medical University in Daqing, Daqing, Heilongjiang, 163319, China
| | - Caihong Sun
- Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Heilongjiang, 150081, China
| | - Jia Wang
- Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Heilongjiang, 150081, China
| | - Wei Xia
- Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Heilongjiang, 150081, China
| | - Yonggang Cao
- Department of Pharmacology, Harbin Medical University in Daqing, Daqing, Heilongjiang, 163319, China
| | - Yanqiu Hao
- Department of pediatrics, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Lijie Wu
- Department of Child and Adolescent Health, School of Public Health, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, Heilongjiang, 150081, China.
| |
Collapse
|
34
|
Lacivita E, Perrone R, Margari L, Leopoldo M. Targets for Drug Therapy for Autism Spectrum Disorder: Challenges and Future Directions. J Med Chem 2017; 60:9114-9141. [PMID: 29039668 DOI: 10.1021/acs.jmedchem.7b00965] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by persistent deficits in social communication and interaction and restricted, repetitive patterns of behavior, interests, and activities. Various factors are involved in the etiopathogenesis of ASD, including genetic factors, environmental toxins and stressors, impaired immune responses, mitochondrial dysfunction, and neuroinflammation. The heterogeneity in the phenotype among ASD patients and the complex etiology of the condition have long impeded the advancement of the development of pharmacological therapies. In the recent years, the integration of findings from mouse models to human genetics resulted in considerable progress toward the understanding of ASD pathophysiology. Currently, strategies to treat core symptoms of ASD are directed to correct synaptic dysfunctions, abnormalities in central oxytocin, vasopressin, and serotonin neurotransmission, and neuroinflammation. Here, we present a survey of the studies that have suggested molecular targets for drug development for ASD and the state-of-the-art of medicinal chemistry efforts in related areas.
Collapse
Affiliation(s)
- Enza Lacivita
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro , via Orabona 4, 70125, Bari, Italy
| | - Roberto Perrone
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro , via Orabona 4, 70125, Bari, Italy
| | - Lucia Margari
- Dipartimento di Scienze Mediche di Base, Neuroscienze e Organi di Senso, Unità di Neuropsichiatria Infantile, Università degli Studi di Bari Aldo Moro , Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Marcello Leopoldo
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro , via Orabona 4, 70125, Bari, Italy
| |
Collapse
|
35
|
Abstract
Human genetic studies have been the driving force in bringing to light the underlying biology of psychiatric conditions. As these studies fill in the gaps in our knowledge of the mechanisms at play, we will be better equipped to design therapies in rational and targeted ways, or repurpose existing therapies in previously unanticipated ways. This review is intended for those unfamiliar with psychiatric genetics as a field and provides a primer on different modes of genetic variation, the technologies currently used to probe them, and concepts that provide context for interpreting the gene-phenotype relationship. Like other subfields in human genetics, psychiatric genetics is moving from microarray technology to sequencing-based approaches as barriers of cost and expertise are removed, and the ramifications of this transition are discussed here. A summary is then given of recent genetic discoveries in a number of neuropsychiatric conditions, with particular emphasis on neurodevelopmental conditions. The general impact of genetics on drug development has been to underscore the extensive etiological heterogeneity in seemingly cohesive diagnostic categories. Consequently, the path forward is not in therapies hoping to reach large swaths of patients sharing a clinically defined diagnosis, but rather in targeting patients belonging to specific "biotypes" defined through a combination of objective, quantifiable data, including genotype.
Collapse
Affiliation(s)
- Jacob J Michaelson
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
- Department of Biomedical Engineering, University of Iowa College of Engineering, Iowa City, IA, USA.
- Department of Communication Sciences and Disorders, University of Iowa College of Liberal Arts and Sciences, Iowa City, IA, USA.
- Iowa Institute of Human Genetics, University of Iowa, Iowa City, IA, USA.
- Genetics Cluster Initiative, University of Iowa, Iowa City, IA, USA.
- The DeLTA Center, University of Iowa, Iowa City, IA, USA.
- University of Iowa Informatics Initiative, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
36
|
Lee E, Lee J, Kim E. Excitation/Inhibition Imbalance in Animal Models of Autism Spectrum Disorders. Biol Psychiatry 2017; 81:838-847. [PMID: 27450033 DOI: 10.1016/j.biopsych.2016.05.011] [Citation(s) in RCA: 330] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 05/02/2016] [Accepted: 05/16/2016] [Indexed: 12/19/2022]
Abstract
Imbalances between excitation and inhibition in synaptic transmission and neural circuits have been implicated in autism spectrum disorders. Excitation and inhibition imbalances are frequently observed in animal models of autism spectrum disorders, and their correction normalizes key autistic-like phenotypes in these animals. These results suggest that excitation and inhibition imbalances may contribute to the development and maintenance of autism spectrum disorders and represent an important therapeutic target.
Collapse
Affiliation(s)
- Eunee Lee
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, Korea
| | - Jiseok Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Eunjoon Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, Korea; Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea.
| |
Collapse
|
37
|
Sethna F, Feng W, Ding Q, Robison AJ, Feng Y, Wang H. Enhanced expression of ADCY1 underlies aberrant neuronal signalling and behaviour in a syndromic autism model. Nat Commun 2017; 8:14359. [PMID: 28218269 PMCID: PMC5321753 DOI: 10.1038/ncomms14359] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 12/20/2016] [Indexed: 12/14/2022] Open
Abstract
Fragile X syndrome (FXS), caused by the loss of functional FMRP, is a leading cause of autism. Neurons lacking FMRP show aberrant mRNA translation and intracellular signalling. Here, we identify that, in Fmr1 knockout neurons, type 1 adenylyl cyclase (Adcy1) mRNA translation is enhanced, leading to excessive production of ADCY1 protein and insensitivity to neuronal stimulation. Genetic reduction of Adcy1 normalizes the aberrant ERK1/2- and PI3K-mediated signalling, attenuates excessive protein synthesis and corrects dendritic spine abnormality in Fmr1 knockout mice. Genetic reduction of Adcy1 also ameliorates autism-related symptoms including repetitive behaviour, defective social interaction and audiogenic seizures. Moreover, peripheral administration of NB001, an experimental compound that preferentially suppresses ADCY1 activity over other ADCY subtypes, attenuates the behavioural abnormalities in Fmr1 knockout mice. These results demonstrate a connection between the elevated Adcy1 translation and abnormal ERK1/2 signalling and behavioural symptoms in FXS. Fragile X syndrome (FXS) is a leading cause of autism and neurons lacking FMRP show aberrant mRNA translation and intracellular signalling. Here, the authors show that neurons from Fmr1 knockout mice have increased levels of ADCY1 protein, producing abnormal ERK1/2 signalling, dysregulated protein synthesis and behavioural symptoms associated with FXS.
Collapse
Affiliation(s)
- Ferzin Sethna
- Genetics Program, Michigan State University, East Lansing, Michigan 48824, USA
| | - Wei Feng
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Qi Ding
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, USA
| | - Alfred J Robison
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, USA.,Neuroscience Program, Michigan State University, East Lansing, Michigan 48824, USA
| | - Yue Feng
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Hongbing Wang
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, USA.,Neuroscience Program, Michigan State University, East Lansing, Michigan 48824, USA
| |
Collapse
|
38
|
Meyza KZ, Blanchard DC. The BTBR mouse model of idiopathic autism - Current view on mechanisms. Neurosci Biobehav Rev 2017; 76:99-110. [PMID: 28167097 DOI: 10.1016/j.neubiorev.2016.12.037] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 12/17/2016] [Accepted: 12/19/2016] [Indexed: 02/07/2023]
Abstract
Autism spectrum disorder (ASD) is the most commonly diagnosed neurodevelopmental disorder, with current estimates of more than 1% of affected children across nations. The patients form a highly heterogeneous group with only the behavioral phenotype in common. The genetic heterogeneity is reflected in a plethora of animal models representing multiple mutations found in families of affected children. Despite many years of scientific effort, for the majority of cases the genetic cause remains elusive. It is therefore crucial to include well-validated models of idiopathic autism in studies searching for potential therapeutic agents. One of these models is the BTBR T+Itpr3tf/J mouse. The current review summarizes data gathered in recent research on potential molecular mechanisms responsible for the autism-like behavioral phenotype of this strain.
Collapse
Affiliation(s)
- K Z Meyza
- Laboratory of Emotions' Neurobiology, Nencki Institute of Experimental Biology, 3 Pasteur Street, Warsaw, 02-093, Poland.
| | - D C Blanchard
- Department of Psychology, University of Hawaii at Manoa,1993 East-West Road, Honolulu, HI 96822, USA
| |
Collapse
|
39
|
Genetic and Pharmacological Reversibility of Phenotypes in Mouse Models of Autism Spectrum Disorder. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2017; 224:189-211. [PMID: 28551757 DOI: 10.1007/978-3-319-52498-6_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
As autism spectrum disorder (ASD) is largely regarded as a neurodevelopmental condition, long-time consensus was that its hallmark features are irreversible. However, several studies from recent years using defined mouse models of ASD have provided clear evidence that in mice neurobiological and behavioural alterations can be ameliorated or even reversed by genetic restoration or pharmacological treatment either before or after symptom onset. Here, we review findings on genetic and pharmacological reversibility of phenotypes in mouse models of ASD. Our review should give a comprehensive overview on both aspects and encourage future studies to better understand the underlying molecular mechanisms that might be translatable from animals to humans.
Collapse
|
40
|
Guo YP, Commons KG. Serotonin neuron abnormalities in the BTBR mouse model of autism. Autism Res 2016; 10:66-77. [PMID: 27478061 DOI: 10.1002/aur.1665] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 06/08/2016] [Accepted: 06/13/2016] [Indexed: 12/21/2022]
Abstract
The inbred mouse strain BTBR T+ Itpr3tf /J (BTBR) is studied as a model of idiopathic autism because they are less social and more resistant to change than other strains. Forebrain serotonin receptors and the response to serotonin drugs are altered in BTBR mice, yet it remains unknown if serotonin neurons themselves are abnormal. In this study, we found that serotonin tissue content and the density of serotonin axons is reduced in the hippocampus of BTBR mice in comparison to C57BL/6J (C57) mice. This was accompanied by possible compensatory changes in serotonin neurons that were most pronounced in regions known to provide innervation to the hippocampus: the caudal dorsal raphe (B6) and the median raphe. These changes included increased numbers of serotonin neurons and hyperactivation of Fos expression. Metrics of serotonin neurons in the rostral 2/3 of the dorsal raphe and serotonin content of the prefrontal cortex were less impacted. Thus, serotonin neurons exhibit region-dependent abnormalities in the BTBR mouse that may contribute to their altered behavioral profile. Autism Res 2017, 10: 66-77. © 2016 International Society for Autism Research, Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yue-Ping Guo
- Department of Anesthesiology, Second Affiliated Hospital, Harbin Medical University, Harbin, China.,Department of Anesthesiology, Perioperative, and Pain Medicine, Boston Children's Hospital; Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts
| | - Kathryn G Commons
- Department of Anesthesiology, Perioperative, and Pain Medicine, Boston Children's Hospital; Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
41
|
Buschler A, Manahan-Vaughan D. Metabotropic glutamate receptor, mGlu5, mediates enhancements of hippocampal long-term potentiation after environmental enrichment in young and old mice. Neuropharmacology 2016; 115:42-50. [PMID: 27267685 DOI: 10.1016/j.neuropharm.2016.06.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 06/01/2016] [Accepted: 06/02/2016] [Indexed: 12/24/2022]
Abstract
The metabotropic glutamate (mGlu) receptor, mGlu5, is of particular relevance for hippocampal function. It is critically required for the expression of long-term potentiation (LTP) and long-term depression (LTD), regulates neuronal oscillations, maintains the stability of place fields and is required for hippocampus-dependent memory. MGlu5-dysfunctions are associated with profound cognitive deficits in humans, and mGlu5 has been targeted as a putative cognitive enhancer. Cognitive enhancement, by means of environmental enrichment (EE) in rodents, results in improved hippocampal synaptic plasticity and memory. Here, we explored whether mGlu5 contributes to these enhancements. MGlu5-antagonism dose-dependently impaired the early phase of LTP (>4 h) in the CA1 region of young(3-4 month old) mice. Late-LTP (>24 h) was also impaired. LTP (>24 h) elicited in old (10-14 month old) mice displayed reduced sensitivity to mGlu5 antagonism. Short-term potentiation (STP, < 2 h) that was elicited by weaker afferent stimulation was unaffected by mGlu5-antagonism in both age-groups. EE significantly amplified STP (<2 h) in old and young animals, but did not increase the duration of synaptic potentiation, or promote induction of LTP. The improvement in STP was prevented by mGlu5-antagonism, in both young and old animals. These results indicate that modifications of the synapse that underlie improvements of LTP by EE require the contribution of mGlu5. Strikingly, although LTP in old mice does not critically depend on mGlu5, improvements in synaptic potentiation resulting from EE are mGlu5-dependent in old mice. Regarded in light of the known role for mGlu5 in hippocampal function and pathophysiology, these data suggest that mGlu5 regulation of synaptic information storage is pivotal to optimal hippocampal function. This article is part of the Special Issue entitled 'Metabotropic Glutamate Receptors, 5 years on'.
Collapse
Affiliation(s)
- Arne Buschler
- Ruhr University Bochum, Medical Faculty, Department of Neurophysiology, Bochum, Germany
| | - Denise Manahan-Vaughan
- Ruhr University Bochum, Medical Faculty, Department of Neurophysiology, Bochum, Germany.
| |
Collapse
|
42
|
Wang Y, Zhao S, Wu Z, Feng Y, Zhao C, Zhang C. Oxytocin in the regulation of social behaviours in medial amygdala-lesioned mice via the inhibition of the extracellular signal-regulated kinase signalling pathway. Clin Exp Pharmacol Physiol 2016; 42:465-74. [PMID: 25707920 DOI: 10.1111/1440-1681.12378] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 01/27/2015] [Accepted: 02/06/2015] [Indexed: 01/22/2023]
Abstract
The neuropeptide oxytocin (OXT) has been implicated in the pathophysiology of behavioural deficits among patients with autism spectrum disorder (ASD). However, the molecular mechanisms underlying its role in ASD remain unclear. In the present study, a murine model with ASD-like phenotypes was induced by intra-medial amygdala injection of N-methyl-d-aspartate, and it was used to investigate the role of OXT in behaviour regulation. Behavioural tests were performed to verify the ASD-like phenotypes of N-methyl-d-aspartate-treated mice, and the results showed that mice with bilateral medial amygdala lesions presented significant behavioural deficits, including impaired learning and memory and increased anxiety and depression. We also observed a notably decreased level of OXT in both the plasma and the hypothalamus of medial amygdala-lesioned mice, and the extracellular signal-regulated kinase (ERK) was activated. Further studies demonstrated that the administration of OXT alleviated ASD-like symptoms and significantly inhibited phosphorylation of ERK; the inhibitory effect was similar to that of U0126, an ERK signalling inhibitor. In addition, OXT administration modulated the expression of downstream proteins of the ERK signalling pathway, such as cyclic adenosine monophosphate response element binding and c-fos. Taken together, our data indicate that OXT plays an important role in ameliorating behavioural deficits in an ASD-like mouse model, which was mediated by inhibiting the ERK signalling pathway and its downstream proteins.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | | | | | | | | | | |
Collapse
|
43
|
Synaptic Wnt/GSK3β Signaling Hub in Autism. Neural Plast 2016; 2016:9603751. [PMID: 26881141 PMCID: PMC4736967 DOI: 10.1155/2016/9603751] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/29/2015] [Accepted: 11/30/2015] [Indexed: 12/29/2022] Open
Abstract
Hundreds of genes have been associated with autism spectrum disorders (ASDs) and the interaction of weak and de novo variants derive from distinct autistic phenotypes thus making up the “spectrum.” The convergence of these variants in networks of genes associated with synaptic function warrants the study of cell signaling pathways involved in the regulation of the synapse. The Wnt/β-catenin signaling pathway plays a central role in the development and regulation of the central nervous system and several genes belonging to the cascade have been genetically associated with ASDs. In the present paper, we review basic information regarding the role of Wnt/β-catenin signaling in excitatory/inhibitory balance (E/I balance) through the regulation of pre- and postsynaptic compartments. Furthermore, we integrate information supporting the role of the glycogen synthase kinase 3β (GSK3β) in the onset/development of ASDs through direct modulation of Wnt/β-catenin signaling. Finally, given GSK3β activity as key modulator of synaptic plasticity, we explore the potential of this kinase as a therapeutic target for ASD.
Collapse
|
44
|
Kazdoba TM, Leach PT, Yang M, Silverman JL, Solomon M, Crawley JN. Translational Mouse Models of Autism: Advancing Toward Pharmacological Therapeutics. Curr Top Behav Neurosci 2016; 28:1-52. [PMID: 27305922 PMCID: PMC5116923 DOI: 10.1007/7854_2015_5003] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Animal models provide preclinical tools to investigate the causal role of genetic mutations and environmental factors in the etiology of autism spectrum disorder (ASD). Knockout and humanized knock-in mice, and more recently knockout rats, have been generated for many of the de novo single gene mutations and copy number variants (CNVs) detected in ASD and comorbid neurodevelopmental disorders. Mouse models incorporating genetic and environmental manipulations have been employed for preclinical testing of hypothesis-driven pharmacological targets, to begin to develop treatments for the diagnostic and associated symptoms of autism. In this review, we summarize rodent behavioral assays relevant to the core features of autism, preclinical and clinical evaluations of pharmacological interventions, and strategies to improve the translational value of rodent models of autism.
Collapse
Affiliation(s)
- Tatiana M Kazdoba
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA
| | - Prescott T Leach
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA
| | - Mu Yang
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA
| | - Jill L Silverman
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA
| | - Marjorie Solomon
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA
| | - Jacqueline N Crawley
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Room 1001A Research 2 Building 96, 4625 2nd Avenue, Sacramento, CA 95817, USA.
| |
Collapse
|
45
|
Daimon CM, Jasien JM, Wood WH, Zhang Y, Becker KG, Silverman JL, Crawley JN, Martin B, Maudsley S. Hippocampal Transcriptomic and Proteomic Alterations in the BTBR Mouse Model of Autism Spectrum Disorder. Front Physiol 2015; 6:324. [PMID: 26635614 PMCID: PMC4656818 DOI: 10.3389/fphys.2015.00324] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 10/27/2015] [Indexed: 12/25/2022] Open
Abstract
Autism spectrum disorders (ASD) are complex heterogeneous neurodevelopmental disorders of an unclear etiology, and no cure currently exists. Prior studies have demonstrated that the black and tan, brachyury (BTBR) T+ Itpr3tf/J mouse strain displays a behavioral phenotype with ASD-like features. BTBR T+ Itpr3tf/J mice (referred to simply as BTBR) display deficits in social functioning, lack of communication ability, and engagement in stereotyped behavior. Despite extensive behavioral phenotypic characterization, little is known about the genes and proteins responsible for the presentation of the ASD-like phenotype in the BTBR mouse model. In this study, we employed bioinformatics techniques to gain a wide-scale understanding of the transcriptomic and proteomic changes associated with the ASD-like phenotype in BTBR mice. We found a number of genes and proteins to be significantly altered in BTBR mice compared to C57BL/6J (B6) control mice controls such as BDNF, Shank3, and ERK1, which are highly relevant to prior investigations of ASD. Furthermore, we identified distinct functional pathways altered in BTBR mice compared to B6 controls that have been previously shown to be altered in both mouse models of ASD, some human clinical populations, and have been suggested as a possible etiological mechanism of ASD, including “axon guidance” and “regulation of actin cytoskeleton.” In addition, our wide-scale bioinformatics approach also discovered several previously unidentified genes and proteins associated with the ASD phenotype in BTBR mice, such as Caskin1, suggesting that bioinformatics could be an avenue by which novel therapeutic targets for ASD are uncovered. As a result, we believe that informed use of synergistic bioinformatics applications represents an invaluable tool for elucidating the etiology of complex disorders like ASD.
Collapse
Affiliation(s)
- Caitlin M Daimon
- Metabolism Unit, National Institute on Aging, National Institutes of Health Baltimore, MD, USA
| | - Joan M Jasien
- Metabolism Unit, National Institute on Aging, National Institutes of Health Baltimore, MD, USA
| | - William H Wood
- Gene Expression and Genomics Unit, National Institutes of Health Baltimore, MD, USA
| | - Yongqing Zhang
- Gene Expression and Genomics Unit, National Institutes of Health Baltimore, MD, USA
| | - Kevin G Becker
- Gene Expression and Genomics Unit, National Institutes of Health Baltimore, MD, USA
| | - Jill L Silverman
- Laboratory of Behavioral Neuroscience, Intramural Research Program, National Institute of Mental Health Bethesda, MD, USA ; MIND Institute, University of California Davis School of Medicine Sacramento, CA, USA
| | - Jacqueline N Crawley
- Laboratory of Behavioral Neuroscience, Intramural Research Program, National Institute of Mental Health Bethesda, MD, USA ; MIND Institute, University of California Davis School of Medicine Sacramento, CA, USA
| | - Bronwen Martin
- Metabolism Unit, National Institute on Aging, National Institutes of Health Baltimore, MD, USA
| | - Stuart Maudsley
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health Baltimore, MD, USA ; Translational Neurobiology Group, VIB Department of Molecular Genetics, University of Antwerp Antwerp, Belgium ; Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp Antwerpen, Belgium
| |
Collapse
|
46
|
|
47
|
Blaser R, Heyser C. Spontaneous object recognition: a promising approach to the comparative study of memory. Front Behav Neurosci 2015; 9:183. [PMID: 26217207 PMCID: PMC4498097 DOI: 10.3389/fnbeh.2015.00183] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 06/29/2015] [Indexed: 01/11/2023] Open
Abstract
Spontaneous recognition of a novel object is a popular measure of exploratory behavior, perception and recognition memory in rodent models. Because of its relative simplicity and speed of testing, the variety of stimuli that can be used, and its ecological validity across species, it is also an attractive task for comparative research. To date, variants of this test have been used with vertebrate and invertebrate species, but the methods have seldom been sufficiently standardized to allow cross-species comparison. Here, we review the methods necessary for the study of novel object recognition in mammalian and non-mammalian models, as well as the results of these experiments. Critical to the use of this test is an understanding of the organism's initial response to a novel object, the modulation of exploration by context, and species differences in object perception and exploratory behaviors. We argue that with appropriate consideration of species differences in perception, object affordances, and natural exploratory behaviors, the spontaneous object recognition test can be a valid and versatile tool for translational research with non-mammalian models.
Collapse
Affiliation(s)
- Rachel Blaser
- Department of Psychological Sciences, University of San DiegoSan Diego, CA, USA
| | - Charles Heyser
- Behavioral Testing Core, Department of Neurosciences, University of California, San DiegoSan Diego, CA, USA
| |
Collapse
|
48
|
Tassin TC, Benavides DR, Plattner F, Nishi A, Bibb JA. Regulation of ERK Kinase by MEK1 Kinase Inhibition in the Brain. J Biol Chem 2015; 290:16319-29. [PMID: 25971971 DOI: 10.1074/jbc.m115.654897] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Indexed: 11/06/2022] Open
Abstract
Metabotropic (slow) and ionotropic (fast) neurotransmission are integrated by intracellular signal transduction mechanisms involving protein phosphorylation/dephosphorylation to achieve experience-dependent alterations in brain circuitry. ERK is an important effector of both slow and fast forms of neurotransmission and has been implicated in normal brain function and CNS diseases. Here we characterize phosphorylation of the ERK-activating protein kinase MEK1 by Cdk5, ERK, and Cdk1 in vitro in intact mouse brain tissue and in the context of an animal behavioral paradigm of stress. Cdk5 only phosphorylates Thr-292, whereas ERK and Cdk1 phosphorylate both Thr-292 and Thr-286 MEK1. These sites interact in a kinase-specific manner and inhibit the ability of MEK1 to activate ERK. Thr-292 and Thr-286 MEK1 are phosphorylated in most mouse brain regions to stoichiometries of ~5% or less. Phosphorylation of Thr-292 MEK1 is regulated by cAMP-dependent signaling in mouse striatum in a manner consistent with negative feedback inhibition in response to ERK activation. Protein phosphatase 1 and 2A contribute to the maintenance of the basal phosphorylation state of both Thr-292 and Thr-286 MEK1 and that of ERK. Activation of the NMDA class of ionotropic glutamate receptors reduces inhibitory MEK1 phosphorylation, whereas forced swim, a paradigm of acute stress, attenuates Thr-292 MEK1 phosphorylation. Together, the data indicate that these inhibitory MEK1 sites phosphorylated by Cdk5 and ERK1 serve as mechanistic points of convergence for the regulation of ERK signaling by both slow and fast neurotransmission.
Collapse
Affiliation(s)
| | - David R Benavides
- the Department of Neurology, The Johns Hopkins Hospital, Baltimore, Maryland 21287, and
| | | | - Akinori Nishi
- the Department of Pharmacology, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - James A Bibb
- From the Departments of Psychiatry and Neurology and Neurotherapeutics and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390,
| |
Collapse
|
49
|
Present and future of developmental neuropsychopharmacology. Eur Neuropsychopharmacol 2015; 25:703-12. [PMID: 25432076 DOI: 10.1016/j.euroneuro.2014.11.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 10/08/2014] [Accepted: 11/04/2014] [Indexed: 11/24/2022]
Abstract
The field of child and adolescent psychiatry has always lagged behind adult psychiatry. With recent evidence that the vast majority of mental disorders, even when they emerge in adulthood, cause abnormal neurodevelopment and resultant emphasis on prevention and early intervention, there is a need to put child psychiatry at the top of the agenda in mental health research. This should also be the case for developmental neuropsychopharmacology. The target of drug discovery should shift toward a population younger than the one that is typically included in clinical trials. This is not only a matter of trying to replicate what has been found in individuals with mature brains; it is about searching for new strategies that address developing brains while the therapeutic window for their effect is still open. At present, major concerns in developmental psychopharmacology are over-prescription rates and use of psychotropic medications for conditions with a particularly underdeveloped evidence base, as well as adverse effects, especially potentially life-shortening cardiometabolic effects and suicidal ideation. The future of research in this area should focus on the use of drugs for primary and secondary prevention that would modify abnormal brain development.
Collapse
|
50
|
Lindemann L, Porter RH, Scharf SH, Kuennecke B, Bruns A, von Kienlin M, Harrison AC, Paehler A, Funk C, Gloge A, Schneider M, Parrott NJ, Polonchuk L, Niederhauser U, Morairty SR, Kilduff TS, Vieira E, Kolczewski S, Wichmann J, Hartung T, Honer M, Borroni E, Moreau JL, Prinssen E, Spooren W, Wettstein JG, Jaeschke G. Pharmacology of basimglurant (RO4917523, RG7090), a unique metabotropic glutamate receptor 5 negative allosteric modulator in clinical development for depression. J Pharmacol Exp Ther 2015; 353:213-33. [PMID: 25665805 DOI: 10.1124/jpet.114.222463] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
Major depressive disorder (MDD) is a serious public health burden and a leading cause of disability. Its pharmacotherapy is currently limited to modulators of monoamine neurotransmitters and second-generation antipsychotics. Recently, glutamatergic approaches for the treatment of MDD have increasingly received attention, and preclinical research suggests that metabotropic glutamate receptor 5 (mGlu5) inhibitors have antidepressant-like properties. Basimglurant (2-chloro-4-[1-(4-fluoro-phenyl)-2,5-dimethyl-1H-imidazol-4-ylethynyl]-pyridine) is a novel mGlu5 negative allosteric modulator currently in phase 2 clinical development for MDD and fragile X syndrome. Here, the comprehensive preclinical pharmacological profile of basimglurant is presented with a focus on its therapeutic potential for MDD and drug-like properties. Basimglurant is a potent, selective, and safe mGlu5 inhibitor with good oral bioavailability and long half-life supportive of once-daily administration, good brain penetration, and high in vivo potency. It has antidepressant properties that are corroborated by its functional magnetic imaging profile as well as anxiolytic-like and antinociceptive features. In electroencephalography recordings, basimglurant shows wake-promoting effects followed by increased delta power during subsequent non-rapid eye movement sleep. In microdialysis studies, basimglurant had no effect on monoamine transmitter levels in the frontal cortex or nucleus accumbens except for a moderate increase of accumbal dopamine, which is in line with its lack of pharmacological activity on monoamine reuptake transporters. These data taken together, basimglurant has favorable drug-like properties, a differentiated molecular mechanism of action, and antidepressant-like features that suggest the possibility of also addressing important comorbidities of MDD including anxiety and pain as well as daytime sleepiness and apathy or lethargy.
Collapse
Affiliation(s)
- Lothar Lindemann
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Richard H Porter
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Sebastian H Scharf
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Basil Kuennecke
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Andreas Bruns
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Markus von Kienlin
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Anthony C Harrison
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Axel Paehler
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Christoph Funk
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Andreas Gloge
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Manfred Schneider
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Neil J Parrott
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Liudmila Polonchuk
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Urs Niederhauser
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Stephen R Morairty
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Thomas S Kilduff
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Eric Vieira
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Sabine Kolczewski
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Juergen Wichmann
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Thomas Hartung
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Michael Honer
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Edilio Borroni
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Jean-Luc Moreau
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Eric Prinssen
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Will Spooren
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Joseph G Wettstein
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| | - Georg Jaeschke
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases (L.L., S.H.S., B.K., A.B., M.v.K., M.H., E.B., E.P., W.S., J.G.W.), Discovery Chemistry (E.V., S.K., J.W., G.J.), Operations for Neuroscience, Ophthalmology, and Rare Diseases (R.H.P., J.-L.M.), Pharmaceutical Sciences (A.C.H., A.P., C.F., A.G., M.S., N.J.P., L.P., U.N.), and Small Molecules Process Research and Synthesis (T.H.), Roche Innovation Center Basel, Basel, Switzerland; and Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California (S.R.M., T.S.K.)
| |
Collapse
|