51
|
Vázquez-Oliver A, Brambilla-Pisoni C, Domingo-Gainza M, Maldonado R, Ivorra A, Ozaita A. Auricular transcutaneous vagus nerve stimulation improves memory persistence in naïve mice and in an intellectual disability mouse model. Brain Stimul 2020; 13:494-498. [DOI: 10.1016/j.brs.2019.12.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 12/10/2019] [Accepted: 12/23/2019] [Indexed: 11/16/2022] Open
|
52
|
Möhrle D, Fernández M, Peñagarikano O, Frick A, Allman B, Schmid S. What we can learn from a genetic rodent model about autism. Neurosci Biobehav Rev 2020; 109:29-53. [DOI: 10.1016/j.neubiorev.2019.12.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/28/2019] [Accepted: 12/10/2019] [Indexed: 12/15/2022]
|
53
|
Gonzalez D, Tomasek M, Hays S, Sridhar V, Ammanuel S, Chang CW, Pawlowski K, Huber KM, Gibson JR. Audiogenic Seizures in the Fmr1 Knock-Out Mouse Are Induced by Fmr1 Deletion in Subcortical, VGlut2-Expressing Excitatory Neurons and Require Deletion in the Inferior Colliculus. J Neurosci 2019; 39:9852-9863. [PMID: 31666356 PMCID: PMC6891051 DOI: 10.1523/jneurosci.0886-19.2019] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 09/06/2019] [Accepted: 10/16/2019] [Indexed: 02/07/2023] Open
Abstract
Fragile X syndrome (FXS) is the most common form of inherited intellectual disability and the leading monogenetic cause of autism. One symptom of FXS and autism is sensory hypersensitivity (also called sensory over-responsivity). Perhaps related to this, the audiogenic seizure (AGS) is arguably the most robust behavioral phenotype in the FXS mouse model-the Fmr1 knock-out (KO) mouse. Therefore, the AGS may be considered a mouse model of sensory hypersensitivity. Hyperactive circuits are hypothesized to underlie dysfunction in a number of brain regions in patients with FXS and Fmr1 KO mice, and the AGS may be a result of this. But the specific cell types and brain regions underlying AGSs in the Fmr1 KO are unknown. We used conditional deletion or expression of Fmr1 in different cell populations to determine whether Fmr1 deletion in those cells was sufficient or necessary, respectively, for the AGS phenotype in males. Our data indicate that Fmr1 deletion in glutamatergic neurons that express vesicular glutamate transporter 2 (VGlut2) and are located in subcortical brain regions is sufficient and necessary to cause AGSs. Furthermore, the deletion of Fmr1 in glutamatergic neurons of the inferior colliculus is necessary for AGSs. When we demonstrate necessity, we show that Fmr1 expression in either the larger population of VGlut2-expressing glutamatergic neurons or the smaller population of inferior collicular glutamatergic neurons-in an otherwise Fmr1 KO mouse-eliminates AGSs. Therefore, targeting these neuronal populations in FXS and autism may be part of a therapeutic strategy to alleviate sensory hypersensitivity.SIGNIFICANCE STATEMENT Sensory hypersensitivity in fragile X syndrome (FXS) and autism patients significantly interferes with quality of life. Audiogenic seizures (AGSs) are arguably the most robust behavioral phenotype in the FXS mouse model-the Fmr1 knockout-and may be considered a model of sensory hypersensitivity in FXS. We provide the clearest and most precise genetic evidence to date for the cell types and brain regions involved in causing AGSs in the Fmr1 knockout and, more broadly, for any mouse mutant. The expression of Fmr1 in these same cell types in an otherwise Fmr1 knockout eliminates AGSs indicating possible cellular targets for alleviating sensory hypersensitivity in FXS and other forms of autism.
Collapse
Affiliation(s)
| | | | - Seth Hays
- Department of Neuroscience, Dallas, and
| | | | | | | | - Karen Pawlowski
- Department of Otolaryngology and Biomedical Engineering Program, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9035
| | | | | |
Collapse
|
54
|
Fleury-Teixeira P, Caixeta FV, Ramires da Silva LC, Brasil-Neto JP, Malcher-Lopes R. Effects of CBD-Enriched Cannabis sativa Extract on Autism Spectrum Disorder Symptoms: An Observational Study of 18 Participants Undergoing Compassionate Use. Front Neurol 2019; 10:1145. [PMID: 31736860 PMCID: PMC6834767 DOI: 10.3389/fneur.2019.01145] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 10/14/2019] [Indexed: 12/17/2022] Open
Abstract
Autism Spectrum Disorders comprise conditions that may affect cognitive development, motor skills, social interaction, communication, and behavior. This set of functional deficits often results in lack of independence for the diagnosed individuals, and severe distress for patients, families, and caregivers. There is a mounting body of evidence indicating the effectiveness of pure cannabidiol (CBD) and CBD-enriched Cannabis sativa extract (CE) for the treatment of autistic symptoms in refractory epilepsy patients. There is also increasing data support for the hypothesis that non-epileptic autism shares underlying etiological mechanisms with epilepsy. Here we report an observational study with a cohort of 18 autistic patients undergoing treatment with compassionate use of standardized CBD-enriched CE (with a CBD to THC ratio of 75/1). Among the 15 patients who adhered to the treatment (10 non-epileptic and five epileptic) only one patient showed lack of improvement in autistic symptoms. Due to adverse effects, three patients discontinued CE use before 1 month. After 6-9 months of treatment, most patients, including epileptic and non-epileptic, showed some level of improvement in more than one of the eight symptom categories evaluated: Attention Deficit/Hyperactivity Disorder; Behavioral Disorders; Motor Deficits; Autonomy Deficits; Communication and Social Interaction Deficits; Cognitive Deficits; Sleep Disorders and Seizures, with very infrequent and mild adverse effects. The strongest improvements were reported for Seizures, Attention Deficit/Hyperactivity Disorder, Sleep Disorders, and Communication and Social Interaction Deficits. This was especially true for the 10 non-epileptic patients, nine of which presented improvement equal to or above 30% in at least one of the eight categories, six presented improvement of 30% or more in at least two categories and four presented improvement equal to or above 30% in at least four symptom categories. Ten out of the 15 patients were using other medicines, and nine of these were able to keep the improvements even after reducing or withdrawing other medications. The results reported here are very promising and indicate that CBD-enriched CE may ameliorate multiple ASD symptoms even in non-epileptic patients, with substantial increase in life quality for both ASD patients and caretakers.
Collapse
Affiliation(s)
| | | | - Leandro Cruz Ramires da Silva
- Clinical Hospital, Federal University of Minas Gerais, Belo Horizonte, Brazil.,Associação Brasileira de Pacientes de Cannabis Medicinal, Belo Horizonte, Brazil
| | | | | |
Collapse
|
55
|
Agarwal R, Burke SL, Maddux M. Current state of evidence of cannabis utilization for treatment of autism spectrum disorders. BMC Psychiatry 2019; 19:328. [PMID: 31664964 PMCID: PMC6819459 DOI: 10.1186/s12888-019-2259-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 08/26/2019] [Indexed: 02/07/2023] Open
Abstract
The core symptoms and co-morbidities associated with autism spectrum disorders (ASD) affect daily living and quality of life. Existing pharmacological interventions are only able to attenuate some related symptoms but are unable to address the underlying etiologies associated with ASD. Anecdotal evidence, which claims benefit from the use of cannabis to treat symptoms among this population, has been gaining popularity as families seek solutions.This paper analyzed recent peer-reviewed literature to identify the current state of evidence regarding cannabis use for the ASD population. Systematic reviews, reports, and experimental studies were assessed to understand the current extent and nature of the evidence on the risks and benefits of cannabis use for ASD. At this time, three large-scale clinical trials are currently at varying stages of progress and publication of results. Only five small studies were identified that have specifically examined cannabis use in ASD. Given the sparse state of evidence directly assessed in this population, studies which examined effects of cannabis on shared pathological symptoms of ASD such as hyperactivity, sleep disorders, self-injury, anxiety, behavioral problems, and communication were also reviewed.Studies revealed mixed and inconclusive findings of cannabis effects for all conditions, except epilepsy. Adverse outcomes were also reported, which included severe psychosis, increased agitation, somnolence, decreased appetite, and irritability. In addition, a wide range of cannabis compositions and dosage were identified within the studies, which impact generalizability.There is currently insufficient evidence for cannabis use in ASD, which creates an urgent need for additional large-scale controlled studies to increase understanding of risks and benefits and also to examine the impact of "entourage effects." This will support discussions of treatment options between health care providers and ASD patients and their families. Evidence may lead to a desired new line of treatment or prevent adverse outcomes from unsubstantiated use amongst families aiming for symptom reduction.
Collapse
Affiliation(s)
- Rumi Agarwal
- FIU Embrace Florida International University, 11200 SW 8th Street, Miami, FL 33199 USA
- BRAINN Lab, Florida International University School of Social Work, 11200 SW 8th Street, Miami, FL 33199 USA
- Florida International University School of Public Health Robert Stempel College of Public Health and Social Work Health Promotion and Disease Prevention, 11200 S.W. 8th Street, Miami, FL 33199 USA
| | - Shanna L. Burke
- BRAINN Lab, Florida International University School of Social Work, 11200 SW 8th Street, Miami, FL 33199 USA
- Florida International University School of Social Work Robert Stempel College of Public Health and Social Work, 11200 S.W. 8th Street, AHC5 585, Miami, FL 33199 USA
| | - Marlaina Maddux
- FIU Embrace Florida International University, 11200 SW 8th Street, Miami, FL 33199 USA
- BRAINN Lab, Florida International University School of Social Work, 11200 SW 8th Street, Miami, FL 33199 USA
- Easterseals Blake Foundation, 750 E Broadway Blvd, Tucson, AZ 85710 USA
| |
Collapse
|
56
|
Martínez-Torres S, Gomis-González M, Navarro-Romero A, Maldonado R, Ozaita A. Use of the Vsoc-maze to Study Sociability and Preference for Social Novelty in Rodents. Bio Protoc 2019; 9:e3393. [PMID: 33654894 DOI: 10.21769/bioprotoc.3393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/24/2019] [Accepted: 09/29/2019] [Indexed: 11/02/2022] Open
Abstract
Studying social behavior in mouse models empowers the understanding of the neurobiological mechanisms involved, which are affected in neuropsychiatric disorders, allowing the evaluation of therapeutic strategies. Behavioral methods available are time-consuming and reducing the length of behavioral sessions may render more manageable experiments and reduce animal stress. We validated a new reliable and sensitive method to study two features of social behavior (sociability and preference for social novelty) in two strains of male mice, the C57BL/6J inbreed strain and the CD1 (ICR) outbreed strain, using a modified version of the V-shaped maze (Vsoc-maze). The Vsoc-maze for sociability and preference for social novelty improves time performance by shortening the length of the sessions, and reduces variability compared to the classical approach performed in the three-chamber apparatus. Altogether, the Vsoc-maze allows evaluating the specific alterations of social behavior in mice in a time-efficient and reproducible manner.
Collapse
Affiliation(s)
- Sara Martínez-Torres
- Laboratory of Neuropharmacology. Department of Experimental and Health Sciences, University Pompeu Fabra, 08003 Barcelona, Spain
| | - Maria Gomis-González
- Laboratory of Neuropharmacology. Department of Experimental and Health Sciences, University Pompeu Fabra, 08003 Barcelona, Spain
| | - Alba Navarro-Romero
- Laboratory of Neuropharmacology. Department of Experimental and Health Sciences, University Pompeu Fabra, 08003 Barcelona, Spain
| | - Rafael Maldonado
- Laboratory of Neuropharmacology. Department of Experimental and Health Sciences, University Pompeu Fabra, 08003 Barcelona, Spain
| | - Andrés Ozaita
- Laboratory of Neuropharmacology. Department of Experimental and Health Sciences, University Pompeu Fabra, 08003 Barcelona, Spain
| |
Collapse
|
57
|
Felgerolle C, Hébert B, Ardourel M, Meyer-Dilhet G, Menuet A, Pinto-Morais K, Bizot JC, Pichon J, Briault S, Perche O. Visual Behavior Impairments as an Aberrant Sensory Processing in the Mouse Model of Fragile X Syndrome. Front Behav Neurosci 2019; 13:228. [PMID: 31680892 PMCID: PMC6797836 DOI: 10.3389/fnbeh.2019.00228] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/12/2019] [Indexed: 12/02/2022] Open
Abstract
Fragile X Syndrome (FXS), the most common inherited form of human intellectual disability (ID) associated with autistic-like behaviors, is characterized by dys-sensitivity to sensory stimuli, especially vision. In the absence of Fragile Mental Retardation Protein (FMRP), both retinal and cerebral structures of the visual pathway are impaired, suggesting that perception and integration of visual stimuli are altered. However, behavioral consequences of these defects remain unknown. In this study, we used male Fmr1−/y mice to further define visual disturbances from a behavioral perspective by focusing on three traits characterizing visual modality: perception of depth, contrasts and movements. We performed specific tests (Optomotor Drum, Visual Cliff) to evaluate these visual modalities, their evolution from youth to adulthood, and to assess their involvement in a cognitive task. We show that Fmr1−/y mice exhibit alteration in their visual skills, displaying impaired perspective perception, a drop in their ability to understand a moving contrasted pattern, and a defect in contrasts discrimination. Interestingly, Fmr1−/y phenotypes remain stable over time from adolescence to late adulthood. Besides, we report that color and shape are meaningful for the achievement of a cognitive test involving object recognition. Altogether, these results underline the significance of visual behavior alterations in FXS conditions and relevance of assessing visual skills in neuropsychiatric models before performing behavioral tasks, such as cognitive assessments, that involve visual discrimination.
Collapse
Affiliation(s)
- Chloé Felgerolle
- UMR7355, CNRS, Orléans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| | - Betty Hébert
- UMR7355, CNRS, Orléans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| | - Maryvonne Ardourel
- UMR7355, CNRS, Orléans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| | - Géraldine Meyer-Dilhet
- UMR7355, CNRS, Orléans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| | - Arnaud Menuet
- UMR7355, CNRS, Orléans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| | - Kimberley Pinto-Morais
- UMR7355, CNRS, Orléans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| | | | - Jacques Pichon
- UMR7355, CNRS, Orléans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| | - Sylvain Briault
- UMR7355, CNRS, Orléans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France.,Department of Genetics, Regional Hospital, Orléans, France
| | - Olivier Perche
- UMR7355, CNRS, Orléans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France.,Department of Genetics, Regional Hospital, Orléans, France
| |
Collapse
|
58
|
Zamberletti E, Gabaglio M, Woolley-Roberts M, Bingham S, Rubino T, Parolaro D. Cannabidivarin Treatment Ameliorates Autism-Like Behaviors and Restores Hippocampal Endocannabinoid System and Glia Alterations Induced by Prenatal Valproic Acid Exposure in Rats. Front Cell Neurosci 2019; 13:367. [PMID: 31447649 PMCID: PMC6696797 DOI: 10.3389/fncel.2019.00367] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 07/29/2019] [Indexed: 12/13/2022] Open
Abstract
Autism spectrum disorder (ASD) is a developmental condition whose primary features include social communication and interaction impairments with restricted or repetitive motor movements. No approved treatment for the core symptoms is available and considerable research efforts aim at identifying effective therapeutic strategies. Emerging evidence suggests that altered endocannabinoid signaling and immune dysfunction might contribute to ASD pathogenesis. In this scenario, phytocannabinoids could hold great pharmacological potential due to their combined capacities to act either directly or indirectly on components of the endocannabinoid system and to modulate immune functions. Among all plant-cannabinoids, the phytocannabinoid cannabidivarin (CBDV) was recently shown to reduce motor impairments and cognitive deficits in animal models of Rett syndrome, a condition showing some degree of overlap with autism, raising the possibility that CBDV might have therapeutic potential in ASD. Here, we investigated the ability of CBDV treatment to reverse or prevent ASD-like behaviors in male rats prenatally exposed to valproic acid (VPA; 500 mg/kg i.p.; gestation day 12.5). The offspring received CBDV according to two different protocols: symptomatic (0.2/2/20/100 mg/kg i.p.; postnatal days 34–58) and preventative (2/20 mg/kg i.p.; postnatal days 19–32). The major efficacy of CBDV was observed at the dose of 20 mg/kg for both treatment schedules. CBDV in symptomatic rats recovered social impairments, social novelty preference, short-term memory deficits, repetitive behaviors and hyperlocomotion whereas preventative treatment reduced sociability and social novelty deficits, short-term memory impairments and hyperlocomotion, without affecting stereotypies. As dysregulations in the endocannabinoid system and neuroinflammatory markers contribute to the development of some ASD phenotypes in the VPA model, neurochemical studies were performed after symptomatic treatment to investigate possible CBDV’s effects on the endocannabinoid system, inflammatory markers and microglia activation in the hippocampus and prefrontal cortex. Prenatal VPA exposure increased CB1 receptor, FAAH and MAGL levels, enhanced GFAP, CD11b, and TNFα levels and triggered microglia activation restricted to the hippocampus. All these alterations were restored after CBDV treatment. These data provide preclinical evidence in support of the ability of CBDV to ameliorate behavioral abnormalities resembling core and associated symptoms of ASD. At the neurochemical level, symptomatic CBDV restores hippocampal endocannabinoid signaling and neuroinflammation induced by prenatal VPA exposure.
Collapse
Affiliation(s)
- Erica Zamberletti
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Marina Gabaglio
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | | | | | - Tiziana Rubino
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Daniela Parolaro
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy.,Zardi-Gori Foundation, Milan, Italy
| |
Collapse
|
59
|
Sossin WS, Costa-Mattioli M. Translational Control in the Brain in Health and Disease. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a032912. [PMID: 30082469 DOI: 10.1101/cshperspect.a032912] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Translational control in neurons is crucially required for long-lasting changes in synaptic function and memory storage. The importance of protein synthesis control to brain processes is underscored by the large number of neurological disorders in which translation rates are perturbed, such as autism and neurodegenerative disorders. Here we review the general principles of neuronal translation, focusing on the particular relevance of several key regulators of nervous system translation, including eukaryotic initiation factor 2α (eIF2α), the mechanistic (or mammalian) target of rapamycin complex 1 (mTORC1), and the eukaryotic elongation factor 2 (eEF2). These pathways regulate the overall rate of protein synthesis in neurons and have selective effects on the translation of specific messenger RNAs (mRNAs). The importance of these general and specific translational control mechanisms is considered in the normal functioning of the nervous system, particularly during synaptic plasticity underlying memory, and in the context of neurological disorders.
Collapse
Affiliation(s)
- Wayne S Sossin
- Montreal Neurological Institute, McGill University, Montreal, Quebec H3A-2B4, Canada
| | - Mauro Costa-Mattioli
- Department of Neuroscience, Memory and Brain Research Center, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
60
|
Martín-Sánchez A, Warnault V, Montagud-Romero S, Pastor A, Mondragón N, De La Torre R, Valverde O. Alcohol-induced conditioned place preference is modulated by CB2 cannabinoid receptors and modifies levels of endocannabinoids in the mesocorticolimbic system. Pharmacol Biochem Behav 2019; 183:22-31. [DOI: 10.1016/j.pbb.2019.06.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 06/06/2019] [Accepted: 06/16/2019] [Indexed: 02/02/2023]
|
61
|
Zamberletti E, Gabaglio M, Piscitelli F, Brodie JS, Woolley-Roberts M, Barbiero I, Tramarin M, Binelli G, Landsberger N, Kilstrup-Nielsen C, Rubino T, Di Marzo V, Parolaro D. Cannabidivarin completely rescues cognitive deficits and delays neurological and motor defects in male Mecp2 mutant mice. J Psychopharmacol 2019; 33:894-907. [PMID: 31084246 DOI: 10.1177/0269881119844184] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Recent evidence suggests that 2-week treatment with the non-psychotomimetic cannabinoid cannabidivarin (CBDV) could be beneficial towards neurological and social deficits in early symptomatic Mecp2 mutant mice, a model of Rett syndrome (RTT). AIM The aim of this study was to provide further insights into the efficacy of CBDV in Mecp2-null mice using a lifelong treatment schedule (from 4 to 9 weeks of age) to evaluate its effect on recognition memory and neurological defects in both early and advanced stages of the phenotype progression. METHODS CBDV 0.2, 2, 20 and 200 mg/kg/day was administered to Mecp2-null mice from 4 to 9 weeks of age. Cognitive and neurological defects were monitored during the whole treatment schedule. Biochemical analyses were carried out in brain lysates from 9-week-old wild-type and knockout mice to evaluate brain-derived neurotrophic factor (BDNF) and insulin-like growth factor-1 (IGF-1) levels as well as components of the endocannabinoid system. RESULTS CBDV rescues recognition memory deficits in Mecp2 mutant mice and delays the appearance of neurological defects. At the biochemical level, it normalizes BDNF/IGF1 levels and the defective PI3K/AKT/mTOR pathway in Mecp2 mutant mice at an advanced stage of the disease. Mecp2 deletion upregulates CB1 and CB2 receptor levels in the brain and these changes are restored after CBDV treatment. CONCLUSIONS CBDV administration exerts an enduring rescue of memory deficits in Mecp2 mutant mice, an effect that is associated with the normalization of BDNF, IGF-1 and rpS6 phosphorylation levels as well as CB1 and CB2 receptor expression. CBDV delays neurological defects but this effect is only transient.
Collapse
Affiliation(s)
- Erica Zamberletti
- 1 Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Varese, Italy
| | - Marina Gabaglio
- 1 Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Varese, Italy
| | - Fabiana Piscitelli
- 2 Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli, Naples, Italy
| | | | | | - Isabella Barbiero
- 1 Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Varese, Italy
| | - Marco Tramarin
- 1 Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Varese, Italy
| | - Giorgio Binelli
- 1 Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Varese, Italy
| | - Nicoletta Landsberger
- 4 Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | | | - Tiziana Rubino
- 1 Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Varese, Italy
| | - Vincenzo Di Marzo
- 2 Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Pozzuoli, Naples, Italy
| | - Daniela Parolaro
- 1 Department of Biotechnology and Life Sciences (DBSV), University of Insubria, Varese, Italy.,5 Zardi Gori Foundation, Milan, Italy
| |
Collapse
|
62
|
Lovastatin, not Simvastatin, Corrects Core Phenotypes in the Fragile X Mouse Model. eNeuro 2019; 6:ENEURO.0097-19.2019. [PMID: 31147392 PMCID: PMC6565377 DOI: 10.1523/eneuro.0097-19.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/11/2019] [Accepted: 05/13/2019] [Indexed: 02/07/2023] Open
Abstract
The cholesterol-lowering drug lovastatin corrects neurological phenotypes in animal models of fragile X syndrome (FX), a commonly identified genetic cause of autism and intellectual disability (ID). The therapeutic efficacy of lovastatin is being tested in clinical trials for FX; however, the structurally similar drug simvastatin has been proposed as an alternative due to an increased potency and brain penetrance. Here, we perform a side-by-side comparison of the effects of lovastatin and simvastatin treatment on two core phenotypes in Fmr1-/y mice versus WT littermates: excessive hippocampal protein synthesis and susceptibility to audiogenic seizures (AGSs). We find that simvastatin does not correct excessive hippocampal protein synthesis in the Fmr1-/y hippocampus at any dose tested. In fact, simvastatin significantly increases protein synthesis in both Fmr1-/y and WT. Moreover, injection of simvastatin does not reduce AGS in the Fmr1-/y mouse, while lovastatin significantly reduces AGS incidence and severity versus vehicle-treated animals. These results show that unlike lovastatin, simvastatin does not correct core phenotypes in the Fmr1-/y mouse model.
Collapse
|
63
|
Cannabidiol (CBD) reduces anxiety-related behavior in mice via an FMRP-independent mechanism. Pharmacol Biochem Behav 2019; 181:93-100. [DOI: 10.1016/j.pbb.2019.05.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/21/2019] [Accepted: 05/01/2019] [Indexed: 11/18/2022]
|
64
|
de la Torre R, de Sola S, Farré M, Xicota L, Cuenca-Royo A, Rodriguez J, León A, Langohr K, Gomis-González M, Hernandez G, Esteba S, Del Hoyo L, Sánchez-Gutiérrez J, Cortés MJ, Ozaita A, Espadaler JM, Novell R, Martínez-Leal R, Milá M, Dierssen M. A phase 1, randomized double-blind, placebo controlled trial to evaluate safety and efficacy of epigallocatechin-3-gallate and cognitive training in adults with Fragile X syndrome. Clin Nutr 2019; 39:378-387. [PMID: 30962103 DOI: 10.1016/j.clnu.2019.02.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 02/13/2019] [Accepted: 02/16/2019] [Indexed: 11/18/2022]
Abstract
BACKGROUND & AIMS Despite the wide spectrum of experimental compounds tested in clinical trials, there is still no proven pharmacological treatment available for Fragile-X syndrome (FXS), since several targeted clinical trials with high expectations of success have failed to demonstrate significant improvements. Here we tested epigallocatechin-3-gallate (EGCG) as a treatment option for ameliorating core cognitive and behavioral features in FXS. METHODS We conducted preclinical studies in Fmr1 knockout mice (Fmr1-/y) using novel object-recognition memory paradigm upon acute EGCG (10 mg/kg) administration. Furthermore we conducted a double-blind placebo-controlled phase I clinical trial (TESXF; NCT01855971). Twenty-seven subjects with FXS (18-55 years) were administered of EGCG (5-7 mg/kg/day) combined with cognitive training (CT) during 3 months with 3 months of follow-up after treatment discontinuation. RESULTS Preclinical studies showed an improvement in memory using the Novel Object Recognition paradigm. We found that FXS patients receiving EGCG + CT significantly improved cognition (visual episodic memory) and functional competence (ABAS II-Home Living skills) in everyday life compared to subjects receiving Placebo + CT. CONCLUSIONS Phase 2 clinical trials in larger groups of subjects are necessary to establish the therapeutic potential of EGCG for the improvement of cognition and daily life competences in FXS.
Collapse
Affiliation(s)
- Rafael de la Torre
- IMIM-Hospital del Mar Medical Research Institute, E-08003 Barcelona, Spain; CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), E-08003 Barcelona, Spain; University Pompeu Fabra (CEXS-UPF), E-08003 Barcelona, Spain.
| | - Susana de Sola
- IMIM-Hospital del Mar Medical Research Institute, E-08003 Barcelona, Spain; CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), E-08003 Barcelona, Spain
| | - Magí Farré
- Autonomous University of Barcelona (UDIMAS-UAB), E-08003 Barcelona, Spain
| | - Laura Xicota
- IMIM-Hospital del Mar Medical Research Institute, E-08003 Barcelona, Spain; CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), E-08003 Barcelona, Spain
| | - Aida Cuenca-Royo
- IMIM-Hospital del Mar Medical Research Institute, E-08003 Barcelona, Spain; CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), E-08003 Barcelona, Spain
| | - Joan Rodriguez
- IMIM-Hospital del Mar Medical Research Institute, E-08003 Barcelona, Spain; CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), E-08003 Barcelona, Spain
| | - Alba León
- Neurofunctionality of Brain and Language Research Group-Neurosciences Program, IMIM-Hospital del Mar Medical Research Institute, E-08003 Barcelona, Spain
| | - Klaus Langohr
- IMIM-Hospital del Mar Medical Research Institute, E-08003 Barcelona, Spain; Polytechnic University of Catalonia, E-08034 Barcelona, Spain
| | | | - Gimena Hernandez
- IMIM-Hospital del Mar Medical Research Institute, E-08003 Barcelona, Spain; CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), E-08003 Barcelona, Spain
| | - Susanna Esteba
- Parc Hospitalari Martí í Julià-Institut d'Assistència Sanitària, E-17190 Salt, Spain
| | - Laura Del Hoyo
- IMIM-Hospital del Mar Medical Research Institute, E-08003 Barcelona, Spain; CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), E-08003 Barcelona, Spain
| | | | - Maria José Cortés
- Intellectual Disabilities and Developmental Disorders Research Unit (UNIVIDD), Fundació Villablanca, IISPV, URV, CIBERSAM, E-43206, Reus, Spain
| | - Andrés Ozaita
- University Pompeu Fabra (CEXS-UPF), E-08003 Barcelona, Spain
| | - Josep María Espadaler
- Neurofunctionality of Brain and Language Research Group-Neurosciences Program, IMIM-Hospital del Mar Medical Research Institute, E-08003 Barcelona, Spain
| | - Ramón Novell
- Parc Hospitalari Martí í Julià-Institut d'Assistència Sanitària, E-17190 Salt, Spain
| | - Rafael Martínez-Leal
- Intellectual Disabilities and Developmental Disorders Research Unit (UNIVIDD), Fundació Villablanca, IISPV, URV, CIBERSAM, E-43206, Reus, Spain
| | - Montserrat Milá
- Biochemistry and Molecular Genetics Department, Hospital Clínic I Provincial de Barcelona, E-08036, Spain
| | - Mara Dierssen
- IMIM-Hospital del Mar Medical Research Institute, E-08003 Barcelona, Spain; University Pompeu Fabra (CEXS-UPF), E-08003 Barcelona, Spain; Center for Genomic Regulation (CRG), E-08003 Barcelona, Spain; CIBER of Rare Diseases (CIBERER), E-08003 Barcelona, Spain; The Barcelona Institute of Science and Technology, E-08003 Barcelona, Spain.
| |
Collapse
|
65
|
Zou M, Li D, Li L, Wu L, Sun C. Role of the endocannabinoid system in neurological disorders. Int J Dev Neurosci 2019; 76:95-102. [PMID: 30858029 DOI: 10.1016/j.ijdevneu.2019.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 01/13/2023] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder that begins in infancy. Although the etiology and pathogenesis are poorly understood, many studies have shown that ASD is closely related to structural and functional defects in the nervous system, especially synaptic transmission. The endocannabinoid (eCB) system is an important regulatory system of the central nervous system that regulates neurotransmission and synaptic plasticity and plays an important role in emotional and social responses and cognitive function. The relationship between eCB system and ASD has attracted increasing attention from scholars. In this review, we discuss the complex lipid signaling network of the eCB system, intracellular transport pathways, abnormal expression and association with various neurological diseases, and direct and indirect evidence for the link between eCB and ASD. Collectively, the findings to date indicate that the eCB system plays a key role in the pathophysiology of ASD and can provide new insights into potential interventions and rehabilitation strategies for ASD.
Collapse
Affiliation(s)
- Mingyang Zou
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin, 150081, China
| | - Dexin Li
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin, 150081, China
| | - Ling Li
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin, 150081, China
| | - Lijie Wu
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin, 150081, China
| | - Caihong Sun
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin, 150081, China
| |
Collapse
|
66
|
Jacquemont S, Pacini L, Jønch AE, Cencelli G, Rozenberg I, He Y, D'Andrea L, Pedini G, Eldeeb M, Willemsen R, Gasparini F, Tassone F, Hagerman R, Gomez-Mancilla B, Bagni C. Protein synthesis levels are increased in a subset of individuals with fragile X syndrome. Hum Mol Genet 2019; 27:2039-2051. [PMID: 29590342 PMCID: PMC5985734 DOI: 10.1093/hmg/ddy099] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/15/2018] [Indexed: 12/15/2022] Open
Abstract
Fragile X syndrome (FXS) is a monogenic form of intellectual disability and autism spectrum disorder caused by the absence of the fragile X mental retardation protein (FMRP). In biological models for the disease, this leads to upregulated mRNA translation and as a consequence, deficits in synaptic architecture and plasticity. Preclinical studies revealed that pharmacological interventions restore those deficits, which are thought to mediate the FXS cognitive and behavioral symptoms. Here, we characterized the de novo rate of protein synthesis in patients with FXS and their relationship with clinical severity. We measured the rate of protein synthesis in fibroblasts derived from 32 individuals with FXS and from 17 controls as well as in fibroblasts and primary neurons of 27 Fmr1 KO mice and 20 controls. Here, we show that levels of protein synthesis are increased in fibroblasts of individuals with FXS and Fmr1 KO mice. However, this cellular phenotype displays a broad distribution and a proportion of fragile X individuals and Fmr1 KO mice do not show increased levels of protein synthesis, having measures in the normal range. Because the same Fmr1 KO animal measures in fibroblasts predict those in neurons we suggest the validity of this peripheral biomarker. Our study offers a potential explanation for the comprehensive drug development program undertaken thus far yielding negative results and suggests that a significant proportion, but not all individuals with FXS, may benefit from the reduction of excessive levels of protein synthesis.
Collapse
Affiliation(s)
- Sébastien Jacquemont
- Sainte-Justine University Hospital Research Centre, Montreal, QC H3T 1C5.,University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Laura Pacini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Aia E Jønch
- Department of Clinical Genetics, Odense University Hospital.,Human Genetics, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Giulia Cencelli
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Izabela Rozenberg
- Neuroscience Translational Medicine, Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Yunsheng He
- Biomarker Development, Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Laura D'Andrea
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Giorgia Pedini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Marwa Eldeeb
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California, Davis Medical Center, Sacramento, CA 95817, USA
| | - Rob Willemsen
- Department of Clinical Genetics, Erasmus Medical Center, 1738, 3000DR Rotterdam, The Netherlands
| | - Fabrizio Gasparini
- Neuroscience Discovery, Novartis Institutes for BioMedical Research, 4002 Basel, Switzerland
| | - Flora Tassone
- Department of Biochemistry and Molecular Medicine and Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, Sacramento, CA 95817, USA
| | - Randi Hagerman
- Department of Pediatric and Medical Investigation of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, School of Medicine, Sacramento, CA 95817, USA
| | - Baltazar Gomez-Mancilla
- Neuroscience Translational Medicine, Novartis Institutes for Biomedical Research, Novartis Pharma AG, 4056 Basel, Switzerland.,Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 0G4, Canada
| | - Claudia Bagni
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy.,Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland
| |
Collapse
|
67
|
Aran A, Eylon M, Harel M, Polianski L, Nemirovski A, Tepper S, Schnapp A, Cassuto H, Wattad N, Tam J. Lower circulating endocannabinoid levels in children with autism spectrum disorder. Mol Autism 2019; 10:2. [PMID: 30728928 PMCID: PMC6354384 DOI: 10.1186/s13229-019-0256-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 01/18/2019] [Indexed: 12/27/2022] Open
Abstract
Background The endocannabinoid system (ECS) is a major regulator of synaptic plasticity and neuromodulation. Alterations of the ECS have been demonstrated in several animal models of autism spectrum disorder (ASD). In some of these models, activating the ECS rescued the social deficits. Evidence for dysregulations of the ECS in human ASD are emerging, but comprehensive assessments and correlations with disease characteristics have not been reported yet. Methods Serum levels of the main endocannabinoids, N-arachidonoylethanolamine (AEA or anandamide) and 2-arachidonoylglycerol (2-AG), and their related endogenous compounds, arachidonic acid (AA), N-palmitoylethanolamine (PEA), and N-oleoylethanolamine (OEA), were analyzed by liquid chromatography/tandem mass spectrometry in 93 children with ASD (age = 13.1 ± 4.1, range 6–21; 79% boys) and 93 age- and gender-matched neurotypical children (age = 11.8 ± 4.3, range 5.5–21; 79% boys). Results were associated with gender and use of medications, and were correlated with age, BMI, and adaptive functioning of ASD participants as reflected by scores of Autism Diagnostic Observation Schedule (ADOS-2), Vineland Adaptive Behavior Scale-II (VABS-II), and Social Responsiveness Scale-II (SRS-2). Results Children with ASD had lower levels (pmol/mL, mean ± SEM) of AEA (0.722 ± 0.045 vs. 1.252 ± 0.072, P < 0.0001, effect size 0.91), OEA (17.3 ± 0.80 vs. 27.8 ± 1.44, P < 0.0001, effect size 0.94), and PEA (4.93 ± 0.32 vs. 7.15 ± 0.37, P < 0.0001, effect size 0.65), but not AA and 2-AG. Serum levels of AEA, OEA, and PEA were not significantly associated or correlated with age, gender, BMI, medications, and adaptive functioning of ASD participants. In children with ASD, but not in the control group, younger age and lower BMI tended to correlate with lower AEA levels. However, these correlations were not statistically significant after a correction for multiple comparisons. Conclusions We found lower serum levels of AEA, PEA, and OEA in children with ASD. Further studies are needed to determine whether circulating endocannabinoid levels can be used as stratification biomarkers that identify clinically significant subgroups within the autism spectrum and if they reflect lower endocannabinoid “tone” in the brain, as found in animal models of ASD. Electronic supplementary material The online version of this article (10.1186/s13229-019-0256-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Adi Aran
- 1Neuropediatric Unit, Shaare Zedek Medical Center, 12 Bayit Street, 91031 Jerusalem, Israel
| | - Maya Eylon
- 2Obesity and Metabolism Laboratory, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Moria Harel
- 1Neuropediatric Unit, Shaare Zedek Medical Center, 12 Bayit Street, 91031 Jerusalem, Israel
| | - Lola Polianski
- 1Neuropediatric Unit, Shaare Zedek Medical Center, 12 Bayit Street, 91031 Jerusalem, Israel
| | - Alina Nemirovski
- 2Obesity and Metabolism Laboratory, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sigal Tepper
- 3Department of Nutritional Sciences, Tel Hai Academic College, Upper Galilee, 1220800 Kiryat Shmona, Israel
| | - Aviad Schnapp
- 1Neuropediatric Unit, Shaare Zedek Medical Center, 12 Bayit Street, 91031 Jerusalem, Israel
| | - Hanoch Cassuto
- 1Neuropediatric Unit, Shaare Zedek Medical Center, 12 Bayit Street, 91031 Jerusalem, Israel
| | - Nadia Wattad
- 1Neuropediatric Unit, Shaare Zedek Medical Center, 12 Bayit Street, 91031 Jerusalem, Israel
| | - Joseph Tam
- 2Obesity and Metabolism Laboratory, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
68
|
Navarro-Romero A, Vázquez-Oliver A, Gomis-González M, Garzón-Montesinos C, Falcón-Moya R, Pastor A, Martín-García E, Pizarro N, Busquets-Garcia A, Revest JM, Piazza PV, Bosch F, Dierssen M, de la Torre R, Rodríguez-Moreno A, Maldonado R, Ozaita A. Cannabinoid type-1 receptor blockade restores neurological phenotypes in two models for Down syndrome. Neurobiol Dis 2019; 125:92-106. [PMID: 30685352 DOI: 10.1016/j.nbd.2019.01.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 12/24/2018] [Accepted: 01/23/2019] [Indexed: 12/31/2022] Open
Abstract
Intellectual disability is the most limiting hallmark of Down syndrome, for which there is no gold-standard clinical treatment yet. The endocannabinoid system is a widespread neuromodulatory system involved in multiple functions including learning and memory processes. Alterations of this system contribute to the pathogenesis of several neurological and neurodevelopmental disorders. However, the involvement of the endocannabinoid system in the pathogenesis of Down syndrome has not been explored before. We used the best-characterized preclinical model of Down syndrome, the segmentally trisomic Ts65Dn model. In male Ts65Dn mice, cannabinoid type-1 receptor (CB1R) expression was enhanced and its function increased in hippocampal excitatory terminals. Knockdown of CB1R in the hippocampus of male Ts65Dn mice restored hippocampal-dependent memory. Concomitant with this result, pharmacological inhibition of CB1R restored memory deficits, hippocampal synaptic plasticity and adult neurogenesis in the subgranular zone of the dentate gyrus. Notably, the blockade of CB1R also normalized hippocampal-dependent memory in female Ts65Dn mice. To further investigate the mechanisms involved, we used a second transgenic mouse model overexpressing a single gene candidate for Down syndrome cognitive phenotypes, the dual specificity tyrosine-phosphorylation-regulated kinase 1A (DYRK1A). CB1R pharmacological blockade similarly improved cognitive performance, synaptic plasticity and neurogenesis in transgenic male Dyrk1A mice. Our results identify CB1R as a novel druggable target potentially relevant for the improvement of cognitive deficits associated with Down syndrome.
Collapse
Affiliation(s)
- Alba Navarro-Romero
- Laboratory of Neuropharmacology-NeuroPhar, Department of Experimental and Health Sciences, University Pompeu Fabra, 08003 Barcelona, Spain
| | - Anna Vázquez-Oliver
- Laboratory of Neuropharmacology-NeuroPhar, Department of Experimental and Health Sciences, University Pompeu Fabra, 08003 Barcelona, Spain
| | - Maria Gomis-González
- Laboratory of Neuropharmacology-NeuroPhar, Department of Experimental and Health Sciences, University Pompeu Fabra, 08003 Barcelona, Spain
| | - Carlos Garzón-Montesinos
- Laboratory of Cellular Neuroscience and Plasticity, Department of Physiology, Anatomy and Cell Biology, University Pablo de Olavide, Ctra Utrera km. 1, 41013 Seville, Spain
| | - Rafael Falcón-Moya
- Laboratory of Cellular Neuroscience and Plasticity, Department of Physiology, Anatomy and Cell Biology, University Pablo de Olavide, Ctra Utrera km. 1, 41013 Seville, Spain
| | - Antoni Pastor
- Integrative Pharmacology and Systems Neuroscience Research Group, Hospital del Mar Medical Research Institute, 08003 Barcelona, Spain; CIBER Pathophysiology of Obesity and Nutrition, Institute of Health Carlos III, 28029 Madrid, Spain
| | - Elena Martín-García
- Laboratory of Neuropharmacology-NeuroPhar, Department of Experimental and Health Sciences, University Pompeu Fabra, 08003 Barcelona, Spain; Department of Psychobiology and Methodology of Health Sciences, Universitat Autònoma de Barcelona, Spain
| | - Nieves Pizarro
- Integrative Pharmacology and Systems Neuroscience Research Group, Hospital del Mar Medical Research Institute, 08003 Barcelona, Spain
| | - Arnau Busquets-Garcia
- Laboratory of Neuropharmacology-NeuroPhar, Department of Experimental and Health Sciences, University Pompeu Fabra, 08003 Barcelona, Spain
| | - Jean-Michel Revest
- INSERM U1215, Neurocentre Magendie, Physiopathology and Therapeutic Approaches of Stress-Related Diseases, 33077 Bordeaux, France
| | - Pier-Vincenzo Piazza
- INSERM U1215, Neurocentre Magendie, Physiopathology and Therapeutic Approaches of Stress-Related Diseases, 33077 Bordeaux, France
| | - Fátima Bosch
- Center of Animal Biotechnology and Gene Therapy (CBATEG), Spain; Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; CIBER Diabetes and Associated Metabolic Disorders (CIBERDEM), 08017 Madrid, Spain
| | - Mara Dierssen
- Cellular & Systems Neurobiology, Systems Biology Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain; CIBER Rare Disorders (CIBERER), Spain; Hospital del Mar Medical Research Institute, 08003 Barcelona, Spain
| | - Rafael de la Torre
- Integrative Pharmacology and Systems Neuroscience Research Group, Hospital del Mar Medical Research Institute, 08003 Barcelona, Spain; CIBER Pathophysiology of Obesity and Nutrition, Institute of Health Carlos III, 28029 Madrid, Spain
| | - Antonio Rodríguez-Moreno
- Laboratory of Cellular Neuroscience and Plasticity, Department of Physiology, Anatomy and Cell Biology, University Pablo de Olavide, Ctra Utrera km. 1, 41013 Seville, Spain
| | - Rafael Maldonado
- Laboratory of Neuropharmacology-NeuroPhar, Department of Experimental and Health Sciences, University Pompeu Fabra, 08003 Barcelona, Spain
| | - Andrés Ozaita
- Laboratory of Neuropharmacology-NeuroPhar, Department of Experimental and Health Sciences, University Pompeu Fabra, 08003 Barcelona, Spain.
| |
Collapse
|
69
|
Bar-Lev Schleider L, Mechoulam R, Saban N, Meiri G, Novack V. Real life Experience of Medical Cannabis Treatment in Autism: Analysis of Safety and Efficacy. Sci Rep 2019; 9:200. [PMID: 30655581 PMCID: PMC6336869 DOI: 10.1038/s41598-018-37570-y] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 11/23/2018] [Indexed: 12/23/2022] Open
Abstract
There has been a dramatic increase in the number of children diagnosed with autism spectrum disorders (ASD) worldwide. Recently anecdotal evidence of possible therapeutic effects of cannabis products has emerged. The aim of this study is to characterize the epidemiology of ASD patients receiving medical cannabis treatment and to describe its safety and efficacy. We analysed the data prospectively collected as part of the treatment program of 188 ASD patients treated with medical cannabis between 2015 and 2017. The treatment in majority of the patients was based on cannabis oil containing 30% CBD and 1.5% THC. Symptoms inventory, patient global assessment and side effects at 6 months were primary outcomes of interest and were assessed by structured questionnaires. After six months of treatment 82.4% of patients (155) were in active treatment and 60.0% (93) have been assessed; 28 patients (30.1%) reported a significant improvement, 50 (53.7%) moderate, 6 (6.4%) slight and 8 (8.6%) had no change in their condition. Twenty-three patients (25.2%) experienced at least one side effect; the most common was restlessness (6.6%). Cannabis in ASD patients appears to be well tolerated, safe and effective option to relieve symptoms associated with ASD.
Collapse
Affiliation(s)
- Lihi Bar-Lev Schleider
- Clinical Cannabis Research Institute, Soroka University Medical Center and Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel
- Research Department, Tikun Olam LTD, Tel Aviv-Yafo, Israel
| | - Raphael Mechoulam
- Institute for Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Naama Saban
- Research Department, Tikun Olam LTD, Tel Aviv-Yafo, Israel
| | - Gal Meiri
- Negev Autism Centre, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Soroka University Medical and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Victor Novack
- Clinical Cannabis Research Institute, Soroka University Medical Center and Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er-Sheva, Israel.
| |
Collapse
|
70
|
García-Rincón D, Díaz-Alonso J, Paraíso-Luna J, Ortega Z, Aguareles J, de Salas-Quiroga A, Jou C, de Prada I, Martínez-Cerdeño V, Aronica E, Guzmán M, Pérez-Jiménez MÁ, Galve-Roperh I. Contribution of Altered Endocannabinoid System to Overactive mTORC1 Signaling in Focal Cortical Dysplasia. Front Pharmacol 2019; 9:1508. [PMID: 30687088 PMCID: PMC6334222 DOI: 10.3389/fphar.2018.01508] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/10/2018] [Indexed: 02/05/2023] Open
Abstract
Alterations of the PI3K/Akt/mammalian target of rapamycin complex 1 (mTORC1) signaling pathway are causally involved in a subset of malformations of cortical development (MCDs) ranging from focal cortical dysplasia (FCD) to hemimegalencephaly and megalencephaly. These MCDs represent a frequent cause of refractory pediatric epilepsy. The endocannabinoid system -especially cannabinoid CB1 receptor- exerts a neurodevelopmental regulatory role at least in part via activation of mTORC1 signaling. Therefore, we sought to characterize the possible contribution of endocannabinoid system signaling to FCD. Confocal microscopy characterization of the CB1 receptor expression and mTORC1 activation was conducted in FCD Type II resection samples. FCD samples were subjected to single nucleotide polymorphism screening for endocannabinoid system elements, as well as CB1 receptor gene sequencing. Cannabinoid CB1 receptor levels were increased in FCD with overactive mTORC1 signaling. CB1 receptors were enriched in phospho-S6-positive cells including balloon cells (BCs) that co-express aberrant markers of undifferentiated cells and dysplastic neurons. Pharmacological regulation of CB1 receptors and the mTORC1 pathway was performed in fresh FCD-derived organotypic cultures. HU-210-evoked activation of CB1 receptors was unable to further activate mTORC1 signaling, whereas CB1 receptor blockade with rimonabant attenuated mTORC1 overactivation. Alterations of the endocannabinoid system may thus contribute to FCD pathological features, and blockade of cannabinoid signaling might be a new therapeutic intervention in FCD.
Collapse
Affiliation(s)
- Daniel García-Rincón
- Instituto Ramón y Cajal de Investigación Sanitaria, Department of Biochemistry and Molecular Biology and Instituto Universitario de Investigación Neuroquímica, Complutense University, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - Javier Díaz-Alonso
- Instituto Ramón y Cajal de Investigación Sanitaria, Department of Biochemistry and Molecular Biology and Instituto Universitario de Investigación Neuroquímica, Complutense University, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - Juan Paraíso-Luna
- Instituto Ramón y Cajal de Investigación Sanitaria, Department of Biochemistry and Molecular Biology and Instituto Universitario de Investigación Neuroquímica, Complutense University, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - Zaira Ortega
- Instituto Ramón y Cajal de Investigación Sanitaria, Department of Biochemistry and Molecular Biology and Instituto Universitario de Investigación Neuroquímica, Complutense University, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - José Aguareles
- Instituto Ramón y Cajal de Investigación Sanitaria, Department of Biochemistry and Molecular Biology and Instituto Universitario de Investigación Neuroquímica, Complutense University, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - Adán de Salas-Quiroga
- Instituto Ramón y Cajal de Investigación Sanitaria, Department of Biochemistry and Molecular Biology and Instituto Universitario de Investigación Neuroquímica, Complutense University, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - Cristina Jou
- Departamento de Anatomía Patológica, Hospital Sant Joan de Déu, Barcelona, Spain
| | | | - Verónica Martínez-Cerdeño
- Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children of Northern California and Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Eleonora Aronica
- Amsterdam UMC, Department of (Neuro)Pathology, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, Netherlands.,Stichting Epilepsie Instellingen Nederland, Heemstede, Netherlands
| | - Manuel Guzmán
- Instituto Ramón y Cajal de Investigación Sanitaria, Department of Biochemistry and Molecular Biology and Instituto Universitario de Investigación Neuroquímica, Complutense University, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | | | - Ismael Galve-Roperh
- Instituto Ramón y Cajal de Investigación Sanitaria, Department of Biochemistry and Molecular Biology and Instituto Universitario de Investigación Neuroquímica, Complutense University, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
| |
Collapse
|
71
|
Lee AW, Ventola P, Budimirovic D, Berry-Kravis E, Visootsak J. Clinical Development of Targeted Fragile X Syndrome Treatments: An Industry Perspective. Brain Sci 2018; 8:E214. [PMID: 30563047 PMCID: PMC6315847 DOI: 10.3390/brainsci8120214] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 11/28/2018] [Accepted: 11/30/2018] [Indexed: 01/03/2023] Open
Abstract
Fragile X syndrome (FXS) is the leading known cause of inherited intellectual disability and autism spectrum disorder. It is caused by a mutation of the fragile X mental retardation 1 (FMR1) gene, resulting in a deficit of fragile X mental retardation protein (FMRP). The clinical presentation of FXS is variable, and is typically associated with developmental delays, intellectual disability, a wide range of behavioral issues, and certain identifying physical features. Over the past 25 years, researchers have worked to understand the complex relationship between FMRP deficiency and the symptoms of FXS and, in the process, have identified several potential targeted therapeutics, some of which have been tested in clinical trials. Whereas most of the basic research to date has been led by experts at academic institutions, the pharmaceutical industry is becoming increasingly involved with not only the scientific community, but also with patient advocacy organizations, as more promising pharmacological agents are moving into the clinical stages of development. The objective of this review is to provide an industry perspective on the ongoing development of mechanism-based treatments for FXS, including identification of challenges and recommendations for future clinical trials.
Collapse
Affiliation(s)
- Anna W Lee
- Ovid Therapeutics Inc., New York, NY 10036, USA.
| | - Pamela Ventola
- Child Study Center, Yale University, New Haven, CT 06520, USA.
| | - Dejan Budimirovic
- Departments of Psychiatry and Behavioral Sciences, Kennedy Krieger Institute and Child Psychiatry, Johns Hopkins University, Baltimore, MD 21205, USA.
| | - Elizabeth Berry-Kravis
- Departments of Pediatrics, Neurological Sciences, Biochemistry, Rush University Medical Center, Chicago, IL 60612, USA.
| | | |
Collapse
|
72
|
Drozd HP, Karathanasis SF, Molosh AI, Lukkes JL, Clapp DW, Shekhar A. From bedside to bench and back: Translating ASD models. PROGRESS IN BRAIN RESEARCH 2018; 241:113-158. [PMID: 30447753 DOI: 10.1016/bs.pbr.2018.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Autism spectrum disorders (ASD) represent a heterogeneous group of disorders defined by deficits in social interaction/communication and restricted interests, behaviors, or activities. Models of ASD, developed based on clinical data and observations, are used in basic science, the "bench," to better understand the pathophysiology of ASD and provide therapeutic options for patients in the clinic, the "bedside." Translational medicine creates a bridge between the bench and bedside that allows for clinical and basic science discoveries to challenge one another to improve the opportunities to bring novel therapies to patients. From the clinical side, biomarker work is expanding our understanding of possible mechanisms of ASD through measures of behavior, genetics, imaging modalities, and serum markers. These biomarkers could help to subclassify patients with ASD in order to better target treatments to a more homogeneous groups of patients most likely to respond to a candidate therapy. In turn, basic science has been responding to developments in clinical evaluation by improving bench models to mechanistically and phenotypically recapitulate the ASD phenotypes observed in clinic. While genetic models are identifying novel therapeutics targets at the bench, the clinical efforts are making progress by defining better outcome measures that are most representative of meaningful patient responses. In this review, we discuss some of these challenges in translational research in ASD and strategies for the bench and bedside to bridge the gap to achieve better benefits to patients.
Collapse
Affiliation(s)
- Hayley P Drozd
- Program in Medical Neurobiology, Stark Neurosciences Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sotirios F Karathanasis
- Program in Medical Neurobiology, Stark Neurosciences Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Andrei I Molosh
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jodi L Lukkes
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
| | - D Wade Clapp
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Anantha Shekhar
- Program in Medical Neurobiology, Stark Neurosciences Institute, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States; Indiana Clinical and Translation Sciences Institute, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
73
|
Activation of autophagy rescues synaptic and cognitive deficits in fragile X mice. Proc Natl Acad Sci U S A 2018; 115:E9707-E9716. [PMID: 30242133 DOI: 10.1073/pnas.1808247115] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Fragile X syndrome (FXS) is the most frequent form of heritable intellectual disability and autism. Fragile X (Fmr1-KO) mice exhibit aberrant dendritic spine structure, synaptic plasticity, and cognition. Autophagy is a catabolic process of programmed degradation and recycling of proteins and cellular components via the lysosomal pathway. However, a role for autophagy in the pathophysiology of FXS is, as yet, unclear. Here we show that autophagic flux, a functional readout of autophagy, and biochemical markers of autophagy are down-regulated in hippocampal neurons of fragile X mice. We further show that enhanced activity of mammalian target of rapamycin complex 1 (mTORC1) and translocation of Raptor, a defining component of mTORC1, to the lysosome are causally related to reduced autophagy. Activation of autophagy by delivery of shRNA to Raptor directly into the CA1 of living mice via the lentivirus expression system largely corrects aberrant spine structure, synaptic plasticity, and cognition in fragile X mice. Postsynaptic density protein (PSD-95) and activity-regulated cytoskeletal-associated protein (Arc/Arg3.1), proteins implicated in spine structure and synaptic plasticity, respectively, are elevated in neurons lacking fragile X mental retardation protein. Activation of autophagy corrects PSD-95 and Arc abundance, identifying a potential mechanism by which impaired autophagy is causally related to the fragile X phenotype and revealing a previously unappreciated role for autophagy in the synaptic and cognitive deficits associated with fragile X syndrome.
Collapse
|
74
|
Ghilan M, Bettio LEB, Noonan A, Brocardo PS, Gil-Mohapel J, Christie BR. Impaired spatial processing in a mouse model of fragile X syndrome. Behav Brain Res 2018; 350:72-79. [PMID: 29778627 DOI: 10.1016/j.bbr.2018.05.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 05/05/2018] [Accepted: 05/14/2018] [Indexed: 01/07/2023]
Abstract
Fragile X syndrome (FXS) is the most common form of inherited intellectual impairment. The Fmr1-/y mouse model has been previously shown to have deficits in context discrimination tasks but not in the elevated plus-maze. To further characterize this FXS mouse model and determine whether hippocampal-mediated behaviours are affected in these mice, dentate gyrus (DG)-dependent spatial processing and Cornu ammonis 1 (CA1)-dependent temporal order discrimination tasks were evaluated. In agreement with previous findings of long-term potentiation deficits in the DG of this transgenic model of FXS, the results reported here demonstrate that Fmr1-/y mice perform poorly in the DG-dependent metric change spatial processing task. However, Fmr1-/y mice did not present deficits in the CA1-dependent temporal order discrimination task, and were able to remember the order in which objects were presented to them to the same extent as their wild-type littermate controls. These data suggest that the previously reported subregional-specific differences in hippocampal synaptic plasticity observed in the Fmr1-/y mouse model may manifest as selective behavioural deficits in hippocampal-dependent tasks.
Collapse
Affiliation(s)
- Mohamed Ghilan
- Graduate Program in Neuroscience, University of Victoria, Victoria, BC, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Luis E B Bettio
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Athena Noonan
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | | | - Joana Gil-Mohapel
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Island Medical Program, University of British Columbia, Victoria, BC, Canada.
| | - Brian R Christie
- Graduate Program in Neuroscience, University of Victoria, Victoria, BC, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Island Medical Program, University of British Columbia, Victoria, BC, Canada
| |
Collapse
|
75
|
Vigli D, Cosentino L, Raggi C, Laviola G, Woolley-Roberts M, De Filippis B. Chronic treatment with the phytocannabinoid Cannabidivarin (CBDV) rescues behavioural alterations and brain atrophy in a mouse model of Rett syndrome. Neuropharmacology 2018; 140:121-129. [DOI: 10.1016/j.neuropharm.2018.07.029] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 05/30/2018] [Accepted: 07/24/2018] [Indexed: 12/26/2022]
|
76
|
Hosie S, Malone DT, Liu S, Glass M, Adlard PA, Hannan AJ, Hill-Yardin EL. Altered Amygdala Excitation and CB1 Receptor Modulation of Aggressive Behavior in the Neuroligin-3 R451C Mouse Model of Autism. Front Cell Neurosci 2018; 12:234. [PMID: 30123111 PMCID: PMC6085410 DOI: 10.3389/fncel.2018.00234] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 07/16/2018] [Indexed: 12/28/2022] Open
Abstract
Understanding neuronal mechanisms underlying aggression in patients with autism spectrum disorder (ASD) could lead to better treatments and prognosis. The Neuroligin-3 (NL3)R451C mouse model of ASD has a heightened aggressive phenotype, however the biological mechanisms underlying this behavior are unknown. It is well established that NL3R451C mice have imbalanced excitatory and inhibitory synaptic activity in the hippocampus and somatosensory cortex. The amygdala plays a role in modulating aggressive behavior, however potential changes in synaptic activity in this region have not previously been assessed in this model. We investigated whether aggressive behavior is robustly present in mice expressing the R451C mutation, following back-crossing onto a congenic background strain. Endocannabinoids influence social interaction and aggressive behavior, therefore we also studied the effects of cannabinoid receptor 1 (CB1) agonist on NL3R451C mice. We report that NL3R451C mice have increased amplitude of miniature excitatory postsynaptic currents (EPSCs) with a concomitant decrease in the amplitude of inhibitory postsynaptic currents (IPSCs) in the basolateral amygdala. Importantly, we demonstrated that NL3R451C mice bred on a C57Bl/6 background strain exhibit an aggressive phenotype. Following non-sedating doses (0.3 and 1.0 mg/kg) of the CB1 receptor agonist WIN55,212-2 (WIN), we observed a significant reduction in aggressive behavior in NL3R451C mice. These findings demonstrate altered synaptic activity in the basolateral amygdala and suggest that the NL3R451C mouse model is a useful preclinical tool to understand the role of CB1 receptor function in aggressive behavior.
Collapse
Affiliation(s)
- Suzanne Hosie
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Daniel T Malone
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Stephanie Liu
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Michelle Glass
- Department of Pharmacology, University of Auckland, Auckland, New Zealand
| | - Paul Anthony Adlard
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Anthony John Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia.,Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC, Australia
| | - Elisa L Hill-Yardin
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia.,Department of Physiology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
77
|
Reducing histone acetylation rescues cognitive deficits in a mouse model of Fragile X syndrome. Nat Commun 2018; 9:2494. [PMID: 29950602 PMCID: PMC6021376 DOI: 10.1038/s41467-018-04869-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 05/23/2018] [Indexed: 12/19/2022] Open
Abstract
Fragile X syndrome (FXS) is the most prevalent inherited intellectual disability, resulting from a loss of fragile X mental retardation protein (FMRP). Patients with FXS suffer lifelong cognitive disabilities, but the function of FMRP in the adult brain and the mechanism underlying age-related cognitive decline in FXS is not fully understood. Here, we report that a loss of FMRP results in increased protein synthesis of histone acetyltransferase EP300 and ubiquitination-mediated degradation of histone deacetylase HDAC1 in adult hippocampal neural stem cells (NSCs). Consequently, FMRP-deficient NSCs exhibit elevated histone acetylation and age-related NSC depletion, leading to cognitive impairment in mature adult mice. Reducing histone acetylation rescues both neurogenesis and cognitive deficits in mature adult FMRP-deficient mice. Our work reveals a role for FMRP and histone acetylation in cognition and presents a potential novel therapeutic strategy for treating adult FXS patients. Loss of fragile X mental retardation protein (FMRP) leads to fragile X syndrome, associated with cognitive dysfunction. Here the authors show that mice lacking FMRP show reduced hippocampal neurogenesis and cognitive deficits, which can be rescued by reducing histone acetylation.
Collapse
|
78
|
Robin LM, Oliveira da Cruz JF, Langlais VC, Martin-Fernandez M, Metna-Laurent M, Busquets-Garcia A, Bellocchio L, Soria-Gomez E, Papouin T, Varilh M, Sherwood MW, Belluomo I, Balcells G, Matias I, Bosier B, Drago F, Van Eeckhaut A, Smolders I, Georges F, Araque A, Panatier A, Oliet SHR, Marsicano G. Astroglial CB 1 Receptors Determine Synaptic D-Serine Availability to Enable Recognition Memory. Neuron 2018; 98:935-944.e5. [PMID: 29779943 DOI: 10.1016/j.neuron.2018.04.034] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 03/20/2018] [Accepted: 04/24/2018] [Indexed: 12/22/2022]
Abstract
Bidirectional communication between neurons and astrocytes shapes synaptic plasticity and behavior. D-serine is a necessary co-agonist of synaptic N-methyl-D-aspartate receptors (NMDARs), but the physiological factors regulating its impact on memory processes are scantly known. We show that astroglial CB1 receptors are key determinants of object recognition memory by determining the availability of D-serine at hippocampal synapses. Mutant mice lacking CB1 receptors from astroglial cells (GFAP-CB1-KO) displayed impaired object recognition memory and decreased in vivo and in vitro long-term potentiation (LTP) at CA3-CA1 hippocampal synapses. Activation of CB1 receptors increased intracellular astroglial Ca2+ levels and extracellular levels of D-serine in hippocampal slices. Accordingly, GFAP-CB1-KO displayed lower occupancy of the co-agonist binding site of synaptic hippocampal NMDARs. Finally, elevation of D-serine levels fully rescued LTP and memory impairments of GFAP-CB1-KO mice. These data reveal a novel mechanism of in vivo astroglial control of memory and synaptic plasticity via the D-serine-dependent control of NMDARs.
Collapse
Affiliation(s)
- Laurie M Robin
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | - José F Oliveira da Cruz
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France; Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95123 Catania, Italy
| | - Valentin C Langlais
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | | | - Mathilde Metna-Laurent
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France; Aelis Farma, 33077 Bordeaux, France
| | - Arnau Busquets-Garcia
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | - Luigi Bellocchio
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | - Edgar Soria-Gomez
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France; Department of Neurosciences, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Thomas Papouin
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | - Marjorie Varilh
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | - Mark W Sherwood
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | - Ilaria Belluomo
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | - Georgina Balcells
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | - Isabelle Matias
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | - Barbara Bosier
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, 95123 Catania, Italy
| | - Ann Van Eeckhaut
- Vrije Universiteit Brussel, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information (FASC), Research group Experimental Pharmacology, Center for Neurosciences (C4N), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Ilse Smolders
- Vrije Universiteit Brussel, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information (FASC), Research group Experimental Pharmacology, Center for Neurosciences (C4N), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Francois Georges
- University of Bordeaux, 33077 Bordeaux, France; Centre National de la Recherche Scientifique, Neurodegenerative Diseases Institute, UMR 5293, 33076 Bordeaux, France
| | - Alfonso Araque
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Aude Panatier
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | - Stéphane H R Oliet
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France
| | - Giovanni Marsicano
- INSERM U1215, NeuroCentre Magendie, 33077 Bordeaux, France; University of Bordeaux, 33077 Bordeaux, France.
| |
Collapse
|
79
|
Perche O, Felgerolle C, Ardourel M, Bazinet A, Pâris A, Rossignol R, Meyer-Dilhet G, Mausset-Bonnefont AL, Hébert B, Laurenceau D, Montécot-Dubourg C, Menuet A, Bizot JC, Pichon J, Ranchon-Cole I, Briault S. Early Retinal Defects in Fmr1-/y Mice: Toward a Critical Role of Visual Dys-Sensitivity in the Fragile X Syndrome Phenotype? Front Cell Neurosci 2018; 12:96. [PMID: 29681800 PMCID: PMC5897671 DOI: 10.3389/fncel.2018.00096] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 03/22/2018] [Indexed: 12/29/2022] Open
Abstract
Fragile X Syndrome (FXS) is caused by a deficiency in Fragile X Mental Retardation Protein (FMRP) leading to global sensorial abnormalities, among which visual defects represent a critical part. These visual defects are associated with cerebral neuron immaturity especially in the primary visual cortex. However, we recently demonstrated that retinas of adult Fmr1−/y mice, the FXS murine model, present molecular, cellular and functional alterations. However, no data are currently available on the evolution pattern of such defects. As retinal stimulation through Eye Opening (EO) is a crucial signal for the cerebral visual system maturation, we questioned the precocity of molecular and functional retinal phenotype. To answer this question, we studied the retinal molecular phenotype of Fmr1−/y mice before EO until adult age and the consequences of the retinal loss of Fmrp on retinal function in young and adult mice. We showed that retinal molecular defects are present before EO and remain stable at adult age, leading to electrophysiological impairments without any underlying structural changes. We underlined that loss of Fmrp leads to a wide range of defects in the retina, settled even before EO. Our work demonstrates a critical role of the sensorial dysfunction in the Fmr1−/y mice overall phenotype, and provides evidence that altered peripheral perception is a component of the sensory processing defect in FXS conditions.
Collapse
Affiliation(s)
- Olivier Perche
- Genetic Department, Centre Hospitalier Régional d'Orléans, Orléans, France.,UMR7355, Immunologie et Neurogénétique Expérimentales et Moléculaires (INEM), Centre National de la Recherche Scientifique, Orléans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| | - Chloé Felgerolle
- UMR7355, Immunologie et Neurogénétique Expérimentales et Moléculaires (INEM), Centre National de la Recherche Scientifique, Orléans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| | - Maryvonne Ardourel
- UMR7355, Immunologie et Neurogénétique Expérimentales et Moléculaires (INEM), Centre National de la Recherche Scientifique, Orléans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| | - Audrey Bazinet
- UMR7355, Immunologie et Neurogénétique Expérimentales et Moléculaires (INEM), Centre National de la Recherche Scientifique, Orléans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| | - Arnaud Pâris
- UMR7355, Immunologie et Neurogénétique Expérimentales et Moléculaires (INEM), Centre National de la Recherche Scientifique, Orléans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| | - Rafaëlle Rossignol
- UMR7355, Immunologie et Neurogénétique Expérimentales et Moléculaires (INEM), Centre National de la Recherche Scientifique, Orléans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| | - Géraldine Meyer-Dilhet
- UMR7355, Immunologie et Neurogénétique Expérimentales et Moléculaires (INEM), Centre National de la Recherche Scientifique, Orléans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| | | | - Betty Hébert
- UMR7355, Immunologie et Neurogénétique Expérimentales et Moléculaires (INEM), Centre National de la Recherche Scientifique, Orléans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| | - David Laurenceau
- Genetic Department, Centre Hospitalier Régional d'Orléans, Orléans, France
| | - Céline Montécot-Dubourg
- UMR7355, Immunologie et Neurogénétique Expérimentales et Moléculaires (INEM), Centre National de la Recherche Scientifique, Orléans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| | - Arnaud Menuet
- UMR7355, Immunologie et Neurogénétique Expérimentales et Moléculaires (INEM), Centre National de la Recherche Scientifique, Orléans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| | | | - Jacques Pichon
- UMR7355, Immunologie et Neurogénétique Expérimentales et Moléculaires (INEM), Centre National de la Recherche Scientifique, Orléans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| | - Isabelle Ranchon-Cole
- Laboratory of Sensorial Biophysical, INSERM UMR1107 Equipe Biophysique Neurosensorielle, University of Clermont 1, Clermont-Ferrand, France
| | - Sylvain Briault
- Genetic Department, Centre Hospitalier Régional d'Orléans, Orléans, France.,UMR7355, Immunologie et Neurogénétique Expérimentales et Moléculaires (INEM), Centre National de la Recherche Scientifique, Orléans, France.,Experimental and Molecular Immunology and Neurogenetics, University of Orléans, Orléans, France
| |
Collapse
|
80
|
Saré RM, Song A, Loutaev I, Cook A, Maita I, Lemons A, Sheeler C, Smith CB. Negative Effects of Chronic Rapamycin Treatment on Behavior in a Mouse Model of Fragile X Syndrome. Front Mol Neurosci 2018; 10:452. [PMID: 29375310 PMCID: PMC5770365 DOI: 10.3389/fnmol.2017.00452] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 12/22/2017] [Indexed: 01/08/2023] Open
Abstract
Fragile X syndrome (FXS), the most common form of inherited intellectual disability, is also highly associated with autism spectrum disorders (ASD). It is caused by expansion of a CGG repeat sequence on the X chromosome resulting in silencing of the FMR1 gene. This is modeled in the mouse by deletion of Fmr1 (Fmr1 KO). Fmr1 KO mice recapitulate many of the behavioral features of the disorder including seizure susceptibility, hyperactivity, impaired social behavior, sleep problems, and learning and memory deficits. The mammalian target of rapamycin pathway (mTORC1) is upregulated in Fmr1 KO mice and is thought to be important for the pathogenesis of this disorder. We treated Fmr1 KO mice chronically with an mTORC1 inhibitor, rapamycin, to determine if rapamycin treatment could reverse behavioral phenotypes. We performed open field, zero maze, social behavior, sleep, passive avoidance, and audiogenic seizure testing. We found that pS6 was upregulated in Fmr1 KO mice and normalized by rapamycin treatment, but, except for an anxiogenic effect, it did not reverse any of the behavioral phenotypes examined. In fact, rapamycin treatment had an adverse effect on sleep and social behavior in both control and Fmr1 KO mice. These results suggest that targeting the mTOR pathway in FXS is not a good treatment strategy and that other pathways should be considered.
Collapse
Affiliation(s)
- Rachel M Saré
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
| | - Alex Song
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
| | - Inna Loutaev
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
| | - Anna Cook
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
| | - Isabella Maita
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
| | - Abigail Lemons
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
| | - Carrie Sheeler
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
| | - Carolyn B Smith
- Section on Neuroadaptation and Protein Metabolism, National Institute of Mental Health, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
| |
Collapse
|
81
|
Busquets-Garcia A, Bains J, Marsicano G. CB 1 Receptor Signaling in the Brain: Extracting Specificity from Ubiquity. Neuropsychopharmacology 2018; 43:4-20. [PMID: 28862250 PMCID: PMC5719111 DOI: 10.1038/npp.2017.206] [Citation(s) in RCA: 193] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/22/2017] [Accepted: 08/14/2017] [Indexed: 02/07/2023]
Abstract
Endocannabinoids (eCBs) are amongst the most ubiquitous signaling molecules in the nervous system. Over the past few decades, observations based on a large volume of work, first examining the pharmacological effects of exogenous cannabinoids, and then the physiological functions of eCBs, have directly challenged long-held and dogmatic views about communication, plasticity and behavior in the central nervous system (CNS). The eCBs and their cognate cannabinoid receptors exhibit a number of unique properties that distinguish them from the widely studied classical amino-acid transmitters, neuropeptides, and catecholamines. Although we now have a loose set of mechanistic rules based on experimental findings, new studies continue to reveal that our understanding of the eCB system (ECS) is continuously evolving and challenging long-held conventions. Here we will briefly summarize findings on the current canonical view of the 'ECS' and will address novel aspects that reveal how a nearly ubiquitous system can determine highly specific functions in the brain. In particular, we will focus on findings that push for an expansion of our ideas around long-held beliefs about eCB signaling that, while clearly true, may be contributing to an oversimplified perspective on how cannabinoid signaling at the microscopic level impacts behavior at the macroscopic level.
Collapse
Affiliation(s)
- Arnau Busquets-Garcia
- INSERM U1215, NeuroCentre Magendie, Team ‘Endocannabinoids and Neuroadaptation’, Bordeaux, France
- Université de Bordeaux, Bordeaux, France
| | - Jaideep Bains
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Giovanni Marsicano
- INSERM U1215, NeuroCentre Magendie, Team ‘Endocannabinoids and Neuroadaptation’, Bordeaux, France
- Université de Bordeaux, Bordeaux, France
| |
Collapse
|
82
|
Drug development for neurodevelopmental disorders: lessons learned from fragile X syndrome. Nat Rev Drug Discov 2017; 17:280-299. [PMID: 29217836 DOI: 10.1038/nrd.2017.221] [Citation(s) in RCA: 219] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Neurodevelopmental disorders such as fragile X syndrome (FXS) result in lifelong cognitive and behavioural deficits and represent a major public health burden. FXS is the most frequent monogenic form of intellectual disability and autism, and the underlying pathophysiology linked to its causal gene, FMR1, has been the focus of intense research. Key alterations in synaptic function thought to underlie this neurodevelopmental disorder have been characterized and rescued in animal models of FXS using genetic and pharmacological approaches. These robust preclinical findings have led to the implementation of the most comprehensive drug development programme undertaken thus far for a genetically defined neurodevelopmental disorder, including phase IIb trials of metabotropic glutamate receptor 5 (mGluR5) antagonists and a phase III trial of a GABAB receptor agonist. However, none of the trials has been able to unambiguously demonstrate efficacy, and they have also highlighted the extent of the knowledge gaps in drug development for FXS and other neurodevelopmental disorders. In this Review, we examine potential issues in the previous studies and future directions for preclinical and clinical trials. FXS is at the forefront of efforts to develop drugs for neurodevelopmental disorders, and lessons learned in the process will also be important for such disorders.
Collapse
|
83
|
Li Y, Stockton ME, Bhuiyan I, Eisinger BE, Gao Y, Miller JL, Bhattacharyya A, Zhao X. MDM2 inhibition rescues neurogenic and cognitive deficits in a mouse model of fragile X syndrome. Sci Transl Med 2017; 8:336ra61. [PMID: 27122614 DOI: 10.1126/scitranslmed.aad9370] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 04/08/2016] [Indexed: 12/15/2022]
Abstract
Fragile X syndrome, the most common form of inherited intellectual disability, is caused by loss of the fragile X mental retardation protein (FMRP). However, the mechanism remains unclear, and effective treatment is lacking. We show that loss of FMRP leads to activation of adult mouse neural stem cells (NSCs) and a subsequent reduction in the production of neurons. We identified the ubiquitin ligase mouse double minute 2 homolog (MDM2) as a target of FMRP. FMRP regulates Mdm2 mRNA stability, and loss of FMRP resulted in elevated MDM2 mRNA and protein. Further, we found that increased MDM2 expression led to reduced P53 expression in adult mouse NSCs, leading to alterations in NSC proliferation and differentiation. Treatment with Nutlin-3, a small molecule undergoing clinical trials for treating cancer, specifically inhibited the interaction of MDM2 with P53, and rescued neurogenic and cognitive deficits in FMRP-deficient mice. Our data reveal a potential regulatory role for FMRP in the balance between adult NSC activation and quiescence, and identify a potential new treatment for fragile X syndrome.
Collapse
Affiliation(s)
- Yue Li
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA. Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA
| | | | - Ismat Bhuiyan
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Brian E Eisinger
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA. Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Yu Gao
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA. Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jessica L Miller
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | | | - Xinyu Zhao
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA. Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
84
|
Castagnola S, Bardoni B, Maurin T. The Search for an Effective Therapy to Treat Fragile X Syndrome: Dream or Reality? Front Synaptic Neurosci 2017; 9:15. [PMID: 29163124 PMCID: PMC5681520 DOI: 10.3389/fnsyn.2017.00015] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/16/2017] [Indexed: 12/22/2022] Open
Abstract
Fragile X Syndrome (FXS) is the most common form of intellectual disability and a primary cause of autism. It originates from the lack of the Fragile X Mental Retardation Protein (FMRP), which is an RNA-binding protein encoded by the Fragile X Mental Retardation Gene 1 (FMR1) gene. Multiple roles have been attributed to this protein, ranging from RNA transport (from the nucleus to the cytoplasm, but also along neurites) to translational control of mRNAs. Over the last 20 years many studies have found a large number of FMRP mRNA targets, but it is still not clear which are those playing a critical role in the etiology of FXS. So far, no therapy for FXS has been found, making the quest for novel targets of considerable importance. Several pharmacological approaches have been attempted, but, despite some promising preclinical results, no strategy gave successful outcomes, due either to the induction of major side effects or to the lack of improvement of the phenotypes. However, these studies suggested that, in order to measure the effectiveness of a specific treatment, trials should be redesigned and new endpoints defined in FXS patients. Nevertheless, the search for new therapeutic targets for FXS is very active. In this context, the advances in animal modeling, coupled with better understanding of neurobiology and physiopathology of FXS, are of crucial importance in developing new selected treatments. Here, we discuss the pathways that were recently linked to the physiopathology of FXS (mGluR, GABAR, insulin, Insulin-like Growth Factor 1 (IGF-1), MPP-9, serotonin, oxytocin and endocannabinoid signaling) and that suggest new approaches to find an effective therapy for this disorder. Our goal with this review article is to summarize some recent relevant findings on FXS treatment strategies in order to have a clearer view of the different pathways analyzed to date emphasizing those shared with other synaptic disorders.
Collapse
Affiliation(s)
- Sara Castagnola
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Valbonne, France
| | - Barbara Bardoni
- Université Côte d'Azur, INSERM, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Valbonne, France
| | - Thomas Maurin
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Valbonne, France
| |
Collapse
|
85
|
Cissé M, Duplan E, Lorivel T, Dunys J, Bauer C, Meckler X, Gerakis Y, Lauritzen I, Checler F. The transcription factor XBP1s restores hippocampal synaptic plasticity and memory by control of the Kalirin-7 pathway in Alzheimer model. Mol Psychiatry 2017; 22:1562-1575. [PMID: 27646263 PMCID: PMC5658671 DOI: 10.1038/mp.2016.152] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 06/14/2016] [Accepted: 07/18/2016] [Indexed: 12/18/2022]
Abstract
Neuronal network dysfunction and cognitive decline constitute the most prominent features of Alzheimer's disease (AD), although mechanisms causing such impairments are yet to be determined. Here we report that virus-mediated delivery of the active spliced transcription factor X-Box binding protein 1s (XBP1s) in the hippocampus rescued spine density, synaptic plasticity and memory function in a mouse model of AD. XBP1s transcriptionally activated Kalirin-7 (Kal7), a protein that controls synaptic plasticity. In addition, we found reduced levels of Kal7 in primary neurons exposed to Aβ oligomers, transgenic mouse models and human AD brains. Short hairpin RNA-mediated knockdown of Kal7 altered synaptic plasticity and memory formation in naive mice. Further, reduction of endogenous Kal7 compromised the beneficial effects of XBP1s in Alzheimer's model. Hence, our findings reveal that XBP1s is neuroprotective through a mechanism that engages Kal7 pathway with therapeutic implications in AD pathology.
Collapse
Affiliation(s)
- M Cissé
- Université de Nice-Sophia-Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS-UMR7275, Sophia-Antipolis, Valbonne, France,Université de Nice-Sophia-Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS-UMR7275, NEUROLOGY, IPMC/CNRS, 660 Route des Lucioles, 06560 Valbonne, France. E-mail:
| | - E Duplan
- Université de Nice-Sophia-Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS-UMR7275, Sophia-Antipolis, Valbonne, France
| | - T Lorivel
- Université de Nice-Sophia-Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS-UMR7275, Sophia-Antipolis, Valbonne, France
| | - J Dunys
- Université de Nice-Sophia-Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS-UMR7275, Sophia-Antipolis, Valbonne, France
| | - C Bauer
- Université de Nice-Sophia-Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS-UMR7275, Sophia-Antipolis, Valbonne, France
| | - X Meckler
- Université de Nice-Sophia-Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS-UMR7275, Sophia-Antipolis, Valbonne, France
| | - Y Gerakis
- Université de Nice-Sophia-Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS-UMR7275, Sophia-Antipolis, Valbonne, France
| | - I Lauritzen
- Université de Nice-Sophia-Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS-UMR7275, Sophia-Antipolis, Valbonne, France
| | - F Checler
- Université de Nice-Sophia-Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS-UMR7275, Sophia-Antipolis, Valbonne, France
| |
Collapse
|
86
|
Altered surface mGluR5 dynamics provoke synaptic NMDAR dysfunction and cognitive defects in Fmr1 knockout mice. Nat Commun 2017; 8:1103. [PMID: 29062097 PMCID: PMC5653653 DOI: 10.1038/s41467-017-01191-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/24/2017] [Indexed: 12/20/2022] Open
Abstract
Metabotropic glutamate receptor subtype 5 (mGluR5) is crucially implicated in the pathophysiology of Fragile X Syndrome (FXS); however, its dysfunction at the sub-cellular level, and related synaptic and cognitive phenotypes are unexplored. Here, we probed the consequences of mGluR5/Homer scaffold disruption for mGluR5 cell-surface mobility, synaptic N-methyl-D-aspartate receptor (NMDAR) function, and behavioral phenotypes in the second-generation Fmr1 knockout (KO) mouse. Using single-molecule tracking, we found that mGluR5 was significantly more mobile at synapses in hippocampal Fmr1 KO neurons, causing an increased synaptic surface co-clustering of mGluR5 and NMDAR. This correlated with a reduced amplitude of synaptic NMDAR currents, a lack of their mGluR5-activated long-term depression, and NMDAR/hippocampus dependent cognitive deficits. These synaptic and behavioral phenomena were reversed by knocking down Homer1a in Fmr1 KO mice. Our study provides a mechanistic link between changes of mGluR5 dynamics and pathological phenotypes of FXS, unveiling novel targets for mGluR5-based therapeutics. Dysfunction of mGluR5 has been implicated in Fragile X syndrome. Here, using a single-molecule tracking technique, the authors found an increased lateral mobility of mGluR5 at the synaptic site in Fmr1 KO hippocampal neurons, leading to abnormal NMDAR-mediated synaptic plasticity and cognitive deficits.
Collapse
|
87
|
Ceolin L, Bouquier N, Vitre-Boubaker J, Rialle S, Severac D, Valjent E, Perroy J, Puighermanal E. Cell Type-Specific mRNA Dysregulation in Hippocampal CA1 Pyramidal Neurons of the Fragile X Syndrome Mouse Model. Front Mol Neurosci 2017; 10:340. [PMID: 29104533 PMCID: PMC5655025 DOI: 10.3389/fnmol.2017.00340] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 10/06/2017] [Indexed: 12/19/2022] Open
Abstract
Fragile X syndrome (FXS) is a genetic disorder due to the silencing of the Fmr1 gene, causing intellectual disability, seizures, hyperactivity, and social anxiety. All these symptoms result from the loss of expression of the RNA binding protein fragile X mental retardation protein (FMRP), which alters the neurodevelopmental program to abnormal wiring of specific circuits. Aberrant mRNAs translation associated with the loss of Fmr1 product is widely suspected to be in part the cause of FXS. However, precise gene expression changes involved in this disorder have yet to be defined. The objective of this study was to identify the set of mistranslated mRNAs that could contribute to neurological deficits in FXS. We used the RiboTag approach and RNA sequencing to provide an exhaustive listing of genes whose mRNAs are differentially translated in hippocampal CA1 pyramidal neurons as the integrative result of FMRP loss and subsequent neurodevelopmental adaptations. Among genes differentially regulated between adult WT and Fmr1-/y mice, we found enrichment in FMRP-binders but also a majority of non-FMRP-binders. Interestingly, both up- and down-regulation of specific gene expression is relevant to fully understand the molecular deficiencies triggering FXS. More importantly, functional genomic analysis highlighted the importance of genes involved in neuronal connectivity. Among them, we show that Klk8 altered expression participates in the abnormal hippocampal dendritic spine maturation observed in a mouse model of FXS.
Collapse
Affiliation(s)
- Laura Ceolin
- IGF, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | | | | | | | - Dany Severac
- Montpellier GenomiX c/o IGF, Montpellier, France
| | | | - Julie Perroy
- IGF, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | | |
Collapse
|
88
|
Pelkey KA, Chittajallu R, Craig MT, Tricoire L, Wester JC, McBain CJ. Hippocampal GABAergic Inhibitory Interneurons. Physiol Rev 2017; 97:1619-1747. [PMID: 28954853 DOI: 10.1152/physrev.00007.2017] [Citation(s) in RCA: 495] [Impact Index Per Article: 70.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 05/16/2017] [Accepted: 05/26/2017] [Indexed: 12/11/2022] Open
Abstract
In the hippocampus GABAergic local circuit inhibitory interneurons represent only ~10-15% of the total neuronal population; however, their remarkable anatomical and physiological diversity allows them to regulate virtually all aspects of cellular and circuit function. Here we provide an overview of the current state of the field of interneuron research, focusing largely on the hippocampus. We discuss recent advances related to the various cell types, including their development and maturation, expression of subtype-specific voltage- and ligand-gated channels, and their roles in network oscillations. We also discuss recent technological advances and approaches that have permitted high-resolution, subtype-specific examination of their roles in numerous neural circuit disorders and the emerging therapeutic strategies to ameliorate such pathophysiological conditions. The ultimate goal of this review is not only to provide a touchstone for the current state of the field, but to help pave the way for future research by highlighting where gaps in our knowledge exist and how a complete appreciation of their roles will aid in future therapeutic strategies.
Collapse
Affiliation(s)
- Kenneth A Pelkey
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Ramesh Chittajallu
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Michael T Craig
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Ludovic Tricoire
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Jason C Wester
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| | - Chris J McBain
- Porter Neuroscience Center, Eunice Kennedy-Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratories, University of Exeter, Exeter, United Kingdom; and Sorbonne Universités, UPMC University of Paris, INSERM, CNRS, Neurosciences Paris Seine-Institut de Biologie Paris Seine, Paris, France
| |
Collapse
|
89
|
Abstract
Fragile X syndrome (FXS) is the leading inherited form of intellectual disability and autism spectrum disorder, and patients can present with severe behavioural alterations, including hyperactivity, impulsivity and anxiety, in addition to poor language development and seizures. FXS is a trinucleotide repeat disorder, in which >200 repeats of the CGG motif in FMR1 leads to silencing of the gene and the consequent loss of its product, fragile X mental retardation 1 protein (FMRP). FMRP has a central role in gene expression and regulates the translation of potentially hundreds of mRNAs, many of which are involved in the development and maintenance of neuronal synaptic connections. Indeed, disturbances in neuroplasticity is a key finding in FXS animal models, and an imbalance in inhibitory and excitatory neuronal circuits is believed to underlie many of the clinical manifestations of this disorder. Our knowledge of the proteins that are regulated by FMRP is rapidly growing, and this has led to the identification of multiple targets for therapeutic intervention, some of which have already moved into clinical trials or clinical practice.
Collapse
|
90
|
Piazza PV, Cota D, Marsicano G. The CB1 Receptor as the Cornerstone of Exostasis. Neuron 2017; 93:1252-1274. [PMID: 28334603 DOI: 10.1016/j.neuron.2017.02.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 01/30/2017] [Accepted: 01/31/2017] [Indexed: 01/07/2023]
Abstract
The type-1 cannabinoid receptor (CB1) is the main effector of the endocannabinoid system (ECS), which is involved in most brain and body functions. In this Perspective, we provide evidence indicating that CB1 receptor functions are key determinants of bodily coordinated exostatic processes. First, we will introduce the concepts of endostasis and exostasis as compensation or accumulation for immediate or future energy needs and discuss how exostasis has been necessary for the survival of species during evolution. Then, we will argue how different specific biological functions of the CB1 receptor in the body converge to provide physiological exostatic processes. Finally, we will introduce the concept of proactive evolution-induced diseases (PEIDs), which helps explain the seeming paradox that an evolutionary-selected physiological function can become the cause of epidemic pathological conditions, such as obesity. We propose here a possible unifying theory of CB1 receptor functions that can be tested by future experimental studies.
Collapse
Affiliation(s)
- Pier Vincenzo Piazza
- INSERM, NeuroCentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33077 Bordeaux, France; University of Bordeaux, NeuroCentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33077 Bordeaux, France.
| | - Daniela Cota
- INSERM, NeuroCentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33077 Bordeaux, France; University of Bordeaux, NeuroCentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33077 Bordeaux, France
| | - Giovanni Marsicano
- INSERM, NeuroCentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33077 Bordeaux, France; University of Bordeaux, NeuroCentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33077 Bordeaux, France.
| |
Collapse
|
91
|
The Endocannabinoid System and Autism Spectrum Disorders: Insights from Animal Models. Int J Mol Sci 2017; 18:ijms18091916. [PMID: 28880200 PMCID: PMC5618565 DOI: 10.3390/ijms18091916] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 08/22/2017] [Accepted: 09/04/2017] [Indexed: 12/27/2022] Open
Abstract
Autism spectrum disorder (ASD) defines a group of neurodevelopmental disorders whose symptoms include impaired communication and social interaction with restricted or repetitive motor movements, frequently associated with general cognitive deficits. Although it is among the most severe chronic childhood disorders in terms of prevalence, morbidity, and impact to the society, no effective treatment for ASD is yet available, possibly because its neurobiological basis is not clearly understood hence specific drugs have not yet been developed. The endocannabinoid (EC) system represents a major neuromodulatory system involved in the regulation of emotional responses, behavioral reactivity to context, and social interaction. Furthermore, the EC system is also affected in conditions often present in subsets of patients diagnosed with ASD, such as seizures, anxiety, intellectual disabilities, and sleep pattern disturbances. Despite the indirect evidence suggestive of an involvement of the EC system in ASD, only a few studies have specifically addressed the role of the EC system in the context of ASD. This review describes the available data on the investigation of the presence of alterations of the EC system as well as the effects of its pharmacological manipulations in animal models of ASD-like behaviors.
Collapse
|
92
|
Boussadia B, Lakhal L, Payrastre L, Ghosh C, Pascussi JM, Gangarossa G, Marchi N. Pregnane X Receptor Deletion Modifies Recognition Memory and Electroencephalographic Activity. Neuroscience 2017; 370:130-138. [PMID: 28743453 DOI: 10.1016/j.neuroscience.2017.07.038] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 05/24/2017] [Accepted: 07/12/2017] [Indexed: 12/17/2022]
Abstract
Nuclear receptors (NR) are emerging as key players in the central nervous system (CNS) with reported implications in physiological and pathophysiological conditions. While a number of NR has been studied, it is unknown whether invalidation of the pregnane xenobiotic receptor (PXR, NR1I2) corresponds to neurological modifications in the adult brain. PXR-/- C57BL/6J and wild-type mice were used to investigate: (i) recognition memory, motor coordination, and anxiety-like behaviors; (ii) longitudinal video-electroencephalographic (EEG) recordings and frequency wave analysis; (iii) neurovascular structures by histological evaluation and expression of the cerebrovascular tight junctions ZO1 and CLDN5. Absence of PXR was associated with anxiety-like behavior and recognition memory impairment in adult mice. The latter was simultaneous to an EEG signature of lower theta frequency during sleep and abnormal delta waves. Neurophysiological changes did not correspond to significant structural changes in the adult brain, expect for a localized and minor increase in the fronto-parietal neurovascular density and reduced ZO1, but not CLDN5, expression in isolated brain capillaries. Our results converge with existing evidence supporting a link between NR expression and brain physiology. Although the exact modalities remain to be elucidated, the possibility that extra-physiological modulation of PXR may constitute a pathophysiological entry point or a molecular target for brain diseases is proposed.
Collapse
Affiliation(s)
- Badreddine Boussadia
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics, (UMR 5203 CNRS - U 1191 INSERM - Univ. Montpellier) Montpellier, France
| | - Laila Lakhal
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Laurence Payrastre
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | | | - Jean-Marc Pascussi
- Laboratory Signalization, Plasticity and Cancer, Department of Cancer Biology, Institute of Functional Genomics, Montpellier, France
| | - Giuseppe Gangarossa
- Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative (BFA), CNRS UMR 8251, F-75205 Paris, France.
| | - Nicola Marchi
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics, (UMR 5203 CNRS - U 1191 INSERM - Univ. Montpellier) Montpellier, France.
| |
Collapse
|
93
|
Araque A, Castillo PE, Manzoni OJ, Tonini R. Synaptic functions of endocannabinoid signaling in health and disease. Neuropharmacology 2017. [PMID: 28625718 DOI: 10.1016/j.neuropharm.2017.06.017] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Endocannabinoids (eCBs) are a family of lipid molecules that act as key regulators of synaptic transmission and plasticity. They are synthetized "on demand" following physiological and/or pathological stimuli. Once released from postsynaptic neurons, eCBs typically act as retrograde messengers to activate presynaptic type 1 cannabinoid receptors (CB1) and induce short- or long-term depression of neurotransmitter release. Besides this canonical mechanism of action, recent findings have revealed a number of less conventional mechanisms by which eCBs regulate neural activity and synaptic function, suggesting that eCB-mediated plasticity is mechanistically more diverse than anticipated. These mechanisms include non-retrograde signaling, signaling via astrocytes, participation in long-term potentiation, and the involvement of mitochondrial CB1. Focusing on paradigmatic brain areas, such as hippocampus, striatum, and neocortex, we review typical and novel signaling mechanisms, and discuss the functional implications in normal brain function and brain diseases. In summary, eCB signaling may lead to different forms of synaptic plasticity through activation of a plethora of mechanisms, which provide further complexity to the functional consequences of eCB signaling. This article is part of the Special Issue entitled "A New Dawn in Cannabinoid Neurobiology".
Collapse
Affiliation(s)
- Alfonso Araque
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Pablo E Castillo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY 10461, USA.
| | - Olivier J Manzoni
- Institut National de la Santé et et de la Recherche Médicale U901 Marseille, France, Université de la Méditerranée UMR S901 Aix-Marseille Marseille, France, INMED Marseille, France.
| | - Raffaella Tonini
- Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Genova, Italy.
| |
Collapse
|
94
|
Badal S, Smith KN, Rajnarayanan R. Analysis of natural product regulation of cannabinoid receptors in the treatment of human disease. Pharmacol Ther 2017; 180:24-48. [PMID: 28583800 DOI: 10.1016/j.pharmthera.2017.06.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The organized, tightly regulated signaling relays engaged by the cannabinoid receptors (CBs) and their ligands, G proteins and other effectors, together constitute the endocannabinoid system (ECS). This system governs many biological functions including cell proliferation, regulation of ion transport and neuronal messaging. This review will firstly examine the physiology of the ECS, briefly discussing some anomalies in the relay of the ECS signaling as these are consequently linked to maladies of global concern including neurological disorders, cardiovascular disease and cancer. While endogenous ligands are crucial for dispatching messages through the ECS, there are also commonalities in binding affinities with copious exogenous ligands, both natural and synthetic. Therefore, this review provides a comparative analysis of both types of exogenous ligands with emphasis on natural products given their putative safer efficacy and the role of Δ9-tetrahydrocannabinol (Δ9-THC) in uncovering the ECS. Efficacy is congruent to both types of compounds but noteworthy is the effect of a combination therapy to achieve efficacy without unideal side-effects. An example is Sativex that displayed promise in treating Huntington's disease (HD) in preclinical models allowing for its transition to current clinical investigation. Despite the in vitro and preclinical efficacy of Δ9-THC to treat neurodegenerative ailments, its psychotropic effects limit its clinical applicability to treating feeding disorders. We therefore propose further investigation of other compounds and their combinations such as the triterpene, α,β-amyrin that exhibited greater binding affinity to CB1 than CB2 and was more potent than Δ9-THC and the N-alkylamides that exhibited CB2 selective affinity; the latter can be explored towards peripherally exclusive ECS modulation. The synthetic CB1 antagonist, Rimonabant was pulled from commercial markets for the treatment of diabetes, however its analogue SR144528 maybe an ideal lead molecule towards this end and HU-210 and Org27569 are also promising synthetic small molecules.
Collapse
Affiliation(s)
- S Badal
- Department of Basic Medical Sciences, Faculty of Medical Sciences, University of the West Indies, Mona, Jamaica.
| | - K N Smith
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - R Rajnarayanan
- Jacobs School of Medicine and Biomedical Sciences, Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY 14228, USA
| |
Collapse
|
95
|
Fowler CJ, Doherty P, Alexander SPH. Endocannabinoid Turnover. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 80:31-66. [PMID: 28826539 DOI: 10.1016/bs.apha.2017.03.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In this review, we consider the biosynthetic, hydrolytic, and oxidative metabolism of the endocannabinoids anandamide and 2-arachidonoylglycerol. We describe the enzymes associated with these events and their characterization. We identify the inhibitor profile for these enzymes and the status of therapeutic exploitation, which to date has been limited to clinical trials for fatty acid amide hydrolase inhibitors. To bring the review to a close, we consider whether point block of a single enzyme is likely to be the most successful approach for therapeutic exploitation of the endocannabinoid system.
Collapse
Affiliation(s)
| | - Patrick Doherty
- Wolfson Centre for Age-Related Disease, King's College London, London, United Kingdom
| | | |
Collapse
|
96
|
Pugin A, Faundes V, Santa María L, Curotto B, Aliaga S, Salas I, Soto P, Bravo P, Peña M, Alliende M. Clinical, molecular, and pharmacological aspects of FMR1 -related disorders. NEUROLOGÍA (ENGLISH EDITION) 2017. [DOI: 10.1016/j.nrleng.2014.10.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
97
|
Abstract
Autism spectrum disorders (ASDs) are diagnosed on the basis of three behavioral features, namely, (1) deficits in social communication, (2) absence or delay in language and (3) stereotypy. The consensus regarding the neurological pathogenesis of ASDs is aberrant synaptogenesis and synapse function. Further, it is now widely accepted that ASD is neurodevelopmental in nature, placing emphasis on derangements occurring at the level of intra- and intercellular signaling during corticogenesis. At present, there is an ever-growing list of mutations in putative susceptibility genes in affected individuals, preventing effective transformation of knowledge gathered from basic science research to the clinic. In response, the focus of ASD biology has shifted toward the identification of cellular signaling pathways that are common to various ASD-related mutations in hopes that these shared pathways may serve as more promising treatment targets than targeting individual genes or proteins. To this end, the endogenous cannabinoid (endocannabinoid, eCB) system has recently emerged as a promising therapeutic target in the field of ASD research. The eCB system is altered in several neurological disorders, but the role of these bioactive lipids in ASD etiology remains poorly understood. In this perspective, we review current evidence linking eCB signaling to ASDs and put forth the notion that continued focus on eCBs in autism research may provide valuable insight into pathophysiology and treatment strategies. In addition to its role in modulating transmitter release at mature synapses, the eCB signaling system plays important roles in many aspects of cortical development, and disruption of these effects of eCBs may also be related to ASD pathophysiology.
Collapse
Affiliation(s)
- Mason L Yeh
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06032, USA
| | - Eric S Levine
- Department of Neuroscience, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06032, USA
| |
Collapse
|
98
|
CB 1 Cannabinoid Receptors Mediate Cognitive Deficits and Structural Plasticity Changes During Nicotine Withdrawal. Biol Psychiatry 2017; 81:625-634. [PMID: 27737762 DOI: 10.1016/j.biopsych.2016.07.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 06/15/2016] [Accepted: 07/07/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Tobacco withdrawal is associated with deficits in cognitive function, including attention, working memory, and episodic memory. Understanding the neurobiological mechanisms involved in these effects is crucial because cognitive deficits during nicotine withdrawal may predict relapse in humans. METHODS We investigated in mice the role of CB1 cannabinoid receptors (CB1Rs) in memory impairment and spine density changes induced by nicotine withdrawal precipitated by the nicotinic antagonist mecamylamine. Drugs acting on the endocannabinoid system and genetically modified mice were used. RESULTS Memory impairment during nicotine withdrawal was blocked by the CB1R antagonist rimonabant or the genetic deletion of CB1R in forebrain gamma-aminobutyric acidergic (GABAergic) neurons (GABA-CB1R). An increase of 2-arachidonoylglycerol (2-AG), but not anandamide, was observed during nicotine withdrawal. The selective inhibitor of 2-AG biosynthesis O7460 abolished cognitive deficits of nicotine abstinence, whereas the inhibitor of 2-AG enzymatic degradation JZL184 did not produce any effect in cognitive impairment. Moreover, memory impairment was prevented by the selective mammalian target of rapamycin inhibitor temsirolimus and the protein synthesis inhibitor anisomycin. Mature dendritic spines on CA1 pyramidal hippocampal neurons decreased 4 days after the precipitation of nicotine withdrawal, when the cognitive deficits were still present. Indeed, a correlation between memory performance and mature spine density was found. Interestingly, these structural plasticity alterations were normalized in GABA-CB1R conditional knockout mice and after subchronic treatment with rimonabant. CONCLUSIONS These findings underline the interest of CB1R as a target to improve cognitive performance during early nicotine withdrawal. Cognitive deficits in early abstinence are associated with increased relapse risk.
Collapse
|
99
|
Cissé M, Duplan E, Checler F. The transcription factor XBP1 in memory and cognition: Implications in Alzheimer disease. Mol Med 2017; 22:905-917. [PMID: 28079229 DOI: 10.2119/molmed.2016.00229] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 12/23/2016] [Indexed: 12/21/2022] Open
Abstract
X-box binding protein 1 (XBP1) is a unique basic region leucine zipper transcription factor isolated two decades ago in a search for regulators of major histocompatibility complex class II gene expression. XBP1 is a very complex protein regulating many physiological functions, including immune system, inflammatory responses, and lipid metabolism. Evidence over the past few years suggests that XBP1 also plays important roles in pathological settings since its activity as transcription factor has profound effects on the prognosis and progression of diseases such as cancer, neurodegeneration, and diabetes. Here we provide an overview on recent advances in our understanding of this multifaceted molecule, particularly in regulating synaptic plasticity and memory function, and the implications in neurodegenerative diseases with emphasis on Alzheimer disease.
Collapse
Affiliation(s)
- Moustapha Cissé
- Université Côte d'Azur, INSERM, CNRS, IPMC, team labeled "Fondation pour la Recherche Médicale" and "Laboratory of Excellence (LABEX) Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| | - Eric Duplan
- Université Côte d'Azur, INSERM, CNRS, IPMC, team labeled "Fondation pour la Recherche Médicale" and "Laboratory of Excellence (LABEX) Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| | - Frédéric Checler
- Université Côte d'Azur, INSERM, CNRS, IPMC, team labeled "Fondation pour la Recherche Médicale" and "Laboratory of Excellence (LABEX) Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| |
Collapse
|
100
|
Faus-Garriga J, Novoa I, Ozaita A. mTOR signaling in proteostasis and its relevance to autism spectrum disorders. AIMS BIOPHYSICS 2017. [DOI: 10.3934/biophy.2017.1.63] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|