1
|
Quadalti C, Sannia M, Humphreys N, Baldassarro V, Gurgone A, Ascolani M, Zanella L, Giardino L, Gross C, Croci S, Meloni I, Giustetto M, Renieri A, Lorenzini L, Calzà L. A new knockin mouse carrying the E364X patient mutation for CDKL5 deficiency disorder: neurological, behavioral and molecular profiling. Heliyon 2024; 10:e40165. [PMID: 39583831 PMCID: PMC11584566 DOI: 10.1016/j.heliyon.2024.e40165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/15/2024] [Accepted: 11/05/2024] [Indexed: 11/26/2024] Open
Abstract
CDKL5 deficiency disorder (CDD) is a rare neurodevelopmental syndrome caused by mutations in the X-linked CDKL5 gene. Hundreds of pathogenic variants have been described, associated with a significant phenotypic heterogeneity observed among patients. To date, different knockout mouse models have been generated. Here we present a new knockin CDKL5 mouse model carrying a humanized, well-characterized nonsense variant (c.1090G > T; p.E364X) described in the C-terminal domain of the CDKL5 protein in a female patient with a milder phenotype. Both male and female Cdkl5 E364X mice were analyzed. The novel Cdkl5 E364X mouse showed altered neurological and motor neuron maturation, hyperactivity, defective coordination and impaired memory and cognition. Gene expression analysis highlighted an unexpected reduction of Cdkl5 expression in Cdkl5 E364X mice brain tissues, with a significant increase in overall neuron-specific gene expression and an area-dependent alteration of astrocyte- and oligodendrocyte-specific transcripts. Moreover, our results showed that the loss of CDKL5 protein had the most significant impact on the cerebellum and hippocampus, compared to other analyzed tissues. A targeted analysis to study synaptic plasticity in cerebellum and hippocampus showed reduced Gabra1 and Gabra5 expression levels in females, whereas Gabra1 expression was increased in males, suggesting an opposite, sex-dependent regulation of the GABA receptor expression already described in humans. In conclusion, the novel Cdkl5E364X mouse model is characterized by robust neurological and neurobehavioral alterations, associated with a molecular profile related to synaptic function indicative of a cerebellar GABAergic hypofunction, pointing to Gabra1 and Gabra5 as novel druggable target candidates for CDD.
Collapse
Affiliation(s)
- C. Quadalti
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - M. Sannia
- IRET Foundation, 40064 Ozzano Emilia (Bologna), Italy
| | - N.E. Humphreys
- Epigenetics & Neurobiology Unit, European Molecular Biology Laboratory (EMBL), Rome, Italy
| | - V.A. Baldassarro
- Department of Veterinary Medical Sciences, University of Bologna, 40064 Bologna, Italy
| | - A. Gurgone
- Department of Neuroscience “Rita Levi-Montalcini”, University of Turin, 10125 Turin, Italy
| | - M. Ascolani
- Epigenetics & Neurobiology Unit, European Molecular Biology Laboratory (EMBL), Rome, Italy
| | - L. Zanella
- Department of Veterinary Medical Sciences, University of Bologna, 40064 Bologna, Italy
| | - L. Giardino
- Department of Veterinary Medical Sciences, University of Bologna, 40064 Bologna, Italy
| | - C.T. Gross
- Epigenetics & Neurobiology Unit, European Molecular Biology Laboratory (EMBL), Rome, Italy
| | - S. Croci
- Medical Genetics, University of Siena, 53100 Siena, Italy
| | - I. Meloni
- Medical Genetics, University of Siena, 53100 Siena, Italy
| | - M. Giustetto
- Department of Neuroscience “Rita Levi-Montalcini”, University of Turin, 10125 Turin, Italy
| | - A. Renieri
- Medical Genetics, University of Siena, 53100 Siena, Italy
- Medical Genetics Department, Siena University Hospital, 53100 Siena, Italy
| | - L. Lorenzini
- Department of Veterinary Medical Sciences, University of Bologna, 40064 Bologna, Italy
| | - L. Calzà
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| |
Collapse
|
2
|
Cesaroni CA, Pisanò G, Trimarchi G, Caraffi SG, Scandolo G, Gnazzo M, Frattini D, Spagnoli C, Rizzi S, Dittadi C, Sigona G, Garavelli L, Fusco C. Severe Neurodevelopmental Disorder in Autosomal Recessive Spinocerebellar Ataxia 13 (SCAR13) Caused by Two Novel Frameshift Variants in GRM1. CEREBELLUM (LONDON, ENGLAND) 2024; 23:1768-1771. [PMID: 37831383 DOI: 10.1007/s12311-023-01617-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/05/2023] [Indexed: 10/14/2023]
Abstract
Autosomal recessive spinocerebellar ataxia 13 (SCAR13) is a neurological disease characterized by psychomotor delay, mild to profound intellectual disability with poor or absent language, nystagmus, stance ataxia, and, if walking is acquired, gait ataxia. Epilepsy and polyneuropathy have also been documented in some patients. Cerebellar atrophy and/or ventriculomegaly may be present on brain MRI. SCAR13 is caused by pathogenic variants in the GRM1 gene encoding the metabotropic receptor of glutamate type 1 (mGlur1), which is highly expressed in Purkinje cerebellar cells, where it plays a fundamental role in cerebellar development. Here we discuss the case of an 8-year-old patient who presented with a severe neurodevelopmental disorder with balance disturbance, absence of independent walking, absence of language, diffuse hypotonia, mild nystagmus, and mild dysphagia. Whole-exome sequencing revealed a compound heterozygosity for two likely pathogenic variants in the GRM1 gene, responsible for the patient's phenotype, and made it possible to diagnose autosomal recessive spinocerebellar ataxia SCAR13. The detected (novel) variants appear to be causative of a particularly severe picture with regard to neurodevelopment, in the context of the typical neurological signs of spinocerebellar ataxia.
Collapse
Affiliation(s)
- Carlo Alberto Cesaroni
- Child Neurology and Psychiatry Unit, Pediatric Neurophysiology Laboratory, Mother-Child Department, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy.
| | - Giulia Pisanò
- Child Neurology and Psychiatry Unit, Pediatric Neurophysiology Laboratory, Mother-Child Department, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Gabriele Trimarchi
- Medical Genetics Unit, Mother-Child Department, Azienda USL-IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | - Stefano Giuseppe Caraffi
- Medical Genetics Unit, Mother-Child Department, Azienda USL-IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | - Giulia Scandolo
- Child Neurology and Psychiatry Unit, Pediatric Neurophysiology Laboratory, Mother-Child Department, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Martina Gnazzo
- Child Neurology and Psychiatry Unit, Pediatric Neurophysiology Laboratory, Mother-Child Department, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Daniele Frattini
- Child Neurology and Psychiatry Unit, Pediatric Neurophysiology Laboratory, Mother-Child Department, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Carlotta Spagnoli
- Child Neurology and Psychiatry Unit, Pediatric Neurophysiology Laboratory, Mother-Child Department, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Susanna Rizzi
- Child Neurology and Psychiatry Unit, Pediatric Neurophysiology Laboratory, Mother-Child Department, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Claudia Dittadi
- Child Neurology and Psychiatry Unit, Pediatric Neurophysiology Laboratory, Mother-Child Department, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Giulia Sigona
- Child Neurology and Psychiatry Unit, Pediatric Neurophysiology Laboratory, Mother-Child Department, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| | - Livia Garavelli
- Medical Genetics Unit, Mother-Child Department, Azienda USL-IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | - Carlo Fusco
- Child Neurology and Psychiatry Unit, Pediatric Neurophysiology Laboratory, Mother-Child Department, Azienda USL-IRCCS Di Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
3
|
Tian Y, Qiao H, Zhu LQ, Man HY. Sexually dimorphic phenotypes and the role of androgen receptors in UBE3A-dependent autism spectrum disorder. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.02.592248. [PMID: 38746146 PMCID: PMC11092617 DOI: 10.1101/2024.05.02.592248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Autism spectrum disorders (ASDs) are characterized by social, communication, and behavioral challenges. UBE3A is one of the most common ASD genes. ASDs display a remarkable sex difference with a 4:1 male to female prevalence ratio; however, the underlying mechanism remains largely unknown. Using the UBE3A-overexpressing mouse model for ASD, we studied sex differences at behavioral, genetic, and molecular levels. We found that male mice with extra copies of Ube3A exhibited greater impairments in social interaction, repetitive self-grooming behavior, memory, and pain sensitivity, whereas female mice with UBE3A overexpression displayed greater olfactory defects. Social communication was impaired in both sexes, with males making more calls and females preferring complex syllables. At the molecular level, androgen receptor (AR) levels were reduced in both sexes due to enhanced degradation mediated by UBE3A. However, AR reduction significantly dysregulated AR target genes only in male, not female, UBE3A-overexpressing mice. Importantly, restoring AR levels in the brain effectively normalized the expression of AR target genes, and rescued the deficits in social preference, grooming behavior, and memory in male UBE3A-overexpressing mice, without affecting females. These findings suggest that AR and its signaling cascade play an essential role in mediating the sexually dimorphic changes in UBE3A-dependent ASD.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Hui Qiao
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Heng-Ye Man
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
- Department of Pharmacology, Physiology & Biophysics, Boston University School of Medicine, 72 East Concord St., Boston, MA 02118, USA
- Center for Systems Neuroscience, Boston University, 610 Commonwealth Ave, Boston, MA 02215, USA
| |
Collapse
|
4
|
Delhaye S, Jarjat M, Boulksibat A, Sanchez C, Tempio A, Turtoi A, Giorgi M, Lacas-Gervais S, Baj G, Rovere C, Trezza V, Pellegrini M, Maurin T, Lalli E, Bardoni B. Defects in AMPAR trafficking and microglia activation underlie socio-cognitive deficits associated to decreased expression of phosphodiesterase 2 a. Neurobiol Dis 2024; 191:106393. [PMID: 38154608 DOI: 10.1016/j.nbd.2023.106393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/21/2023] [Accepted: 12/21/2023] [Indexed: 12/30/2023] Open
Abstract
Phosphodiesterase 2 A (PDE2A) is an enzyme involved in the homeostasis of cAMP and cGMP and is the most highly expressed PDE in human brain regions critical for socio-cognitive behavior. In cerebral cortex and hippocampus, PDE2A expression level is upregulated in Fmr1-KO mice, a model of the Fragile X Syndrome (FXS), the most common form of inherited intellectual disability (ID) and autism spectrum disorder (ASD). Indeed, PDE2A translation is negatively modulated by FMRP, whose functional absence causes FXS. While the pharmacological inhibition of PDE2A has been associated to its pro-cognitive role in normal animals and in models of ID and ASD, homozygous PDE2A mutations have been identified in patients affected by ID, ASD and epilepsy. To clarify this apparent paradox about the role of PDE2A in brain development, we characterized here Pde2a+/- mice (homozygote animals being not viable) at the behavioral, cellular, molecular and electrophysiological levels. Pde2a+/- females display a milder form of the disorder with reduced cognitive performance in adulthood, conversely males show severe socio-cognitive deficits throughout their life. In males, these phenotypes are associated with microglia activation, elevated glutathione levels and increased externalization of Glutamate receptor (GluR1) in CA1, producing reduced mGluR-dependent Long-term Depression. Overall, our results reveal molecular targets of the PDE2A-dependent pathway underlying socio-cognitive performance. These results clarify the mechanism of action of pro-cognitive drugs based on PDE2A inactivation, which have been shown to be promising therapeutic approaches for Alzheimer's disease, schizophrenia, FXS as well as other forms of ASD.
Collapse
Affiliation(s)
- Sébastien Delhaye
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Marielle Jarjat
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Asma Boulksibat
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Clara Sanchez
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Alessandra Tempio
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Andrei Turtoi
- Inserm U1194, Université Montpellier, Institut de Recherche en Cancérologie de Montpellier, 34298 Montpellier Cedex 5, France
| | - Mauro Giorgi
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, DAHFMO, Sapienza University of Rome, 00161 Rome, Italy
| | - Sandra Lacas-Gervais
- Université Côte d'Azur, Centre Commun de Microscopie Appliquée, 06100 Nice, France
| | - Gabriele Baj
- Department of Life Science, University of Trieste, 34100 Trieste, Italy
| | - Carole Rovere
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | | | - Manuela Pellegrini
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, DAHFMO, Sapienza University of Rome, 00161 Rome, Italy; Institute of Biochemistry and Cell Biology, IBBC-CNR, 00015 Monterotondo Scalo, Rome, Italy
| | - Thomas Maurin
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Enzo Lalli
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| | - Barbara Bardoni
- CNRS UMR7275, Inserm U1323, Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France.
| |
Collapse
|
5
|
Frare C, Pitt SK, Hewett SJ. Sex- and age-dependent contribution of System x c- to cognitive, sensory, and social behaviors revealed by comprehensive behavioral analyses of System x c- null mice. Front Behav Neurosci 2023; 17:1238349. [PMID: 37649973 PMCID: PMC10462982 DOI: 10.3389/fnbeh.2023.1238349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 07/31/2023] [Indexed: 09/01/2023] Open
Abstract
Background System xc- (Sxc-) is an important heteromeric amino acid cystine/glutamate exchanger that plays a pivotal role in the CNS by importing cystine into cells while exporting glutamate. Although certain behaviors have been identified as altered in Sxc- null mutant mice, our understanding of the comprehensive impact of Sxc- on behavior remains incomplete. Methods To address this gap, we compared motor, sensory and social behaviors of male and female mice in mice null for Sxc- (SLC7A11sut/sut) with wildtype littermates (SLC7A11+/+) in a comprehensive and systematic manner to determine effects of genotype, sex, age, and their potential interactions. Results Motor performance was not affected by loss of Sxc- in both males and females, although it was impacted negatively by age. Motor learning was specifically disrupted in female mice lacking Sxc- at both 2 and 6 months of age. Further, female SLC7A11sut/sut mice at both ages exhibited impaired sociability, but normal spatial and recognition memory, as well as sensorimotor gating. Finally, pronounced open-space anxiety was displayed by female SLC7A11sut/sut when they were young. In contrast, young SLC7A11sut/sut male mice demonstrated normal sociability, delayed spatial learning, increased open-space anxiety and heightened sensitivity to noise. As they aged, anxiety and noise sensitivity abated but hyperactivity emerged. Discussion We find that the behavioral phenotypes of female SLC7A11sut/sut are similar to those observed in mouse models of autism spectrum disorder, while behaviors of male SLC7A11sut/sut resemble those seen in mouse models of attention deficit hyperactivity disorder. These results underscore the need for further investigation of SLC7A11 in neurodevelopment. By expanding our understanding of the potential involvement of Sxc-, we may gain additional insights into the mechanisms underlying complex neurodevelopmental conditions.
Collapse
Affiliation(s)
| | | | - Sandra J. Hewett
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, NY, United States
| |
Collapse
|
6
|
Malik AM, Wu JJ, Gillies CA, Doctrove QA, Li X, Huang H, Tank EHM, Shakkottai VG, Barmada S. Neuronal activity regulates Matrin 3 abundance and function in a calcium-dependent manner through calpain-mediated cleavage and calmodulin binding. Proc Natl Acad Sci U S A 2023; 120:e2206217120. [PMID: 37011198 PMCID: PMC10104577 DOI: 10.1073/pnas.2206217120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 02/14/2023] [Indexed: 04/05/2023] Open
Abstract
RNA-binding protein (RBP) dysfunction is a fundamental hallmark of amyotrophic lateral sclerosis (ALS) and related neuromuscular disorders. Abnormal neuronal excitability is also a conserved feature in ALS patients and disease models, yet little is known about how activity-dependent processes regulate RBP levels and functions. Mutations in the gene encoding the RBP Matrin 3 (MATR3) cause familial disease, and MATR3 pathology has also been observed in sporadic ALS, suggesting a key role for MATR3 in disease pathogenesis. Here, we show that glutamatergic activity drives MATR3 degradation through an NMDA receptor-, Ca2+-, and calpain-dependent mechanism. The most common pathogenic MATR3 mutation renders it resistant to calpain degradation, suggesting a link between activity-dependent MATR3 regulation and disease. We also demonstrate that Ca2+ regulates MATR3 through a nondegradative process involving the binding of Ca2+/calmodulin to MATR3 and inhibition of its RNA-binding ability. These findings indicate that neuronal activity impacts both the abundance and function of MATR3, underscoring the effect of activity on RBPs and providing a foundation for further study of Ca2+-coupled regulation of RBPs implicated in ALS and related neurological diseases.
Collapse
Affiliation(s)
- Ahmed M. Malik
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI48109
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI48109
- Department of Neurology, University of Michigan, Ann Arbor, MI48109
| | - Josephine J. Wu
- Department of Neurology, University of Michigan, Ann Arbor, MI48109
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI48109
| | - Christie A. Gillies
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI48109
| | - Quinlan A. Doctrove
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI48109
- Postbac Research Education Program, University of Michigan, Ann Arbor, MI48109
| | - Xingli Li
- Department of Neurology, University of Michigan, Ann Arbor, MI48109
| | - Haoran Huang
- University of Texas Southwestern Medical Center, Dallas, TX 75390
| | | | | | - Sami Barmada
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI48109
- Department of Neurology, University of Michigan, Ann Arbor, MI48109
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI48109
| |
Collapse
|
7
|
Sefik E, Boamah M, Addington J, Bearden CE, Cadenhead KS, Cornblatt BA, Keshavan MS, Mathalon DH, Perkins DO, Stone WS, Tsuang MT, Woods SW, Cannon TD, Walker EF. Sex- and Age-Specific Deviations in Cerebellar Structure and Their Link With Symptom Dimensions and Clinical Outcome in Individuals at Clinical High Risk for Psychosis. Schizophr Bull 2023; 49:350-363. [PMID: 36394426 PMCID: PMC10016422 DOI: 10.1093/schbul/sbac169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND The clinical high-risk (CHR) period offers a temporal window into neurobiological deviations preceding psychosis onset, but little attention has been given to regions outside the cerebrum in large-scale studies of CHR. Recently, the North American Prodrome Longitudinal Study (NAPLS)-2 revealed altered functional connectivity of the cerebello-thalamo-cortical circuitry among individuals at CHR; however, cerebellar morphology remains underinvestigated in this at-risk population, despite growing evidence of its involvement in psychosis. STUDY DESIGN In this multisite study, we analyzed T1-weighted magnetic resonance imaging scans obtained from N = 469 CHR individuals (61% male, ages = 12-36 years) and N = 212 healthy controls (52% male, ages = 12-34 years) from NAPLS-2, with a focus on cerebellar cortex and white matter volumes separately. Symptoms were rated by the Structured Interview for Psychosis-Risk Syndromes (SIPS). The outcome by two-year follow-up was categorized as in-remission, symptomatic, prodromal-progression, or psychotic. General linear models were used for case-control comparisons and tests for volumetric associations with baseline SIPS ratings and clinical outcomes. STUDY RESULTS Cerebellar cortex and white matter volumes differed between the CHR and healthy control groups at baseline, with sex moderating the difference in cortical volumes, and both sex and age moderating the difference in white matter volumes. Baseline ratings for major psychosis-risk dimensions as well as a clinical outcome at follow-up had tissue-specific associations with cerebellar volumes. CONCLUSIONS These findings point to clinically relevant deviations in cerebellar cortex and white matter structures among CHR individuals and highlight the importance of considering the complex interplay between sex and age when studying the neuromaturational substrates of psychosis risk.
Collapse
Affiliation(s)
- Esra Sefik
- Department of Psychology, Emory University, Atlanta, GA, USA
- Department of Human Genetics, Emory University, Atlanta, GA, USA
| | - Michelle Boamah
- Department of Psychology, Emory University, Atlanta, GA, USA
| | - Jean Addington
- Department of Psychiatry, University of Calgary, Calgary, AB, Canada
| | - Carrie E Bearden
- Department of Psychology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Kristin S Cadenhead
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | | | - Matcheri S Keshavan
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Daniel H Mathalon
- Department of Psychiatry, University of California San Francisco, San Francisco, CA, USA
- Mental Health Service, San Francisco VA Medical Center, San Francisco, CA, USA
| | - Diana O Perkins
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - William S Stone
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Ming T Tsuang
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Scott W Woods
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Tyrone D Cannon
- Department of Psychiatry, Yale University, New Haven, CT, USA
- Department of Psychology, Yale University, New Haven, CT, USA
| | - Elaine F Walker
- Department of Psychology, Emory University, Atlanta, GA, USA
| |
Collapse
|
8
|
Fabian CB, Seney ML, Joffe ME. Sex differences and hormonal regulation of metabotropic glutamate receptor synaptic plasticity. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 168:311-347. [PMID: 36868632 PMCID: PMC10392610 DOI: 10.1016/bs.irn.2022.10.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Striking sex differences exist in presentation and incidence of several psychiatric disorders. For example, major depressive disorder is more prevalent in women than men, and women who develop alcohol use disorder progress through drinking milestones more rapidly than men. With regards to psychiatric treatment responses, women respond more favorably to selective serotonin reuptake inhibitors than men, whereas men have better outcomes when prescribed tricyclic antidepressants. Despite such well-documented biases in incidence, presentation, and treatment response, sex as a biological variable has long been neglected in preclinical and clinical research. An emerging family of druggable targets for psychiatric diseases, metabotropic glutamate (mGlu) receptors are G-protein coupled receptors broadly distributed throughout the central nervous system. mGlu receptors confer diverse neuromodulatory actions of glutamate at the levels of synaptic plasticity, neuronal excitability, and gene transcription. In this chapter, we summarize the current preclinical and clinical evidence for sex differences in mGlu receptor function. We first highlight basal sex differences in mGlu receptor expression and function and proceed to describe how gonadal hormones, notably estradiol, regulate mGlu receptor signaling. We then describe sex-specific mechanisms by which mGlu receptors differentially modulate synaptic plasticity and behavior in basal states and models relevant for disease. Finally, we discuss human research findings and highlight areas in need of further research. Taken together, this review emphasizes how mGlu receptor function and expression can differ across sex. Gaining a more complete understanding of how sex differences in mGlu receptor function contribute to psychiatric diseases will be critical in the development of novel therapeutics that are effective in all individuals.
Collapse
Affiliation(s)
- Carly B Fabian
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States; Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, United States
| | - Marianne L Seney
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States; Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, United States
| | - Max E Joffe
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States; Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
9
|
Deficits in Cerebellum-Dependent Learning and Cerebellar Morphology in Male and Female BTBR Autism Model Mice. NEUROSCI 2022. [DOI: 10.3390/neurosci3040045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Recently, there has been increased interest in the role of the cerebellum in autism spectrum disorder (ASD). To better understand the pathophysiological role of the cerebellum in ASD, it is necessary to have a variety of mouse models that have face validity for cerebellar disruption in humans. Here, we add to the literature on the cerebellum in mouse models of autism with the characterization of the cerebellum in the idiopathic BTBR T + Itpr3tf/J (BTBR) inbred mouse strain, which has behavioral phenotypes that are reminiscent of ASD in patients. When we examined both male and female BTBR mice in comparison to C57BL/6J (C57) controls, we noted that both sexes of BTBR mice showed motor coordination deficits characteristic of cerebellar dysfunction, but only the male mice showed differences in delay eyeblink conditioning, a cerebellum-dependent learning task that is known to be disrupted in ASD patients. Both male and female BTBR mice showed considerable expansion of, and abnormal foliation in, the cerebellum vermis—including a significant expansion of specific lobules in the anterior cerebellum. In addition, we found a slight but significant decrease in Purkinje cell density in both male and female BTBR mice, irrespective of the lobule. Finally, there was a marked reduction of Purkinje cell dendritic spine density in both male and female BTBR mice. These findings suggest that, for the most part, the BTBR mouse model phenocopies many of the characteristics of the subpopulation of ASD patients that have a hypertrophic cerebellum. We discuss the significance of strain differences in the cerebellum as well as the importance of this first effort to identify both similarities and differences between male and female BTBR mice with regard to the cerebellum.
Collapse
|
10
|
Martinez ME, Stohn JP, Mutina EM, Whitten RJ, Hernandez A. Thyroid hormone elicits intergenerational epigenetic effects on adult social behavior and fetal brain expression of autism susceptibility genes. Front Neurosci 2022; 16:1055116. [PMID: 36419462 PMCID: PMC9676973 DOI: 10.3389/fnins.2022.1055116] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/24/2022] [Indexed: 11/09/2022] Open
Abstract
Genetic mutations identified in genome-wide association studies can only explain a small percentage of the cases of complex, highly heritable human conditions, including neurological and neurodevelopmental disorders. This suggests that intergenerational epigenetic effects, possibly triggered by environmental circumstances, may contribute to their etiology. We previously described altered DNA methylation signatures in the sperm of mice that experienced developmental overexposure to thyroid hormones as a result of a genetic defect in hormone clearance (DIO3 deficiency). Here we studied fetal brain gene expression and adult social behavior in genetically normal F2 generation descendants of overexposed mice. The brain of F2 generation E13.5 fetuses exhibited abnormal expression of genes associated with autism in humans, including Auts2, Disc1, Ldlr, Per2, Shank3, Oxtr, Igf1, Foxg1, Cd38, Grid2, Nrxn3, and Reln. These abnormal gene expression profiles differed depending on the sex of the exposed ancestor. In the three-chamber social box test, adult F2 generation males manifested significantly decreased interest in social interaction and social novelty, as revealed by decrease total time, distance traveled and time immobile in the area of interaction with novel strangers. F1 generation mice, compared to appropriate controls also exhibited altered profiles in fetal brain gene expression, although these profiles were substantially different to those in the F2 generation. Likewise adult F1 generation mice showed some abnormalities in social behavior that were sexually dimorphic and milder than those in F2 generation mice. Our results indicate that developmental overexposure to thyroid hormone causes intergenerational epigenetic effects impacting social behavior and the expression of autism-related genes during early brain development. Our results open the possibility that altered thyroid hormone states, by eliciting changes in the epigenetic information of the germ line, contribute to the susceptibility and the missing-but heriTables-etiology of complex neurodevelopmental conditions characterized by social deficits, including autism and schizophrenia.
Collapse
Affiliation(s)
- Maria Elena Martinez
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, United States
| | - Julia Patrizia Stohn
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, United States
| | - Elizabeth M. Mutina
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, United States
| | - Rayne J. Whitten
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, United States
| | - Arturo Hernandez
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, United States
- Graduate School for Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States
- Department of Medicine, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
11
|
Su LD, Wang N, Han J, Shen Y. Group 1 Metabotropic Glutamate Receptors in Neurological and Psychiatric Diseases: Mechanisms and Prospective. Neuroscientist 2021; 28:453-468. [PMID: 34088252 PMCID: PMC9449437 DOI: 10.1177/10738584211021018] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Metabotropic glutamate receptors (mGluRs) are G-protein coupled receptors
that are activated by glutamate in the central nervous system (CNS).
Basically, mGluRs contribute to fine-tuning of synaptic efficacy and
control the accuracy and sharpness of neurotransmission. Among eight
subtypes, mGluR1 and mGluR5 belong to group 1 (Gp1) family, and are
implicated in multiple CNS disorders, such as Alzheimer’s disease,
autism, Parkinson’s disease, and so on. In the present review, we
systematically discussed underlying mechanisms and prospective of Gp1
mGluRs in a group of neurological and psychiatric diseases, including
Alzheimer’s disease, Parkinson’s disease, autism spectrum disorder,
epilepsy, Huntington’s disease, intellectual disability, Down’s
syndrome, Rett syndrome, attention-deficit hyperactivity disorder,
addiction, anxiety, nociception, schizophrenia, and depression, in
order to provide more insights into the therapeutic potential of Gp1
mGluRs.
Collapse
Affiliation(s)
- Li-Da Su
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Na Wang
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Junhai Han
- School of Life Science and Technology, the Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Ying Shen
- Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
12
|
Rudolph S, Guo C, Pashkovski SL, Osorno T, Gillis WF, Krauss JM, Nyitrai H, Flaquer I, El-Rifai M, Datta SR, Regehr WG. Cerebellum-Specific Deletion of the GABA A Receptor δ Subunit Leads to Sex-Specific Disruption of Behavior. Cell Rep 2021; 33:108338. [PMID: 33147470 PMCID: PMC7700496 DOI: 10.1016/j.celrep.2020.108338] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 08/04/2020] [Accepted: 10/08/2020] [Indexed: 12/19/2022] Open
Abstract
Granule cells (GCs) of the cerebellar input layer express high-affinity δ GABAA subunit-containing GABAA receptors (δGABAARs) that respond to ambient GABA levels and context-dependent neuromodulators like steroids. We find that GC-specific deletion of δGABAA (cerebellar [cb] δ knockout [KO]) decreases tonic inhibition, makes GCs hyperexcitable, and in turn, leads to differential activation of cb output regions as well as many cortical and subcortical brain areas involved in cognition, anxiety-like behaviors, and the stress response. Cb δ KO mice display deficits in many behaviors, but motor function is normal. Strikingly, δGABAA deletion alters maternal behavior as well as spontaneous, stress-related, and social behaviors specifically in females. Our findings establish that δGABAARs enable the cerebellum to control diverse behaviors not previously associated with the cerebellum in a sex-dependent manner. These insights may contribute to a better understanding of the mechanisms that underlie behavioral abnormalities in psychiatric and neurodevelopmental disorders that display a gender bias. Rudolph et al. show that deletion of the neuromodulator and hormone-sensitive δGABAA receptor subunit from cerebellar granule cells results in anxiety-like behaviors and female-specific deficits in social behavior and maternal care. δGABAA deletion is associated with hyperexcitability of the cerebellar input layer and altered activation of many stress-related brain regions.
Collapse
Affiliation(s)
- Stephanie Rudolph
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Chong Guo
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Stan L Pashkovski
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Tomas Osorno
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Winthrop F Gillis
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Jeremy M Krauss
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Hajnalka Nyitrai
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Isabella Flaquer
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Mahmoud El-Rifai
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Wade G Regehr
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
13
|
van der Heijden ME, Kizek DJ, Perez R, Ruff EK, Ehrlich ME, Sillitoe RV. Abnormal cerebellar function and tremor in a mouse model for non-manifesting partially penetrant dystonia type 6. J Physiol 2021; 599:2037-2054. [PMID: 33369735 PMCID: PMC8559601 DOI: 10.1113/jp280978] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/16/2020] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS Loss-of-function mutations in the Thap1 gene cause partially penetrant dystonia type 6 (DYT6). Some non-manifesting DYT6 mutation carriers have tremor and abnormal cerebello-thalamo-cortical signalling. We show that Thap1 heterozygote mice have action tremor, a reduction in cerebellar neuron number, and abnormal electrophysiological signals in the remaining neurons. These results underscore the importance of Thap1 levels for cerebellar function. These results uncover how cerebellar abnormalities contribute to different dystonia-associated motor symptoms. ABSTRACT Loss-of-function mutations in the Thanatos-associated domain-containing apoptosis-associated protein 1 (THAP1) gene cause partially penetrant autosomal dominant dystonia type 6 (DYT6). However, the neural abnormalities that promote the resultant motor dysfunctions remain elusive. Studies in humans show that some non-manifesting DYT6 carriers have altered cerebello-thalamo-cortical function with subtle but reproducible tremor. Here, we uncover that Thap1 heterozygote mice have action tremor that rises above normal baseline values even though they do not exhibit overt dystonia-like twisting behaviour. At the neural circuit level, we show using in vivo recordings in awake Thap1+/- mice that Purkinje cells have abnormal firing patterns and that cerebellar nuclei neurons, which connect the cerebellum to the thalamus, fire at a lower frequency. Although the Thap1+/- mice have fewer Purkinje cells and cerebellar nuclei neurons, the number of long-range excitatory outflow projection neurons is unaltered. The preservation of interregional connectivity suggests that abnormal neural function rather than neuron loss instigates the network dysfunction and the tremor in Thap1+/- mice. Accordingly, we report an inverse correlation between the average firing rate of cerebellar nuclei neurons and tremor power. Our data show that cerebellar circuitry is vulnerable to Thap1 mutations and that cerebellar dysfunction may be a primary cause of tremor in non-manifesting DYT6 carriers and a trigger for the abnormal postures in manifesting patients.
Collapse
Affiliation(s)
- Meike E. van der Heijden
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Dominic J. Kizek
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Ross Perez
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Elena K. Ruff
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Michelle E. Ehrlich
- Department of Neurology and Pediatrics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Roy V. Sillitoe
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, USA
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| |
Collapse
|
14
|
Gating by Functionally Indivisible Cerebellar Circuits: a Hypothesis. THE CEREBELLUM 2021; 20:518-532. [PMID: 33464470 PMCID: PMC8360902 DOI: 10.1007/s12311-020-01223-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 12/01/2020] [Indexed: 11/08/2022]
Abstract
The attempt to understand the cerebellum has been dominated for years by supervised learning models. The central idea is that a learning algorithm modifies transmission strength at repeatedly co-active synapses, creating memories stored as finely calibrated synaptic weights. As a result, Purkinje cells, usually the de facto output cells of these models, acquire a modified response to input in a remembered pattern. This paper proposes an alternative model of pattern memory in which the function of a match is permissive, allowing but not driving output, and accordingly controlling the timing of output but not the rate of firing by Purkinje cells. Learning does not result in graded synaptic weights. There is no supervised learning algorithm or memory of individual patterns, which, like graded weights, are unnecessary to explain the evidence. Instead, patterns are classed as simply either known or not, at the level of input to a functional population of 100s of Purkinje cells (a microzone). The standard is strict. If only a handful of Purkinje cells receive a mismatch output of the whole circuit is blocked. Only if there is a full and accurate match are projection neurons in deep nuclei, which carry the output of most circuits, released from default inhibitory restraint. Purkinje cell firing at those times is a linear function of input rates. There is no effect of modification of synaptic transmission except to either allow or block output.
Collapse
|
15
|
Gandhi T, Lee CC. Neural Mechanisms Underlying Repetitive Behaviors in Rodent Models of Autism Spectrum Disorders. Front Cell Neurosci 2021; 14:592710. [PMID: 33519379 PMCID: PMC7840495 DOI: 10.3389/fncel.2020.592710] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 12/09/2020] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorder (ASD) is comprised of several conditions characterized by alterations in social interaction, communication, and repetitive behaviors. Genetic and environmental factors contribute to the heterogeneous development of ASD behaviors. Several rodent models display ASD-like phenotypes, including repetitive behaviors. In this review article, we discuss the potential neural mechanisms involved in repetitive behaviors in rodent models of ASD and related neuropsychiatric disorders. We review signaling pathways, neural circuits, and anatomical alterations in rodent models that display robust stereotypic behaviors. Understanding the mechanisms and circuit alterations underlying repetitive behaviors in rodent models of ASD will inform translational research and provide useful insight into therapeutic strategies for the treatment of repetitive behaviors in ASD and other neuropsychiatric disorders.
Collapse
Affiliation(s)
- Tanya Gandhi
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | | |
Collapse
|
16
|
Corain L, Grisan E, Graïc JM, Carvajal-Schiaffino R, Cozzi B, Peruffo A. Multi-aspect testing and ranking inference to quantify dimorphism in the cytoarchitecture of cerebellum of male, female and intersex individuals: a model applied to bovine brains. Brain Struct Funct 2020; 225:2669-2688. [PMID: 32989472 PMCID: PMC7674367 DOI: 10.1007/s00429-020-02147-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 09/08/2020] [Indexed: 11/28/2022]
Abstract
The dimorphism among male, female and freemartin intersex bovines, focusing on the vermal lobules VIII and IX, was analyzed using a novel data analytics approach to quantify morphometric differences in the cytoarchitecture of digitalized sections of the cerebellum. This methodology consists of multivariate and multi-aspect testing for cytoarchitecture-ranking, based on neuronal cell complexity among populations defined by factors, such as sex, age or pathology. In this context, we computed a set of shape descriptors of the neural cell morphology, categorized them into three domains named size, regularity and density, respectively. The output and results of our methodology are multivariate in nature, allowing an in-depth analysis of the cytoarchitectonic organization and morphology of cells. Interestingly, the Purkinje neurons and the underlying granule cells revealed the same morphological pattern: female possessed larger, denser and more irregular neurons than males. In the Freemartin, Purkinje neurons showed an intermediate setting between males and females, while the granule cells were the largest, most regular and dense. This methodology could be a powerful instrument to carry out morphometric analysis providing robust bases for objective tissue screening, especially in the field of neurodegenerative pathologies.
Collapse
Affiliation(s)
- L Corain
- Department of Management and Engineering, University of Padova, 36100, Vicenza, VI, Italy
| | - E Grisan
- Department of Information Engineering, University of Padova, 35131, Padua, PD, Italy
- School of Engineering, London South Bank University, London, SE1 0AA, UK
| | - J-M Graïc
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020, Legnaro, PD, Italy.
| | - R Carvajal-Schiaffino
- Department of Mathematics and Computer Science, University of Santiago de Chile, Santiago, Chile
| | - B Cozzi
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020, Legnaro, PD, Italy
| | - A Peruffo
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020, Legnaro, PD, Italy
| |
Collapse
|
17
|
Vaaga CE, Brown ST, Raman IM. Cerebellar modulation of synaptic input to freezing-related neurons in the periaqueductal gray. eLife 2020; 9:e54302. [PMID: 32207681 PMCID: PMC7124251 DOI: 10.7554/elife.54302] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/24/2020] [Indexed: 01/23/2023] Open
Abstract
Innate defensive behaviors, such as freezing, are adaptive for avoiding predation. Freezing-related midbrain regions project to the cerebellum, which is known to regulate rapid sensorimotor integration, raising the question of cerebellar contributions to freezing. Here, we find that neurons of the mouse medial (fastigial) cerebellar nuclei (mCbN), which fire spontaneously with wide dynamic ranges, send glutamatergic projections to the ventrolateral periaqueductal gray (vlPAG), which contains diverse cell types. In freely moving mice, optogenetically stimulating glutamatergic vlPAG neurons that express Chx10 reliably induces freezing. In vlPAG slices, mCbN terminals excite ~20% of neurons positive for Chx10 or GAD2 and ~70% of dopaminergic TH-positive neurons. Stimulating either mCbN afferents or TH neurons augments IPSCs and suppresses EPSCs in Chx10 neurons by activating postsynaptic D2 receptors. The results suggest that mCbN activity regulates dopaminergic modulation of the vlPAG, favoring inhibition of Chx10 neurons. Suppression of cerebellar output may therefore facilitate freezing.
Collapse
Affiliation(s)
| | - Spencer T Brown
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
| | - Indira M Raman
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
| |
Collapse
|
18
|
Asher M, Rosa JG, Rainwater O, Duvick L, Bennyworth M, Lai RY, Kuo SH, Cvetanovic M. Cerebellar contribution to the cognitive alterations in SCA1: evidence from mouse models. Hum Mol Genet 2020; 29:117-131. [PMID: 31696233 PMCID: PMC8216071 DOI: 10.1093/hmg/ddz265] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 09/30/2019] [Accepted: 10/23/2019] [Indexed: 11/13/2022] Open
Abstract
Spinocerebellar ataxia type 1 (SCA1) is a fatal neurodegenerative disease caused by abnormal expansion of glutamine (Q) encoding CAG repeats in the gene Ataxin-1 (ATXN1). Although motor and balance deficits are the core symptoms of SCA1, cognitive decline is also commonly observed in patients. While mutant ATXN1 is expressed throughout the brain, pathological findings reveal severe atrophy of cerebellar cortex in SCA1 patients. The cerebellum has recently been implicated in diverse cognitive functions, yet to what extent cerebellar neurodegeneration contributes to cognitive alterations in SCA1 remains poorly understood. Much of our understanding of the mechanisms underlying pathogenesis of motor symptoms in SCA1 comes from mouse models. Reasoning that mouse models could similarly offer important insights into the mechanisms of cognitive alterations in SCA1, we tested cognition in several mouse lines using Barnes maze and fear conditioning. We confirmed cognitive deficits in Atxn1154Q/2Q knock-in mice with brain-wide expression of mutant ATXN1 and in ATXN1 null mice. We found that shorter polyQ length and haploinsufficiency of ATXN1 do not cause significant cognitive deficits. Finally, ATXN1[82Q ] transgenic mice-with cerebellum limited expression of mutant ATXN1-demonstrated milder impairment in most aspects of cognition compared to Atxn1154Q/2Q mice, supporting the concept that cognitive deficits in SCA1 arise from a combination of cerebellar and extra-cerebellar dysfunctions.
Collapse
Affiliation(s)
- Melissa Asher
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Juao-Guilherme Rosa
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Orion Rainwater
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Lisa Duvick
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michael Bennyworth
- Mouse Behavior Core, University of Minnesota, Minneapolis, 55455 NY 10032-3784, USA
| | - Ruo-Yah Lai
- Department of Neurology, Columbia University, New York, NY 10032-3784, USA
| | - CRC-SCA
- Clinical Research Consortium for Spinocerebellar Ataxia (CRC-SCA)#
| | - Sheng-Han Kuo
- Department of Neurology, Columbia University, New York, NY 10032-3784, USA
| | - Marija Cvetanovic
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
- Mouse Behavior Core, University of Minnesota, Minneapolis, 55455 NY 10032-3784, USA
| |
Collapse
|
19
|
Mossa A, Manzini MC. Molecular causes of sex-specific deficits in rodent models of neurodevelopmental disorders. J Neurosci Res 2019; 99:37-56. [PMID: 31872500 PMCID: PMC7754327 DOI: 10.1002/jnr.24577] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 11/02/2019] [Accepted: 12/09/2019] [Indexed: 12/12/2022]
Abstract
Neurodevelopmental disorders (NDDs) such as intellectual disability and autism spectrum disorder consistently show a male bias in prevalence, but it remains unclear why males and females are affected with different frequency. While many behavioral studies of transgenic NDD models have focused only on males, the requirement by the National Institutes of Health to consider sex as a biological variable has promoted the comparison of male and female performance in wild-type and mutant animals. Here, we review examples of rodent models of NDDs in which sex-specific deficits were identified in molecular, physiological, and/or behavioral responses, showing sex differences in susceptibility to disruption of genes mutated in NDDs. Haploinsufficiency in genes involved in mechanisms such as synaptic function (GABRB3 and NRXN1), chromatin remodeling (CHD8, EMHT1, and ADNP), and intracellular signaling (CC2D1A and ERK1) lead to more severe behavioral outcomes in males. However, in the absence of behavioral deficits, females can still present with cellular and electrophysiological changes that could be due to compensatory mechanisms or differential allocation of molecular and cellular functions in the two sexes. By contrasting these findings with mouse models where females are more severely affected (MTHFR and AMBRA1), we propose a framework to approach the study of sex-specific deficits possibly leading to sex bias in NDDs.
Collapse
Affiliation(s)
- Adele Mossa
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - M Chiara Manzini
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.,Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
20
|
Stay TL, Miterko LN, Arancillo M, Lin T, Sillitoe RV. In vivo cerebellar circuit function is disrupted in an mdx mouse model of Duchenne muscular dystrophy. Dis Model Mech 2019; 13:dmm040840. [PMID: 31704708 PMCID: PMC6906634 DOI: 10.1242/dmm.040840] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 10/30/2019] [Indexed: 12/20/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a debilitating and ultimately lethal disease involving progressive muscle degeneration and neurological dysfunction. DMD is caused by mutations in the dystrophin gene, which result in extremely low or total loss of dystrophin protein expression. In the brain, dystrophin is heavily localized to cerebellar Purkinje cells, which control motor and non-motor functions. In vitro experiments in mouse Purkinje cells revealed that loss of dystrophin leads to low firing rates and high spiking variability. However, it is still unclear how the loss of dystrophin affects cerebellar function in the intact brain. Here, we used in vivo electrophysiology to record Purkinje cells and cerebellar nuclear neurons in awake and anesthetized female mdx (also known as Dmd) mice. Purkinje cell simple spike firing rate is significantly lower in mdx mice compared to controls. Although simple spike firing regularity is not affected, complex spike regularity is increased in mdx mutants. Mean firing rate in cerebellar nuclear neurons is not altered in mdx mice, but their local firing pattern is irregular. Based on the relatively well-preserved cytoarchitecture in the mdx cerebellum, our data suggest that faulty signals across the circuit between Purkinje cells and cerebellar nuclei drive the abnormal firing activity. The in vivo requirements of dystrophin during cerebellar circuit communication could help explain the motor and cognitive anomalies seen in individuals with DMD.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Trace L Stay
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX 77030, USA
| | - Lauren N Miterko
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX 77030, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Marife Arancillo
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX 77030, USA
| | - Tao Lin
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX 77030, USA
| | - Roy V Sillitoe
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute of Texas Children's Hospital, 1250 Moursund Street, Suite 1325, Houston, TX 77030, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
21
|
Castagna C, Merighi A, Lossi L. Decreased Expression of Synaptophysin 1 (SYP1 Major Synaptic Vesicle Protein p38) and Contactin 6 (CNTN6/NB3) in the Cerebellar Vermis of reln Haplodeficient Mice. Cell Mol Neurobiol 2019; 39:833-856. [PMID: 31098770 DOI: 10.1007/s10571-019-00683-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 05/02/2019] [Indexed: 01/17/2023]
Abstract
Reeler heterozygous mice (reln+/-) are seemingly normal but haplodeficient in reln, a gene implicated in autism. Structural/neurochemical alterations in the reln+/- brain are subtle and difficult to demonstrate. Therefore, the usefulness of these mice in translational research is still debated. As evidence implicated several synapse-related genes in autism and the cerebellar vermis is structurally altered in the condition, we have investigated the expression of synaptophysin 1 (SYP1) and contactin 6 (CNTN6) within the vermis of reln+/- mice. Semi-thin plastic sections of the vermis from adult mice of both sexes and different genotypes (reln+/- and reln+/+) were processed with an indirect immunofluorescence protocol. Immunofluorescence was quantified on binary images and statistically analyzed. Reln+/- males displayed a statistically significant reduction of 11.89% in the expression of SYP1 compared to sex-matched wild-type animals, whereas no differences were observed between reln+/+ and reln+/- females. In reln+/- male mice, reductions were particularly evident in the molecular layer: 10.23% less SYP1 than reln+/+ males and 5.84% < reln+/+ females. In reln+/- females, decrease was 9.84% versus reln+/+ males and 5.43% versus reln+/+ females. Both reln+/- males and females showed a stronger decrease in CNTN6 expression throughout all the three cortical layers of the vermis: 17-23% in the granular layer, 24-26% in the Purkinje cell layer, and 9-14% in the molecular layer. Altogether, decrease of vermian SYP1 and CNTN6 in reln+/- mice displayed patterns compatible with the structural modifications of the autistic cerebellum. Therefore, these mice may be a good model in translational studies.
Collapse
Affiliation(s)
- Claudia Castagna
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, I-10095, Turin, Grugliasco (TO), Italy.
| | - Adalberto Merighi
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, I-10095, Turin, Grugliasco (TO), Italy
| | - Laura Lossi
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, I-10095, Turin, Grugliasco (TO), Italy
| |
Collapse
|
22
|
Giatti S, Diviccaro S, Garcia-Segura LM, Melcangi RC. Sex differences in the brain expression of steroidogenic molecules under basal conditions and after gonadectomy. J Neuroendocrinol 2019; 31:e12736. [PMID: 31102564 DOI: 10.1111/jne.12736] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 05/09/2019] [Accepted: 05/15/2019] [Indexed: 01/19/2023]
Abstract
The brain is a steroidogenic tissue. It expresses key molecules involved in the synthesis and metabolism of neuroactive steroids, such as steroidogenic acute regulatory protein (StAR), translocator protein 18 kDa (TSPO), cytochrome P450 cholesterol side-chain cleavage enzyme (P450scc), 3β-hydroxysteroid dehydrogenases (3β-HSD), 5α-reductases (5α-R) and 3α-hydroxysteroid oxidoreductases (3α-HSOR). Previous studies have shown that the levels of brain steroids are different in male and female rats under basal conditions and after gonadectomy. In the present study, we assessed gene expression of key neurosteroidogenic molecules in the cerebral cortex and cerebellum of gonadally intact and gonadectomised adult male and female rats. In the cerebellum, the basal mRNA levels of StAR and 3α-HSOR were significantly higher in females than in males. By contrast, the mRNA levels of TSPO and 5α-R were significantly higher in males. In the cerebral cortex, all neurosteroidogenic molecules analysed showed similar mRNA levels in males and females. Gonadectomy increased the expression of 5α-R in the brain of both sexes, although it affected the brain expression of StAR, TSPO, P450scc and 3α-HSOR in females only and with regional differences. Although protein levels were not investigated in the present study, our findings indicate that mRNA expression of steroidogenic molecules in the adult rat brain is sexually dimorphic and presents regional specificity, both under basal conditions and after gonadectomy. Thus, local steroidogenesis may contribute to the reported sex and regional differences in the levels of brain neuroactive steroids and may be involved in the generation of sex differences in the adult brain function.
Collapse
Affiliation(s)
- Silvia Giatti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Silvia Diviccaro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Luis Miguel Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Roberto Cosimo Melcangi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
23
|
Hyperexcitability and Hyperplasticity Disrupt Cerebellar Signal Transfer in the IB2 KO Mouse Model of Autism. J Neurosci 2019; 39:2383-2397. [PMID: 30696733 DOI: 10.1523/jneurosci.1985-18.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 12/22/2018] [Accepted: 01/08/2019] [Indexed: 12/25/2022] Open
Abstract
Autism spectrum disorders (ASDs) are pervasive neurodevelopmental conditions that often involve mutations affecting synaptic mechanisms. Recently, the involvement of cerebellum in ASDs has been suggested, but the underlying functional alterations remained obscure. We investigated single-neuron and microcircuit properties in IB2 (Islet Brain-2) KO mice of either sex. The IB2 gene (chr22q13.3 terminal region) deletion occurs in virtually all cases of Phelan-McDermid syndrome, causing autistic symptoms and a severe delay in motor skill acquisition. IB2 KO granule cells showed a larger NMDA receptor-mediated current and enhanced intrinsic excitability, raising the excitatory/inhibitory balance. Furthermore, the spatial organization of granular layer responses to mossy fibers shifted from a "Mexican hat" to a "stovepipe hat" profile, with stronger excitation in the core and weaker inhibition in the surround. Finally, the size and extension of long-term synaptic plasticity were remarkably increased. These results show for the first time that hyperexcitability and hyperplasticity disrupt signal transfer in the granular layer of IB2 KO mice, supporting cerebellar involvement in the pathogenesis of ASD.SIGNIFICANCE STATEMENT This article shows for the first time a complex set of alterations in the cerebellum granular layer of a mouse model [IB2 (Islet Brain-2) KO] of autism spectrum disorders. The IB2 KO in mice mimics the deletion of the corresponding gene in the Phelan-McDermid syndrome in humans. The changes reported here are centered on NMDA receptor hyperactivity, hyperplasticity, and hyperexcitability. These, in turn, increase the excitatory/inhibitory balance and alter the shape of center/surround structures that emerge in the granular layer in response to mossy fiber activity. These results support recent theories suggesting the involvement of cerebellum in autism spectrum disorders.
Collapse
|
24
|
Bachmann SO, Sledziowska M, Cross E, Kalbassi S, Waldron S, Chen F, Ranson A, Baudouin SJ. Behavioral training rescues motor deficits in Cyfip1 haploinsufficiency mouse model of autism spectrum disorders. Transl Psychiatry 2019; 9:29. [PMID: 30664619 PMCID: PMC6341103 DOI: 10.1038/s41398-018-0338-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/15/2018] [Accepted: 11/25/2018] [Indexed: 12/27/2022] Open
Abstract
Deletions in the 15q11.2 region of the human genome are associated with neurobehavioral deficits, and motor development delay, as well as in some cases, symptoms of autism or schizophrenia. The cytoplasmic FMRP-interacting protein 1 (CYFIP1) is one of the four genes contained within this locus and has been associated with other genetic forms of autism spectrum disorders (ASD). In mice, Cyfip1 haploinsufficiency leads to alteration of dendritic spine morphology and defects in synaptic plasticity, two pathophysiological hallmarks of mouse models of ASD. At the behavioral level, however, Cyfip1 haploinsufficiency leads to minor phenotypes, not directly relevant for 15q11.2 deletion syndrome or ASD. A fundamental question is whether neuronal phenotypes caused by the mutation of Cyfip1 are relevant for the human condition. Here, we describe a synaptic cluster of ASD-associated proteins centered on CYFIP1 and the adhesion protein Neuroligin-3. Cyfip1 haploinsufficiency in mice led to decreased dendritic spine density and stability associated with social behavior and motor learning phenotypes. Behavioral training early in development resulted in alleviating the motor learning deficits caused by Cyfip1 haploinsufficiency. Altogether, these data provide new insight into the neuronal and behavioral phenotypes caused by Cyfip1 mutation and proof-of-concept for the development of a behavioral therapy to treat phenotypes associated with 15q11.2 syndromes and ASD.
Collapse
Affiliation(s)
- Sven O. Bachmann
- 0000 0001 0807 5670grid.5600.3School of Biosciences, Cardiff University, Cardiff, CF10 3AX Wales UK
| | - Monika Sledziowska
- 0000 0001 0807 5670grid.5600.3School of Biosciences, Cardiff University, Cardiff, CF10 3AX Wales UK
| | - Ellen Cross
- 0000 0001 0807 5670grid.5600.3School of Biosciences, Cardiff University, Cardiff, CF10 3AX Wales UK
| | - Shireene Kalbassi
- 0000 0001 0807 5670grid.5600.3School of Biosciences, Cardiff University, Cardiff, CF10 3AX Wales UK
| | - Sophie Waldron
- 0000 0001 0807 5670grid.5600.3School of Biosciences, Cardiff University, Cardiff, CF10 3AX Wales UK
| | - Fangli Chen
- 0000 0001 0807 5670grid.5600.3Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff, CF24 4HQ UK
| | - Adam Ranson
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK. .,Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Barcelona, Spain.
| | - Stéphane J. Baudouin
- 0000 0001 0807 5670grid.5600.3School of Biosciences, Cardiff University, Cardiff, CF10 3AX Wales UK
| |
Collapse
|
25
|
Roux S, Lohof A, Ben-Ari Y, Poulain B, Bossu JL. Maturation of GABAergic Transmission in Cerebellar Purkinje Cells Is Sex Dependent and Altered in the Valproate Model of Autism. Front Cell Neurosci 2018; 12:232. [PMID: 30104962 PMCID: PMC6077203 DOI: 10.3389/fncel.2018.00232] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/16/2018] [Indexed: 12/20/2022] Open
Abstract
Brain development is accompanied by a shift in gamma-aminobutyric acid (GABA) response from depolarizing-excitatory to hyperpolarizing-inhibitory, due to a reduction of intracellular chloride concentration. This sequence is delayed in Autism Spectrum Disorders (ASD). We now report a similar alteration of this shift in the cerebellum, a structure implicated in ASD. Using single GABAA receptor channel recordings in cerebellar Purkinje cells (PCs), we found two conductance levels (18 and 10 pS), the former being dominant in newborns and the latter in young-adults. This conductance shift and the depolarizing/excitatory to hyperpolarizing/inhibitory GABA shift occurred 4 days later in females than males. Our data support a sex-dependent developmental shift of GABA conductance and chloride gradient, leading to different developmental timing in males and females. Because these developmental sequences are altered in ASD, this study further stresses the importance of developmental timing in pathological neurodevelopment.
Collapse
Affiliation(s)
- Sébastien Roux
- Institut des Neurosciences Cellulaires et Intégratives (INCI)-CNRS, UPR 3212, Strasbourg, France
| | - Ann Lohof
- Sorbonne Université, CNRS UMR 8256, Biological Adaptation and Ageing, Paris, France
| | - Yehezkel Ben-Ari
- Neurochlore, Ben-Ari Institute of Neuroarcheology, Campus Scientifique de Luminy, Aix Marseille Université, Marseille, France
| | - Bernard Poulain
- Institut des Neurosciences Cellulaires et Intégratives (INCI)-CNRS, UPR 3212, Strasbourg, France
| | - Jean-Louis Bossu
- Institut des Neurosciences Cellulaires et Intégratives (INCI)-CNRS, UPR 3212, Strasbourg, France
| |
Collapse
|
26
|
Perineuronal Nets in the Deep Cerebellar Nuclei Regulate GABAergic Transmission and Delay Eyeblink Conditioning. J Neurosci 2018; 38:6130-6144. [PMID: 29858484 DOI: 10.1523/jneurosci.3238-17.2018] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 04/28/2018] [Accepted: 05/28/2018] [Indexed: 11/21/2022] Open
Abstract
Perineuronal nets (PNNs), composed mainly of chondroitin sulfate proteoglycans, are the extracellular matrix that surrounds cell bodies, proximal dendrites, and axon initial segments of adult CNS neurons. PNNs are known to regulate neuronal plasticity, although their physiological roles in cerebellar functions have yet to be elucidated. Here, we investigated the contribution of PNNs to GABAergic transmission from cerebellar Purkinje cells (PCs) to large glutamatergic neurons in the deep cerebellar nuclei (DCN) in male mice by recording IPSCs from cerebellar slices, in which PNNs were depleted with chondroitinase ABC (ChABC). We found that PNN depletion increased the amplitude of evoked IPSCs and enhanced the paired-pulse depression. ChABC treatment also facilitated spontaneous IPSCs and increased the miniature IPSC frequency without changing not only the amplitude but also the density of PC terminals, suggesting that PNN depletion enhances presynaptic GABA release. We also demonstrated that the enhanced GABAergic transmission facilitated rebound firing in large glutamatergic DCN neurons, which is expected to result in the efficient induction of synaptic plasticity at synapses onto DCN neurons. Furthermore, we tested whether PNN depletion affects cerebellar motor learning. Mice having received the enzyme into the interpositus nuclei, which are responsible for delay eyeblink conditioning, exhibited the conditioned response at a significantly higher rate than control mice. Therefore, our results suggest that PNNs of the DCN suppress GABAergic transmission between PCs and large glutamatergic DCN neurons and restrict synaptic plasticity associated with motor learning in the adult cerebellum.SIGNIFICANCE STATEMENT Perineuronal nets (PNNs) are one of the extracellular matrices of adult CNS neurons and implicated in regulating various brain functions. Here we found that enzymatic PNN depletion in the mouse deep cerebellar nuclei (DCN) reduced the paired-pulse ratio of IPSCs and increased the miniature IPSC frequency without changing the amplitude, suggesting that PNN depletion enhances GABA release from the presynaptic Purkinje cell (PC) terminals. Mice having received the enzyme in the interpositus nuclei exhibited a higher conditioned response rate in delay eyeblink conditioning than control mice. These results suggest that PNNs regulate presynaptic functions of PC terminals in the DCN and functional plasticity of synapses on DCN neurons, which influences the flexibility of adult cerebellar functions.
Collapse
|
27
|
Sarnaik R, Raman IM. Control of voluntary and optogenetically perturbed locomotion by spike rate and timing of neurons of the mouse cerebellar nuclei. eLife 2018; 7:29546. [PMID: 29659351 PMCID: PMC5902160 DOI: 10.7554/elife.29546] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 03/30/2018] [Indexed: 11/13/2022] Open
Abstract
Neurons of the cerebellar nuclei (CbN), which generate cerebellar output, are inhibited by Purkinje cells. With extracellular recordings during voluntary locomotion in head-fixed mice, we tested how the rate and coherence of inhibition influence CbN cell firing and well-practiced movements. Firing rates of Purkinje and CbN cells were modulated systematically through the stride cycle (~200–300 ms). Optogenetically stimulating ChR2-expressing Purkinje cells with light steps or trains evoked either asynchronous or synchronous inhibition of CbN cells. Steps slowed CbN firing. Trains suppressed CbN cell firing less effectively, but consistently altered millisecond-scale spike timing. Steps or trains that perturbed stride-related modulation of CbN cell firing rates correlated well with irregularities of movement, suggesting that ongoing locomotion is sensitive to alterations in modulated CbN cell firing. Unperturbed locomotion continued more often during trains than steps, however, suggesting that stride-related modulation of CbN spiking is less readily disrupted by synchronous than asynchronous inhibition.
Collapse
Affiliation(s)
- Rashmi Sarnaik
- Department of Neurobiology, Northwestern University, Evanston, United States
| | - Indira M Raman
- Department of Neurobiology, Northwestern University, Evanston, United States
| |
Collapse
|
28
|
Bushart DD, Chopra R, Singh V, Murphy GG, Wulff H, Shakkottai VG. Targeting potassium channels to treat cerebellar ataxia. Ann Clin Transl Neurol 2018; 5:297-314. [PMID: 29560375 PMCID: PMC5846455 DOI: 10.1002/acn3.527] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/14/2017] [Accepted: 12/18/2017] [Indexed: 12/22/2022] Open
Abstract
Objective Purkinje neuron dysfunction is associated with cerebellar ataxia. In a mouse model of spinocerebellar ataxia type 1 (SCA1), reduced potassium channel function contributes to altered membrane excitability resulting in impaired Purkinje neuron spiking. We sought to determine the relationship between altered membrane excitability and motor dysfunction in SCA1 mice. Methods Patch-clamp recordings in acute cerebellar slices and motor phenotype testing were used to identify pharmacologic agents which improve Purkinje neuron physiology and motor performance in SCA1 mice. Additionally, we retrospectively reviewed records of patients with SCA1 and other autosomal-dominant SCAs with prominent Purkinje neuron involvement to determine whether currently approved potassium channel activators were tolerated. Results Activating calcium-activated and subthreshold-activated potassium channels improved Purkinje neuron spiking impairment in SCA1 mice (P < 0.05). Additionally, dendritic hyperexcitability was improved by activating subthreshold-activated potassium channels but not calcium-activated potassium channels (P < 0.01). Improving spiking and dendritic hyperexcitability through a combination of chlorzoxazone and baclofen produced sustained improvements in motor dysfunction in SCA1 mice (P < 0.01). Retrospective review of SCA patient records suggests that co-treatment with chlorzoxazone and baclofen is tolerated. Interpretation Targeting both altered spiking and dendritic membrane excitability is associated with sustained improvements in motor performance in SCA1 mice, while targeting altered spiking alone produces only short-term improvements in motor dysfunction. Potassium channel activators currently in clinical use are well tolerated and may provide benefit in SCA patients. Future clinical trials with potassium channel activators are warranted in cerebellar ataxia.
Collapse
Affiliation(s)
- David D Bushart
- Department of Molecular & Integrative Physiology University of Michigan Ann Arbor Michigan
| | - Ravi Chopra
- Department of Neurology University of Michigan Ann Arbor Michigan
| | - Vikrant Singh
- Department of Pharmacology University of California Davis California
| | - Geoffrey G Murphy
- Department of Molecular & Integrative Physiology University of Michigan Ann Arbor Michigan.,Molecular & Behavioral Neuroscience Institute University of Michigan Ann Arbor Michigan
| | - Heike Wulff
- Department of Pharmacology University of California Davis California
| | - Vikram G Shakkottai
- Department of Molecular & Integrative Physiology University of Michigan Ann Arbor Michigan.,Department of Neurology University of Michigan Ann Arbor Michigan
| |
Collapse
|
29
|
Takahashi Y, Wu J, Suzuki K, Martinez-Redondo P, Li M, Liao HK, Wu MZ, Hernández-Benítez R, Hishida T, Shokhirev MN, Esteban CR, Sancho-Martinez I, Belmonte JCI. Integration of CpG-free DNA induces de novo methylation of CpG islands in pluripotent stem cells. Science 2018; 356:503-508. [PMID: 28473583 DOI: 10.1126/science.aag3260] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 12/12/2016] [Accepted: 04/06/2017] [Indexed: 12/31/2022]
Abstract
CpG islands (CGIs) are primarily promoter-associated genomic regions and are mostly unmethylated within highly methylated mammalian genomes. The mechanisms by which CGIs are protected from de novo methylation remain elusive. Here we show that insertion of CpG-free DNA into targeted CGIs induces de novo methylation of the entire CGI in human pluripotent stem cells (PSCs). The methylation status is stably maintained even after CpG-free DNA removal, extensive passaging, and differentiation. By targeting the DNA mismatch repair gene MLH1 CGI, we could generate a PSC model of a cancer-related epimutation. Furthermore, we successfully corrected aberrant imprinting in induced PSCs derived from an Angelman syndrome patient. Our results provide insights into how CpG-free DNA induces de novo CGI methylation and broaden the application of targeted epigenome editing for a better understanding of human development and disease.
Collapse
Affiliation(s)
- Yuta Takahashi
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.,Life Science Center, Tsukuba Advanced Research Alliance, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8577, Japan
| | - Jun Wu
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.,Universidad Católica San Antonio de Murcia (UCAM) Campus de los Jerónimos, N° 135 Guadalupe 30107, Murcia, Spain
| | - Keiichiro Suzuki
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Paloma Martinez-Redondo
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Mo Li
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.,King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Hsin-Kai Liao
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Min-Zu Wu
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.,Universidad Católica San Antonio de Murcia (UCAM) Campus de los Jerónimos, N° 135 Guadalupe 30107, Murcia, Spain
| | - Reyna Hernández-Benítez
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.,King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Tomoaki Hishida
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Maxim Nikolaievich Shokhirev
- Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Concepcion Rodriguez Esteban
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Ignacio Sancho-Martinez
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Juan Carlos Izpisua Belmonte
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
30
|
Najac M, Raman IM. Synaptic excitation by climbing fibre collaterals in the cerebellar nuclei of juvenile and adult mice. J Physiol 2017; 595:6703-6718. [PMID: 28795396 PMCID: PMC5663862 DOI: 10.1113/jp274598] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 08/08/2017] [Indexed: 01/24/2023] Open
Abstract
KEY POINTS The inferior olive sends instructive motor signals to the cerebellum via the climbing fibre projection, which sends collaterals directly to large premotor neurons of the mouse cerebellar nuclei (CbN cells). Optogenetic activation of inferior olivary axons in vitro evokes EPSCs in CbN cells of several hundred pA to more than 1 nA. The inputs are three-fold larger at younger ages, 12 to 14 days old, than at 2 months old, suggesting a strong functional role for this pathway earlier in development. The EPSCs are multipeaked, owing to burst firing in several olivary afferents that fire asynchronously. The convergence of climbing fibre collaterals onto CbN cells decreases from ∼40 to ∼8, which is consistent with the formation of closed-loop circuits in which each CbN neuron receives input from 4-7 collaterals from inferior olivary neurons as well as from all 30-50 Purkinje cells that are innervated by those olivary neurons. ABSTRACT The inferior olive conveys instructive signals to the cerebellum that drive sensorimotor learning. Inferior olivary neurons transmit their signals via climbing fibres, which powerfully excite Purkinje cells, evoking complex spikes and depressing parallel fibre synapses. Additionally, however, these climbing fibres send collaterals to the cerebellar nuclei (CbN). In vivo and in vitro data suggest that climbing fibre collateral excitation is weak in adult mice, raising the question of whether the primary role of this pathway may be developmental. We therefore examined climbing fibre collateral input to large premotor CbN cells over development by virally expressing channelrhodopsin in the inferior olive. In acute cerebellar slices from postnatal day (P)12-14 mice, light-evoked EPSCs were large (> 1 nA at -70 mV). The amplitude of these EPSCs decreased over development, reaching a plateau of ∼350 pA at P20-60. Trains of EPSCs (5 Hz) depressed strongly throughout development, whereas convergence estimates indicated that the total number of functional afferents decreased with age. EPSC waveforms consisted of multiple peaks, probably resulting from action potential bursts in single collaterals and variable times to spike threshold in converging afferents. Activating climbing fibre collaterals evoked well-timed increases in firing probability in CbN neurons, especially in younger mice. The initially strong input, followed by the decrement in synaptic strength coinciding with the pruning of climbing fibres in the cerebellar cortex, implicates the climbing fibre collateral pathway in early postnatal development. Additionally, the persistence of substantial synaptic input at least to P60 suggests that this pathway may function in cerebellar processing into adulthood.
Collapse
Affiliation(s)
- Marion Najac
- Department of NeurobiologyNorthwestern UniversityEvanstonILUSA
| | - Indira M. Raman
- Department of NeurobiologyNorthwestern UniversityEvanstonILUSA
| |
Collapse
|
31
|
Considering Sex as a Biological Variable Will Be Valuable for Neuroscience Research. J Neurosci 2017; 36:11817-11822. [PMID: 27881768 DOI: 10.1523/jneurosci.1390-16.2016] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 08/08/2016] [Accepted: 08/16/2016] [Indexed: 01/09/2023] Open
Abstract
The recently implemented National Institutes of Health policy requiring that grant applicants consider sex as a biological variable in the design of basic and preclinical animal research studies has prompted considerable discussion within the neuroscience community. Here, we present reasons to be optimistic that this new policy will be valuable for neuroscience, and we suggest some ways for neuroscientists to think about incorporating sex as a variable in their research.
Collapse
|
32
|
Wu Y, Raman IM. Facilitation of mossy fibre-driven spiking in the cerebellar nuclei by the synchrony of inhibition. J Physiol 2017; 595:5245-5264. [PMID: 28513836 PMCID: PMC5538193 DOI: 10.1113/jp274321] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 05/11/2017] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Large premotor neurons of the cerebellar nuclei (CbN cells) integrate synaptic inhibition from Purkinje neurons and synaptic excitation from mossy fibres to generate cerebellar output. We find that mossy fibre inputs to CbN cells generate unitary AMPA receptor EPSCs of ∼1 nS that decay in ∼1 ms and mildly voltage-dependent NMDA receptor EPSCs of ∼0.6 nS that decay in ∼7 ms. A few hundred mossy fibres active at a few tens of spikes s-1 must converge on CbN cells to generate physiological CbN spike rates (∼60 spikes s-1 ) during convergent inhibition from spontaneously active Purkinje cells. Dynamic clamp studies in cerebellar slices from weanling mice demonstrate that synaptic excitation from mossy fibres becomes more effective at increasing the rate of CbN cell spiking when the coherence (synchrony) of convergent inhibition is increased. ABSTRACT Large projection neurons of the cerebellar nuclei (CbN cells), whose activity generates movement, are inhibited by Purkinje cells and excited by mossy fibres. The high convergence, firing rates and strength of Purkinje inputs predict powerful suppression of CbN cell spiking, raising the question of what activity patterns favour excitation over inhibition. Recording from CbN cells at near-physiological temperatures in cerebellar slices from weanling mice, we measured the amplitude, kinetics, voltage dependence and short-term plasticity of mossy fibre-mediated EPSCs. Unitary EPSCs were small and brief (AMPA receptor, ∼1 nS, ∼1 ms; NMDA receptor, ∼0.6 nS, ∼7 ms) and depressed moderately. Using these experimentally measured parameters, we applied combinations of excitation and inhibition to CbN cells with dynamic clamp. Because Purkinje cells can fire coincident simple spikes during cerebellar behaviours, we varied the proportion (0-20 of 40) and precision (0-4 ms jitter) of synchrony of inhibitory inputs, along with the rates (0-100 spikes s-1 ) and number (0-800) of excitatory inputs. Even with inhibition constant, when inhibitory synchrony was higher, excitation increased CbN cell firing rates more effectively. Partial inhibitory synchrony also dictated CbN cell spike timing, even with physiological rates of excitation. These effects were present with ≥10 inhibitory inputs active within 2-4 ms of each other. Conversely, spiking was most effectively suppressed when inhibition was maximally asynchronous. Thus, the rate and relative timing of Purkinje-mediated inhibition set the rate and timing of cerebellar output. The results suggest that increased coherence of Purkinje cell activity can facilitate mossy fibre-driven spiking by CbN cells, in turn driving movements.
Collapse
Affiliation(s)
- Yeechan Wu
- Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL, 60208, USA
- Department of Neurobiology, Northwestern University, Evanston, IL, 60208, USA
| | - Indira M Raman
- Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL, 60208, USA
- Department of Neurobiology, Northwestern University, Evanston, IL, 60208, USA
| |
Collapse
|
33
|
Chahrour M, O'Roak BJ, Santini E, Samaco RC, Kleiman RJ, Manzini MC. Current Perspectives in Autism Spectrum Disorder: From Genes to Therapy. J Neurosci 2016; 36:11402-11410. [PMID: 27911742 PMCID: PMC5125207 DOI: 10.1523/jneurosci.2335-16.2016] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 08/23/2016] [Accepted: 08/25/2016] [Indexed: 12/17/2022] Open
Abstract
Autism spectrum disorder (ASD) is a constellation of neurodevelopmental presentations with high heritability and both phenotypic and genetic heterogeneity. To date, mutations in hundreds of genes have been associated to varying degrees with increased ASD risk. A better understanding of the functions of these genes and whether they fit together in functional groups or impact similar neuronal circuits is needed to develop rational treatment strategies. We will review current areas of emphasis in ASD research, starting from human genetics and exploring how mouse models of human mutations have helped identify specific molecular pathways (protein synthesis and degradation, chromatin remodeling, intracellular signaling), which are linked to alterations in circuit function and cognitive/social behavior. We will conclude by discussing how we can leverage the findings on molecular and cellular alterations found in ASD to develop therapies for neurodevelopmental disorders.
Collapse
Affiliation(s)
- Maria Chahrour
- Eugene McDermott Center for Human Growth and Development, Departments of Neuroscience and Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas 75390,
| | - Brian J O'Roak
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, Oregon 97239
| | - Emanuela Santini
- Center for Neural Science, New York University, New York, New York 10003
| | - Rodney C Samaco
- Department of Molecular and Human Genetics, Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030
| | - Robin J Kleiman
- Translational Neuroscience Center, F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, and
| | - M Chiara Manzini
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037
| |
Collapse
|