Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters

Therapeutic Methods and Therapies TCIM
Database
Language
Publication year range
1.
J Neurosci ; 41(42): 8801-8814, 2021 10 20.
Article in English | MEDLINE | ID: mdl-34475199

ABSTRACT

Angelman syndrome (AS) is a rare genetic neurodevelopmental disorder characterized by intellectual disabilities, motor and balance deficits, impaired communication, and a happy, excitable demeanor with frequent laughter. We sought to elucidate a preclinical outcome measure in male and female rats that addressed communication abnormalities of AS and other neurodevelopmental disorders in which communication is atypical and/or lack of speech is a core feature. We discovered, and herein report for the first time, excessive laughter-like 50 kHz ultrasonic emissions in the Ube3amat-/pat+ rat model of AS, which suggests an excitable, playful demeanor and elevated positive affect, similar to the demeanor of individuals with AS. Also in line with the AS phenotype, Ube3amat-/pat+ rats demonstrated aberrant social interactions with a novel partner, distinctive gait abnormalities, impaired cognition, an underlying LTP deficit, and profound reductions in brain volume. These unique, robust phenotypes provide advantages compared with currently available mouse models and will be highly valuable as outcome measures in the evaluation of therapies for AS.SIGNIFICANCE STATEMENT Angelman syndrome (AS) is a severe neurogenetic disorder for which there is no cure, despite decades of research using mouse models. This study used a recently developed rat model of AS to delineate disease-relevant outcome measures to facilitate therapeutic development. We found the rat to be a strong model of AS, offering several advantages over mouse models by exhibiting numerous AS-relevant phenotypes, including overabundant laughter-like vocalizations, reduced hippocampal LTP, and volumetric anomalies across the brain. These findings are unconfounded by detrimental motor abilities and background strain, issues plaguing mouse models. This rat model represents an important advancement in the field of AS, and the outcome metrics reported herein will be central to the therapeutic pipeline.


Subject(s)
Angelman Syndrome/genetics , Disease Models, Animal , Laughter/physiology , Microcephaly/genetics , Ubiquitin-Protein Ligases/genetics , Vocalization, Animal/physiology , Angelman Syndrome/metabolism , Angelman Syndrome/psychology , Animals , Brain/metabolism , Female , Gene Deletion , Laughter/psychology , Male , Microcephaly/metabolism , Microcephaly/psychology , Organ Culture Techniques , Protein Biosynthesis/physiology , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Reflex, Startle/physiology , Social Behavior , Ubiquitin-Protein Ligases/deficiency
2.
Neurotoxicology ; 87: 106-119, 2021 12.
Article in English | MEDLINE | ID: mdl-34509511

ABSTRACT

Organophosphate (OP) nerve agents and pesticides are a class of neurotoxic compounds that can cause status epilepticus (SE), and death following acute high-dose exposures. While the standard of care for acute OP intoxication (atropine, oxime, and high-dose benzodiazepine) can prevent mortality, survivors of OP poisoning often experience long-term brain damage and cognitive deficits. Preclinical studies of acute OP intoxication have primarily used rat models to identify candidate medical countermeasures. However, the mouse offers the advantage of readily available knockout strains for mechanistic studies of acute and chronic consequences of OP-induced SE. Therefore, the main objective of this study was to determine whether a mouse model of acute diisopropylfluorophosphate (DFP) intoxication would produce acute and chronic neurotoxicity similar to that observed in rat models and humans following acute OP intoxication. Adult male C57BL/6J mice injected with DFP (9.5 mg/kg, s.c.) followed 1 min later with atropine sulfate (0.1 mg/kg, i.m.) and 2-pralidoxime (25 mg/kg, i.m.) developed behavioral and electrographic signs of SE within minutes that continued for at least 4 h. Acetylcholinesterase inhibition persisted for at least 3 d in the blood and 14 d in the brain of DFP mice relative to vehicle (VEH) controls. Immunohistochemical analyses revealed significant neurodegeneration and neuroinflammation in multiple brain regions at 1, 7, and 28 d post-exposure in the brains of DFP mice relative to VEH controls. Deficits in locomotor and home-cage behavior were observed in DFP mice at 28 d post-exposure. These findings demonstrate that this mouse model replicates many of the outcomes observed in rats and humans acutely intoxicated with OPs, suggesting the feasibility of using this model for mechanistic studies and therapeutic screening.


Subject(s)
Brain/pathology , Isoflurophate/toxicity , Status Epilepticus/chemically induced , Acetylcholinesterase/metabolism , Animals , Brain/drug effects , Brain/enzymology , Cholinesterase Inhibitors/pharmacology , Disease Models, Animal , Electroencephalography , Female , Male , Mice , Mice, Inbred C57BL , Nesting Behavior/drug effects , Neuroinflammatory Diseases/chemically induced , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/psychology , Open Field Test , Status Epilepticus/pathology , Status Epilepticus/psychology
3.
Hum Mol Genet ; 28(22): 3842-3852, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31625566

ABSTRACT

Ubiquitin E3 ligase 3A (UBE3A) encodes an E3 ubiquitin ligase whose loss from the maternal allele causes the neurodevelopmental disorder Angelman syndrome (AS). Previous studies of UBE3A function have not examined full Ube3a deletion in mouse, the complexity of imprinted gene networks in brain nor the molecular basis of systems-level cognitive dysfunctions in AS. We therefore utilized a systems biology approach to elucidate how UBE3A loss impacts the early postnatal brain in a novel CRISPR/Cas9-engineered rat Angelman model of a complete Ube3a deletion. Strand-specific transcriptome analysis of offspring from maternally or paternally inherited Ube3a deletions revealed the expected parental expression patterns of Ube3a sense and antisense transcripts by postnatal day 2 (P2) in hypothalamus and day 9 (P9) in cortex, compared to wild-type littermates. The dependency of genome-wide effects on parent-of-origin, Ube3a genotype and time (P2 and P9) was investigated through transcriptome (RNA sequencing of cortex and hypothalamus) and methylome (whole-genome bisulfite sequencing of hypothalamus). Weighted gene co-expression and co-methylation network analyses identified co-regulated networks in maternally inherited Ube3a deletion offspring enriched in postnatal developmental processes including Wnt signaling, synaptic regulation, neuronal and glial functions, epigenetic regulation, ubiquitin, circadian entrainment and splicing. Furthermore, we showed that loss of the paternal Ube3a antisense transcript resulted in both unique and overlapping dysregulated gene pathways with maternal loss, predominantly at the level of differential methylation. Together, these results provide a holistic examination of the molecular impacts of UBE3A loss in brain, supporting the existence of interactive epigenetic networks between maternal and paternal transcripts at the Ube3a locus.


Subject(s)
Genomic Imprinting , Ubiquitin-Protein Ligases/genetics , Angelman Syndrome/genetics , Angelman Syndrome/metabolism , Animals , Brain/metabolism , Cerebral Cortex/metabolism , Epigenesis, Genetic , Female , Gene Expression , Gene Expression Profiling/methods , Gene Regulatory Networks/genetics , Hypothalamus/metabolism , Neuroglia/metabolism , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Synapses/genetics , Synapses/metabolism , Systems Biology , Transcriptome , Ubiquitin-Protein Ligases/metabolism , Wnt Signaling Pathway
4.
PLoS One ; 7(7): e40914, 2012.
Article in English | MEDLINE | ID: mdl-22829897

ABSTRACT

ENGRAILED 2 (En2), a homeobox transcription factor, functions as a patterning gene in the early development and connectivity of rodent hindbrain and cerebellum, and regulates neurogenesis and development of monoaminergic pathways. To further understand the neurobiological functions of En2, we conducted neuroanatomical expression profiling of En2 wildtype mice. RTQPCR assays demonstrated that En2 is expressed in adult brain structures including the somatosensory cortex, hippocampus, striatum, thalamus, hypothalamus and brainstem. Human genetic studies indicate that EN2 is associated with autism. To determine the consequences of En2 mutations on mouse behaviors, including outcomes potentially relevant to autism, we conducted comprehensive phenotyping of social, communication, repetitive, and cognitive behaviors. En2 null mutants exhibited robust deficits in reciprocal social interactions as juveniles and adults, and absence of sociability in adults, replicated in two independent cohorts. Fear conditioning and water maze learning were impaired in En2 null mutants. High immobility in the forced swim test, reduced prepulse inhibition, mild motor coordination impairments and reduced grip strength were detected in En2 null mutants. No genotype differences were found on measures of ultrasonic vocalizations in social contexts, and no stereotyped or repetitive behaviors were observed. Developmental milestones, general health, olfactory abilities, exploratory locomotor activity, anxiety-like behaviors and pain responses did not differ across genotypes, indicating that the behavioral abnormalities detected in En2 null mutants were not attributable to physical or procedural confounds. Our findings provide new insight into the role of En2 in complex behaviors and suggest that disturbances in En2 signaling may contribute to neuropsychiatric disorders marked by social and cognitive deficits, including autism spectrum disorders.


Subject(s)
Autistic Disorder/physiopathology , Cognition Disorders/physiopathology , Animals , Autistic Disorder/genetics , Brain/physiology , Brain Stem/physiology , Cognition Disorders/genetics , Hippocampus/physiology , Homeodomain Proteins , Hypothalamus/physiology , Maze Learning/physiology , Mice , Mice, Knockout , Nerve Tissue Proteins , Signal Transduction , Social Behavior , Somatosensory Cortex/physiology
5.
Learn Mem ; 18(8): 534-44, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21791566

ABSTRACT

BDNF regulates components of cognitive processes and has been implicated in psychiatric disorders. Here we report that genetic overexpression of the BDNF mature isoform (BDNF-tg) in female mice impaired working memory functions while sparing components of fear conditioning. BDNF-tg mice also displayed reduced breeding efficiency, higher anxiety-like scores, high self-grooming, impaired prepulse inhibition, and higher susceptibility to seizures when placed in a new empty cage, as compared with wild-type (WT) littermate controls. Control measures of general health, locomotor activity, motor coordination, depression-related behaviors, and sociability did not differ between genotypes. The present findings, indicating detrimental effects of life-long increased BDNF in mice, may inform human studies evaluating the role of BDNF functional genetic variations on cognitive abilities and vulnerability to psychiatric disorders.


Subject(s)
Anxiety/physiopathology , Brain-Derived Neurotrophic Factor/metabolism , Memory Disorders/metabolism , Memory, Short-Term/physiology , Seizures/genetics , Acoustic Stimulation , Analysis of Variance , Animals , Anxiety/genetics , Brain-Derived Neurotrophic Factor/genetics , Dark Adaptation/genetics , Disease Models, Animal , Electroshock/adverse effects , Enzyme-Linked Immunosorbent Assay/methods , Exploratory Behavior/physiology , Fear/psychology , Female , Hindlimb Suspension/methods , Humans , Inhibition, Psychological , Male , Maze Learning , Memory Disorders/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pain Measurement , Rotarod Performance Test , Seizures/physiopathology , Social Behavior , Swimming/psychology
SELECTION OF CITATIONS
SEARCH DETAIL