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










Publication year range
1.
Mamm Genome ; 23(5-6): 367-77, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22290451

ABSTRACT

Variability and modification of the symptoms of Huntington's disease (HD) are commonly observed in both patient populations and animal models of the disease. Utilizing a stable line of the R6/2 HD mouse model, the present study investigated the role of genetic background in the onset and severity of HD symptoms in a transgenic mouse. R6/2 congenic C57BL/6J and C57BL/6J×DBA/2J F1 (B6D2F1) mice were evaluated for survival and a number of behavioral phenotypes. This study reports that the presence of the DBA/2J allele results in amelioration or exacerbation of several HD-like phenotypes characteristic of the R6/2 mouse model and indicates the presence of dominant genetic modifiers of HD symptoms. This study is the first step in identifying genes that confer natural genetic variation and modify the HD symptoms. This identification may lead to novel targets for treatment and help elucidate the molecular mechanisms of HD pathogenesis.


Subject(s)
Behavior , Huntington Disease/genetics , Animals , Disease Models, Animal , Female , Humans , Huntington Disease/pathology , Huntington Disease/physiopathology , Huntington Disease/psychology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Motor Activity
2.
Behav Brain Res ; 228(1): 1-8, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22123412

ABSTRACT

INTRODUCTION: The G-protein coupled muscarinic acetylcholine receptors, widely expressed in the CNS, have been implicated in fragile X syndrome (FXS). Recent studies have reported an overactive signaling through the muscarinic receptors in the Fmr1KO mouse model. Hence, it was hypothesized that reducing muscarinic signaling might modulate behavioral phenotypes in the Fmr1KO mice. Pharmacological studies from our lab have provided evidence for this hypothesis, with subtype-preferring muscarinic M1 and M4 receptor antagonists modulating select behaviors in the Fmr1KO mice. Since the pharmacological antagonists were not highly specific, we investigated the specific role of M4 receptors in the Fmr1KO mouse model, using a genetic approach. METHODS: We created a double mutant heterozygous for the M4 receptor gene and hemizygous for the Fmr1 gene and examined the mutants on various behaviors. Each animal was tested on a behavior battery comprising of open-field activity (activity), light-dark (anxiety), marble burying (perseverative behavior), prepulse inhibition (sensorimotor gating), rotarod (motor coordination), passive avoidance (learning and memory) and hotplate (analgesia). Animals were also tested on the audiogenic seizure protocol and testis weights were measured. RESULTS: Reduction of M4 receptor expression in the heterozygotes completely rescued the analgesic response and partly rescued the acoustic startle response phenotype in the Fmr1KO mice. However, no modulation was observed in a number of behaviors including learning and memory, activity, perseverative behavior and audiogenic seizures. CONCLUSION: Reducing M4 receptor signaling altered only select behavioral phenotypes in the Fmr1KO mouse model, suggesting that other targets are involved in the modulation of fragile X behaviors.


Subject(s)
Fragile X Syndrome/physiopathology , Fragile X Syndrome/psychology , Receptor, Muscarinic M4/physiology , Reflex, Startle/physiology , Animals , Behavior, Animal/physiology , Disease Models, Animal , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Organ Size , Pain Measurement/methods , Receptor, Muscarinic M4/biosynthesis , Receptor, Muscarinic M4/genetics , Reflex, Startle/genetics , Testis/pathology
3.
Psychopharmacology (Berl) ; 219(1): 47-58, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21656124

ABSTRACT

RATIONALE: Studies in the Fmr1 knockout (KO) mouse, a model of fragile X syndrome (FXS), suggest that excessive signaling through group I metabotropic glutamate receptors (mGluRs), comprised of subtypes mGluR1 and mGluR5, may play a role in the pathogenesis of FXS. Currently, no studies have assessed the effect of mGluR1 modulation on Fmr1 KO behavior, and there has not been an extensive behavioral analysis of mGluR5 manipulation in Fmr1 KO mice. OBJECTIVES: The goals for this study were to determine if pharmacologic blockade of mGluR1 may affect Fmr1 KO behavior as well as to expand on the current literature regarding pharmacologic blockade of mGluR5 on Fmr1 KO behavior. METHODS: Reduction of mGluR1 or mGluR5 activity was evaluated on a variety of behavioral assays in wild-type (WT) and Fmr1 KO mice through the use of antagonists: JNJ16259685 (JNJ, mGluR1 antagonist) and MPEP (mGluR5 antagonist). RESULTS: JNJ and MPEP decreased marble burying in both WT and Fmr1 KO mice without reductions in activity. Neither JNJ nor MPEP affected the prepulse inhibition in either WT or Fmr1 KO mice. JNJ did not affect Fmr1 KO motor coordination but did impair WT performance. MPEP improved a measure of motor learning in Fmr1 KO but not WT mice. While both JNJ and MPEP decreased the audiogenic seizures in the Fmr1 KO, MPEP completely abolished the manifestation of seizures. CONCLUSION: These data illustrate that, while the manipulation of either mGluR1 or mGluR5 can affect select behaviors in the Fmr1 KO, we observe greater effects upon mGluR5 reduction.


Subject(s)
Disease Models, Animal , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Acoustic Stimulation/methods , Animals , Dose-Response Relationship, Drug , Fragile X Syndrome/psychology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Quinolines/pharmacology , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/physiology , Reflex, Startle/drug effects , Reflex, Startle/physiology
4.
PLoS One ; 6(12): e28409, 2011.
Article in English | MEDLINE | ID: mdl-22163300

ABSTRACT

In the present study we report on the use of speed congenics to generate a C57BL/6J congenic line of HD-model R6/2 mice carrying 110 CAG repeats, which uniquely exhibits minimal intergenerational instability. We also report the first identification of the R6/2 transgene insertion site. The relatively stable line of 110 CAG R6/2 mice was characterized for the onset of behavioral impairments in motor, cognitive and psychiatric-related phenotypes as well as the progression of disease-related impairments from 4 to 10 weeks of age. 110Q mice exhibited many of the phenotypes commonly associated with the R6/2 model including reduced activity and impairments in rotarod performance. The onset of many of the phenotypes occurred around 6 weeks and was progressive across age. In addition, some phenotypes were observed in mice as early as 4 weeks of age. The present study also reports the onset and progression of changes in several molecular phenotypes in the novel R6/2 mice and the association of these changes with behavioral symptom onset and progression. Data from TR-FRET suggest an association of mutant protein state changes (soluble versus aggregated) in disease onset and progression.


Subject(s)
Huntington Disease/genetics , Animals , Avoidance Learning , Behavior, Animal , Crosses, Genetic , Disease Models, Animal , Disease Progression , Female , Genotype , Humans , Huntingtin Protein , Male , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Nerve Tissue Proteins/genetics , Phenotype , Transgenes , Trinucleotide Repeat Expansion
5.
Behav Neurosci ; 125(5): 783-90, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21942438

ABSTRACT

Muscarinic acetylcholine receptors (mAChR) are G protein-coupled receptors (M1-M5), grouped together into two functional classes, based on their G protein interaction. Although ubiquitously expressed in the CNS, the M4 protein shows highest expression in the neostriatum, cortex, and hippocampus. Electrophysiological and biochemical studies have provided evidence for overactive mAChR signaling in the fragile X knock-out (Fmr1KO) mouse model, and this has been hypothesized to contribute to the phenotypes seen in Fmr1KO mice. To address this hypothesis we used an M4 antagonist, tropicamide, to reduce the activity through the M4 mAChR and investigated the behavioral response in the Fmr1KO animals. Data from the marble-burying assay have shown that tropicamide treatment resulted in a decreased number of marbles buried in the wild-type (WT) and in the knockout (KO) animals. Results from the open field assay indicated that tropicamide increases activity in both the WT and KO mice. In the passive avoidance assay, tropicamide treatment resulted in the improvement of performance in both the WT and the KO animals at the lower doses (2 and 5 mg/kg), and the drug was shown to be important for the acquisition and not the consolidation process. Lastly, we observed that tropicamide causes a significant decrease in the percentage of audiogenic seizures in the Fmr1KO animals. These results suggest that pharmacological antagonism of the M4 receptor modulates select behavioral responses in the Fmr1KO mice.


Subject(s)
Fragile X Mental Retardation Protein/physiology , Fragile X Syndrome/drug therapy , Muscarinic Antagonists/therapeutic use , Receptor, Muscarinic M4/antagonists & inhibitors , Receptor, Muscarinic M4/physiology , Tropicamide/therapeutic use , Animals , Dose-Response Relationship, Drug , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/metabolism , Male , Mice , Mice, Knockout , Muscarinic Antagonists/pharmacology , Tropicamide/pharmacology
6.
Behav Brain Res ; 223(2): 310-21, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21571007

ABSTRACT

INTRODUCTION: Genetic heterogeneity likely contributes to variability in the symptoms among individuals with fragile X syndrome (FXS). Studies in the Fmr1 knockout (KO) mouse model for FXS suggest that excessive signaling through group I metabotropic glutamate receptors (Gp1 mGluRs), comprised of subtypes mGluR1 and mGluR5, may play a role. Hence, Gp1 mGluRs may act as modifiers of FXS. Currently no studies have addressed whether manipulation of mGluR1 activity may alter Fmr1 KO behavioral responses, and only a few have reported the effects of mGluR5 manipulation. Therefore, the goals for this study were to extend our understanding of the effects of modulating Gp1 mGluR activity on Fmr1 KO behavioral responses. METHODS: The present study determined if genetically reducing mGluR1 or mGluR5 by 50% affects an extensive array of behaviors in the Fmr1 KO. RESULTS: Reduction of mGluR1 moderately decreased Fmr1 KO activity. Reduction of mGluR5 caused an analgesic response in the Fmr1 KO and decreased active social behavior. Modulation of either mGluR1 or mGluR5 did not significantly alter audiogenic seizures, anxiety- and perseverative-related responses, sensorimotor gating, memory, or motor responses. CONCLUSIONS: Genetic reduction of mGluR1 or mGluR5 modified a few select Fmr1 KO behaviors, although these modifications appeared to be subtle in nature and/or limited to select behaviors. This may indicate that 50% reduction of either mGluR1 or mGluR5 is insufficient to produce behavioral changes, and therefore, these receptors may not be dominant modifiers of a number of Fmr1 KO behavioral phenotypes.


Subject(s)
Behavior, Animal/physiology , Fragile X Syndrome/genetics , Fragile X Syndrome/psychology , Receptors, Metabotropic Glutamate/genetics , Animals , Blotting, Western , Conditioning, Psychological , Disease Models, Animal , Epilepsy, Reflex/genetics , Epilepsy, Reflex/psychology , Exploratory Behavior/physiology , Fear/psychology , Female , Fragile X Mental Retardation Protein/genetics , Genotype , Hot Temperature , Interpersonal Relations , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Pain Measurement/psychology , Postural Balance/genetics , Postural Balance/physiology , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/biosynthesis , Reflex, Startle/physiology , Testis/anatomy & histology , Testis/physiology
7.
Psychopharmacology (Berl) ; 217(1): 143-51, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21487657

ABSTRACT

RATIONALE: Muscarinic acetylcholine receptors (mAChR) are G protein-coupled receptors, widely expressed in the CNS. Electrophysiological and molecular studies have provided evidence for overactive M1 receptor signaling in the fragile X knockout (Fmr1 KO) mouse model, suggesting the involvement of the M1 receptors in fragile X syndrome. Overactive signaling through the M1 receptor has been hypothesized to contribute to the phenotypes seen in fragile X mice. OBJECTIVE: We investigated the modulation of behavioral responses in the Fmr1 KO animals by reducing the activity through the muscarinic M1 receptor using the pharmacological agent dicyclomine, an M1 antagonist. METHODS: The behavioral assays used to investigate the pharmacological effects include marble burying (perseverative behavior), open-field exploration (activity), passive avoidance (learning and memory), prepulse inhibition (sensorimotor gating), and audiogenic seizures. RESULTS: Data from the marble-burying assay suggests that treatment with dicyclomine results in a decrease in the number of marbles buried in the wild-type and in the KO animals. To examine the possibility of drug-induced sedation, overall activity was measured in an open-field chamber. Dicyclomine only increases activity at a dose of 20 mg/kg in the wild-type mice but did not affect exploration in the KO animals. Lastly, we observed that dicyclomine causes a significant decrease in the percentage of audiogenic seizures in the Fmr1 KO animals. CONCLUSION: Our findings suggest that pharmacologically reducing the activity through the mAChR M1 alters select behavioral responses in the Fmr1 KO mice.


Subject(s)
Behavior, Animal/drug effects , Dicyclomine/therapeutic use , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/physiopathology , Muscarinic Antagonists/therapeutic use , Receptor, Muscarinic M1/antagonists & inhibitors , Animals , Disease Models, Animal , Epilepsy, Reflex/genetics , Epilepsy, Reflex/physiopathology , Epilepsy, Reflex/prevention & control , Exploratory Behavior/drug effects , Female , Fragile X Syndrome/complications , Fragile X Syndrome/genetics , Male , Mice , Mice, Knockout , Motor Activity/drug effects , Reflex, Startle/drug effects
8.
Autism Res ; 4(1): 40-56, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21268289

ABSTRACT

Fragile X syndrome (FXS) is the most common inherited form of intellectual disability in humans. In addition to cognitive impairment, patients may exhibit hyperactivity, attention deficits, social difficulties and anxiety, and autistic-like behaviors. The degree to which patients display these behaviors varies considerably and is influenced by family history, suggesting that genetic modifiers play a role in the expression of behaviors in FXS. Several studies have examined behavior in a mouse model of FXS in which the Fmr1 gene has been ablated. Most of those studies were done in Fmr1 knockout mice on a pure C57BL/6 or FVB strain background. To gain a better understanding of the effects of genetic background on behaviors resulting from the loss of Fmr1 gene expression, we generated F1 hybrid lines from female Fmr1 heterozygous mice on a pure C57BL/6J background bred with male Fmr1 wild-type (WT) mice of various background strains (A/J, DBA/2J, FVB/NJ, 129S1/SvImJ and CD-1). Male Fmr1 knockout and WT littermates from each line were examined in an extensive behavioral test battery. Results clearly indicate that multiple behavioral responses are dependent on genetic background, including autistic-like traits that are present on limited genetic backgrounds. This approach has allowed us to identify improved models for different behavioral symptoms present in FXS including autistic-like traits.


Subject(s)
Child Development Disorders, Pervasive/genetics , Disease Models, Animal , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Phenotype , Social Behavior , Animals , Animals, Newborn/genetics , Child , DNA Mutational Analysis , Exploratory Behavior , Female , Genetic Carrier Screening , Genotype , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Motor Skills , Psychomotor Performance , Sensory Gating/genetics , Stereotyped Behavior
9.
Behav Brain Res ; 218(1): 29-41, 2011 Mar 17.
Article in English | MEDLINE | ID: mdl-21093492

ABSTRACT

Autism spectrum disorder (ASD) diagnoses are behaviorally based with no defined universal biomarkers, occur at a 1:110 ratio in the population, and predominantly affect males compared to females at approximately a 4:1 ratio. One approach to investigate and identify causes of ASD is to use organisms that display abnormal behavioral responses that model ASD-related impairments. This study describes a novel transgenic mouse, MALTT, which was generated using a forward genetics approach. It was determined that the transgene integrated within a non-coding region on the X chromosome. The MALTT line exhibited a complete repertoire of ASD-like behavioral deficits in all three domains required for an ASD diagnosis: reciprocal social interaction, communication, and repetitive or inflexible behaviors. Specifically, MALTT male mice showed deficits in social interaction and interest, abnormalities in pup and juvenile ultrasonic vocalization communications, and exhibited a repetitive stereotypy. Abnormalities were also observed in the domain of sensory function, a secondary phenotype prevalently associated with ASD. Mapping and expression studies suggested that the Fam46 gene family may be linked to the observed ASD-related behaviors. The MALTT line provides a unique genetic model for examining the underlying biological mechanisms involved in ASD-related behaviors.


Subject(s)
Aggression/psychology , Autistic Disorder/psychology , Disease Models, Animal , Social Behavior , Analysis of Variance , Animals , Autistic Disorder/genetics , Female , Male , Mice , Mice, Transgenic , Sensory Gating , Stereotyped Behavior , Vocalization, Animal
10.
PLoS Genet ; 5(12): e1000758, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20011099

ABSTRACT

The mental retardation, autistic features, and behavioral abnormalities characteristic of the Fragile X mental retardation syndrome result from the loss of function of the RNA-binding protein FMRP. The disease is usually caused by a triplet repeat expansion in the 5'UTR of the FMR1 gene. This leads to loss of function through transcriptional gene silencing, pointing to a key function for FMRP, but precluding genetic identification of critical activities within the protein. Moreover, antisense transcripts (FMR4, ASFMR1) in the same locus have been reported to be silenced by the repeat expansion. Missense mutations offer one means of confirming a central role for FMRP in the disease, but to date, only a single such patient has been described. This patient harbors an isoleucine to asparagine mutation (I304N) in the second FMRP KH-type RNA-binding domain, however, this single case report was complicated because the patient harbored a superimposed familial liver disease. To address these issues, we have generated a new Fragile X Syndrome mouse model in which the endogenous Fmr1 gene harbors the I304N mutation. These mice phenocopy the symptoms of Fragile X Syndrome in the existing Fmr1-null mouse, as assessed by testicular size, behavioral phenotyping, and electrophysiological assays of synaptic plasticity. I304N FMRP retains some functions, but has specifically lost RNA binding and polyribosome association; moreover, levels of the mutant protein are markedly reduced in the brain specifically at a time when synapses are forming postnatally. These data suggest that loss of FMRP function, particularly in KH2-mediated RNA binding and in synaptic plasticity, play critical roles in pathogenesis of the Fragile X Syndrome and establish a new model for studying the disorder.


Subject(s)
Disease Models, Animal , Fragile X Syndrome/genetics , Mutation, Missense , Animals , Behavior, Animal , Fragile X Mental Retardation Protein/genetics , Hippocampus/metabolism , Humans , Mice , Neuronal Plasticity , Phenotype
11.
Psychopharmacology (Berl) ; 204(2): 361-73, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19189082

ABSTRACT

RATIONALE: An increasing number of investigators utilize the marble-burying assay despite the paucity of information available regarding what underlies the behavior. OBJECTIVES: We tested the possibility that a genetic component underlies marble burying in mice and if there is a genetic correlation with other anxiety-like traits. Since findings reported in the literature indicate that marble-burying behavior reflects an anxiety-like response, we explored the assumption that the novel nature of a marble induces this anxiety. Finally, we investigated how the natural response of a mouse to dig relates to the marble-burying phenomenon. METHODS: We examined ten different inbred mouse strains to determine if marble-burying behavior is genetically regulated and correlated with anxiety-like traits in two other assays. We employed multiple variants of the "traditional" marble-burying assay to address how issues such as the novelty of marbles and digging behavior contribute to marble burying. RESULTS: Marble-burying behavior varied across strain and did not correlate with anxiety measures in other assays. Multiple tests conducted to reduce the novelty of marbles failed to alter burying behavior. Additionally, digging behavior correlated with marble burying, and the presence of marbles did not significantly impact the digging response. CONCLUSIONS: Our results indicate that mouse marble burying is genetically regulated, not correlated with other anxiety-like traits, not stimulated by novelty, and is a repetitive behavior that persists/perseveres with little change across multiple exposures. Marble burying is related to digging behavior and may in fact be more appropriately considered as an indicative measure of repetitive digging.


Subject(s)
Anxiety/psychology , Behavior, Animal/physiology , Obsessive Behavior/psychology , Animals , Environment , Exploratory Behavior/physiology , Feeding Behavior , Food , Male , Mice , Mice, Inbred Strains , Motor Activity/physiology , Species Specificity
12.
Behav Neurosci ; 122(6): 1371-7, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19045956

ABSTRACT

Fragile X syndrome is caused by a CGG trinucleotide repeat expansion of the FMR1 gene. Individuals with fragile X display several behavioral abnormalities including hyperactivity, social anxiety, autistic-like features, impaired cognitive processing, and impaired sensorimotor gating. The Fmr1KO mouse model of fragile X exhibits several related behavioral phenotypes such as increased activity and altered social interactions. Individuals with fragile X also have impaired sensorimotor gating as measured using the prepulse inhibition of startle response. The authors have recently shown that Fmr1KO mice with a yeast artificial chromosome containing the human FMR1 gene have corrected or overcorrected abnormal behaviors including hyperactivity and altered social interactions. Here the authors present results from a study examining abnormal sensorimotor gating in Fmr1KO mice. Consistent with previous findings, Fmr1KO mice have increased prepulse inhibition. The KO mice with the yeast artificial chromosome containing the human FMR1 gene had levels of prepulse inhibition comparable to WT mice, indicating not only a correction of this phenotype, but also clearly demonstrating that in mice levels of the fragile X mental retardation protein regulate sensorimotor gating.


Subject(s)
Fragile X Mental Retardation Protein/genetics , Neural Inhibition/genetics , Sensory Gating/genetics , Acoustic Stimulation/methods , Animals , Behavior, Animal , Genotype , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Inhibition/physiology , Psychophysics , Reaction Time
13.
Behav Neurosci ; 122(3): 710-5, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18513141

ABSTRACT

Fragile X syndrome (FXS) results from the loss of expression of the fragile X mental retardation (FMR1) gene. Individuals affected by FXS experience many behavioral problems, including cognitive impairment, hyperactivity, social anxiety, and autistic-like behaviors. A mouse model of Fmr1 deficiency (Fmr1KO) exhibits several similar behavioral phenotypes, including alterations in social behavior. In an earlier study, Fmr1 knockout mice carrying a yeast-artificial chromosome (YAC) transgene that over-expresses normal human FMRP (KOYAC) showed a correction or overcorrection of some behavioral responses, such as hyperactivity and anxiety-related responses. This report presents results from a study examining transgenic rescue of abnormal social responses in Fmr1KO mice. Relative to their wild-type (WT) littermates, Fmr1KO mice made more active social approaches to standard C57BL/6 partner mice in a direct social interaction test, a result consistent with a previous study. KOYAC mice showed fewer active approaches to partners than the WT or Fmr1KO littermates, indicating correction of this phenotype. This finding expands the number of murine behavioral responses caused by Fmr1 deficiency and corrected by overexpression of human FMRP.


Subject(s)
Fragile X Mental Retardation Protein/genetics , Interpersonal Relations , Mice, Transgenic/physiology , Phenotype , Animals , Appetitive Behavior/physiology , Behavior, Animal/physiology , Exploratory Behavior/physiology , Humans , Locomotion/genetics , Male , Mice , Mice, Inbred C57BL
14.
Hum Mol Genet ; 16(15): 1802-13, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17517686

ABSTRACT

Smith-Magenis syndrome (SMS) is associated with an approximately 3.7 Mb common deletion in 17p11.2 and characterized by its craniofacial and neurobehavioral abnormalities. The reciprocal duplication leads to dup(17)(p11.2p11.2) associated with the Potocki-Lupski syndrome (PLS), a neurological disorder whose features include autism. Retinoic acid induced 1 (RAI1) appears to be responsible for the majority of clinical features in both SMS and PLS. Mouse models of these syndromes harboring an approximately 2 Mb chromosome engineered deletion and duplication, respectively, displayed abnormal locomotor activity and/or learning deficits. To determine the contribution of RAI1 in the neurobehavioral traits in SMS, we performed a battery of behavioral tests on Rai1 mutant mice and the Df(11)17-1/+ mice that have a small deletion of approximately 590 kb. The mice with the small deletion were hypoactive like the large deletion mice and they also showed learning deficits. The Rai1+/- mice exhibited normal locomotor activity. However, they had an abnormal electroencephalogram with overt seizure observed in a subset of mice. The few surviving Rai1-/- mice displayed more severe neurobehavioral abnormalities including hind limb clasping, overt seizures, motor impairment and context- and tone-dependant learning deficits. X-gal staining of the Rai1+/- mice suggests that Rai1 is predominantly expressed in neurons of the hippocampus and the cerebellum. Our results suggest that Rai1 is a critical gene in the central nervous system functioning in a dosage sensitive manner and that the neurobehavioral phenotype is modified by regulator(s) in the approximately 590 kb genomic interval, wherein the major modifier affecting the craniofacial penetrance resides.


Subject(s)
Gene Deletion , Learning Disabilities/genetics , Psychomotor Performance/physiology , Trans-Activators/genetics , Abnormalities, Multiple/genetics , Animals , Central Nervous System/metabolism , Craniofacial Abnormalities/genetics , Disease Models, Animal , Electroencephalography , Female , Heterozygote , Immunohistochemistry , Learning Disabilities/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Trans-Activators/deficiency , Trans-Activators/metabolism
15.
Hum Mol Genet ; 15(12): 1984-94, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16675531

ABSTRACT

Individuals affected by Fragile X syndrome (FXS) experience cognitive impairment, hyperactivity, attention deficits, social anxiety and autistic-like behaviors. FXS results from the loss of expression of the Fragile X mental retardation (FMR1) gene, whose protein product FMRP is thought to play an important role in neuronal function and synaptic plasticity. Two paralogs of FMRP, FXR1P and FXR2P, have been identified, forming the Fragile X-related (FXR) family of proteins. Although the functions of FXR1P and FXR2P are not well understood, there are similarities among all three FXR proteins in gene structure, amino acid sequence, expression pattern and cellular functions. Mouse models have been described for loss of Fmrp, Fxr1p and Fxr2p, the mouse homologs of FMRP, FXR1P and FXR2P. In earlier studies, we found that Fmr1 knockout (KO) mice, which do not express Fmrp, and Fxr2 KO mice, which do not express Fxr2p, show similarities in some behavioral responses such as hyperactivity. To better understand the functional relationship between FMRP and FXR2P, we generated Fmr1 KO, Fxr2 KO, Fmr1/Fxr2 double KO and wild-type control mice as littermates on the same genetic background and examined them in several behavioral assays. Results show that Fmr1/Fxr2 double KO mice have exaggerated behavioral phenotypes in open-field activity, prepulse inhibition of acoustic startle response and contextual fear conditioning when compared with Fmr1 KO mice, Fxr2 KO mice or wild-type littermates. Our findings suggest that Fmr1 and Fxr2 genes contribute in a cooperative manner to pathways controlling locomotor activity, sensorimotor gating and cognitive processes.


Subject(s)
Behavior, Animal , Fragile X Mental Retardation Protein/genetics , RNA-Binding Proteins/genetics , Acoustic Stimulation , Animals , Anxiety , Conditioning, Psychological , Fear , Female , Learning , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Phenotype , Psychomotor Performance , Reflex, Startle , Somatosensory Disorders
16.
Physiol Behav ; 87(1): 95-102, 2006 Jan 30.
Article in English | MEDLINE | ID: mdl-16197969

ABSTRACT

Test batteries are commonly used to assess the behavioral phenotype of genetically modified and inbred strains of mice. However, few systematic studies have been employed to address several key issues concerning the use of a test battery. The current study was designed to address whether inter-test interval affects behavioral performance. Male mice of 3 different inbred strains and one F1 hybrid strain were randomly assigned to either a test battery with 1 week inter-test intervals, or a rapid test battery with 1-2 day inter-test intervals. The test battery included a neurological exam, open-field activity, light-dark exploration, rotarod test, prepulse inhibition, and startle habituation. The experiment was repeated with female animals of 2 different strains. As expected, there were strain differences on each of the behavioral assays; however, there was no major difference in performance between mice of the standard test battery and the rapid test battery. Similar results were found with females. These results indicate that the interval between most tests could be as little as 1-2 days, with little significant effect on overall performance. Thus, it is possible with the current test battery to reduce the inter-test interval to facilitate the rate of studying and identifying behavioral phenotypes in mice.


Subject(s)
Animal Experimentation , Behavior, Animal/physiology , Behavioral Research/instrumentation , Behavioral Research/methods , Motor Activity/physiology , Psychomotor Performance/physiology , Analysis of Variance , Animals , Exploratory Behavior/physiology , Female , Genetics, Behavioral/methods , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Models, Animal , Motor Activity/genetics , Phenotype , Random Allocation , Reflex, Startle/genetics , Reflex, Startle/physiology , Rotarod Performance Test , Species Specificity , Time Factors
17.
Neuron ; 35(2): 243-54, 2002 Jul 18.
Article in English | MEDLINE | ID: mdl-12160743

ABSTRACT

Mutations in the methyl-CpG binding protein 2 (MECP2) gene cause Rett syndrome (RTT), a neurodevelopmental disorder characterized by the loss of language and motor skills during early childhood. We generated mice with a truncating mutation similar to those found in RTT patients. These mice appeared normal and exhibited normal motor function for about 6 weeks, but then developed a progressive neurological disease that includes many features of RTT: tremors, motor impairments, hypoactivity, increased anxiety-related behavior, seizures, kyphosis, and stereotypic forelimb motions. Additionally, we show that although the truncated MeCP2 protein in these mice localizes normally to heterochromatic domains in vivo, histone H3 is hyperacetylated, providing evidence that the chromatin architecture is abnormal and that gene expression may be misregulated in this model of Rett syndrome.


Subject(s)
Central Nervous System/metabolism , Chromosomal Proteins, Non-Histone , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Histones/metabolism , Mutation/genetics , Repressor Proteins , Rett Syndrome/genetics , Acetylation , Aging/genetics , Aging/metabolism , Animals , Behavior, Animal/physiology , Biomarkers/analysis , Central Nervous System/abnormalities , Central Nervous System/physiopathology , Conditioning, Psychological/physiology , Disease Models, Animal , Fear/physiology , Female , Humans , Male , Maze Learning/physiology , Methyl-CpG-Binding Protein 2 , Mice , Mice, Knockout , Movement Disorders/genetics , Movement Disorders/metabolism , Movement Disorders/physiopathology , Neurons/metabolism , Neurons/pathology , Protein Structure, Tertiary/genetics , Rett Syndrome/metabolism , Rett Syndrome/physiopathology , Social Behavior
18.
Neuron ; 34(6): 905-19, 2002 Jun 13.
Article in English | MEDLINE | ID: mdl-12086639

ABSTRACT

To faithfully recreate the features of the human neurodegenerative disease spinocerebellar ataxia type 1 (SCA1) in the mouse, we targeted 154 CAG repeats into the endogenous mouse locus. Sca1(154Q/2Q) mice developed a progressive neurological disorder that resembles human SCA1, featuring motor incoordination, cognitive deficits, wasting, and premature death, accompanied by Purkinje cell loss and age-related hippocampal synaptic dysfunction. Mutant ataxin-1 solubility varied with brain region, being most soluble in the neurons most vulnerable to degeneration. Solubility decreased overall as the mice aged; Purkinje cells, the most affected in SCA1, did not form aggregates of mutant protein until an advanced stage of disease. It appears that those neurons that cannot sequester the mutant protein efficiently and thereby curb its toxicity suffer the worst damage from polyglutamine-induced toxicity.


Subject(s)
Disease Models, Animal , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Spinocerebellar Ataxias/genetics , Trinucleotide Repeats/genetics , Animals , Ataxin-1 , Ataxins , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/chemistry , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Nuclear Proteins/biosynthesis , Nuclear Proteins/chemistry , Purkinje Cells/metabolism , Purkinje Cells/pathology , Solubility , Spinocerebellar Ataxias/metabolism , Spinocerebellar Ataxias/physiopathology
19.
Hum Mol Genet ; 11(5): 487-98, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11875043

ABSTRACT

Fragile X syndrome is a common form of mental retardation caused by the absence of the FMR1 protein, FMRP. Fmr1 knockout mice exhibit a phenotype with some similarities to humans, such as macro-orchidism and behavioral abnormalities. Two homologs of FMRP have been identified, FXR1P and FXR2P. These proteins show high sequence similarity, including all functional domains identified in FMRP, such as RNA binding domains. They have an overlap in tissue distribution to that of FMRP. Interactions between the three FXR proteins have also been described. FXR2P shows high expression in brain and testis, like FMRP. To study the function of FXR2P, we generated an Fxr2 knockout mouse model. No pathological differences between knockout and wild-type mice were found in brain or testis. Given the behavioral phenotype in fragile X patients and the phenotype previously reported for the Fmr1 knockout mouse, we performed a thorough evaluation of the Fxr2 knockout phenotype using a behavioral test battery. Fxr2 knockout mice were hyperactive (i.e. traveled a greater distance, spent more time moving and moved faster) in the open-field test, impaired on the rotarod test, had reduced levels of prepulse inhibition, displayed less contextual conditioned fear, impaired at locating the hidden platform in the Morris water task and were less sensitive to a heat stimulus. Interestingly, there are some behavioral phenotypes in Fxr2 knockout mice which are similar to those observed in Fmr1 knockout mice, but there are also some different behavioral abnormalities that are only observed in the Fxr2 mutant mice. The findings implicate a role for Fxr2 in central nervous system function.


Subject(s)
Intellectual Disability/genetics , RNA-Binding Proteins/genetics , Animals , Behavior, Animal , Brain Chemistry , Conditioning, Psychological , Fear/psychology , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Fragile X Syndrome/pathology , Fragile X Syndrome/psychology , Hot Temperature , Hyperkinesis , Male , Maze Learning , Mice , Mice, Knockout , Phenotype , Testis/chemistry , Tissue Distribution
20.
J Neurosci ; 22(7): 2753-63, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11923441

ABSTRACT

FRAXE mental retardation results from expansion and methylation of a CCG trinucleotide repeat located in exon 1 of the X-linked FMR2 gene, which results in transcriptional silencing. The product of FMR2 is a member of a family of proteins rich in serine and proline, members of which have been associated with transcriptional activation. We have developed a murine Fmr2 gene knock-out model by replacing a fragment containing parts of exon 1 and intron 1 with the Escherichia coli lacZ gene, placing lacZ under control of the Fmr2 promoter. Expression of lacZ in the knock-out animals indicates that Fmr2 is expressed in several tissues, including brain, bone, cartilage, hair follicles, lung, tongue, tendons, salivary glands, and major blood vessels. In the CNS, Fmr2 expression begins at the time that cells in the neuroepithelium differentiate into neuroblasts. Mice lacking Fmr2 showed a delay-dependent conditioned fear impairment. Long-term potentiation (LTP) was found to be enhanced in hippocampal slices of Fmr2 knock-out compared with wild-type littermates. To our knowledge, this mouse knock-out is the first example of an animal model of human mental retardation with impaired learning and memory performance and increased LTP. Thus, although a number of studies have suggested that diminished LTP is associated with memory impairment, our data suggest that increased LTP may be a mechanism that leads to impaired cognitive processing as well.


Subject(s)
Fear , Fragile X Syndrome/physiopathology , Long-Term Potentiation , Nuclear Proteins/deficiency , Trans-Activators/deficiency , Animals , Behavior, Animal , Blotting, Southern , Chimera , Chromosome Mapping , Conditioning, Classical , Disease Models, Animal , Fear/physiology , Fragile X Syndrome/complications , Gene Expression , Gene Targeting , In Vitro Techniques , Intellectual Disability/etiology , Intellectual Disability/physiopathology , Learning Disabilities/etiology , Learning Disabilities/physiopathology , Long-Term Potentiation/physiology , Male , Memory Disorders/etiology , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/genetics , Nuclear Proteins/genetics , Organ Specificity , Phenotype , Promoter Regions, Genetic , Survival Rate , Trans-Activators/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...