Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters










Publication year range
1.
Genes Brain Behav ; 5(6): 467-71, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16923151

ABSTRACT

Fragile X syndrome (FXS) is the most common form of inherited mental retardation (MR). FXS is typically caused by a mutation of the Fmr1 gene (Verkerk et al. 1991, Cell 65, 905-914). To better understand the role of Fmr1 and its gene product fragile X mental-retardation protein (FMRP) in central nervous system function, researchers have turned to the use of animal model systems to generate an Fmr1 knockout (KO) mouse that is deficient in FMRP (Bakker et al. 1994, Cell 78, 23-33). Unfortunately, a number of studies have found no consistent, robust learning and memory impairment in the Fmr1 KO mice. We conducted a study to assess the performance of Fmr1 KO and wildtype (WT) animals in a leverpress escape/avoidance paradigm. Fmr1 KO and WT littermates were studied in four daily 1-h sessions. The Fmr1 KO mice performed fewer avoidance and total responses than WT mice. The KO animals were not simply deficient in avoidance, but a within-factor ANOVA revealed that they did not acquire the leverpress response to any appreciable degree. Observation during the sessions indicated that the Fmr1 KO animals clearly responded to the shock, eliminating an obvious sensory explanation for the deficit. The fact that other studies have found that the KO mice displayed increased exploratory and locomotor activity compared with WT controls argues against a motoric deficit. Future studies will attempt to delineate the nature of the behavioral deficit as well as attempt to rescue the response with glutamatergic or dopaminergic agents.


Subject(s)
Avoidance Learning/physiology , Brain/metabolism , Cognition Disorders/genetics , Fragile X Mental Retardation Protein/genetics , Learning Disabilities/genetics , Animals , Brain/physiopathology , Cognition Disorders/metabolism , Cognition Disorders/psychology , Disease Models, Animal , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Fragile X Syndrome/physiopathology , Learning Disabilities/metabolism , Learning Disabilities/psychology , Male , Memory Disorders/genetics , Memory Disorders/metabolism , Memory Disorders/psychology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropsychological Tests
2.
Genes Brain Behav ; 4(7): 420-30, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16176388

ABSTRACT

The loss of fragile X mental retardation (FMR1) gene function causes fragile X syndrome (FXS), a common mental retardation syndrome. Anxiety and abnormal social behaviors are prominent features of FXS in humans. To better understand the role of FMR1 in these behaviors, we analyzed anxiety-related and social behaviors in Fmr1 knockout (KO) mice. In the mirrored chamber test, Fmr1 KO mice showed greater aversion to the central mirrored chamber than wild-type (WT) littermates, suggesting increased anxiety-like responses to reflected images of mice. Fmr1 KO mice exhibited abnormal social interactions in a tube test of social dominance, winning fewer matches than WT littermates. In a partition test, Fmr1 KO mice had normal levels of social interest and social recognition. However, during direct interaction tests, Fmr1 KO mice showed significant increases in sniffing behaviors. We further tested the influence of environmental familiarity on the social responses of Fmr1 KO mice to unfamiliar partners. In unfamiliar partitioned cages, Fmr1 KO mice did not differ from WT mice in investigation of unfamiliar partners. However, in familiar partitioned cages, Fmr1 KO mice showed less investigation of a newly introduced partner during the first 5 min and more investigation during the last 5 min of a 20-min partition test, behaviors consistent with initial social anxiety followed by enhanced social investigation. Our findings indicate that the loss of Fmr1 gene function results in altered anxiety and social behavior in mice and demonstrate that the Fmr1 KO mouse is a relevant animal model for the abnormal social responses seen in FXS.


Subject(s)
Anxiety/metabolism , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Social Behavior , Social Environment , Animals , Anxiety/genetics , Anxiety/psychology , Disease Models, Animal , Dominance-Subordination , Fragile X Syndrome/genetics , Fragile X Syndrome/psychology , Genetics, Behavioral/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
3.
Neuroscience ; 129(3): 575-82, 2004.
Article in English | MEDLINE | ID: mdl-15541879

ABSTRACT

The nicotinic cholinergic system influences cognition, anxiety, locomotion, and addiction by acting upon nicotinic acetylcholine receptors (nAChRs). To date, there are 12 known neuronal mammalian nAChR subunits leading to a rich pharmacological diversity that is difficult to attribute to specific subunits. We generated alpha7-beta2 nAChR double mutant mice by breeding to investigate the effect of a minimal number of nAChRs in the CNS. These mice have been used to determine the role these receptor subunits play in a variety of behaviors. A battery of behavioral tests was used to determine the effect of the mutation in anxiety, locomotor activity, startle response, pre-pulse inhibition, motor coordination and learning and memory. Mice lacking both the alpha7 and the beta2 nAChR subunits displayed impaired learning and memory performance in a passive avoidance test and showed enhanced motor performance on the rotarod.


Subject(s)
Avoidance Learning/physiology , Brain/cytology , Neurons/metabolism , Receptors, Nicotinic/deficiency , Analysis of Variance , Animals , Behavior, Animal , Exploratory Behavior/physiology , Inhibition, Psychological , Male , Mice , Mice, Knockout , Motor Activity/genetics , Protein Subunits/deficiency , Protein Subunits/genetics , Psychomotor Performance/physiology , Receptors, Nicotinic/genetics , Receptors, Nicotinic/physiology , Reflex, Acoustic/physiology
4.
Neuroscience ; 129(1): 11-24, 2004.
Article in English | MEDLINE | ID: mdl-15489024

ABSTRACT

Nicotine can enhance contextual learning while ethanol impairs some forms of learning. Nicotine can overcome some of the impairing effects of ethanol when the two drugs are co-administered. The specific brain nicotinic acetylcholine receptors (nAChRs) that mediate nicotine's effects on contextual learning and whether any of ethanol's actions are mediated by nAChRs are unknown. The potential roles of nAChRs in contextual and cued fear conditioning as well as the effects of nicotine, ethanol, or co-administration of nicotine and ethanol were examined in wild type and homozygous null mutant mice from alpha7, beta2, beta3, and beta4 mouse lines at 24 h after training. Nicotine was given prior to training and testing, whereas ethanol was given only before training. Nicotine enhanced contextual learning in both alpha7 wild types and mutants when mice were trained at 0.17 mA, but not 0.35 mA. Mutants lacking the alpha7 subunit were less sensitive to the memory impairing effects of ethanol trained at 0.35 mA. beta2 Null mutants receiving saline showed a small, but significant, impairment in contextual learning compared with wild type littermates when the shock stimulus was 0.35 mA. Beta2 Null mutant mice also did not respond to the cognitive enhancing effects of nicotine alone, or after ethanol administration. beta3 and beta4 null mutants did not differ from wild types either after saline or any of drug treatments. These results show that beta2-containing nAChRs, but not beta3- or beta4-containing receptors, mediate the enhancing effects of nicotine on contextual learning and confirm previous studies implicating beta2 in other forms of learning. A new role for alpha7 nAChRs in regulating sensitivity to the cognitive disrupting effects of ethanol is proposed.


Subject(s)
Ethanol/pharmacology , Fear/physiology , Nicotine/pharmacology , Receptors, Nicotinic/physiology , Animals , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Learning/drug effects , Learning/physiology , Mice , Mice, Knockout , Mutation , Neurons
5.
Genes Brain Behav ; 3(1): 51-62, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14960015

ABSTRACT

Dvl1 is one of three murine Dishevelled genes widely expressed in embryonic development and in the adult central nervous system. Dishevelled proteins are a necessary component of the Wnt and planar cell polarity developmental signaling pathways. We reported previously that mice deficient in Dvl1 exhibited abnormal social interaction and sensorimotor gating. To assess the validity of our earlier findings, we replicated the previous behavioral tests and included several new assays. The behaviors assessed included: social interaction, sensorimotor reflexes, motor activity, nociception, prepulse inhibition of acoustic startle (PPI) and learning and memory. Assessments with an explicit social component included: social dominance test, whisker trimming, nest building, home-cage huddling and ultrasonic vocalization rate analysis in pups. In addition, separate cohorts of wildtype and Dvl1-null mice were assessed for social recognition of a conspecific. Replicating the original report, Dvl1-null mice were impaired in several tasks containing an explicit social component. However, no impairment was observed in the social memory task. A previously observed deficit in PPI did not replicate in two institutions. In conclusion, we provide evidence that the social interaction phenotype of Dvl1-deficient mice has a strong genetic influence, but the sensorimotor gating deficit was subject to environmental influences. The specificity of observed social interaction deficits also suggests that lack of Dvl1 is associated with deficits in the recognition of social hierarchy and dominance.


Subject(s)
Neural Inhibition/genetics , Phosphoproteins/genetics , Reflex, Startle/genetics , Social Behavior , Adaptor Proteins, Signal Transducing , Animals , Behavior, Animal/physiology , Dishevelled Proteins , Female , Gene Expression Regulation, Developmental , Male , Mice , Mice, Mutant Strains , Nesting Behavior/physiology , Phosphoproteins/physiology , Psychomotor Performance/physiology , Vibrissae
6.
Mol Psychiatry ; 7(10): 1039-53, 2002.
Article in English | MEDLINE | ID: mdl-12476318

ABSTRACT

Prepulse inhibition (PPI) is the phenomenon in which a weak prepulse stimulus attenuates the response to a subsequent startling stimulus. Patients with schizophrenia and some other neuropsychiatric disorders have impaired PPI. Impaired PPI in these patient populations is thought to reflect dysfunctional sensorimotor gating mechanisms. Recently, various inbred mouse strains and genetically modified mouse lines have been examined to investigate the potential genetic basis of sensorimotor gating. This review provides a synopsis of the use of mouse models to explore genetic and neurochemical influences on PPI. Studies describing the PPI responses of various inbred strains of mice, mice with genetic mutations, and mice treated with various drugs prior to July 2001 are reviewed. The continuous nature of the distribution of PPI responses among inbred strains of mice indicates that PPI is a polygenic trait. Findings from spontaneous and gene-targeted mutants suggest that mutant mice are important tools for dissecting and studying the role of single genes and their products, and chromosomal regions in regulating PPI. Pharmacological studies of PPI have typically confirmed effects in mice that are similar to those reported previously in rats, with some important exceptions. The use of mice to study PPI is increasing at a dramatic rate and is helping to increase our understanding of the biological basis for sensorimotor gating.


Subject(s)
Mutation , Reflex, Startle/genetics , Animals , Brain/physiology , Brain/physiopathology , DiGeorge Syndrome/genetics , Humans , Mice , Mice, Neurologic Mutants , Models, Animal , Models, Genetic , Receptors, Serotonin/genetics , Receptors, Serotonin/physiology , Schizophrenia/genetics
7.
Hum Mol Genet ; 10(23): 2645-50, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11726551

ABSTRACT

Del22q11 syndrome is caused by heterozygous deletion of an approximately 3 Mb segment of chromosome 22q11.2. Children diagnosed with del22q11 syndrome commonly have learning difficulties, deficits of motor development, cognitive defects and attention deficit disorder. They also have a higher than normal risk for developing psychiatric disorders, mainly schizophrenia, schizoaffective disorder and bipolar disorder. Here, we show that mice that are heterozygously deleted for a subset of the genes that are deleted in patients have deficits in sensorimotor gating and learning and memory. The finding of sensorimotor gating deficits is particularly significant because patients with schizophrenia and schizotypal personality disorder show similar deficits. Thus, our deletion mouse models at least two major features of the del22q11-associated behavioral phenotype, and as such, represents an animal model of this complex behavioral phenotype. These findings not only open the way to pharmacological analyses that may lead to improved treatments, but also to the identification of gene/s that modulate these specific behaviors in humans.


Subject(s)
Abnormalities, Multiple/genetics , Chromosome Deletion , DiGeorge Syndrome/genetics , Abnormalities, Multiple/pathology , Abnormalities, Multiple/physiopathology , Animals , Behavior, Animal/physiology , Chromosomes, Human, Pair 22/genetics , DiGeorge Syndrome/physiopathology , Disease Models, Animal , Female , Gait Disorders, Neurologic/physiopathology , Genotype , Humans , Learning Disabilities/physiopathology , Male , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Mutant Strains , Reflex, Startle/physiology , Syndrome
8.
Psychopharmacology (Berl) ; 156(2-3): 284-90, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11549230

ABSTRACT

RATIONALE: Prepulse inhibition (PPI) of the startle response in mice is increasingly used as a paradigm of sensory gating with potential predictive and construct validity towards schizophrenia. OBJECTIVES: Establishment of a mouse PPI paradigm in which typical and atypical antipsychotic drugs directly improve a low performance PPI. METHODS: Three strains of mice--C57Bl/6J, 129S6/SvEvTac and DBA/2J--were tested in a startle paradigm with three prepulse intensities, 2, 4 and 8 dB above background. RESULTS: Under these conditions, risperidone (0, 0.25, 0.5 and 1 mg/kg i.p.) and clozapine (0, 1, 3 and 9 mg/kg i.p.) improved PPI in all three strains, with order of effect in DBA/2J > 129S6SvEvTac > C57Bl/6J. The DBA/2J strain showed larger PPI-enhancing effects, without disturbing the basal startle response. Two alpha7 nicotinic receptor agonists, GTS-21 (1-10 mg/kg i.p.) and AR-R17779 (1-10 mg/kg i.p.) were inactive in the PPI procedure in DBA/2J mice. CONCLUSIONS: DBA/2J mice were very sensitive to the antipsychotic-like effects of atypical (clozapine) and typical (risperidone) antipsychotics, and this strain is proposed as a model to directly measure sensory gating properties of drugs. Alpha7 Nicotinergic receptor agonists were ineffective in this PPI paradigm.


Subject(s)
Antipsychotic Agents/pharmacology , Reflex, Startle/drug effects , Animals , Benzylidene Compounds/pharmacology , Bridged-Ring Compounds/pharmacology , Clozapine/pharmacology , Male , Mice , Mice, Inbred DBA , Models, Psychological , Nicotinic Agonists/pharmacology , Pyridines/pharmacology , Receptors, Nicotinic/drug effects , Risperidone/pharmacology , Spiro Compounds/pharmacology , alpha7 Nicotinic Acetylcholine Receptor
9.
Physiol Behav ; 73(5): 705-17, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11566205

ABSTRACT

Our laboratory uses a specific test battery for the initial assessment of phenotypic behavioral differences of transgenic, knockout, and inbred strains of mice. Our standard battery includes: open field activity, light-dark exploration, rotarod, prepulse inhibition (PPI), acoustic startle habituation, conditioned fear, Morris water maze, and hot plate. Tests are run in the order listed, from least invasive to most invasive, to decrease the chance that behavioral responses are altered by prior test history. The studies presented here were designed around two questions. The first study asks if differences exist between mice that have undergone testing on different tasks and mice that are naïve to the test experience. The second study asks if the test order affects how an animal performs on subsequent tests. In the first experiment, C57BL/6J male mice were evaluated on all of the tests described above. The behavior of these 'test battery' mice was compared to aged matched naïve mice that were only tested on one test from the battery. Results indicate that on some tests, the behavior of 'test battery' mice was significantly different from the behavior of naïve mice, while on other tests there were no differences. For example, test battery mice responded differently in the open-field, rotarod, and hot-plate test, but behaved similar on the PPI and conditioned fear. Experiments in the second study were performed on male 129/SvEvTac (129S6) and C57BL/6J male mice. An abbreviated battery of tasks was used and the results suggest that certain test variables are sensitive to test order, whereas others are resistant. These two studies demonstrate that some behavioral tests appear to be sensitive to previous testing experience, while other tests are immune.


Subject(s)
Behavior, Animal/physiology , Mice, Inbred Strains/genetics , Mice, Knockout/genetics , Mice, Transgenic/genetics , Phenotype , Animals , Arousal/genetics , Genetics, Behavioral , Male , Mice , Mice, Inbred C57BL , Species Specificity
10.
Physiol Behav ; 73(5): 781-92, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11566211

ABSTRACT

Lhx5 is a member of the LIM homeobox gene family that regulates development of the nervous system. Adult mice generated with a mutation in Lhx5 were found to display absent or disorganized hippocampal neuroanatomy. The pyramidal cell layer in Ammon's horn and the granule cell layer in the dentate gyrus were absent or poorly defined in the hippocampus of adult Lhx5 knockout mice. Behavioral phenotyping of Lhx5 null mutants detected deficits on learning and memory tasks, including the Barnes maze spatial learning task, spontaneous alternation recognition memory, and contextual and cued fear conditioning. General health, neurological reflexes, and sensory abilities appeared to be normal in Lhx5 knockout mice. Motor tests showed impaired performance on some measures of motor activity, coordination, balance, and gait. These results reveal functional outcomes of Lhx5 gene deletion on the integrity of hippocampal neuroanatomy and behavior in the adult mouse.


Subject(s)
Hippocampus/physiology , Homeodomain Proteins/genetics , Mice, Neurologic Mutants/genetics , Motor Activity/physiology , Motor Skills/physiology , Nerve Tissue Proteins/genetics , Animals , Brain Mapping , Female , LIM-Homeodomain Proteins , Male , Mice , Phenotype , Transcription Factors
11.
Brain Res ; 906(1-2): 107-14, 2001 Jul 06.
Article in English | MEDLINE | ID: mdl-11430867

ABSTRACT

Somatostatin was first identified as a hypothalamic factor which inhibits the release of growth hormone from the anterior pituitary (somatotropin release inhibitory factor, SRIF). Both SRIF and its receptors were subsequently found widely distributed within and outside the nervous system, in the adult as well as in the developing organism. Reflecting this wide distribution, somatostatin has been implicated regulating a diverse array of biological processes. These include body growth, homeostasis, sensory perception, autonomous functions, rate of intestinal absorption, behavior, including cognition and memory, and developmental processes. We produced null mutant mice lacking somatostatin through targeted mutagenesis. The mutant mice are healthy, fertile, and superficially indistinguishable from their heterozygous and wildtype littermates. A 'first round' phenotype screen revealed that mice lacking somatostatin have elevated plasma growth hormone levels, despite normal body size, and have elevated basal plasma corticosterone levels. In order to uncover subtle and unexpected differences, we carried out a systematic behavioral phenotype screen which identified a significant impairment in motor learning revealed when increased demands were made on motor coordination. Motor coordination and motor learning require an intact cerebellum. While somatostatin is virtually absent from the adult cerebellum, the ligand and its receptor(s) are transiently expressed at high levels in the developing cerebellum. This result suggests the functional significance of transient expression of SRIF and its receptors in the development of the cerebellum.


Subject(s)
Cerebellum/physiopathology , Learning/physiology , Mice, Neurologic Mutants/physiology , Motor Activity/physiology , Neurons/metabolism , Somatostatin/deficiency , Animals , Cerebellum/metabolism , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiopathology , Mice , Mice, Knockout , Mice, Neurologic Mutants/genetics , Mice, Neurologic Mutants/metabolism , Movement Disorders/genetics , Movement Disorders/metabolism , Movement Disorders/physiopathology , Psychomotor Performance/physiology , Somatostatin/genetics
13.
Learn Mem ; 8(1): 11-9, 2001.
Article in English | MEDLINE | ID: mdl-11160759

ABSTRACT

The extracellular signal-regulated kinases (ERKs) are members of the mitogen-activated protein kinase (MAPK) superfamily of enzymes and have recently garnered considerable attention in the field of learning and memory. ERK activation has been shown to be required for the induction of long-term potentiation (LTP) in the rat hippocampus and for the formation of associative and spatial memories in both the rat and the mouse. However, the individual roles for the two isoforms of ERK have yet to be deciphered. To investigate the specific contribution of the ERK1 (p44) isoform of MAPK to mammalian learning, we performed a general behavioral and physiological characterization of mice lacking the ERK1 gene. The ERK1-null animals demonstrated significantly higher levels of activity in the open field test. However, we observed no other discernible deficits in the ERK1 knockout mice in our behavioral testing. Specifically, no differences were observed in the acquisition or retention (24 h and 2 wk after training) of either contextual or cue fear conditioning between the ERK1(-/-) and their wild-type littermate controls. In addition, no learning phenotype was observed in the passive avoidance test. When hippocampal slices were analyzed, we found no deficits in baseline synaptic transmission or in tetanus-induced LTP in hippocampal area CA1. We found no apparent compensatory changes in the expression of ERK2 (p42 MAPK). We conclude that hippocampus- and amygdala-dependent emotional learning does not depend critically on the activity of ERK1.


Subject(s)
Emotions/physiology , Learning/physiology , Mitogen-Activated Protein Kinases/physiology , Animals , Hippocampus/physiology , Mice , Mice, Knockout/genetics , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/genetics , Motor Activity/physiology , Reference Values , Sensation/physiology
14.
J Neurosci ; 20(24): 9071-85, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11124984

ABSTRACT

Voltage-gated K(+) channels of the Kv3 subfamily have unusual electrophysiological properties, including activation at very depolarized voltages (positive to -10 mV) and very fast deactivation rates, suggesting special roles in neuronal excitability. In the brain, Kv3 channels are prominently expressed in select neuronal populations, which include fast-spiking (FS) GABAergic interneurons of the neocortex, hippocampus, and caudate, as well as other high-frequency firing neurons. Although evidence points to a key role in high-frequency firing, a definitive understanding of the function of these channels has been hampered by a lack of selective pharmacological tools. We therefore generated mouse lines in which one of the Kv3 genes, Kv3.2, was disrupted by gene-targeting methods. Whole-cell electrophysiological recording showed that the ability to fire spikes at high frequencies was impaired in immunocytochemically identified FS interneurons of deep cortical layers (5-6) in which Kv3.2 proteins are normally prominent. No such impairment was found for FS neurons of superficial layers (2-4) in which Kv3.2 proteins are normally only weakly expressed. These data directly support the hypothesis that Kv3 channels are necessary for high-frequency firing. Moreover, we found that Kv3.2 -/- mice showed specific alterations in their cortical EEG patterns and an increased susceptibility to epileptic seizures consistent with an impairment of cortical inhibitory mechanisms. This implies that, rather than producing hyperexcitability of the inhibitory interneurons, Kv3.2 channel elimination suppresses their activity. These data suggest that normal cortical operations depend on the ability of inhibitory interneurons to generate high-frequency firing.


Subject(s)
Cerebral Cortex/physiopathology , Genetic Predisposition to Disease , Neural Inhibition , Neuropeptides/deficiency , Potassium Channels, Voltage-Gated , Potassium Channels/deficiency , Seizures/physiopathology , Action Potentials/genetics , Animals , Behavior, Animal , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Cloning, Molecular , Electroencephalography , Female , Gene Targeting , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Inhibition/drug effects , Neural Inhibition/genetics , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Patch-Clamp Techniques , Phenotype , Potassium Channels/genetics , Potassium Channels/metabolism , Seizures/genetics , Shaw Potassium Channels , Stem Cells , Tetraethylammonium/pharmacology
15.
J Neurosci ; 20(16): 5906-14, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10934237

ABSTRACT

The protein kinase C family of enzymes has been implicated in synaptic plasticity and memory in a wide range of animal species, but to date little information has been available concerning specific roles for individual isoforms of this category of kinases. To investigate the role of the beta isoform of PKC in mammalian learning, we characterized mice deficient in the PKC beta gene using anatomical, biochemical, physiological, and behavioral approaches. In our studies we observed that PKC beta was predominantly expressed in the neocortex, in area CA1 of the hippocampus, and in the basolateral nucleus of the amygdala. Mice deficient in PKC beta showed normal brain anatomy and normal hippocampal synaptic transmission, paired pulse facilitation, and long-term potentiation and normal sensory and motor responses. The PKC beta knock-out animals exhibited a loss of learning, however; they suffered deficits in both cued and contextual fear conditioning. The PKC expression pattern and behavioral phenotype in the PKC beta knock-out animals indicate a critical role for the beta isoform of PKC in learning-related signal transduction mechanisms, potentially in the basolateral nucleus of the amygdala.


Subject(s)
Avoidance Learning/physiology , Brain/metabolism , Conditioning, Psychological/physiology , Fear/physiology , Isoenzymes/genetics , Memory/physiology , Protein Isoforms/genetics , Protein Kinase C/genetics , Amygdala/cytology , Amygdala/metabolism , Animals , Behavior, Animal/physiology , Brain/cytology , Electrophysiology , Hippocampus/cytology , Hippocampus/metabolism , Isoenzymes/metabolism , Long-Term Potentiation/physiology , Mice , Mice, Knockout , Neuronal Plasticity/physiology , Protein Isoforms/metabolism , Protein Kinase C/metabolism , Protein Kinase C beta , Synaptic Transmission/physiology
16.
Hum Mol Genet ; 9(8): 1145-59, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10767339

ABSTRACT

Fragile X syndrome is a common cause of mental retardation involving loss of expression of the FMR1 gene. The role of FMR1 remains undetermined but the protein appears to be involved in RNA metabolism. Fmr1 knockout mice exhibit a phenotype with some similarities to humans, such as macroorchidism and behavioral abnormalities. As a step toward understanding the function of FMR1 and the determination of the potential for therapeutic approaches to fragile X syndrome, yeast artificial chromosome (YAC) transgenic mice were generated in order to determine whether the Fmr1 knockout mouse phenotype could be rescued. Several transgenic lines were generated that carried the entire FMR1 locus with extensive amounts of flanking sequence. We observed that the YAC transgene supported production of the human protein (FMRP) which was present at levels 10 to 15 times that of endogenous protein and was expressed in a cell- and tissue-specific manner. Macro-orchidism was absent in knockout mice carrying the YAC transgene indicating functional rescue by the human protein. Given the complex behavioral phenotype in fragile X patients and the mild phenotype previously reported for the Fmr1 knockout mouse, we performed a more thorough evaluation of the Fmr1 knockout phenotype using additional behavioral assays that had not previously been reported for this animal model. The mouse displayed reduced anxiety-related responses with increased exploratory behavior. FMR1 YAC transgenic mice overexpressing the human protein did produce opposing behavioral responses and additional abnormal behaviors were also observed. These findings have significant implications for gene therapy for fragile X syndrome since overexpression of the gene may harbor its own phenotype.


Subject(s)
Fragile X Syndrome/genetics , Nerve Tissue Proteins/physiology , RNA-Binding Proteins , Animals , Brain/metabolism , Chromosomes, Artificial, Yeast , Exons , Fragile X Mental Retardation Protein , Fragile X Syndrome/therapy , Genetic Therapy , Humans , Male , Mental Disorders/genetics , Mice , Mice, Knockout , Mice, Transgenic , Motor Activity , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Phenotype , Recombinant Proteins/biosynthesis , Testis/metabolism
17.
Hum Mol Genet ; 9(4): 477-88, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10699171

ABSTRACT

Microphthalmia with linear skin defects (MLS) is an X-linked dominant, male-lethal syndrome characterized by microphthalmia, aplastic skin and agenesis of the corpus callosum, and is caused by the deletion of a 500 kb critical region in Xp22.3. Our laboratory isolated a novel rho GTPase-activating protein (rhoGAP) gene named ARHGAP6 from the MLS region. ARHGAP6 contains 14 exons encoding a 974 amino acid protein with three putative SH3-binding domains. Because exons 2-14 are deleted in all MLS patients, we hypothesized that ARHGAP6 may be responsible for some of the phenotypic features of MLS. We pursued two approaches to study the function of ARHGAP6 and its role in the pathogenesis of MLS: gene targeting of the rhoGAP domain in mouse embryonic stem cells and in vitro expression studies. Surprisingly, loss of the rhoGAP function of Arhgap6 does not cause any detectable phenotypic or behavioral abnormalities in the mutant mice. Transfected mammalian cells expressing ARHGAP6 lose their actin stress fibers, retract from the growth surface and extend thin, branching processes resembling filopodia. The ARHGAP6 protein co-localizes with actin filaments through an N-terminal domain and recruits F-actin into the growing processes. Mutation of a conserved arginine residue in the rhoGAP domain prevents the loss of stress fibers but has little effect on process outgrowth. These results suggest that ARHGAP6 has two independent functions: one as a GAP with specificity for RhoA and the other as a cytoskeletal protein that promotes actin remodeling.


Subject(s)
GTPase-Activating Proteins/physiology , rhoA GTP-Binding Protein/physiology , Actins/metabolism , Actins/physiology , Alternative Splicing , Amino Acid Sequence , Animals , Behavior, Animal , Cytoplasm/physiology , Cytoskeleton/metabolism , Cytoskeleton/physiology , Exons , Female , GTPase-Activating Proteins/deficiency , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/isolation & purification , Humans , Introns , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microphthalmos/genetics , Microphthalmos/pathology , Microphthalmos/physiopathology , Molecular Sequence Data , Muscle, Skeletal/abnormalities , Muscle, Skeletal/pathology , Peptide Fragments/physiology , cdc42 GTP-Binding Protein/physiology , rac1 GTP-Binding Protein/physiology , rhoA GTP-Binding Protein/deficiency , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/isolation & purification
18.
Learn Mem ; 6(5): 478-90, 1999.
Article in English | MEDLINE | ID: mdl-10541468

ABSTRACT

Although the biochemical mechanisms underlying learning and memory have not yet been fully elucidated, mounting evidence suggests that activation of protein kinases and phosphorylation of their downstream effectors plays a major role. Recent findings in our laboratory have shown a requirement for the mitogen-activated protein kinase (MAPK) cascade in hippocampal synaptic plasticity. Therefore, we used an inhibitor of MAPK activation, SL327, to test the role of the MAPK cascade in hippocampus-dependent learning in mice. SL327, which crosses the blood-brain barrier, was administered intraperitoneally at several concentrations to animals prior to cue and contextual fear conditioning. Administration of SL327 completely blocked contextual fear conditioning and significantly attenuated cue learning when measured 24 hr after training. To determine whether MAPK activation is required for spatial learning, we administered SL327 to mice prior to training in the Morris water maze. Animals treated with SL327 exhibited significant attenuation of water maze learning; they took significantly longer to find a hidden platform compared with vehicle-treated controls and also failed to use a selective search strategy during subsequent probe trials in which the platform was removed. These impairments cannot be attributed to nonspecific effects of the drug during the training phase; no deficit was seen in the visible platform task, and injection of SL327 following training produced no effect on the performance of these mice in the hidden platform task. These findings indicate that the MAPK cascade is required for spatial and contextual learning in mice.


Subject(s)
MAP Kinase Signaling System/physiology , Maze Learning/physiology , Mitogen-Activated Protein Kinase Kinases/metabolism , Animals , Conditioning, Operant/physiology , Cues , Enzyme Activation , Enzyme Inhibitors/pharmacology , Fear , Mice
19.
Learn Mem ; 6(5): 521-37, 1999.
Article in English | MEDLINE | ID: mdl-10541472

ABSTRACT

Heterozygous mutation or deletion of Pafab1b1 (LIS1) in humans is associated with syndromes with type 1 lissencephaly, a severe brain developmental disorder resulting from abnormal neuronal migration. We have created Lis1 heterozygous mutant mice by gene targeting. Heterozygous mutant mice are viable and fertile, but display global organizational brain defects as a result of impaired neuronal migration. To assess the functional impact of the mutation, Lis1 heterozygous mice and their wild-type littermates were evaluated on a wide variety of behavioral tests. Lis1 mutant mice displayed abnormal hindpaw clutching responses and were impaired on a rotarod test. Lis1 heterozygous mice were also impaired in the spatial learning version of the Morris water task. Impaired motor behavior and spatial learning and memory in Lis1 mutant mice indicates that impaired neuronal migration can have functional effects on complex behavioral responses. The behavioral findings also support the use of the Lis1 mutant mice as a model from human type 1 lissencephaly.


Subject(s)
Brain/abnormalities , Heterozygote , Learning Disabilities/physiopathology , Microtubule-Associated Proteins/genetics , Motor Activity/physiology , 1-Alkyl-2-acetylglycerophosphocholine Esterase , Acoustic Stimulation , Animals , Developmental Disabilities/genetics , Disease Models, Animal , Female , Humans , Male , Maze Learning/physiology , Mice , Mice, Mutant Strains , Neurologic Examination , Phenotype , Psychomotor Performance/physiology , Reflex, Startle/physiology
20.
Hum Mol Genet ; 8(12): 2229-37, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10545603

ABSTRACT

Deletions or rearrangements of human chromosome 22q11 lead to a variety of related clinical syndromes such as DiGeorge syndrome (DGS) and velo--cardiofacial syndrome (VCFS). In addition, patients with 22q11 deletions have an increased incidence of schizophrenia and several studies have mapped susceptibility loci for schizophrenia to this region. Human molecular genetic studies have so far failed to identify the crucial genes or disruption mechanisms that result in these disorders. We have used gene targeting in the mouse to delete a defined region within the conserved DGS critical region (DGCR) on mouse chromosome 16 to prospectively investigate the role of the mouse DGCR in 22q11 syndromes. The deletion spans a conserved portion ( approximately 150 kb) of the proximal region of the DGCR, containing at least seven genes ( Znf74l, Idd, Tsk1, Tsk2, Es2, Gscl and Ctp ). Mice heterozygous for this deletion display no findings of DGS/VCFS in either inbred or mixed backgrounds. However, heterozygous mice display an increase in prepulse inhibition of the startle response, a manifestation of sensorimotor gating that is reduced in humans with schizophrenia. Homozygous deleted mice die soon after implantation, demonstrating that the deleted region contains genes essential for early post-implantation embryonic development. These results suggest that heterozygous deletion of this portion of the DGCR is sufficient for sensorimotor gating abnormalities, but not sufficient to produce the common features of DGS/VCFS in the mouse.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 22 , DiGeorge Syndrome/genetics , Animals , Base Sequence , DNA Primers , Heterozygote , Humans , Male , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...