Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
BMC Genomics ; 16: 1079, 2015 Dec 21.
Article in English | MEDLINE | ID: mdl-26691352

ABSTRACT

BACKGROUND: The YAC128 model of Huntington's disease (HD) shows substantial deficits in motor, learning and memory tasks and alterations in its transcriptional profile. We examined the changes in the transcriptional profile in the YAC128 mouse model of HD at 6, 12 and 18 months and compared these with those seen in other models and human HD caudate. RESULTS: Differential gene expression by genotype showed that genes related to neuronal function, projection outgrowth and cell adhesion were altered in expression. A Time-course ANOVA revealed that genes downregulated with increased age in wild-type striata were likely to be downregulated in the YAC128 striata. There was a substantial overlap of concordant gene expression changes in the YAC128 striata compared with those in human HD brain. Changes in gene expression over time showed fewer striatal YAC128 RNAs altered in abundance than in the HdhQ150 striata but there was a very marked overlap in transcriptional changes at all time points. Despite the similarities in striatal expression changes at 18 months the HdhQ150 mice showed widespread mHTT and ubiquitin positive inclusion staining in the striatum whereas this was absent in the YAC128 striatum. CONCLUSIONS: The gene expression changes in YAC128 striata show a very closely matched profile to that of HdhQ150 striata and are already significantly different between genotypes by six months of age, implying that the temporal molecular gene expression profiles of these models match very closely, despite differences in the prevalence of brain inclusion formation between the models. The YAC128 gene expression changes appear to correlate well with gene expression differences caused by ageing. A relatively small number of genes showed significant differences in expression between the striata of the two models and these could explain some of the phenotypic differences between the models.


Subject(s)
Brain/growth & development , Huntington Disease/epidemiology , Huntington Disease/genetics , Transcriptome , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Female , Gene Expression Regulation, Developmental , Humans , Huntington Disease/pathology , Male , Mice , Prevalence
2.
Neurobiol Learn Mem ; 105: 20-30, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23850596

ABSTRACT

It is well-known that Notch signaling plays a critical role in brain development and growing evidence implicates this signaling pathway in adult synaptic plasticity and memory formation. The Notch1 receptor is activated by two subclasses of ligands, Delta-like (including Dll1 and Dll4) and Jagged (including Jag1 and Jag2). Ligand-induced Notch1 receptor signaling is modulated by a family of Fringe proteins, including Lunatic fringe (Lfng). Although Dll1, Jag1 and Lfng are critical regulators of Notch signaling, their relative contribution to memory formation in the adult brain is unknown. To investigate the roles of these important components of Notch signaling in memory formation, we examined spatial and fear memory formation in adult mice with reduced expression of Dll1, Jag1, Lfng and Dll1 plus Lfng. We also examined motor activity, anxiety-like behavior and sensorimotor gating using the acoustic startle response in these mice. Of the lines of mutant mice tested, we found that only mice with reduced Jag1 expression (mice heterozygous for a null mutation in Jag1, Jag1(+/-)) showed a selective impairment in spatial memory formation. Importantly, all other behavior including open field activity, conditioned fear memory (both context and discrete cue), acoustic startle response and prepulse inhibition, was normal in this line of mice. These results provide the first in vivo evidence that Jag1-Notch signaling is critical for memory formation in the adult brain.

3.
Neurobiol Learn Mem ; 103: 39-49, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23567106

ABSTRACT

It is well-known that Notch signaling plays a critical role in brain development and growing evidence implicates this signaling pathway in adult synaptic plasticity and memory formation. The Notch1 receptor is activated by two subclasses of ligands, Delta-like (including Dll1 and Dll4) and Jagged (including Jag1 and Jag2). Ligand-induced Notch1 receptor signaling is modulated by a family of Fringe proteins, including Lunatic fringe (Lfng). Although Dll1, Jag1 and Lfng are critical regulators of Notch signaling, their relative contribution to memory formation in the adult brain is unknown. To investigate the roles of these important components of Notch signaling in memory formation, we examined spatial and fear memory formation in adult mice with reduced expression of Dll1, Jag1, Lfng and Dll1 plus Lfng. We also examined motor activity, anxiety-like behavior and sensorimotor gating using the acoustic startle response in these mice. Of the lines of mutant mice tested, we found that only mice with reduced Jag1 expression (mice heterozygous for a null mutation in Jag1, Jag1(+/-)) showed a selective impairment in spatial memory formation. Importantly, all other behavior including open field activity, conditioned fear memory (both context and discrete cue), acoustic startle response and prepulse inhibition, was normal in this line of mice. These results provide the first in vivo evidence that Jag1-Notch signaling is critical for memory formation in the adult brain.


Subject(s)
Calcium-Binding Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Memory/physiology , Receptors, Notch/metabolism , Signal Transduction/physiology , Space Perception/physiology , Animals , Anxiety/genetics , Anxiety/metabolism , Calcium-Binding Proteins/genetics , Fear/physiology , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein , Jagged-2 Protein , Membrane Proteins/genetics , Mice , Motor Activity/physiology , Receptors, Notch/genetics , Reflex, Startle/genetics , Serrate-Jagged Proteins
4.
Front Behav Neurosci ; 7: 209, 2013.
Article in English | MEDLINE | ID: mdl-24391565

ABSTRACT

Neurons may compete against one another for integration into a memory trace. Specifically, neurons in the lateral nucleus of the amygdala with relatively higher levels of cAMP Responsive Element Binding Protein (CREB) seem to be preferentially allocated to a fear memory trace, while neurons with relatively decreased CREB function seem to be excluded from a fear memory trace. CREB is a ubiquitous transcription factor that modulates many diverse cellular processes, raising the question as to which of these CREB-mediated processes underlie memory allocation. CREB is implicated in modulating dendritic spine number and morphology. As dendritic spines are intimately involved in memory formation, we investigated whether manipulations of CREB function alter spine number or morphology of neurons at the time of fear conditioning. We used viral vectors to manipulate CREB function in the lateral amygdala (LA) principal neurons in mice maintained in their homecages. At the time that fear conditioning normally occurs, we observed that neurons with high levels of CREB had more dendritic spines, while neurons with low CREB function had relatively fewer spines compared to control neurons. These results suggest that the modulation of spine density provides a potential mechanism for preferential allocation of a subset of neurons to the memory trace.

5.
Brain Res Bull ; 88(2-3): 121-9, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-21624441

ABSTRACT

People with HD have a demonstrated early extra-dimensional set-shifting deficit. In the present study, we use a novel water T-maze set-shifting procedure and demonstrate its validity as a set-shifting task in a mouse model of Huntington's disease. Three groups of YAC128 mice of different ages (27, 69 and 117 weeks) were run on the task, which incorporated six distinct stages in which the mice must learn a rule and then switch to a different rule. The six stages were: directional learning, directional learning reversal, light discrimination, light discrimination reversal, return to place learning and a maze rotation spatial learning test. Rule changes from place learning to light discrimination and back constitute extra-dimensional shifts. The results of the study demonstrate robust light/dark discrimination reversal learning deficits in transgenic mice from 27 weeks of age, and a directional learning to light discrimination extra-dimensional set-shifting deficit from 69 weeks of age. The extra-dimensional shift deficit was confirmed with control trials demonstrating the validity of the deficit and the task. The onset of reversal learning and extra-dimensional shift deficits corresponded with the development of mutant huntingtin N-terminal fragment aggregates in neurons of relevant forebrain regions.


Subject(s)
Behavior, Animal , Huntington Disease/physiopathology , Animals , Brain/pathology , Brain/physiopathology , Discrimination Learning , Disease Models, Animal , Female , Huntington Disease/genetics , Huntington Disease/pathology , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Reversal Learning
6.
Brain Res Bull ; 88(2-3): 199-209, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22001697

ABSTRACT

Substantial transcriptional changes are seen in Huntington's disease (HD) brain and parallel early changes in gene expression are observed in mouse models of HD. Analysis of behaviour in such models also shows substantial deficits in motor, learning and memory tasks. We examined the changes in the transcriptional profile in the HdhQ150 mouse model of HD at 6, 12 and 18 months and correlated these changes with the behavioural tasks the animals had undertaken. Changes in gene expression over time showed a significant enrichment of RNAs altered in abundance that related to cognition in both HdhQ150 and wild-type animals. The most significantly down-regulated mRNA between genotypes over the whole time-course was Htt itself. Other changes between genotypes identified at 6 months related to chromatin organization and structure, whilst at 18 months changes related mainly to intracellular signalling. Correlation of the changes in gene product abundance with phenotypic changes revealed that weight and detection of the opposite position of the platform in the water maze seemed to correlate with the chromatin alterations whereas changes in the rotarod performance related mainly to intracellular signalling and homeostasis. These results implicate alterations in specific molecular pathways that may underpin changes in different behavioural tasks.


Subject(s)
Behavior, Animal/physiology , Gene Expression , Huntington Disease/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Visual Cortex/metabolism , Animals , Disease Models, Animal , Female , Huntingtin Protein , Huntington Disease/metabolism , Longitudinal Studies , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
7.
Brain Res Bull ; 88(2-3): 148-55, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-20457229

ABSTRACT

Huntington's disease is caused by a single mutation resulting in an expanded polyglutamine sequence which causes the production of a mutant variant of the protein huntingtin, which is ultimately responsible for the motor, cognitive and emotional symptoms and early death of the individual. Several mouse models have been created that seek to recapitulate the features of the disease. The present study sought to characterise the Hdh(Q92) mouse line longitudinally, to determine the nature, extent and age of onset of any behavioural deficits. On each of the tests used the Hdh(Q92/Q92) mice demonstrated poorer performance than their wildtype littermates, and these performance deficits were age dependent. Of the tests applied acoustic startle and prepulse inhibition proved to be the most sensitive with differences between the mouse groups appearing ∼4 months of age, an age where grip strength differences were also found. Male Hdh(Q92/Q92) mice started losing weight relative their wildtype littermates from 10 months of age, and water maze performance began to deteriorate from 14 months. There were slight differences in rotarod ability with advancing age, with the Hdh(Q92/Q92) demonstrating greater variability in performance than their wildtype littermates. Analysis of body weight and the initial stage of the water maze procedure produced clear between group differences, whereas the grip strength, rotarod and acoustic startle tests demonstrated significance only when age was a factor in the analyses, suggesting that changes in the pattern of performance over time were responsible for the differences, rather than overall group effects per se. The results highlight the necessity for the longitudinal assessment of mouse lines to detect subtle behavioural differences experimental groups.


Subject(s)
Behavior, Animal/physiology , Huntington Disease/physiopathology , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Aging , Animals , Disease Models, Animal , Female , Gene Knock-In Techniques , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/metabolism , Longitudinal Studies , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/physiology , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Phenotype , Psychomotor Performance/physiology , Sex Characteristics
8.
Brain Res Bull ; 88(2-3): 182-8, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-20457230

ABSTRACT

In people with Huntington's disease, an expanded CAG repeat sequence on the HTT gene confers a toxic gain function resulting in a progressive and fatal neurodegeneration. The Hdh((CAG)Q150) Huntington's disease mouse line is a knock-in model of the disease that carries ∼150 CAG repeats on the normal mouse Htt locus. To determine that these mice are a useful model of the disease, they were assessed longitudinally for motor and cognitive deficits relevant to the human disease state. Each test was conducted bi-monthly across the lifespan of the animal. The results indicate that the Hdh(Q150/Q150) mice were impaired on each of the measures used, with deficits appearing on a 3-stage water maze test at 4 months of age and on prepulse inhibition at 6 months of age, both of which were prior to the manifestation of motor abnormalities. Grip strength, as measured by the inverted cage lid test, was reduced in the Hdh(Q150/Q150) mice from 10 months of age, when the male mice also exhibited weight loss relative to their wildtype littermates. On the accelerating rotarod, deficits in the carrier mice did not appear until they were 21 months old. Our results demonstrate that the Hdh((CAG)150) is a valid model of HD that displays early and progressive cognitive deficits that precede the onset of motor abnormalities.


Subject(s)
Behavior, Animal/physiology , Huntington Disease/physiopathology , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Brain/metabolism , Brain/physiology , Disease Models, Animal , Female , Gene Knock-In Techniques , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/metabolism , Longitudinal Studies , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Phenotype
9.
Brain Res Bull ; 88(2-3): 113-20, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-20460143

ABSTRACT

To determine the suitability of mouse models of disease for therapeutic trials, the models must be characterised to determine their similarity to the human condition, and utility for specific therapeutic approaches. The YAC128 mouse model of HD has been bred on to C57BL/6J background in order to provide a mouse model of the disease better suited to behavioural testing, than the visually impaired original line on the FVB background. In the present study, the C57BL/6J YAC128 mice were assessed on several behavioural tasks bi-monthly between 4 and 24 months of age. On the rotarod early and stable deficits were demonstrated in the YAC128 mice from 4 months of age indicating an early abnormality in motor coordination. Early and stable deficits were also found on the balance beam measures of latency to orientate towards the beam and time to traverse it. Measures of fore and hind limb footslips on the balance beam demonstrated early and progressive limb use deficits in the YAC128 mice. On a 3-stage Morris water maze protocol, the YAC128 mice took longer and travelled further to find the hidden platform in each of the 3 locations, indicative of a spatial learning deficit. The YAC128 mice were also less reactive to the primary startle stimuli and the effects of the prepulse which may suggest striatal dysfunction. As a measure of general well being, the body weights of the mice were recorded and demonstrated increased weight in the YAC128 mice until 14 months of age, when they became comparable to that of their wildtype littermates. The YAC128 mouse on the C57BL/6J background has an early, robust and severe behavioural phenotype that shares some similarity to human HD symptomatology.


Subject(s)
Behavior, Animal , Huntington Disease/pathology , Huntington Disease/physiopathology , Animals , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Disease Models, Animal , Female , Genotype , Huntington Disease/genetics , Longitudinal Studies , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/genetics , Motor Activity/physiology , Phenotype , Rotarod Performance Test
10.
Circ Res ; 93(6): 531-40, 2003 Sep 19.
Article in English | MEDLINE | ID: mdl-12919952

ABSTRACT

Ca2+ release from the sarcoplasmic reticulum mediated by the cardiac ryanodine receptor (RyR2) is a fundamental event in cardiac muscle contraction. RyR2 mutations suggested to cause defective Ca2+ channel function have recently been identified in catecholaminergic polymorphic ventricular tachycardia (CPVT) and arrhythmogenic right ventricular dysplasia (ARVD) affected individuals. We report expression of three CPVT-linked human RyR2 (hRyR2) mutations (S2246L, N4104K, and R4497C) in HL-1 cardiomyocytes displaying correct targeting to the endoplasmic reticulum. N4104K also localized to the Golgi apparatus. Phenotypic characteristics including intracellular Ca2+ handling, proliferation, viability, RyR2:FKBP12.6 interaction, and beat rate in resting HL-1 cells expressing mutant hRyR2 were indistinguishable from wild-type (WT) hRyR2. However, Ca2+ release was augmented in cells expressing mutant hRyR2 after RyR activation (caffeine and 4-chloro-m-cresol) or beta-adrenergic stimulation (isoproterenol). RyR2:FKBP12.6 interaction remained intact after caffeine or 4-CMC activation, but was dramatically disrupted by isoproterenol or forskolin, an activator of adenylate cyclase. Isoproterenol and forskolin elevated cyclic-AMP to similar magnitudes in all cells and were associated with equivalent hyperphosphorylation of mutant and WT hRyR2. CPVT-linked mutations in hRyR2 did not alter resting cardiomyocyte phenotype but mediated augmented Ca2+ release on RyR-agonist or beta-AR stimulation. Furthermore, equivalent interaction between mutant and WT hRyR2 and FKBP12.6 was demonstrated.


Subject(s)
Calcium/metabolism , Mutation , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Adenylyl Cyclases/metabolism , Adrenergic beta-Agonists/pharmacology , Caffeine/pharmacology , Cell Line , Cresols/pharmacology , Humans , Isoproterenol/pharmacology , Myocytes, Cardiac/drug effects , Phenotype , Ryanodine Receptor Calcium Release Channel/metabolism , Stress, Physiological/complications , Tachycardia, Ventricular/etiology , Tachycardia, Ventricular/genetics , Tacrolimus Binding Proteins/metabolism
11.
J Biol Chem ; 278(31): 28856-64, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12754204

ABSTRACT

Ca2+ homeostasis is a vital cellular control mechanism in which Ca2+ release from intracellular stores plays a central role. Ryanodine receptor (RyR)-mediated Ca2+ release is a key modulator of Ca2+ homeostasis, and the defective regulation of RyR is pathogenic. However, the molecular events underlying RyR-mediated pathology remain undefined. Cells stably expressing recombinant human RyR2 (Chinese hamster ovary cells, CHOhRyR2) had similar resting cytoplasmic Ca2+ levels ([Ca2+]c) to wild-type CHO cells (CHOWT) but exhibited increased cytoplasmic Ca2+ flux associated with decreased cell viability and proliferation. Intracellular Ca2+ flux increased with human RyR2 (hRyR2) expression levels and determined the extent of phenotypic modulation. Co-expression of FKBP12.6, but not FKBP12, or incubation of cells with ryanodine suppressed intracellular Ca2+ flux and restored normal cell viability and proliferation. Restoration of normal phenotype was independent of the status of resting [Ca2+]c or ER Ca2+ load. Heparin inhibition of endogenous inositol trisphosphate receptors (IP3R) had little effect on intracellular Ca2+ handling or viability. However, purinergic stimulation of endogenous IP3R resulted in apoptotic cell death mediated by hRyR2 suggesting functional interaction occurred between IP3R and hRyR2 Ca2+ release channels. These data demonstrate that defective regulation of RyR causes altered cellular phenotype via profound perturbations in intracellular Ca2+ signaling and highlight a key modulatory role of FKBP12.6 in hRyR2 Ca2+ channel function.


Subject(s)
Cell Death , Phenotype , Ryanodine Receptor Calcium Release Channel/physiology , Tacrolimus Binding Proteins/physiology , Adenosine Triphosphate/pharmacology , Animals , Apoptosis/drug effects , CHO Cells , Calcium/metabolism , Calcium Channels/drug effects , Cell Division , Cresols/pharmacology , Cricetinae , Cytoplasm/metabolism , Gene Expression , Heparin/pharmacology , Humans , Inositol 1,4,5-Trisphosphate Receptors , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Purinergic P2/drug effects , Receptors, Purinergic P2/physiology , Recombinant Proteins , Ryanodine Receptor Calcium Release Channel/genetics , Signal Transduction , Tacrolimus Binding Proteins/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...