Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Am J Med Genet A ; 191(10): 2508-2517, 2023 10.
Article in English | MEDLINE | ID: mdl-37353954

ABSTRACT

TBCK-related encephalopathy is a rare pediatric neurodegenerative disorder caused by biallelic loss-of-function variants in the TBCK gene. After receiving anecdotal reports of neurologic phenotypes in both human and mouse TBCK heterozygotes, we quantified if TBCK haploinsufficiency causes a phenotype in mice and humans. Using the tbck+/- mouse model, we performed a battery of behavioral assays and mTOR pathway analysis to investigate potential alterations in neurophysiology. We conducted as well a phenome-wide association study (PheWAS) analysis in a large adult biobank to determine the presence of potential phenotypes associated to this variant. The tbck+/- mouse model demonstrates a reduction of exploratory behavior in animals with significant sex and genotype interactions. The concurrent PheWAS analysis of 10,900 unrelated individuals showed that patients with one copy of a TBCK loss-of-function allele had a significantly higher rate of acquired toe and foot deformities, likely indicative of a mild peripheral neuropathy phenotype. This study presents an example of what may be the underappreciated occurrence of mild neurogenic symptoms in heterozygote individuals of recessive neurogenetic syndromes.


Subject(s)
Brain Diseases , Protein Serine-Threonine Kinases , Humans , Child , Animals , Mice , Protein Serine-Threonine Kinases/genetics , Heterozygote , Syndrome , Brain Diseases/genetics , Phenotype
2.
Genet Med ; 25(7): 100861, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37087635

ABSTRACT

PURPOSE: This study aimed to establish variants in CBX1, encoding heterochromatin protein 1ß (HP1ß), as a cause of a novel syndromic neurodevelopmental disorder. METHODS: Patients with CBX1 variants were identified, and clinician researchers were connected using GeneMatcher and physician referrals. Clinical histories were collected from each patient. To investigate the pathogenicity of identified variants, we performed in vitro cellular assays and neurobehavioral and cytological analyses of neuronal cells obtained from newly generated Cbx1 mutant mouse lines. RESULTS: In 3 unrelated individuals with developmental delay, hypotonia, and autistic features, we identified heterozygous de novo variants in CBX1. The identified variants were in the chromodomain, the functional domain of HP1ß, which mediates interactions with chromatin. Cbx1 chromodomain mutant mice displayed increased latency-to-peak response, suggesting the possibility of synaptic delay or myelination deficits. Cytological and chromatin immunoprecipitation experiments confirmed the reduction of mutant HP1ß binding to heterochromatin, whereas HP1ß interactome analysis demonstrated that the majority of HP1ß-interacting proteins remained unchanged between the wild-type and mutant HP1ß. CONCLUSION: These collective findings confirm the role of CBX1 in developmental disabilities through the disruption of HP1ß chromatin binding during neurocognitive development. Because HP1ß forms homodimers and heterodimers, mutant HP1ß likely sequesters wild-type HP1ß and other HP1 proteins, exerting dominant-negative effects.


Subject(s)
Chromobox Protein Homolog 5 , Heterochromatin , Animals , Mice , Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , Histones/genetics , Histones/metabolism
3.
Curr Biol ; 31(23): 5238-5248.e7, 2021 12 06.
Article in English | MEDLINE | ID: mdl-34653361

ABSTRACT

Many aspects of sleep are heritable, but only a few sleep-regulating genes have been reported. Here, we leverage mouse models to identify and confirm a previously unreported gene affecting sleep duration-dihydropyrimidine dehydrogenase (Dpyd). Using activity patterns to quantify sleep in 325 Diversity Outbred (DO) mice-a population with high genetic and phenotypic heterogeneity-a linkage peak for total sleep in the active lights off period was identified on chromosome 3 (LOD score = 7.14). Mice with the PWK/PhJ ancestral haplotype at this location demonstrated markedly reduced sleep. Among the genes within the linkage region, available RNA sequencing data in an independent sample of DO mice supported a highly significant expression quantitative trait locus for Dpyd, wherein reduced expression was associated with the PWK/PhJ allele. Validation studies were performed using activity monitoring and EEG/EMG recording in Collaborative Cross mouse strains with and without the PWK/PhJ haplotype at this location, as well as EEG and EMG recording of sleep and wake in Dpyd knockout mice and wild-type littermate controls. Mice lacking Dpyd had 78.4 min less sleep during the lights-off period than wild-type mice (p = 0.007; Cohen's d = -0.94). There was no difference in other measured behaviors in knockout mice, including assays evaluating cognitive-, social-, and affective-disorder-related behaviors. Dpyd encodes the rate-limiting enzyme in the metabolic pathway that catabolizes uracil and thymidine to ß-alanine, an inhibitory neurotransmitter. Thus, data support ß-alanine as a neurotransmitter that promotes sleep in mice.


Subject(s)
Dihydrouracil Dehydrogenase (NADP) , Sleep , Animals , Dihydrouracil Dehydrogenase (NADP)/genetics , Haplotypes , Mice , Mice, Knockout , Sleep/genetics , beta-Alanine/genetics
4.
Brain ; 143(4): 1114-1126, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32293671

ABSTRACT

Congenital disorders of glycosylation are a growing group of rare genetic disorders caused by deficient protein and lipid glycosylation. Here, we report the clinical, biochemical, and molecular features of seven patients from four families with GALNT2-congenital disorder of glycosylation (GALNT2-CDG), an O-linked glycosylation disorder. GALNT2 encodes the Golgi-localized polypeptide N-acetyl-d-galactosamine-transferase 2 isoenzyme. GALNT2 is widely expressed in most cell types and directs initiation of mucin-type protein O-glycosylation. All patients showed loss of O-glycosylation of apolipoprotein C-III, a non-redundant substrate for GALNT2. Patients with GALNT2-CDG generally exhibit a syndrome characterized by global developmental delay, intellectual disability with language deficit, autistic features, behavioural abnormalities, epilepsy, chronic insomnia, white matter changes on brain MRI, dysmorphic features, decreased stature, and decreased high density lipoprotein cholesterol levels. Rodent (mouse and rat) models of GALNT2-CDG recapitulated much of the human phenotype, including poor growth and neurodevelopmental abnormalities. In behavioural studies, GALNT2-CDG mice demonstrated cerebellar motor deficits, decreased sociability, and impaired sensory integration and processing. The multisystem nature of phenotypes in patients and rodent models of GALNT2-CDG suggest that there are multiple non-redundant protein substrates of GALNT2 in various tissues, including brain, which are critical to normal growth and development.


Subject(s)
Apolipoprotein C-III/blood , Developmental Disabilities/genetics , N-Acetylgalactosaminyltransferases/genetics , Adolescent , Animals , Apolipoprotein C-III/genetics , Child , Child, Preschool , Female , Glycosylation , Humans , Loss of Function Mutation , Male , Mice , Pedigree , Rats , Young Adult , Polypeptide N-acetylgalactosaminyltransferase
5.
Neurosci Lett ; 704: 67-72, 2019 06 21.
Article in English | MEDLINE | ID: mdl-30940476

ABSTRACT

Chronic lithium treatment stimulates adult hippocampal neurogenesis, but whether increased neurogenesis contributes to its therapeutic mechanism remains unclear. We use a genetic model of neural progenitor cell (NPC) ablation to test whether a lithium-sensitive behavior requires hippocampal neurogenesis. NPC-ablated mice were treated with lithium and assessed in the forced swim test (FST). Lithium reduced time immobile in the FST in NPC-ablated and control mice but had no effect on activity in the open field, a control for the locomotion-based FST. These findings show that hippocampal NPCs that proliferate in response to chronic lithium are not necessary for the behavioral response to lithium in the FST. We further show that 4-6 week old immature hippocampal neurons are not required for this response. These data suggest that increased hippocampal neurogenesis does not contribute to the response to lithium in the forced swim test and may not be an essential component of its therapeutic mechanism.


Subject(s)
Hippocampus/drug effects , Lithium Compounds/pharmacology , Neural Stem Cells/drug effects , Neurons/drug effects , Animals , Cell Proliferation/drug effects , Female , Hippocampus/cytology , Hippocampus/physiology , Male , Mice, Inbred C57BL , Motor Activity/drug effects , Neural Stem Cells/cytology , Neurogenesis/drug effects , Neurons/cytology , Neurons/physiology , Swimming
6.
Autism Res ; 10(4): 572-584, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27739237

ABSTRACT

Sleep disturbances and hyperactivity are prevalent in several neurodevelopmental disorders, including autism spectrum disorders (ASDs) and attention deficit-hyperactivity disorder (ADHD). Evidence from genome-wide association studies indicates that chromosomal copy number variations (CNVs) are associated with increased prevalence of these neurodevelopmental disorders. In particular, CNVs in chromosomal region 16p11.2 profoundly increase the risk for ASD and ADHD, disorders that are more common in males than females. We hypothesized that mice hemizygous for the 16p11.2 deletion (16p11.2 del/+) would exhibit sex-specific sleep and activity alterations. To test this hypothesis, we recorded activity patterns using infrared beam breaks in the home-cage of adult male and female 16p11.2 del/+ and wildtype (WT) littermates. In comparison to controls, we found that both male and female 16p11.2 del/+ mice exhibited robust home-cage hyperactivity. In additional experiments, sleep was assessed by polysomnography over a 24-hr period. 16p11.2 del/+ male, but not female mice, exhibited significantly more time awake and significantly less time in non-rapid-eye-movement (NREM) sleep during the 24-hr period than wildtype littermates. Analysis of bouts of sleep and wakefulness revealed that 16p11.2 del/+ males, but not females, spent a significantly greater proportion of wake time in long bouts of consolidated wakefulness (greater than 42 min in duration) compared to controls. These changes in hyperactivity, wake time, and wake time distribution in the males resemble sleep disturbances observed in human ASD and ADHD patients, suggesting that the 16p11.2 del/+ mouse model may be a useful genetic model for studying sleep and activity problems in human neurodevelopmental disorders. Autism Res 2016. © 2016 International Society for Autism Research, Wiley Periodicals, Inc. Autism Res 2017, 10: 572-584. © 2016 International Society for Autism Research, Wiley Periodicals, Inc.


Subject(s)
Autism Spectrum Disorder/diagnosis , Autism Spectrum Disorder/genetics , Chromosome Deletion , Disease Models, Animal , Genetic Predisposition to Disease/genetics , Hyperkinesis/diagnosis , Hyperkinesis/genetics , Models, Genetic , Adult , Animals , Attention Deficit Disorder with Hyperactivity/diagnosis , Attention Deficit Disorder with Hyperactivity/genetics , Circadian Rhythm/genetics , DNA Copy Number Variations , Female , Genome-Wide Association Study , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Polysomnography , Sex Factors , Sleep Stages/genetics
7.
Hum Gene Ther ; 27(11): 906-915, 2016 11.
Article in English | MEDLINE | ID: mdl-27510804

ABSTRACT

Mucopolysaccharidosis type II (MPS II) is a rare X-linked genetic disorder caused by deficiency of the lysosomal enzyme iduronate-2-sulfatase (IDS), leading to impaired catabolism of ubiquitous polysaccharides and abnormal accumulation of these undegraded substrates in the lysosome. Like many lysosomal storage diseases, MPS II is characterized by both somatic and central nervous system (CNS) involvement. Intravenous enzyme replacement therapy can improve somatic manifestations of MPS II, but systemic IDS does not cross the blood-brain barrier and therefore cannot address CNS disease. In this study, an adeno-associated virus serotype 9 vector carrying the IDS gene was injected into the cerebrospinal fluid (CSF) of IDS deficient mice, a model of MPS II. Treated mice exhibited dose-dependent IDS expression and resolution of brain storage lesions, as well as improvement in long-term memory in a novel object recognition test. These findings suggest that delivery of adeno-associated virus vectors into CSF could serve as a platform for efficient, long-term enzyme delivery to the CNS, potentially addressing this critical unmet need for patients with MPS II and many related lysosomal enzyme deficiencies.


Subject(s)
Central Nervous System Diseases/therapy , Dependovirus/genetics , Genetic Therapy , Genetic Vectors/administration & dosage , Glycoproteins/genetics , Iduronidase/genetics , Mucopolysaccharidosis II/physiopathology , Animals , Blood-Brain Barrier , Central Nervous System Diseases/genetics , Cerebrospinal Fluid/metabolism , Disease Models, Animal , Drug Delivery Systems , Enzyme Replacement Therapy , Humans , Male , Mice , Mice, Inbred C57BL , Mucopolysaccharidosis II/cerebrospinal fluid
8.
Front Behav Neurosci ; 8: 349, 2014.
Article in English | MEDLINE | ID: mdl-25339878

ABSTRACT

Classifying behavior patterns in mouse models of neurological, psychiatric and neurodevelopmental disorders is critical for understanding disease causality and treatment. However, complete characterization of behavior is time-intensive, prone to subjective scoring, and often requires specialized equipment. Although several reports describe automated home-cage monitoring and individual task scoring methods, we report the first open source, comprehensive toolbox for automating the scoring of several common behavior tasks used by the neuroscience community. We show this new toolbox is robust and achieves equal or better consistency when compared to manual scoring methods. We use this toolbox to study the alterations in behavior that occur following blast-induced traumatic brain injury (bTBI), and study if these behavior patterns are altered following genetic deletion of the transcription factor Ets-like kinase 1 (Elk-1). Due to the role of Elk-1 in neuronal survival and proposed role in synaptic plasticity, we hypothesized that Elk-1 deletion would improve some neurobehavioral deficits, while impairing others, following blast exposure. In Elk-1 knockout (KO) animals, deficits in open field, spatial object recognition (SOR) and elevated zero maze performance after blast exposure disappeared, while new significant deficits appeared in spatial and associative memory. These are the first data suggesting a molecular mediator of anxiety deficits following bTBI, and represent the utility of the broad screening tool we developed. More broadly, we envision this open-source toolbox will provide a more consistent and rapid analysis of behavior across many neurological diseases, promoting the rapid discovery of novel pathways mediating disease progression and treatment.

9.
Mol Endocrinol ; 28(4): 490-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24552589

ABSTRACT

The regulation of behavior by the molecular components of the circadian clock is not well understood. Here we report that mice lacking the nuclear receptor Rev-erbα, a potent transcriptional repressor and core clock component, displayed marked hyperactivity and impaired response habituation in novel environments. In addition, Rev-erbα knockout (KO) mice were deficient in short-term, long-term, and contextual memories and also showed impairment in nest-building ability. Together, these results suggest that Rev-erbα KO mice manifest defective hippocampal function. Interestingly, the changes in novelty-induced locomotor activity of Rev-erbα KO mice were comparable at multiple times of day, potentially due to the muted amplitude of Rev-erbα oscillation in the hippocampus of wild-type mice. Hippocampal dopamine turnover was increased in Rev-erbα KO mice, due to up-regulation of tyrosine hydroxylase, the rate-limiting enzyme in dopamine production, and pharmacologic inhibition of tyrosine hydroxylase activity partially rescued locomotor hyperactivity. These findings reveal a novel, nonredundant function for Rev-erbα that links a core component of the circadian gene-regulatory network to the control of dopaminergic and hippocampus-dependent behaviors.


Subject(s)
Behavior, Animal , Dopamine/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/deficiency , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Animals , Circadian Rhythm/genetics , Cognition , Exploratory Behavior , Habituation, Psychophysiologic/genetics , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Tyrosine 3-Monooxygenase/metabolism , Up-Regulation/genetics
10.
J Clin Invest ; 121(9): 3756-62, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21821916

ABSTRACT

Lithium is the first-line therapy for bipolar disorder. However, its therapeutic target remains controversial. Candidates include inositol monophosphatases, glycogen synthase kinase-3 (GSK-3), and a ß-arrestin-2/AKT/protein phosphatase 2A (ß-arrestin-2/AKT/PP2A) complex that is known to be required for lithium-sensitive behaviors. Defining the direct target(s) is critical for the development of new therapies and for elucidating the molecular pathogenesis of this major psychiatric disorder. Here, we show what we believe to be a new link between GSK-3 and the ß-arrestin-2 complex in mice and propose an integrated mechanism that accounts for the effects of lithium on multiple behaviors. GSK-3ß (Gsk3b) overexpression reversed behavioral defects observed in lithium-treated mice and similar behaviors observed in Gsk3b+/- mice. Furthermore, immunoprecipitation of striatial tissue from WT mice revealed that lithium disrupted the ß-arrestin-2/Akt/PP2A complex by directly inhibiting GSK-3. GSK-3 inhibitors or loss of one copy of the Gsk3b gene reduced ß-arrestin-2/Akt/PP2A complex formation in mice, while overexpression of Gsk3b restored complex formation in lithium-treated mice. Thus, GSK-3 regulates the stability of the ß-arrestin-2/Akt/PP2A complex, and lithium disrupts the complex through direct inhibition of GSK-3. We believe these findings reveal a new role for GSK-3 within the ß-arrestin complex and demonstrate that GSK-3 is a critical target of lithium in mammalian behaviors.


Subject(s)
Arrestins/metabolism , Behavior, Animal/drug effects , Glycogen Synthase Kinase 3/metabolism , Lithium/pharmacology , Animals , Arrestins/genetics , Behavior, Animal/physiology , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Humans , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , beta-Arrestin 2 , beta-Arrestins
11.
Nat Cell Biol ; 12(10): 1007-13, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20852629

ABSTRACT

Stem cells reside in specialized microenvironments or 'niches' that regulate their function. In vitro studies using hypoxic culture conditions (<5% O2) have revealed strong regulatory links between O2 availability and functions of stem and precursor cells. Although some stem cells are perivascular, others may occupy hypoxic niches and be regulated by O2 gradients. However, the underlying mechanisms remain unclear. Here, we show that hypoxia inducible factor-1α (HIF-1α), a principal mediator of hypoxic adaptations, modulates Wnt/ß-catenin signalling in hypoxic embryonic stem (ES) cells by enhancing ß-catenin activation and expression of the downstream effectors LEF-1 and TCF-1. This regulation extends to primary cells, including isolated neural stem cells (NSCs), and is not observed in differentiated cells. In vivo, Wnt/ß-catenin activity is closely associated with low O2 regions in the subgranular zone of the hippocampus, a key NSC niche. Hif-1α deletion impairs hippocampal Wnt-dependent processes, including NSC proliferation, differentiation and neuronal maturation. This decline correlates with reduced Wnt/ß-catenin signalling in the subgranular zone. O2 availability, therefore, may have a direct role in stem cell regulation through HIF-1α modulation of Wnt/ß-catenin signalling.


Subject(s)
Embryonic Stem Cells/metabolism , Oxygen/metabolism , Signal Transduction , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Apoptosis , Cell Differentiation , Cell Proliferation , Cells, Cultured , Embryonic Stem Cells/cytology , Hepatocyte Nuclear Factor 1-alpha , Hippocampus/cytology , Hippocampus/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lymphoid Enhancer-Binding Factor 1/metabolism , Mice , Neurogenesis , Neurons/cytology , Neurons/metabolism , T Cell Transcription Factor 1/metabolism
12.
J Clin Invest ; 119(12): 3519-29, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19959876

ABSTRACT

Hematopoietic stem cell (HSC) homeostasis depends on the balance between self renewal and lineage commitment, but what regulates this decision is not well understood. Using loss-of-function approaches in mice, we found that glycogen synthase kinase-3 (Gsk3) plays a pivotal role in controlling the decision between self renewal and differentiation of HSCs. Disruption of Gsk3 in BM transiently expanded phenotypic HSCs in a betta-catenin-dependent manner, consistent with a role for Wnt signaling in HSC homeostasis. However, in assays of long-term HSC function, disruption of Gsk3 progressively depleted HSCs through activation of mammalian target of rapamycin (mTOR). This long-term HSC depletion was prevented by mTOR inhibition and exacerbated by betta-catenin knockout. Thus, GSK-3 regulated both Wnt and mTOR signaling in mouse HSCs, with these pathways promoting HSC self renewal and lineage commitment, respectively, such that inhibition of Gsk3 in the presence of rapamycin expanded the HSC pool in vivo. These findings identify unexpected functions for GSK-3 in mouse HSC homeostasis, suggest a therapeutic approach to expand HSCs in vivo using currently available medications that target GSK-3 and mTOR, and provide a compelling explanation for the clinically prevalent hematopoietic effects observed in individuals prescribed the GSK-3 inhibitor lithium.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Hematopoietic Stem Cells/enzymology , Animals , Bone Marrow Transplantation , Carrier Proteins/metabolism , Enzyme Inhibitors/pharmacology , Female , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/deficiency , Glycogen Synthase Kinase 3/genetics , Hematopoiesis/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Homeostasis , Humans , Lithium/pharmacology , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Biological , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Signal Transduction , TOR Serine-Threonine Kinases , Wnt Proteins/metabolism , beta Catenin/deficiency , beta Catenin/genetics , beta Catenin/metabolism
13.
Biochem Soc Trans ; 37(Pt 5): 1133-8, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19754466

ABSTRACT

Lithium is widely used to treat bipolar disorder, but its mechanism of action in this disorder is unknown. Lithium directly inhibits GSK3 (glycogen synthase kinase 3), a critical regulator of multiple signal transduction pathways. Inhibition of GSK3 provides a compelling explanation for many of the known effects of lithium, including effects on early development and insulin signalling/glycogen synthesis. However, lithium also inhibits inositol monophosphatase, several structurally related phosphomonoesterases, phosphoglucomutase and the scaffolding function of beta-arrestin-2. It is not known which of these targets is responsible for the behavioural or therapeutic effects of lithium in vivo. The present review discusses basic criteria that can be applied to model systems to validate a proposed direct target of lithium. In this context, we describe a set of simple behaviours in mice that are robustly affected by chronic lithium treatment and are similarly affected by structurally diverse GSK3 inhibitors and by removing one copy of the Gsk3b gene. These observations, from several independent laboratories, support a central role for GSK3 in mediating behavioural responses to lithium.


Subject(s)
Antimanic Agents/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Lithium Compounds/metabolism , Animals , Antimanic Agents/pharmacology , Antimanic Agents/therapeutic use , Behavior, Animal/drug effects , Glycogen Synthase Kinase 3/metabolism , Humans , Lithium Compounds/pharmacology , Lithium Compounds/therapeutic use , Neurons/drug effects , Neurons/metabolism , Neuropsychological Tests , Phosphoric Monoester Hydrolases/metabolism , Reproducibility of Results , Signal Transduction/drug effects
15.
Mol Genet Metab ; 88(4): 384-8, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16644257

ABSTRACT

Two leading hypotheses to explain lithium action in bipolar disorder propose either inositol depletion or inhibition of GSK-3 as mechanisms of action. Behavioral effects of lithium are mimicked in Gsk-3beta+/- mice, but the contribution of inositol depletion to these behaviors has not been tested. According to the inositol depletion hypothesis, lithium-sensitive behavior is secondary to impaired phosphatidylinositol synthesis caused by inositol deficiency. By disrupting the sodium myo-inositol transporter1 gene, SMIT1, we show that depletion of brain myo-inositol in SMIT1+/- mice has no effect on lithium-sensitive behavior. These findings, taken together with our previous work showing that SMIT-/- mice have an even greater depletion of inositol in brain with no reduction in phosphatidylinositol levels, are difficult to reconcile with the current formulation of the inositol depletion hypothesis.


Subject(s)
Behavior, Animal/drug effects , Bipolar Disorder/drug therapy , Brain/metabolism , Inositol/metabolism , Lithium/pharmacology , Symporters/genetics , Animals , Bipolar Disorder/genetics , Bipolar Disorder/metabolism , Bipolar Disorder/psychology , Genotype , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Inositol/deficiency , Lithium/therapeutic use , Mice
16.
J Neurosci ; 24(30): 6791-8, 2004 Jul 28.
Article in English | MEDLINE | ID: mdl-15282284

ABSTRACT

Lithium is widely used to treat bipolar disorder, but its mechanism of action in this disorder is unknown. Several molecular targets of lithium have been identified, but these putative targets have not been shown to be responsible for the behavioral effects of lithium in vivo. A robust model for the effects of chronic lithium on behavior in mice would greatly facilitate the characterization of lithium action. We describe behaviors in mice that are robustly affected by chronic lithium. Remarkably, these lithium-sensitive behaviors are also observed in mice lacking one copy of the gene encoding glycogen synthase kinase-3beta (Gsk-3beta), a well established direct target of lithium. In addition, chronic lithium induces molecular changes consistent with inhibition of GSK-3 within regions of the brain that are paralleled in Gsk-3beta+/- heterozygous mice. We also show that lithium therapy activates Wnt signaling in vivo, as measured by increased Wnt-dependent gene expression in the amygdala, hippocampus, and hypothalamus. These observations support a central role for GSK-3beta in mediating behavioral responses to lithium.


Subject(s)
Behavior, Animal/drug effects , Brain Chemistry/drug effects , Brain/enzymology , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/physiology , Lithium Chloride/pharmacology , Nerve Tissue Proteins/physiology , Psychotropic Drugs/pharmacology , Animals , Cytoskeletal Proteins/analysis , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Exploratory Behavior/drug effects , Female , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/deficiency , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Grooming/drug effects , Habituation, Psychophysiologic/drug effects , Heterozygote , Intercellular Signaling Peptides and Proteins/physiology , Lithium Chloride/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Motor Activity/drug effects , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Psychotropic Drugs/administration & dosage , Random Allocation , Reflex, Startle/drug effects , Social Behavior , Swimming , Trans-Activators/analysis , Transcription, Genetic/drug effects , Wnt Proteins , beta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...