Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
Add more filters










Publication year range
1.
J Appl Physiol (1985) ; 129(6): 1431-1438, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33054660

ABSTRACT

Dopamine is often used to treat hypotension in preterm infants who are at risk of hypoxic-ischemic (HI) brain injury due to cerebral hypoperfusion and impaired autoregulation. There is evidence that systemically administered dopamine crosses the preterm blood-brain barrier. However, the effects of exogenous dopamine and cerebral HI on dopaminergic signaling in the immature brain are unknown. We determined the effect of HI and dopamine on D1 and D2 receptor binding and expressions of dopamine transporter (DAT) and tyrosine hydroxylase (TH) in the striatum of the preterm fetal sheep. Fetal sheep (99 days of gestation, term = 147days) were unoperated controls (n = 6) or exposed to severe HI using umbilical cord occlusion and saline infusion (UCO + saline, n = 8) or to HI with dopamine infusion (UCO + dopamine, 10 µg/kg/min, n = 7) for 74 h. D1 and D2 receptor densities were measured by autoradiography in vitro. DAT, TH, and cell death were measured using immunohistochemistry. HI resulted in cell death in the caudate nucleus and putamen, and dopamine infusion started before HI did not exacerbate or ameliorate these effects. HI led to reduced D1 and D2 receptor densities in the caudate nucleus and reduction in DAT protein expression in the caudate and putamen. Fetal brains exposed to dopamine in addition to HI were not different from those exposed to HI alone in these changes in dopaminergic parameters. We conclude that dopamine infusion does not alter the striatal cell death or the reductions in D1 and D2 receptor densities and DAT protein expression induced by HI in the preterm brain.NEW & NOTEWORTHY This is the first study on the effects of hypoxia-ischemia and dopamine treatment on the dopaminergic pathway in the preterm brain. In the striatum of fetal sheep (equivalent to ∼26-28 wk of human gestation), we demonstrate that hypoxia-ischemia leads to cell death, reduces D1 and D2 receptors, and reduces dopamine transporter. Intravenous dopamine infusion at clinical dosage used in preterm human infants does not alter the striatal cell death, D1 and D2 receptor density levels, and DAT protein expressions after hypoxia-ischemia in the preterm brain.


Subject(s)
Dopamine , Hypoxia-Ischemia, Brain , Animals , Brain , Humans , Hypoxia , Hypoxia-Ischemia, Brain/drug therapy , Infant, Newborn , Infant, Premature , Ischemia , Receptors, Dopamine , Sheep
2.
Horm Behav ; 112: 32-41, 2019 06.
Article in English | MEDLINE | ID: mdl-30928609

ABSTRACT

RATIONALE: Males are more prone to psychosis, schizophrenia and substance abuse and addiction in adolescence and early adulthood than females. However, the role of androgens during this developmental period is poorly understood. OBJECTIVES: This study aimed to examine how androgens in adolescence influence psychosis-like behaviour in adulthood and whether brain-derived neurotrophic factor (BDNF) is a mediator of these developmental effects. METHODS: Wild-type and BDNF heterozygous male mice were castrated at pre-pubescence and implanted with testosterone or dihydrotestosterone (DHT). In adulthood, we assessed amphetamine- and MK-801-induced hyperlocomotion as a model of psychosis-like behaviour. Western blot analysis was used to quantify levels of the dopamine transporter (DAT) and N-methyl-d-aspartate (NMDA) receptor subunits. RESULTS: While castration itself had little effect on behaviour, adolescent testosterone, but not DHT, significantly reduced amphetamine-induced hyperlocomotion, whereas both testosterone and DHT reduced the effect of MK-801. These effects were similar in mice of either genotype. In wildtype mice, both testosterone and DHT treatment reduced DAT expression in the medial prefrontal cortex (mPFC) but these effects were absent in BDNF heterozygous mice. There were no effects on NMDA receptor subunit levels. CONCLUSIONS: The differential effect of adolescent testosterone and DHT on amphetamine-induced hyperlocomotion in adulthood suggests involvement of conversion of testosterone to estrogen and subsequent modulation of dopaminergic signalling. In contrast, the similar effect of testosterone and DHT treatment on NMDA receptor-mediated hyperlocomotion indicates it is mediated by androgen receptors. The involvement of BDNF in these hormone effects remains to be elucidated. These results demonstrate that, during adolescence, androgens significantly influence key pathways related to various mental illnesses prevalent in adolescence.


Subject(s)
Androgens/pharmacology , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/genetics , Psychotic Disorders , Sexual Maturation/physiology , Age Factors , Animals , Dihydrotestosterone/pharmacology , Dizocilpine Maleate/pharmacology , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Heterozygote , Male , Mice , Mice, Inbred C57BL , Orchiectomy , Psychotic Disorders/genetics , Psychotic Disorders/metabolism , Psychotic Disorders/physiopathology , Receptors, Androgen/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Testosterone/metabolism , Testosterone/pharmacology
3.
Transl Psychiatry ; 7(9): e1233, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28926000

ABSTRACT

The BDNF Val66Met polymorphism has been associated with sensitivity to stress and affective disorders. We therefore sought to model the inter-causality of these relationships under controlled laboratory conditions. We subjected humanized BDNF Val66Met (hBDNFVal66Met) transgenic mice to a history of stress, modeled by chronic late-adolescent corticosterone (CORT) exposure, before evaluating affective-related behavior using the forced-swim test (FST) in adulthood. While hBDNFMet/Met mice had a depression-like phenotype in the FST irrespective of CORT, hBDNFVal/Val wildtype mice had a resilient phenotype but developed an equally robust depressive-like phenotype following CORT. A range of stress-sensitive molecules were studied across the corticohippocampal axis, and where genotype differences occurred following CORT they tended to inversely coincide with the behavior of the hBDNFVal/Val group. Notably, tyrosine hydroxylase was markedly down-regulated in the mPFC of hBDNFVal/Val mice as a result of CORT treatment, which mimicked expression levels of hBDNFMet/Met mice and the FST behavior of both groups. The expression of calretinin, PSD-95, and truncated TrkB were also concomitantly reduced in the mPFC of hBDNFVal/Val mice by CORT. This work establishes BDNFVal66Met genotype as a regulator of behavioral despair, and identifies new biological targets of BDNF genetic variation relevant to stress-inducible disorders such as depression.


Subject(s)
Behavior, Animal , Brain-Derived Neurotrophic Factor/genetics , Corticosterone/pharmacology , Depression , Hippocampus/drug effects , Hippocampus/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Resilience, Psychological , Stress, Psychological , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Depression/chemically induced , Depression/genetics , Depression/physiopathology , Disease Models, Animal , Mice , Mice, Transgenic , Polymorphism, Single Nucleotide , Resilience, Psychological/drug effects , Stress, Psychological/chemically induced , Stress, Psychological/genetics , Stress, Psychological/physiopathology
4.
Transl Psychiatry ; 6: e794, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-27115125

ABSTRACT

Clinical evidence indicates that serotonin-1A receptor (5-HT1AR) gene polymorphisms are associated with anxiety disorders and deficits in cognition. In animal models, exercise (Ex) and environmental enrichment (EE) can change emotionality-related behaviours, as well as enhance some aspects of cognition and hippocampal neurogenesis. We investigated the effects of Ex and EE (which does not include running wheels) on cognition and anxiety-like behaviours in wild-type (WT) and 5-HT1AR knock-out (KO) mice. Using an algorithm-based classification of search strategies in the Morris water maze, we report for we believe the first time that Ex increased the odds for mice to select more hippocampal-dependent strategies. In the retention probe test, Ex (but not EE) corrected long-term spatial memory deficits displayed by KO mice. In agreement with these findings, only Ex increased hippocampal cell survival and BDNF protein levels. However, only EE (but not Ex) modified anxiety-like behaviours, demonstrating dissociation between improvements in cognition and innate anxiety. EE enhanced hippocampal cell proliferation in WT mice only, suggesting a crucial role for intact serotonergic signalling in mediating this effect. Together, these results demonstrate differential effects of Ex vs EE in a mouse model of anxiety with cognitive impairment. Overall, the 5-HT1AR does not seem to be critical for those behavioural effects to occur. These findings will have implications for our understanding of how Ex and EE enhance experience-dependent plasticity, as well as their differential impacts on anxiety and cognition.


Subject(s)
Anxiety/physiopathology , Behavior, Animal/physiology , Cognitive Dysfunction/physiopathology , Environment , Physical Conditioning, Animal , Animals , Blotting, Western , Disease Models, Animal , Hippocampus/physiopathology , Maze Learning/physiology , Memory, Long-Term/physiology , Mice , Mice, Knockout , Polymerase Chain Reaction
6.
Mol Psychiatry ; 20(8): 916-30, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25824305

ABSTRACT

Brain-derived neurotrophic factor (BDNF) has a primary role in neuronal development, differentiation and plasticity in both the developing and adult brain. A single-nucleotide polymorphism in the proregion of BDNF, termed the Val66Met polymorphism, results in deficient subcellular translocation and activity-dependent secretion of BDNF, and has been associated with impaired neurocognitive function in healthy adults and in the incidence and clinical features of several psychiatric disorders. Research investigating the Val66Met polymorphism has increased markedly in the past decade, and a gap in integration exists between and within academic subfields interested in the effects of this variant. Here we comprehensively review the role and relevance of the Val66Met polymorphism in psychiatric disorders, with emphasis on suicidal behavior and anxiety, eating, mood and psychotic disorders. The cognitive and molecular neuroscience of the Val66Met polymorphism is also concisely reviewed to illustrate the effects of this genetic variant in healthy controls, and is complemented by a commentary on the behavioral neuroscience of BDNF and the Val66Met polymorphism where relevant to specific disorders. Lastly, a number of controversies and unresolved issues, including small effect sizes, sampling of allele inheritance but not genotype and putative ethnicity-specific effects of the Val66Met polymorphism, are also discussed to direct future research.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Genetic Predisposition to Disease , Mental Disorders/genetics , Polymorphism, Single Nucleotide , Animals , Brain-Derived Neurotrophic Factor/metabolism , Humans , Mental Disorders/metabolism
7.
Transl Psychiatry ; 5: e498, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25603414

ABSTRACT

The recent use of estrogen-based therapies as adjunctive treatments for the cognitive impairments of schizophrenia has produced promising results; however the mechanism behind estrogen-based cognitive enhancement is relatively unknown. Brain-derived neurotrophic factor (BDNF) regulates learning and memory and its expression is highly responsive to estradiol. We recently found that estradiol modulates the expression of hippocampal parvalbumin-positive GABAergic interneurons, known to regulate neuronal synchrony and cognitive function. What is unknown is whether disruptions to the aforementioned estradiol-parvalbumin pathway alter learning and memory, and whether BDNF may mediate these events. Wild-type (WT) and BDNF heterozygous (+/-) mice were ovariectomized (OVX) at 5 weeks of age and simultaneously received empty, estradiol- or progesterone-filled implants for 7 weeks. At young adulthood, mice were tested for spatial and recognition memory in the Y-maze and novel-object recognition test, respectively. Hippocampal protein expression of BDNF and GABAergic interneuron markers, including parvalbumin, were assessed. WT OVX mice show impaired performance on Y-maze and novel-object recognition test. Estradiol replacement in OVX mice prevented the Y-maze impairment, a Behavioral abnormality of dorsal hippocampal origin. BDNF and parvalbumin protein expression in the dorsal hippocampus and parvalbumin-positive cell number in the dorsal CA1 were significantly reduced by OVX in WT mice, while E2 replacement prevented these deficits. In contrast, BDNF(+/-) mice showed either no response or an opposite response to hormone manipulation in both behavioral and molecular indices. Our data suggest that BDNF status is an important biomarker for predicting responsiveness to estrogenic compounds which have emerged as promising adjunctive therapeutics for schizophrenia patients.


Subject(s)
Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/genetics , CA1 Region, Hippocampal/drug effects , Estradiol/pharmacology , Estrogens/pharmacology , Maze Learning/drug effects , Spatial Memory/drug effects , Animals , Behavior, Animal/physiology , CA1 Region, Hippocampal/metabolism , Female , Gene-Environment Interaction , Heterozygote , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Ovariectomy , Parvalbumins/drug effects , Parvalbumins/metabolism , Spatial Memory/physiology
8.
Neuroscience ; 284: 297-310, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25445195

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is a widely expressed neurotrophin involved in neurodevelopment, neuroprotection and synaptic plasticity. It is also implicated in a range of psychiatric disorders such as schizophrenia, depression and post-traumatic stress disorder. Stress during adolescence/young adulthood can have long-term psychiatric and cognitive consequences, however it is unknown how altered BDNF signaling is involved in such effects. Here we investigated whether a congenital deficit in BDNF availability in rats increases vulnerability to the long-term effects of the stress hormone, corticosterone (CORT). Compared to wildtype (WT) littermates, BDNF heterozygous (HET) rats showed higher body weights and minor developmental changes, such as reduced relative brain and pituitary weight. These animals furthermore showed deficits in short-term spatial memory in the Y-maze and in prepulse inhibition and startle, but not in object-recognition memory. CORT treatment induced impairments in novel-object recognition memory in both genotypes but disrupted fear conditioning extinction learning in BDNF HET rats only. These results show selective behavioral changes in BDNF HET rats, at baseline or after chronic CORT treatment and add to our understanding of the role of BDNF and its interaction with stress. Importantly, this study demonstrates the utility of the BDNF HET rat in investigations into the pathophysiology of various psychiatric disorders.


Subject(s)
Brain-Derived Neurotrophic Factor/deficiency , Corticosterone/toxicity , Memory Disorders/physiopathology , Stress, Psychological/physiopathology , Animals , Brain/pathology , Brain-Derived Neurotrophic Factor/genetics , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Disease Models, Animal , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Fear/drug effects , Fear/physiology , Genetic Predisposition to Disease , Memory Disorders/pathology , Memory, Short-Term/drug effects , Memory, Short-Term/physiology , Mutation , Organ Size , Rats, Sprague-Dawley , Rats, Transgenic , Receptor, trkB/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Reflex, Startle/drug effects , Reflex, Startle/physiology , Spatial Memory/drug effects , Spatial Memory/physiology , Stress, Psychological/pathology
9.
Transl Psychiatry ; 3: e327, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24301645

ABSTRACT

Dopamine (DA) neurotransmission requires a complex series of enzymatic reactions that are tightly linked to catecholamine exocytosis and receptor interactions on pre- and postsynaptic neurons. Regulation of dopaminergic signalling is primarily achieved through reuptake of extracellular DA by the DA transporter (DAT) on presynaptic neurons. Aberrant regulation of DA signalling, and in particular hyperactivation, has been proposed as a key insult in the presentation of schizophrenia and related neuropsychiatric disorders. We recently identified 14-3-3ζ as an essential component of neurodevelopment and a central risk factor in the schizophrenia protein interaction network. Our analysis of 14-3-3ζ-deficient mice now shows that baseline hyperactivity of knockout (KO) mice is rescued by the antipsychotic drug clozapine. 14-3-3ζ KO mice displayed enhanced locomotor hyperactivity induced by the DA releaser amphetamine. Consistent with 14-3-3ζ having a role in DA signalling, we found increased levels of DA in the striatum of 14-3-3ζ KO mice. Although 14-3-3ζ is proposed to modulate activity of the rate-limiting DA biosynthesis enzyme, tyrosine hydroxylase (TH), we were unable to identify any differences in total TH levels, TH localization or TH activation in 14-3-3ζ KO mice. Rather, our analysis identified significantly reduced levels of DAT in the absence of notable differences in RNA or protein levels of DA receptors D1-D5. Providing insight into the mechanisms by which 14-3-3ζ controls DAT stability, we found a physical association between 14-3-3ζ and DAT by co-immunoprecipitation. Taken together, our results identify a novel role for 14-3-3ζ in DA neurotransmission and provide support to the hyperdopaminergic basis of pathologies associated with schizophrenia and related disorders.


Subject(s)
14-3-3 Proteins/genetics , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine/metabolism , Motor Activity/genetics , Neostriatum/metabolism , Schizophrenia/genetics , Synaptic Transmission/genetics , Amphetamine/pharmacology , Animals , Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Clozapine/pharmacology , Disease Models, Animal , Dopamine Agents/pharmacology , Dopamine Plasma Membrane Transport Proteins/drug effects , Mice , Mice, Knockout , Motor Activity/drug effects , Schizophrenia/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Synaptic Transmission/drug effects
10.
Neuroscience ; 239: 67-83, 2013 Jun 03.
Article in English | MEDLINE | ID: mdl-23085218

ABSTRACT

Schizophrenia is a severe psychiatric disorder with a complex and variable set of symptoms. Both genetic and environmental mechanisms are involved in the development of the illness and lead to structural and neurochemical abnormalities in the brain. An intriguing facet of schizophrenia is sex differences, which have been described for nearly all features of the illness, including the peak age of onset, symptoms and treatment response. The ovarian hormone, estrogen, may be protective against schizophrenia and evidence is accumulating that estrogen may exert this effect via an interaction with brain-derived neurotrophic factor (BDNF). Both estrogen and BDNF have trophic effects on the developing brain and promote synaptic plasticity and maintain neurons well into adulthood. Major neurotransmitter systems including dopaminergic, serotonergic and glutamatergic pathways are modulated and supported by estrogen and BDNF. Despite their commonalities, estrogen and BDNF have mostly been examined independently but increasing evidence suggests an interaction between the two in brain regions pertinent to schizophrenia. This review will focus on the role of estrogen and BDNF in clinical and animal studies of schizophrenia. We include animal models of neurotransmitter dysfunction and genetic manipulation to show how estrogen may provide a protective effect in schizophrenia, including through mediating BDNF expression and activity. This posited estrogen-BDNF interaction could play a key role in modulating sex-dependent results reported in animal work as well as sex differences in clinical aspects of schizophrenia.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Estrogens/metabolism , Schizophrenia/metabolism , Sex Characteristics , Animals , Disease Models, Animal , Female , Humans , Male
11.
J Neuroendocrinol ; 24(5): 774-88, 2012 May.
Article in English | MEDLINE | ID: mdl-22221196

ABSTRACT

Sex steroid hormones and neurotrophic factors are involved in pruning and shaping the adolescent brain and have been implicated in the pathogenesis of neurodevelopmental disorders, including mental illness. We aimed to determine the association between altered levels of sex steroid hormones during adolescent development and neurotrophic signalling in the C57Bl/6 mouse. We first performed a week by week analysis from pre-pubescence to adulthood in male and female C57Bl/6 mice, measuring serum levels of testosterone and oestradiol in conjunction with western blot analysis of neurotrophin expression in the forebrain and hippocampal regions. Second, we manipulated adolescent sex steroid hormone levels by gonadectomy and hormone replacement at the pre-pubescent age of 5 weeks. Young-adult forebrain and hippocampal neurotrophin expression was then determined. Male mice showed significant changes in brain-derived neurotrophic factor (BDNF) expression in the forebrain regions during weeks 7-10, which corresponded significantly with a surge in serum testosterone. Castration and testosterone or di-hydrotestosterone replacement experiments revealed an androgen receptor-dependent effect on BDNF-tyrosine kinase (Trk) B signalling in the forebrain and hippocampal regions during adolescence. Female mice showed changes in BDNF-TrkB signalling at a much earlier time point (weeks 4-8) in the forebrain and hippocampal regions and these did not correspond with changes in serum oestradiol. Ovariectomy actually increased BDNF expression but decreased TrkB phosphorylation in the forebrain regions. 17ß-Oestradiol replacement had no effect, suggesting a role for other ovarian hormones in regulating BDNF-TrkB signalling in the adolescent female mouse brain. These results suggest the differential actions of sex steroid hormones in modulating BDNF-TrkB signalling during adolescence. These data provide insight into how the male and female brain changes in response to altered levels of circulating sex steroid hormones and could help to explain some of the developmental sex differences in the pathogenesis of neurodevelopmental disorders, including mental illness.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Gonadal Steroid Hormones/pharmacology , Receptor, trkB/metabolism , Sexual Maturation/drug effects , Animals , Brain-Derived Neurotrophic Factor/blood , Female , Gene Expression Regulation, Developmental/drug effects , Growth and Development/drug effects , Male , Mice , Mice, Inbred C57BL , Orchiectomy , Ovariectomy , Sex Characteristics , Testosterone/blood
12.
Mol Psychiatry ; 17(4): 451-66, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22124272

ABSTRACT

Complex neuropsychiatric disorders are believed to arise from multiple synergistic deficiencies within connected biological networks controlling neuronal migration, axonal pathfinding and synapse formation. Here, we show that deletion of 14-3-3ζ causes neurodevelopmental anomalies similar to those seen in neuropsychiatric disorders such as schizophrenia, autism spectrum disorder and bipolar disorder. 14-3-3ζ-deficient mice displayed striking behavioural and cognitive deficiencies including a reduced capacity to learn and remember, hyperactivity and disrupted sensorimotor gating. These deficits are accompanied by subtle developmental abnormalities of the hippocampus that are underpinned by aberrant neuronal migration. Significantly, 14-3-3ζ-deficient mice exhibited abnormal mossy fibre navigation and glutamatergic synapse formation. The molecular basis of these defects involves the schizophrenia risk factor, DISC1, which interacts isoform specifically with 14-3-3ζ. Our data provide the first evidence of a direct role for 14-3-3ζ deficiency in the aetiology of neurodevelopmental disorders and identifies 14-3-3ζ as a central risk factor in the schizophrenia protein interaction network.


Subject(s)
14-3-3 Proteins/physiology , Brain/growth & development , Genetic Predisposition to Disease/genetics , Hippocampus/growth & development , Neurogenesis/physiology , Schizophrenia/genetics , 14-3-3 Proteins/genetics , Animals , Brain/metabolism , Cell Movement/genetics , Cell Movement/physiology , Cells, Cultured , Disease Models, Animal , Female , Hippocampus/metabolism , Hippocampus/pathology , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Neurogenesis/genetics , Neurons/metabolism , Neurons/physiology , Recognition, Psychology/physiology , Sensory Gating/physiology
13.
Behav Brain Res ; 223(2): 336-41, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21605597

ABSTRACT

Evidence suggests that the heterozygous transmembrane domain mutant mouse model for the schizophrenia candidate gene neuregulin 1 (Nrg1 HET) exhibits a deficit in prepulse inhibition (PPI). However, not all mouse models for Nrg1 exhibit PPI deficits. Thus, our study intended to clarify the severity of the initially described PPI deficit in Nrg1 HET mice. For this, Nrg1 mutant mice and wild type-like littermates of one breeding colony were tested for PPI in four different phenotyping facilities in Australia employing a variety of different PPI protocols with fixed and variable interstimulus intervals (ISIs). Testing mutant and wild type-like mice in three Australian phenotyping facilities using PPI protocols with variable ISIs revealed no effect of mutant transmembrane domain Nrg1 on sensorimotor gating. Changes to the startle response and startle response habituation were site/protocol-specific. The employment of two different PPI protocols at the same phenotyping facility revealed a protocol-dependent and site-specific facilitation of PPI in Nrg1 mutant mice compared to wild type-like mice. In conclusion, the often-noted PPI phenotype of the transmembrane domain Nrg1 mutant mouse model is highly PPI protocol-specific and appears sensitive to the particular conditions of the test laboratory. Our study describes wild type-like PPI under most test conditions and across three different laboratories. The research suggests that analysing one of the alleged hallmarks of animal models for schizophrenia must be done carefully: to obtain reliable PPI data it seems necessary to use more than one particular PPI protocol.


Subject(s)
Mutation/physiology , Neuregulin-1/genetics , Neuregulin-1/physiology , Sensory Gating/genetics , Sensory Gating/physiology , Acoustic Stimulation , Animals , Disease Models, Animal , Habituation, Psychophysiologic/genetics , Habituation, Psychophysiologic/physiology , Male , Membrane Potentials/physiology , Mice , Reflex, Startle/physiology , Schizophrenia
14.
Curr Med Chem ; 16(23): 2965-76, 2009.
Article in English | MEDLINE | ID: mdl-19689277

ABSTRACT

The tripeptide, glutathione (gamma-glutamylcysteinylglycine) is the primary endogenous free radical scavenger in the human body. When glutathione (GSH) levels are reduced there is an increased potential for cellular oxidative stress, characterised by an increase and accruement of reactive oxygen species (ROS). Oxidative stress has been implicated in the pathology of schizophrenia and bipolar disorder. This could partly be caused by alterations in dopaminergic and glutamatergic activity that are implicated in these illnesses. Glutamate and dopamine are highly redox reactive molecules and produce ROS during normal neurotransmission. Alterations to these neurotransmitter pathways may therefore increase the oxidative burden in the brain. Furthermore, mitochondrial dysfunction, as a source of oxidative stress, has been documented in both schizophrenia and bipolar disorder. The combination of altered neurotransmission and this mitochondrial dysfunction leading to oxidative damage may ultimately contribute to illness symptoms. Animal models have been established to investigate the involvement of glutathione depletion in aspects of schizophrenia and bipolar disorder to further characterise the role of oxidative stress in psychopathology. Stemming from preclinical evidence, clinical studies have recently shown antioxidant precursor treatment to be effective in schizophrenia and bipolar disorder, providing a novel clinical angle to augment often suboptimal conventional treatments.


Subject(s)
Bipolar Disorder/etiology , Glutathione/physiology , Schizophrenia/etiology , Animals , Bipolar Disorder/drug therapy , Bipolar Disorder/metabolism , Disease Models, Animal , Free Radical Scavengers/pharmacology , Humans , Oxidative Stress , Reactive Oxygen Species/metabolism , Schizophrenia/drug therapy , Schizophrenia/metabolism
15.
Br J Pharmacol ; 154(2): 417-28, 2008 May.
Article in English | MEDLINE | ID: mdl-18475255

ABSTRACT

BACKGROUND AND PURPOSE: We investigated possible differences in the impact of chronic amphetamine administration during adolescence and adulthood on aspects of behaviour and brain chemistry. EXPERIMENTAL APPROACH: Adult (n=32) and adolescent (n=32) male Sprague-Dawley rats were given either D-amphetamine sulphate (10 mg kg(-1) daily, i.p.) or saline (1 mL kg(-1), i.p.) for 10 days. Rats were subsequently tested for anxiety-like behaviour, learning and memory, and sensorimotor gating. Nine weeks later, rats received saline (1 mL kg(-1)) or acute amphetamine challenge (1.5 mg kg(-1)) and the expression levels of mRNA for tyrosine kinase B (TrkB) or cAMP response element-binding protein (CREB) were measured in the hippocampus. KEY RESULTS: The adolescent amphetamine pretreated group revealed a deficit in exploration on the Y-maze during a 6 h retention test. The frequency of visits to the novel arm was 35% lower for the amphetamine group compared with controls. In parallel, a 43% decrease in hippocampal CREB mRNA, but not TrkB mRNA, was observed in periadolescent rats treated chronically with amphetamine 9 weeks earlier. None of the effects were detected in the adult treated cohort. CONCLUSIONS AND IMPLICATIONS: Chronic amphetamine treatment during periadolescence resulted in altered behaviour on the Y-maze and persistent downregulation of hippocampal CREB mRNA expression. Given that this group had intact spatial learning and reference memory, it would appear that the deficits observed on the Y-maze reflect a dysfunction in response to novelty. Because no effects of amphetamine treatment were observed in the adult cohort, these data suggest idiosyncratic sensitivity of periadolescence to the long-term effects of psychostimulants.


Subject(s)
Amphetamine/administration & dosage , Behavior, Animal/drug effects , Central Nervous System Stimulants/administration & dosage , Cyclic AMP Response Element-Binding Protein/metabolism , Exploratory Behavior/drug effects , Hippocampus/drug effects , RNA, Messenger/metabolism , Age Factors , Amphetamine/adverse effects , Animals , Anxiety/chemically induced , Central Nervous System Stimulants/adverse effects , Cyclic AMP Response Element-Binding Protein/genetics , Down-Regulation , Drug Administration Schedule , Habituation, Psychophysiologic , Hippocampus/enzymology , Hippocampus/metabolism , Injections, Intraperitoneal , Learning/drug effects , Male , Memory/drug effects , Rats , Rats, Sprague-Dawley , Receptor, trkB/metabolism , Reflex, Startle/drug effects , Sexual Maturation , Time Factors
16.
Mol Psychiatry ; 13(7): 661-72, 2008 Jul.
Article in English | MEDLINE | ID: mdl-17667964

ABSTRACT

Phospholipase C-beta1 (PLC-beta1) is a rate-limiting enzyme implicated in postnatal-cortical development and neuronal plasticity. PLC-beta1 transduces intracellular signals from specific muscarinic, glutamate and serotonin receptors, all of which have been implicated in the pathogenesis of schizophrenia. Here, we present data to show that PLC-beta1 knockout mice display locomotor hyperactivity, sensorimotor gating deficits as well as cognitive impairment. These changes in behavior are regarded as endophenotypes homologous to schizophrenia-like symptoms in rodents. Importantly, the locomotor hyperactivity and sensorimotor gating deficits in PLC-beta1 knockout mice are subject to beneficial modulation by environmental enrichment. Furthermore, clozapine but not haloperidol (atypical and typical antipsychotics, respectively) rescues the sensorimotor gating deficit in these animals, suggesting selective predictive validity. We also demonstrate a relationship between the beneficial effects of environmental enrichment and levels of M1/M4 muscarinic acetylcholine receptor binding in the neocortex and hippocampus. Thus we have demonstrated a novel mouse model, displaying disruption of multiple postsynaptic signals implicated in the pathogenesis of schizophrenia, a relevant behavioral phenotype and associated gene-environment interactions.


Subject(s)
Clozapine/therapeutic use , Phospholipase C beta/deficiency , Schizophrenia/genetics , Schizophrenia/rehabilitation , Animals , Antipsychotic Agents/therapeutic use , Disease Models, Animal , Environment , Hippocampus/physiopathology , Mice , Mice, Knockout , Motor Activity , Neocortex/physiopathology , Phenotype , Receptors, Muscarinic/physiology , Schizophrenia/drug therapy , Schizophrenia/enzymology , Schizophrenic Psychology
17.
Br J Pharmacol ; 150(6): 750-7, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17279088

ABSTRACT

BACKGROUND AND PURPOSE: Prepulse inhibition (PPI) of the acoustic startle response is a model of sensorimotor gating which is disrupted in schizophrenia and other mental illnesses. We and others have shown that treatment with the 5-hydroxytryptamine-1A (5-HT(1A)) receptor agonist, 8-OH-DPAT, disrupts PPI in rats. In the present study, we highlight the importance of baseline levels on the effect of 8-OH-DPAT on PPI. EXPERIMENTAL APPROACH: Adult male and female Sprague-Dawley rats were gonadectomised. These rats were treated with saline, 0.02 and 0.5 mg kg(-1) of 8-OH-DPAT using a random-sequence, repeated-measures protocol. The rats were allocated into high and low baseline groups depending on their baseline PPI observed after saline treatment. KEY RESULTS: Treatment with 0.5 mg kg(-1) of 8-OH-DPAT significantly disrupted PPI in both male and female rats. In male rats only, 0.02 mg kg(-1) 8-OH-DPAT caused a small, but significant, increase in PPI. When these male rats were allocated to either a high or low baseline PPI group, 0.5 mg kg(-1) 8-OH-DPAT disrupted PPI in the high baseline group only. In contrast, treatment with 0.02 mg kg(-1) 8-OH-DPAT increased PPI only in the low baseline PPI group. There were no changes in the effect of 8-OH-DPAT administration in female rats when they were divided into high and low baseline PPI groups. CONCLUSIONS AND IMPLICATIONS: The level of baseline PPI is an important variable that can influence the direction of drug effects induced by 8-OH-DPAT. The explanation for this phenomenon could be differential activation of pre- and postsynaptic 5-HT(1A) receptors.


Subject(s)
8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Reflex, Startle/drug effects , Acoustic Stimulation , Animals , Female , Humans , Male , Orchiectomy , Ovariectomy , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1A/physiology , Reflex, Startle/physiology , Serotonin 5-HT1 Receptor Agonists , Serotonin Receptor Agonists/pharmacology
19.
Psychopharmacology (Berl) ; 183(3): 358-67, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16220329

ABSTRACT

RATIONALE: G(z) is a member of the G(i) G protein family associated with dopamine D2-like receptors; however, its functions remain relatively unknown. The aim of the present study was to investigate prepulse inhibition (PPI) of acoustic startle, locomotor hyperactivity and dopamine D2 receptor binding in mice deficient in the alpha subunit of G(z). METHODS: We used automated startle boxes to assess startle and PPI after treatment with saline, amphetamine, apomorphine or MK-801. We used photocell cages to quantitate locomotor activity after amphetamine treatment. Dopamine D2 receptor density was determined by autoradiography. RESULTS: Startle responses and baseline PPI were not different between the Galpha(z) knockout mice and wild-type controls (average PPI 46+/-4 vs 49+/-3%, respectively). Amphetamine treatment caused a marked disruption of PPI in Galpha(z) knockouts (average PPI 22+/-2%), but less so in controls (average PPI 42+/-3%). Similar genotype-dependent responses were seen after apomorphine treatment (average PPI 23+/-3% vs 40+/-3%), but not after MK-801 treatment (average PPI 29+/-5 vs 33+/-2%). Amphetamine-induced locomotor hyperactivity was greater in Galpha(z) knockouts than in controls. There was no difference in the density of dopamine D2 receptors in nucleus accumbens. CONCLUSIONS: Mice deficient in the alpha subunit of G(z) show enhanced sensitivity to the disruption of PPI and locomotor hyperactivity caused by dopaminergic stimulation. These results suggest a possible role for G(z) in neuropsychiatric illnesses with presumed dopaminergic hyperactivity, such as schizophrenia.


Subject(s)
GTP-Binding Protein alpha Subunits/physiology , Receptors, Dopamine D1/physiology , Receptors, Dopamine D2/physiology , Reflex, Startle , Amphetamine/pharmacology , Animals , Apomorphine/pharmacology , Autoradiography , Dizocilpine Maleate/pharmacology , Dopamine/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects
20.
Aust N Z J Psychiatry ; 39(7): 550-7, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15996135

ABSTRACT

OBJECTIVE: This review aims to summarize the importance of animal models for research on psychiatric illnesses, particularly schizophrenia. METHOD AND RESULTS: Several aspects of animal models are addressed, including animal experimentation ethics and theoretical considerations of different aspects of validity of animal models. A more specific discussion is included on two of the most widely used behavioural models, psychotropic drug-induced locomotor hyperactivity and prepulse inhibition, followed by comments on the difficulty of modelling negative symptoms of schizophrenia. Furthermore, we emphasize the impact of new developments in molecular biology and the generation of genetically modified mice, which have generated the concept of behavioural phenotyping. CONCLUSIONS: Complex psychiatric illnesses, such as schizophrenia, cannot be exactly reproduced in species such as rats and mice. Nevertheless, by providing new information on the role of neurotransmitter systems and genes in behavioural function, animal 'models' can be an important tool in unravelling mechanisms involved in the symptoms and development of such illnesses, alongside approaches such as post-mortem studies, cognitive and psychophysiological studies, imaging and epidemiology.


Subject(s)
Disease Models, Animal , Schizophrenia , Animals , Locomotion/drug effects , Mice , Mice, Inbred C57BL , Neural Inhibition/physiology , Psychotropic Drugs/adverse effects , Rats , Rats, Sprague-Dawley , Schizophrenia/drug therapy , Schizophrenia/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...