Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
BMC Biol ; 21(1): 186, 2023 09 05.
Article in English | MEDLINE | ID: mdl-37667240

ABSTRACT

BACKGROUND: Studies have shown that paternal stress prior to conception can influence the innate behaviours of their offspring. The evolutionary impacts of such intergenerational effects are therefore of considerable interest. Our group previously showed in a model of daily stress that glucocorticoid treatment of adult male mouse breeders prior to conception leads to increased anxiety-related behaviours in male offspring. Here, we aimed to understand the transgenerational effects of paternal stress exposure on the social behaviour of progeny and its potential influence on reproductive success. RESULTS: We assessed social parameters including social reward, male attractiveness and social dominance, in the offspring (F1) and grand-offspring (F2). We report that paternal corticosterone treatment was associated with increased display of subordination towards other male mice. Those mice were unexpectedly more attractive to female mice while expressing reduced levels of the key rodent pheromone Darcin, contrary to its conventional role in driving female attraction. We investigated the epigenetic regulation of major urinary protein (Mup) expression by performing the first Oxford Nanopore direct methylation of sperm DNA in a mouse model of stress, but found no differences in Mup genes that could be attributed to corticosterone-treatment. Furthermore, no overt differences of the prefrontal cortex transcriptome were found in F1 offspring, implying that peripheral mechanisms are likely contributing to the phenotypic differences. Interestingly, no phenotypic differences were observed in the F2 grand-offspring. CONCLUSIONS: Overall, our findings highlight the potential of moderate paternal stress to affect intergenerational (mal)adaptive responses, informing future studies of adaptiveness in rodents, humans and other species.


Subject(s)
Corticosterone , Epigenesis, Genetic , Adult , Humans , Male , Female , Animals , Mice , Semen , Research Design , Pheromones
2.
FASEB J ; 36(1): e21981, 2022 01.
Article in English | MEDLINE | ID: mdl-34907601

ABSTRACT

The global consumption of highly processed, calorie-dense foods has contributed to an epidemic of overweight and obesity, along with negative consequences for metabolic dysfunction and disease susceptibility. As it becomes apparent that overweight and obesity have ripple effects through generations, understanding of the processes involved is required, in both maternal and paternal epigenetic inheritance. We focused on the patrilineal effects of a Western-style high-fat (21%) and high-sugar (34%) diet (WD) compared to control diet (CD) during adolescence and investigated F0 and F1 mice for physiological and behavioral changes. F0 males (fathers) showed increased body weight, impaired glycemic control, and decreased attractiveness to females. Paternal WD caused significant phenotypic changes in F1 offspring, including higher body weights of pups, increased Actinobacteria abundance in the gut microbiota (ascertained using 16S microbiome profiling), a food preference for WD pellets, increased male dominance and attractiveness to females, as well as decreased behavioral despair. These results collectively demonstrate the long-term intergenerational effects of a Western-style diet during paternal adolescence. The behavioral and physiological alterations in F1 offspring provide evidence of adaptive paternal programming via epigenetic inheritance. These findings have important implications for understanding paternally mediated intergenerational inheritance, and its relevance to offspring health and disease susceptibility.


Subject(s)
Behavior, Animal , Diet, Western , Gastrointestinal Microbiome , Paternal Inheritance , Social Behavior , Stress, Physiological , Animals , Female , Male , Mice
3.
Gen Comp Endocrinol ; 280: 47-53, 2019 09 01.
Article in English | MEDLINE | ID: mdl-30981703

ABSTRACT

There is increasing evidence that one's risk for psychiatric disturbances and metabolic syndromes is influenced by their parents' own health history, lifestyle and living environment. For example, paternal high fat diet is strongly linked to neuroendocrine dysregulation in offspring and increased risk for diabetes. The potential intergenerational impact of paternal stress has only just begun to emerge, with the initial evidence suggestive of greater risk for anxiety-related disorders. The hypothalamic-pituitary-adrenal (HPA)-axis is a key neuroendocrine signalling system involved in physiological homeostasis and stress response. In individuals, dysregulation of this system is closely associated with behavioral deficits and mood disorders. Various preclinical models of paternal stress have demonstrated robust behavioral shifts but little is known about the intergenerational modification of HPA axis function. This review will present evidence drawn from a range of laboratory mouse and rat models that the intergenerational influence of paternal stress on offspring behavioral phenotypes involve some level of HPA axis dysregulation. It makes the case that further investigations to comprehensively profile HPA axis function in offspring generations is warranted.


Subject(s)
Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Stress, Psychological/metabolism , Wounds and Injuries/psychology , Animals , Disease Models, Animal , Humans , Male , Mice, Inbred C57BL , Rats , Stress, Psychological/complications , Wounds and Injuries/complications , Wounds and Injuries/genetics
4.
Hum Mol Genet ; 25(14): 2923-2933, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27179791

ABSTRACT

Glutamatergic dysfunction has been implicated in the pathogenesis of depressive disorders and Huntington's disease (HD), in which depression is the most common psychiatric symptom. Synaptic glutamate homeostasis is regulated by cystine-dependent glutamate transporters, including GLT-1 and system xc- In HD, the enzyme regulating cysteine (and subsequently cystine) production, cystathionine-γ-lygase, has recently been shown to be lowered. The aim of the present study was to establish whether cysteine supplementation, using N-acetylcysteine (NAC) could ameliorate glutamate pathology through the cystine-dependent transporters, system xc- and GLT-1. We demonstrate that the R6/1 transgenic mouse model of HD has lower basal levels of cystine, and showed depressive-like behaviors in the forced-swim test. Administration of NAC reversed these behaviors. This effect was blocked by co-administration of the system xc- and GLT-1 inhibitors CPG and DHK, showing that glutamate transporter activity was required for the antidepressant effects of NAC. NAC was also able to specifically increase glutamate in HD mice, in a glutamate transporter-dependent manner. These in vivo changes reflect changes in glutamate transporter protein in HD mice and human HD post-mortem tissue. Furthermore, NAC was able to rescue changes in key glutamate receptor proteins related to excitotoxicity in HD, including NMDAR2B. Thus, we have shown that baseline reductions in cysteine underlie glutamatergic dysfunction and depressive-like behavior in HD and these changes can be rescued by treatment with NAC. These findings have implications for the development of new therapeutic approaches for depressive disorders.


Subject(s)
Acetylcysteine/administration & dosage , Depression/drug therapy , Excitatory Amino Acid Transporter 2/genetics , Huntington Disease/drug therapy , Receptors, N-Methyl-D-Aspartate/genetics , Animals , Autopsy , Behavior, Animal/drug effects , Chromosome Pairing/drug effects , Chromosome Pairing/genetics , Cystathionine gamma-Lyase/biosynthesis , Cystathionine gamma-Lyase/genetics , Cystine/biosynthesis , Depression/genetics , Depression/physiopathology , Disease Models, Animal , Excitatory Amino Acid Transporter 2/biosynthesis , Glutamic Acid/genetics , Glutamic Acid/metabolism , Humans , Huntington Disease/genetics , Huntington Disease/physiopathology , Mice , Mice, Transgenic
6.
J Physiol ; 600(20): 4419-4420, 2022 10.
Article in English | MEDLINE | ID: mdl-36190175
7.
Hum Mol Genet ; 23(15): 4051-63, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24634145

ABSTRACT

All cases of Huntington's disease (HD) are caused by mutant huntingtin protein (mhtt), yet the molecular mechanisms that link mhtt to disease symptoms are not fully elucidated. Given glycogen synthase kinase-3 (GSK3) is implicated in several neurodegenerative diseases as a molecular mediator of neuronal decline and widely touted as a therapeutic target, we investigated GSK3 in cells expressing mhtt, brains of R6/1 HD mice and post-mortem human brain samples. Consistency in data across the two models and the human brain samples indicate decreased GSK3 signalling contributes to neuronal dysfunction in HD. Inhibitory phosphorylation of GSK3 (pGSK3) was elevated in mhtt cells and this appeared related to an overall energy metabolism deficit as the mhtt cells had less ATP and inhibiting ATP production in control cells expressing non-pathogenic htt with paraquat also increased pGSK3. pGSK3 was increased and ATP levels decreased in the frontal cortex and striatum of R6/1 mice and levels of cortical pGSK3 inversely correlated with cognitive function of the mice. Consistent with decreased GSK3 activity in the R6/1 mouse brain, ß-catenin levels were increased and phosphorylation of collapsin response mediator protein-2 (CRMP2) decreased in the frontal cortex where inhibitory phosphorylation of GSK3 was the greatest. pGSK3 was predominantly undetectable in HD and healthy control human brain samples, but levels of total GSK3 were decreased in the HD-affected frontal cortex and this correlated with decreased pCRMP2. Thus, disruptions to cortical GSK3 signalling, possibly due to localized energy metabolism deficits, appear to contribute to the cognitive symptoms of HD.


Subject(s)
Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Glycogen Synthase Kinase 3/genetics , Huntington Disease/genetics , Intercellular Signaling Peptides and Proteins/genetics , Nerve Tissue Proteins/genetics , Adenosine Triphosphate/biosynthesis , Aged , Aged, 80 and over , Animals , Cell Line , Cerebral Cortex/pathology , Corpus Striatum/pathology , Disease Models, Animal , Female , Gene Expression Regulation , Glycogen Synthase Kinase 3/metabolism , Humans , Huntingtin Protein , Huntington Disease/metabolism , Huntington Disease/pathology , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Transgenic , Middle Aged , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Paraquat/pharmacology , Phosphorylation , Signal Transduction , beta Catenin/genetics , beta Catenin/metabolism
8.
Neurobiol Dis ; 69: 248-62, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24825316

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder caused by a tandem repeat mutation in the huntingtin gene. Lifestyle factors, such as lack of activity may contribute to the variability in the age of disease onset. Therefore, better understanding of environmental modifiers may uncover potential therapeutic approaches to delay disease onset and progression. Recent data suggest that HD patients and transgenic mouse models show a dysregulated stress response. In this present study, we elevated stress hormone levels through oral corticosterone (CORT) treatment and assessed its impact on the development of motor impairment and cognitive deficits using the R6/1 transgenic mouse model of HD. We found that CORT consumption did not alter rotarod performance of R6/1 HD or wild-type (WT) littermates. However, the onset of hippocampal-dependent Y-maze deficits was accelerated in male R6/1 mice by 5days of CORT treatment, whereas short term memory of WT and female R6/1 mice was unaffected. We then further investigated the male HD susceptibility to CORT by measuring TrkB activation, BDNF and glucocorticoid receptor expression as well as the level of cell proliferation in the hippocampus. CORT treatment increased the levels of phosphorylated TrkB in male R6/1 mice only. There were no effects of CORT on hippocampal BDNF protein or mRNA levels; nor on expression of the glucocorticoid receptors in any group. Hippocampal cell proliferation was decreased in male R6/1 mice and this was further reduced in CORT-drinking male R6/1 mice. Female mice (WT and R6/1) appeared to be protected from the impacts of CORT treatment in all our hippocampal measures. Overall, our data demonstrate that treatment with corticosterone is able to modulate the onset of HD symptomatology. We present the first evidence of a male-specific vulnerability to stress impacting on the development of short-term memory deficits in HD. More generally, we found that female mice were protected from the detrimental effects of CORT treatment on a variety of hippocampus-based measures. Hippocampal plasticity and memory in HD may be more susceptible to the impacts of stress in a sex-dependent manner. We propose clinical investigations of stress as a key environmental modifier of HD symptom onset.


Subject(s)
Corticosterone/metabolism , Huntington Disease/physiopathology , Memory Disorders/physiopathology , Memory, Short-Term/physiology , Age of Onset , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cell Proliferation/physiology , Disease Models, Animal , Hippocampus/physiopathology , Male , Maze Learning/physiology , Mice, Transgenic , Motor Activity/physiology , RNA, Messenger/metabolism , Receptor, trkB/metabolism , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/metabolism , Rotarod Performance Test , Sex Characteristics
9.
Psychopharmacology (Berl) ; 241(3): 555-567, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38170320

ABSTRACT

RATIONALE: Atypical attention orienting has been associated with some autistic symptoms, but the neural mechanisms remain unclear. The human Posner task, a classic attention orienting paradigm, was recently adapted for use with mice, supporting the investigation of the neurobiological underpinnings of atypical attention orienting in preclinical mouse models. OBJECTIVE: The current study tested mice expressing the autism-associated R451C gene mutation in neuroligin-3 (NL3) on the mouse-Posner (mPosner) task. METHODS: NL3R451C and wild-type (WT) mice were trained to respond to a validly or invalidly cued target on a touchscreen. The cue was a peripheral non-predictive flash in the exogenous task and a central spatially predictive image in the endogenous task. The effects of dopaminergic- and noradrenergic-modulating drugs, methylphenidate and atomoxetine, on task performance were assessed. RESULTS: In both tasks, mice were quicker and more accurate in the validly versus invalidly cued trials, consistent with results in the human Posner task. NL3R451C and WT mice showed similar response times and accuracy but responded differently when treated with methylphenidate and atomoxetine. Methylphenidate impaired exogenous attention disengagement in NL3R451C mice but did not significantly affect WT mice. Atomoxetine impaired endogenous orienting in WT mice but did not significantly affect NL3R451C mice. CONCLUSIONS: NL3R451C mice demonstrated intact attention orienting but altered responses to the pharmacological manipulation of the dopaminergic and noradrenergic networks. These findings expand our understanding of the NL3R451C mutation by suggesting that this mutation may lead to selective alterations in attentional processes.


Subject(s)
Autistic Disorder , Mice , Humans , Animals , Atomoxetine Hydrochloride/pharmacology , Neuroligins , Mutation/genetics , Attention
10.
Andrology ; 12(5): 1038-1057, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38576152

ABSTRACT

BACKGROUND: The epididymis has long been of interest owing to its role in promoting the functional maturation of the male germline. More recent evidence has also implicated the epididymis as an important sensory tissue responsible for remodeling of the sperm epigenome, both under physiological conditions and in response to diverse forms of environmental stress. Despite this knowledge, the intricacies of the molecular pathways involved in regulating the adaptation of epididymal tissue to paternal stressors remains to be fully resolved. OBJECTIVE: The overall objective of this study was to investigate the direct impact of corticosterone challenge on a tractable epididymal epithelial cell line (i.e., mECap18 cells), in terms of driving adaptation of the cellular proteome and phosphoproteome signaling networks. MATERIALS AND METHODS: The newly developed phosphoproteomic platform EasyPhos coupled with sequencing via an Orbitrap Exploris 480 mass spectrometer, was applied to survey global changes in the mECap18 cell (phospho)proteome resulting from sub-chronic (10-day) corticosterone challenge. RESULTS: The imposed corticosterone exposure regimen elicited relatively subtle modifications of the global mECap18 proteome (i.e., only 73 out of 4171 [∼1.8%] proteins displayed altered abundance). By contrast, ∼15% of the mECap18 phosphoproteome was substantially altered following corticosterone challenge. In silico analysis of the corresponding parent proteins revealed an activation of pathways linked to DNA damage repair and oxidative stress responses as well as a reciprocal inhibition of pathways associated with organismal death. Corticosterone challenge also induced the phosphorylation of several proteins linked to the biogenesis of microRNAs. Accordingly, orthogonal validation strategies confirmed an increase in DNA damage, which was ameliorated upon selective kinase inhibition, and an altered abundance profile of a subset of microRNAs in corticosterone-treated cells. CONCLUSIONS: Together, these data confirm that epididymal epithelial cells are reactive to corticosterone challenge, and that their response is tightly coupled to the opposing action of cellular kinases and phosphatases.


Subject(s)
Corticosterone , Epididymis , Epithelial Cells , Proteomics , Male , Epididymis/metabolism , Epididymis/drug effects , Animals , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Corticosterone/pharmacology , Proteomics/methods , Cell Line , Proteome/metabolism , Phosphoproteins/metabolism , Signal Transduction/drug effects
11.
J Physiol ; 591(1): 41-55, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23045340

ABSTRACT

Psychiatric disorders such as depression and anxiety are reported in patients with Huntington's disease (HD). Recent studies suggest beneficial effects of environmental enrichment (EE) on HD progression possibly through the serotonergic system. We investigated the potential effectiveness of EE in correcting the affective-like phenotype of female R6/1 HD mice. In addition to a behavioural battery of tests assessing depression and anxiety-related endophenotypes, we recorded physiological measures, including body temperature regulation and defecation rate as indices of stress reactivity. Finally, following identification of changes in serotonin (5-HT) receptor gene expression we measured the function of 5-HT(1A) auto- and hetero-receptors. We found that 8-week-old female HD mice exhibited higher immobility time in the forced swimming test and a decreased preference for saccharin solution. EE did not correct those depressive-like behaviours but reduced anxiety-related measures in unconditioned approach/avoidance conflict situations. Defecation rate in a large open field and change in temperature during exposure to the tail suspension test were both enhanced in HD compared to wild-type animals. Despite the enhanced hypothermic response to the 5-HT(1A) receptor agonist 8-OH-DPAT exhibited by HD mice, we found a reduction in 5-HT(1A) receptor-mediated stimulation of [(35)S]GTP-γ-S binding in the dorsal raphe nucleus and the hippocampus of HD animals. EE did not change 5-HT(1A) receptor function. Our data suggest that early EE has beneficial effects on the anxiety-like, but not on depression-like, behaviours in HD. This is the first evidence that these affective endophenotypes can be dissociated via this form of environmental stimulation. As 5-HT(1A) receptor dysfunction was not affected by EE, this receptor is unlikely to underlie the anxiety-related phenotype of HD. However, the specific regulatory role of the 5-HT(1A) autoreceptor in mediating depressive-like behaviour in HD remains to be elucidated. Interestingly, by comparing in vivo and in vitro results, our findings suggest that 8-OH-DPAT-induced hypothermia could be mediated by other targets besides the 5-HT(1A) autoreceptor, including hippocampal 5-HT(7) receptors.


Subject(s)
Environment , Huntington Disease/physiopathology , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Anxiety/physiopathology , Behavior, Animal/physiology , Depression/physiopathology , Disease Models, Animal , Emotions/physiology , Female , Hypothermia/chemically induced , Mice , Mice, Transgenic , Serotonin 5-HT1 Receptor Agonists/pharmacology , Stress, Psychological
12.
Neurobiol Dis ; 57: 12-27, 2013 Sep.
Article in English | MEDLINE | ID: mdl-22525570

ABSTRACT

Mental illness is the leading cause of disability worldwide. We are only just beginning to reveal and comprehend the complex interaction that exists between the genetic makeup of an organism and the potential modifying effect of the environment in which it lives, and how this translates into mediating susceptibility to neurological and psychiatric conditions. The capacity to address this issue experimentally has been facilitated by the availability of rodent models which allow the precise manipulation of genetic and environmental factors. In this review, we discuss the valuable nature of animal models in furthering our understanding of the relationship between genetic and environmental factors in affective illnesses, such as anxiety and depressive disorders. We first highlight the behavioral impairments exhibited by genetically targeted animal models of affective disorders, and then provide a discussion of the underlying neurobiology, focusing on animal models that involve exposure to stress. This is followed by a review of recent studies that report of beneficial effects of environmental manipulations such as environmental enrichment and enhanced physical activity and discuss the likely mechanisms that mediate those benefits.


Subject(s)
Disease Models, Animal , Gene-Environment Interaction , Mood Disorders/genetics , Animals , Hypothalamo-Hypophyseal System/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Mood Disorders/metabolism , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Neurotransmitter Agents/genetics , Neurotransmitter Agents/metabolism , Pituitary-Adrenal System/metabolism
13.
Eur J Neurosci ; 37(11): 1803-10, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23551162

ABSTRACT

Withdrawal from a chronic period of alcohol consumption is commonly associated with the manifestation of depression, potentially exerting a significant influence on treatment prospects and increasing the likelihood of relapse. Better therapeutic strategies need to be developed to assist with rehabilitation. Here, we report the detection of depression-related behaviours in a mouse model of 6-week free-choice ethanol (10%, v/v) consumption followed by 2-week abstinence. Mice abstinent from alcohol showed increased immobility time on the forced-swim test, reduced saccharin consumption and increased latency to feed in the novelty-suppressed feeding test. By comparison, there was no significant effect on anxiety-related behaviours as determined by testing on the light-dark box and elevated plus maze. We found that the provision of running-wheels through the duration of abstinence attenuated depressive behaviour in the forced-swim and novelty-suppressed feeding tests, and increased saccharin consumption. Given the link between withdrawal from addictive substances and depression, this model will be useful for the study of the pathophysiology underlying alcohol-related depression. The findings of this study establish an interaction between physical activity and the development of behavioural changes following cessation of alcohol consumption that could have implications for the development of rehabilitative therapies.


Subject(s)
Alcohol Drinking/physiopathology , Depression/therapy , Ethanol/toxicity , Physical Exertion , Animals , Anxiety/etiology , Anxiety/therapy , Depression/etiology , Depression/physiopathology , Female , Mice , Mice, Inbred C57BL , Substance Withdrawal Syndrome/complications , Substance Withdrawal Syndrome/physiopathology
14.
Brain Res ; 1807: 148319, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36898476

ABSTRACT

White-matter brain abnormalities have been found across a variety of psychiatric disorders. The extent of white matter pathology is proposed to be predictive of the severity of anxiety disorders. However, it is still unknown whether disruptions of white matter integrity precede, and are sufficient to give rise to, the behavioural symptoms. Interestingly, mood disturbances feature prominently in central demyelinating diseases such as multiple sclerosis. It is unclear whether the greater frequency of neuropsychiatric symptoms is linked to underlying neuropathology. In this study, we characterised male and female Tyro3 knockout (KO) mice using a variety of behavioural paradigms. Anxiety-related behaviours were assessed with the elevated-plus maze and light-dark box. Fear memory processing was assessed using fear conditioning and extinction paradigms. Finally, we assessed immobility time in the Porsolt swim test as a measure of depression-related behavioural despair. Surprisingly, loss of Tyro3 did not lead to manifestation of major shifts in baseline behaviour. We noted significant differences in habituation to novel environments and post-conditioning freezing levels of female Tyro3 KO mice, which are consistent with the female bias in anxiety disorders and could be indicative of maladaptive stress-responses. This study has demonstrated that white matter pathology related to a loss of Tyro3 is associated with pro-anxiety behavioural responses of female mice. Future studies could probe their contribution to increased risk for neuropsychiatric disorders when combined with stressful triggering events.


Subject(s)
Anxiety , Fear , Mice , Male , Female , Animals , Anxiety/psychology , Fear/physiology , Brain , Anxiety Disorders/genetics , Mice, Knockout , Behavior, Animal , Maze Learning/physiology
15.
Neuronal Signal ; 7(2): NS20220097, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37152245

ABSTRACT

Paternal preconceptional health factors, such as exposures to stress, diet and exercise, have been found to significantly influence offspring phenotypes in a range of animal models. Preclinical studies have provided evidence that paternal stress is associated with increased stress responsivity and anxiety-related traits, particularly in male offspring. It was previously reported that a paternal history of maternal separation (MS) led to male offspring (PatMS) displaying reduced cautious behavior during exploration of a novel environment. The neural basis for that absence of behavioral moderation is unclear. Here, we investigated the adaptive behavioral responses of control and PatMS male offspring in the predator odor risk-assessment task (PORT). PatMS mice failed to moderate their behaviors in the presence of a predator odor 2,4,5-trimethylthiazoline (TMT). c-Fos mapping revealed reduced cellular activation in fear-regulating brain regions of PatMS mice, such as in the cingulate cortex, dentate gyrus of the hippocampus and the basolateral amygdala. Expression of the paternally imprinted gene Grb10 (previously identified as a key molecular regulator of risk-taking behavior) was unaltered in PatMS mice. However, other paternal imprinted genes such as Igf2 and PEG3 were differentially expressed in PatMS mice. Overall, our study provides the first evidence of an intergenerational influence of preconceptional paternal stress exposure on offspring brain zunction relevant to risk-taking behavior, which is also independent of Grb10 gene expression.

16.
Neurobiol Dis ; 45(3): 913-22, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22198572

ABSTRACT

Insufficiency of the transcriptional regulator GTF2IRD1 has become a strong potential explanation for some of the major characteristic features of the neurodevelopmental disorder Williams-Beuren syndrome (WBS). Genotype/phenotype correlations in humans indicate that the hemizygous loss of the GTF2IRD1 gene and an adjacent paralogue, GTF2I, play crucial roles in the neurocognitive and craniofacial aspects of the disease. In order to explore this genetic relationship in greater detail, we have generated a targeted Gtf2ird1 mutation in mice that blocks normal GTF2IRD1 protein production. Detailed analyses of homozygous null Gtf2ird1 mice have revealed a series of phenotypes that share some intriguing parallels with WBS. These include reduced body weight, a facial deformity resulting from localised epidermal hyperplasia, a motor coordination deficit, alterations in exploratory activity and, in response to specific stress-inducing stimuli; a novel audible vocalisation and increased serum corticosterone. Analysis of Gtf2ird1 expression patterns in the brain using a knock-in LacZ reporter and c-fos activity mapping illustrates the regions where these neurological abnormalities may originate. These data provide new mechanistic insight into the clinical genetic findings in WBS patients and indicate that insufficiency of GTF2IRD1 protein contributes to abnormalities of facial development, motor function and specific behavioural disorders that accompany this disease.


Subject(s)
Focal Epithelial Hyperplasia/etiology , Motor Skills Disorders/etiology , Muscle Proteins/genetics , Mutation/genetics , Nuclear Proteins/genetics , Trans-Activators/genetics , Vocalization, Animal/physiology , Williams Syndrome/complications , Analysis of Variance , Animals , Animals, Newborn/blood , Body Temperature/genetics , Body Weight/genetics , Brain/metabolism , Circadian Rhythm/genetics , Corticosterone/blood , Disease Models, Animal , Exploratory Behavior/physiology , Fats , Female , Focal Epithelial Hyperplasia/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Skills Disorders/genetics , Muscle Strength , Muscle, Skeletal/pathology , Phenotype , Sex Factors , Sleep/genetics , Sound Spectrography , Stress, Psychological/genetics , Swimming/psychology , Williams Syndrome/genetics , Williams Syndrome/pathology
17.
Behav Brain Res ; 432: 113983, 2022 08 26.
Article in English | MEDLINE | ID: mdl-35777551

ABSTRACT

Stroke continues to be a major cause of mortality globally. Post-stroke treatment is complicated by the heterogenous nature of pathology and the emergence of secondary psychological symptoms are an additional challenge to the recovery process. Poststroke depression (PSD) is a common co-morbidity and is a major impediment to recovery. While selective serotonin reuptake inhibitors (SSRIs) have proven to be clinically efficacious in treating PSD, the pathogenic processes that underlie the manifestation of depressive mood post-stroke remains unclear. Furthermore, the use of SSRIs is associated with risks of intracerebral haemorrhage, so alternative treatment options need to be continuously explored. Exercise has been demonstrated to be beneficial for improving mood in humans and preclinical models of neurological conditions. Little is known of the mood-related benefits of physical exercise post-stroke. Using the middle cerebral artery occlusion (MCAO) mouse model of cerebral ischaemia, we investigated whether behavioural deficits emerge post-MCAO and could be rescued by voluntary wheel-running. We report that MCAO induced hypo-locomotion and anhedonia-related behaviours, with some improvements conferred by wheel-running. Serotonin transporter gene expression was increased in the MCAO hippocampus and frontal cortex, but this increase remained despite wheel-running. Wheel-running associated up-regulation of BDNF gene expression was unaffected in MCAO mice, reflecting conservation of key neuroplasticity molecular pathways. Taken together, our results highlight the need for further research into serotonergic modulation of the affective symptoms of stroke.


Subject(s)
Anxiety , Depression , Infarction, Middle Cerebral Artery , Physical Conditioning, Animal , Stroke , Animals , Anxiety/physiopathology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Depression/physiopathology , Disease Models, Animal , Gene Expression , Infarction, Middle Cerebral Artery/complications , Mice , Physical Conditioning, Animal/psychology , Receptors, Serotonin , Selective Serotonin Reuptake Inhibitors/pharmacology , Selective Serotonin Reuptake Inhibitors/therapeutic use , Stroke/complications , Stroke/drug therapy
18.
Neuronal Signal ; 6(1): NS20210053, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35036000

ABSTRACT

Cognitive impairments associated with advanced age involve alterations in the hippocampus that changes with experience throughout life. The hippocampus is critical for cognitive flexibility involved with extinction and reinstatement of conditioned fear. It is widely accepted that regular exercise can be beneficial for hippocampal function. Therefore, we asked whether chronic voluntary exercise in middle-aged mice can improve extinction and/or reinstatement of conditioned fear compared with standard-housing. Eight-month-old male and female C57Bl/6J mice had access to a running wheel or remained in standard-housing until 11 months of age. Alongside control standard-housed young adult (3-month-old) mice, they received tone-footshock pairings, which were subsequently extinguished with tone-alone presentations the next day. Half of the mice then received a reminder in the form of a single footshock. Male and female 11-month-old mice housed in standard conditions exhibited impaired reinstatement compared with young adult mice. However, for males that had access to a running wheel from 8 months of age, the reminder treatment rescued reinstatement ability. This was not observed in females. Additionally, exercise during middle age in both sexes increased expression of brain-derived neurotrophic factor (Bdnf) mRNA in the hippocampus, specifically exon 4 mRNA. These results show that, at least for males, physical exercise is beneficial for reducing age-related decline in cognitive abilities. Despite not affecting reinstatement, exercise also increased Bdnf gene expression in the female hippocampus, which could potentially benefit other forms of hippocampus-dependent cognition.

19.
Hum Mol Genet ; 18(4): 753-66, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19008301

ABSTRACT

Dysregulation of the serotonergic signaling system has been implicated in the pathology of mood disorders including depression, and various rodent models of disrupted serotonergic signaling display depression-related behavioral phenotypes. Depression is a common neuropsychiatric feature of preclinical Huntington's disease (HD) but the underlying changes in the HD brain contributing to the development of depression are unknown. Using the R6/1 transgenic mouse model of HD, we show that pre-motor symptomatic HD mice display sex-specific depressive-related behaviors on the forced-swim (FST), tail-suspension (TST) and novelty-suppressed feeding (NSFT) tests while having muted responses to acute anti-depressant administration. The baseline behaviors of HD mice were similar to the behavioral phenotypes of serotonin (5-HT) receptor and transporter null mutants, and gene expression of specific serotonin receptors were subsequently found to be reduced in the hippocampus and cortex of HD mice. Female HD mice had an additional deficit in cortical expression of serotonin transporter (SerT). Environmental enrichment normalized the FST behavioral response of female HD mice corresponding with increased gene expression of specific 5-HT receptors in the hippocampus and cortex. Our findings implicate altered serotonergic signaling as the basis for the development of depression during the preclinical stages of HD.


Subject(s)
Depression/physiopathology , Gene Expression , Huntington Disease/psychology , Receptor, Serotonin, 5-HT1B/metabolism , Receptors, Serotonin/metabolism , Animals , Antidepressive Agents/administration & dosage , Behavior, Animal , Body Weight , Brain/metabolism , Brain/physiopathology , Disease Models, Animal , Female , Humans , Huntington Disease/drug therapy , Huntington Disease/genetics , Huntington Disease/physiopathology , Male , Mice , Mice, Transgenic , Motor Activity , Receptor, Serotonin, 5-HT1B/genetics , Receptors, Serotonin/genetics , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Species Specificity
20.
World J Psychiatry ; 11(10): 711-735, 2021 Oct 19.
Article in English | MEDLINE | ID: mdl-34733638

ABSTRACT

Mental illness remains the greatest chronic health burden globally with few in-roads having been made despite significant advances in genomic knowledge in recent decades. The field of psychiatry is constantly challenged to bring new approaches and tools to address and treat the needs of vulnerable individuals and subpopulations, and that has to be supported by a continuous growth in knowledge. The majority of neuropsychiatric symptoms reflect complex gene-environment interactions, with epigenetics bridging the gap between genetic susceptibility and environmental stressors that trigger disease onset and drive the advancement of symptoms. It has more recently been demonstrated in preclinical models that epigenetics underpins the transgenerational inheritance of stress-related behavioural phenotypes in both paternal and maternal lineages, providing further supporting evidence for heritability in humans. However, unbiased prospective studies of this nature are practically impossible to conduct in humans so preclinical models remain our best option for researching the molecular pathophysiologies underlying many neuropsychiatric conditions. While rodents will remain the dominant model system for preclinical studies (especially for addressing complex behavioural phenotypes), there is scope to expand current research of the molecular and epigenetic pathologies by using invertebrate models. Here, we will discuss the utility and advantages of two alternative model organisms-Caenorhabditis elegans and Drosophila melanogaster-and summarise the compelling insights of the epigenetic regulation of transgenerational inheritance that are potentially relevant to human psychiatry.

SELECTION OF CITATIONS
SEARCH DETAIL