Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Trends Genet ; 38(7): 662-675, 2022 07.
Article in English | MEDLINE | ID: mdl-35410793

ABSTRACT

Maternal immune activation (MIA) and infection during pregnancy are known to reprogramme offspring phenotypes. However, the epigenetic effects of preconceptual paternal infection and paternal immune activation (PIA) are not currently well understood. Recent reports show that paternal infection and immune activation can affect offspring phenotypes, particularly brain function, behaviour, and immune system functioning, across multiple generations without re-exposure to infection. Evidence from other environmental exposures indicates that epigenetic inheritance also occurs in humans. Given the growing impact of the coronavirus disease 2019 (COVID-19) pandemic, it is imperative that we investigate all of the potential epigenetic mechanisms and multigenerational phenotypes that may arise from both maternal and paternal severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, as well as associated MIA, PIA, and inflammation. This will allow us to understand and, if necessary, mitigate any potential changes in disease susceptibility in the children, and grandchildren, of affected parents.


Subject(s)
COVID-19 , SARS-CoV-2 , COVID-19/genetics , Disease Susceptibility , Epigenesis, Genetic , Female , Humans , Parents , Pregnancy , SARS-CoV-2/genetics
2.
Brain Behav Immun ; 119: 520-538, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38636562

ABSTRACT

Paternal pre-conceptual exposures, including stress, diet, substance abuse, parasite infection, and viral immune activation via Poly I:C, have been reported to influence the brains and behavior of offspring through sperm epigenetic changes. However, the effects of paternal (F0) pre-conceptual exposure to bacterial-induced immune activation on the behavior and physiology of F1 and F2 generations remain unexplored. We examined this using C57BL/6J mice. Eight-week-old males (F0) received a single intraperitoneal injection of the bacterial mimetic lipopolysaccharide (LPS: 5 mg/kg) or 0.9 % saline (vehicle control) before mating with naïve females at four weeks post-injection. Comprehensive behavioral assessments were conducted to investigate anxiety, social behaviors, depressive-like behaviors and cognition in both the F1 and F2 generations within the age range of 8 to 14 weeks. Results demonstrated that only female offspring of LPS-exposed fathers exhibited reduced anxiety levels in the light/dark box, large open field, and novelty-suppressed feeding test. These F1 female offspring also exhibited heightened sociability in the 3-chambered social interaction test and a reduced preference for saccharin in the saccharin preference test. Additionally, the F1 male offspring of LPS-challenged males demonstrated an increased total distance traveled in the light/dark box and a longer distance covered in the light zone. They also exhibited diminished preference for social novelty in the 3-chambered social interaction test and an elevated novel arm preference index in the Y-maze. In the F2 generation, male descendants of LPS-treated fathers showed reduced latency to feed in the novelty-suppressed feeding test. Additionally, the F2 generation of LPS-challenged fathers, but not the F1 generation, displayed enhanced immune response in both sexes after an acute LPS immune challenge (5 mg/kg). Analysis of sperm small noncoding RNA profiles from LPS-treated F0 mice revealed significant changes at 4 weeks after administration of LPS. These changes included three microRNAs, eight PIWI-interacting RNAs, and two transfer RNAs, exhibiting significant upregulation (mmu-miR-146a-5p, mmu-piR-27082 and mmu-piR-29102) or downregulation (mmu-miR-5110, mmu-miR-467e-3p, mmu-piR-22583, mmu-piR-23548, mmu-piR-36341, mmu-piR-50293, mmu-piR-16583, mmu-piR-36507, Mus_musculus_tRNA-Ile-AAT-2-1 and Mus_musculus_tRNA-Tyr-GTA-1-1). Additionally, we detected 52 upregulated small noncoding RNAs (including 9 miRNAs, 41 piRNAs, and 2 tRNAs) and 7 downregulated small noncoding RNAs (3 miRNAs, 3 piRNAs, and 1 tRNA) in the sperm of F1 offspring from LPS-treated males. These findings provide compelling evidence for the involvement of epigenetic mechanisms in the modulation of brain function and immunity, and associated behavioral and immunological traits, across generations, in response to bacterial infection.


Subject(s)
Anxiety , Behavior, Animal , Lipopolysaccharides , Mice, Inbred C57BL , Spermatozoa , Animals , Male , Female , Mice , Lipopolysaccharides/pharmacology , Spermatozoa/metabolism , Behavior, Animal/physiology , Social Behavior , Bacterial Infections/immunology , Depression/metabolism , Epigenesis, Genetic , MicroRNAs/metabolism , MicroRNAs/genetics , Paternal Exposure/adverse effects
3.
Brain Behav Immun ; 115: 258-279, 2024 01.
Article in English | MEDLINE | ID: mdl-37820975

ABSTRACT

Paternal pre-conceptual environmental experiences, such as stress and diet, can affect offspring brain and behavioral phenotypes via epigenetic modifications in sperm. Furthermore, maternal immune activation due to infection during gestation can reprogram offspring behavior and brain functioning in adulthood. However, the effects of paternal pre-conceptual exposure to immune activation on the behavior and physiology of offspring (F1) and grand-offspring (F2) are not currently known. We explored effects of paternal pre-conceptual exposure to viral-like immune activation on F1 and F2 behavioral and physiological phenotypes using a C57BL/6J mouse model. Males were treated with a single injection (intraperitoneal) of the viral mimetic polyinosinic:polycytidylic acid (Poly I:C: 12 mg/kg) then bred with naïve female mice four weeks after the Poly I:C (or 0.9% saline control) injection. The F1 offspring of Poly I:C treated fathers displayed increased depression-like behavior in the Porsolt swim test, an altered stress response in the novelty-suppressed feeding test, and significant transcriptomic changes in their hippocampus. Additionally, the F1 male offspring of Poly I:C treated F0 males showed significantly increased immune responsivity after a Poly I:C immune challenge (12 mg/kg). Furthermore, the F2 male grand-offspring took longer to enter and travelled significantly shorter distances in the light zone of the light/dark box. An analysis of the small noncoding RNA profiles in sperm from Poly I:C treated males and their male offspring revealed significant effects of Poly I:C on the sperm microRNA content at the time of conception and on the sperm PIWI-interacting RNA content of the male offspring. Notably, eight miRNAs with an FDR < 0.05 (miR-141-3p, miR-126b-5p, miR-669o-5p, miR-10b-3p, miR-471-5p, miR-463-5p, miR-148b-3p, and miR-181c-5p) were found to be significantly downregulated in the sperm of Poly I:C treated males. Collectively, we demonstrate that paternal pre-conceptual exposure to a viral immune challenge results in both intergenerational and transgenerational effects on brain and behavior that may be mediated by alterations in the sperm small noncoding RNA content.


Subject(s)
MicroRNAs , RNA, Small Untranslated , Male , Female , Mice , Animals , Humans , Mice, Inbred C57BL , Semen , Spermatozoa , Fathers , MicroRNAs/genetics , MicroRNAs/pharmacology , RNA, Small Untranslated/pharmacology , Poly I/pharmacology
4.
Brain Behav Immun ; 116: 404-418, 2024 02.
Article in English | MEDLINE | ID: mdl-38142919

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder involving psychiatric, cognitive and motor deficits, as well as peripheral symptoms, including gastrointestinal dysfunction. The R6/1 HD mouse model expresses a mutant human huntingtin transgene and has been shown to provide an accurate disease model. Recent evidence of gut microbiome disruption was shown in preclinical and clinical HD. Therefore, we aimed to assess the potential role of gut microbial modulation in the treatment of HD. The R6/1 HD mice and wild-type littermate controls were randomised to receive diets containing different amounts of fibre: high-fibre (10 % fibre), control (5 % fibre), or zero-fibre (0 % fibre), from 6 to 20 weeks of age. We characterized the onset and progression of motor, cognitive and affective deficits, as well as gastrointestinal function and gut morphological changes. Faeces were collected for gut microbiome profiling using 16S rRNA sequencing, at 14 and 20 weeks of age. When compared to the control diet, high-fibre diet improved the performance of HD mice in behavioral tests of cognitive and affective function, as well as the gastrointestinal function of both HD and wild-type mice. While the diets changed the beta diversity of wild-type mice, no statistical significance was observed at 14 or 20 weeks of age within the HD mice. Analysis of Composition of Microbiomes with Bias Correction (ANCOM-BC) models were performed to evaluate microbiota composition, which identified differences, including a decreased relative abundance of the phyla Actinobacteriota, Campylobacterota and Proteobacteria and an increased relative abundance of the families Bacteroidaceae, Oscillospiraceae and Ruminococcaceae in HD mice when compared to wild-type mice after receiving high-fibre diet. PICRUSt2 revealed that high-fibre diet also decreased potentially pathogenic functional pathways in HD. In conclusion, high-fibre intake was effective in enhancing gastrointestinal function, cognition and affective behaviors in HD mice. These findings indicate that dietary fibre interventions may have therapeutic potential in Huntington's disease to delay clinical onset, and have implications for related disorders exhibiting dysfunction of the gut-brain axis.


Subject(s)
Huntington Disease , Humans , Mice , Animals , Huntington Disease/therapy , Huntington Disease/genetics , Mice, Transgenic , RNA, Ribosomal, 16S , Cognition , Disease Models, Animal , Dietary Fiber
5.
Brain Behav Immun ; 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39293692

ABSTRACT

The paternal environment prior to conception has been demonstrated to influence offspring physiology and behavior, with the sperm epigenome (including noncoding RNAs) proposed as a potential facilitator of non-genetic inheritance. Whilst the maternal gut microbiome has been established as an important influence on offspring development, the impact of the paternal gut microbiota on offspring development, health and behavior is largely unknown. Gut microbiota have major influences on immunity, and thus we hypothesized that it may be relevant to paternal immune activation modulating epigenetic inheritance in mice. Therefore, male C57BL/6J mice (F0) were orally administered non-absorbable antibiotics via drinking water in order to substantially deplete their gut microbiota. Four weeks after administration of the antibiotics (gut microbiome depletion), F0 male mice were then mated with naïve female mice. The F1 offspring of the microbiome-depleted males had reduced body weight as well as altered gut morphology (shortened colon length). F1 females showed significant alterations in affective behaviors, including measures of anxiety and depressive-like behaviors, indicating altered development. Analysis of small noncoding RNAs in the sperm of F0 mice revealed that gut microbiome depletion is associated with differential expression of 8 different PIWI-interacting RNAs (piRNAs), each of which has the potential to modulate the expression of multiple downstream gene targets, and thus influence epigenetic inheritance and offspring development. This study demonstrates that the gut-germline axis influences sperm small RNA profiles, offspring physiology, with specific impacts on offspring affective and/or coping behaviors. These findings may have broader implications for other animal species with comparable gut microbiota, intergenerational epigenetics and developmental biology, including humans.

6.
Neurobiol Dis ; 142: 104958, 2020 08.
Article in English | MEDLINE | ID: mdl-32526274

ABSTRACT

Huntington's disease (HD) is an extraordinary disorder that usually strikes when individuals are in the prime of their lives, as was the case for the influential 20th century musician Woody Guthrie. HD demonstrates the exceptionally fine line between life and death in such 'genetic diseases', as the only difference between those who suffer horribly and die slowly of this disease is often just a handful of extra tandem repeats (beyond the normal polymorphic range) in a genome that constitutes over 3 billion paired nucleotides of DNA. Furthermore, HD presents as a complex and heterogenous combination of psychiatric, cognitive and motor symptoms, so can appear as an unholy trinity of 'three disorders in one'. The autosomal dominant nature of the disorder is also extremely challenging for affected families, as a 'flip of a coin' dictates which children inherit the mutation from their affected parent, and the gene-negative family members bear the burden of caring for the other half of the family that is affected. In this review, we will focus on one of the earliest, and most devastating, symptoms associated with HD, depression, which has been reported to affect approximately half of gene-positive HD family members. We will discuss the pathogenesis of HD, and depressive symptoms in particular, including molecular and cellular mechanisms, and potential genetic and environmental modifiers. This expanding understanding of HD pathogenesis may not only lead to novel therapeutic options for HD families, but may also provide insights into depression in the wider population, which has the greatest burden of disease of any disorder and an enormous unmet need for new therapies.


Subject(s)
Depression/psychology , Depressive Disorder/psychology , Huntington Disease/psychology , Depression/genetics , Depressive Disorder/genetics , Humans , Huntington Disease/genetics
7.
Neurobiol Dis ; 134: 104621, 2020 02.
Article in English | MEDLINE | ID: mdl-31628992

ABSTRACT

The last decade has witnessed an exponentially growing interest in gut microbiota and the gut-brain axis in health and disease. Accumulating evidence from preclinical and clinical research indicate that gut microbiota, and their associated microbiomes, may influence pathogenic processes and thus the onset and progression of various diseases, including neurological and psychiatric disorders. In fact, gut dysbiosis (microbiota dysregulation) has been associated with a range of neurodegenerative diseases, including Alzheimer's, Parkinson's, Huntington's and motor neuron disease, as well as multiple sclerosis. The gut microbiota constitutes a dynamic microbial system constantly challenged by many biological variables, including environmental factors. Since the gut microbiota constitute a changeable and experience-dependent ecosystem, they provide potential therapeutic targets that can be modulated as new interventions for dysbiosis-related disorders, including neurodegenerative diseases. This article reviews the evidence for environmental modulation of gut microbiota and its relevance to brain disorders, exploring in particular the implications for neurodegenerative diseases. We will focus on three major environmental factors that are known to influence the onset and progression of those diseases, namely exercise, diet and stress. Further exploration of environmental modulation, acting via both peripheral (e.g. gut microbiota and associated metabolic dysfunction or 'metabolopathy') and central (e.g. direct effects on CNS neurons and glia) mechanisms, may lead to the development of novel therapeutic approaches, such as enviromimetics, for a wide range of neurological and psychiatric disorders.


Subject(s)
Diet , Exercise , Gastrointestinal Microbiome , Neurodegenerative Diseases , Stress, Psychological , Animals , Dysbiosis , Humans , Neurodegenerative Diseases/microbiology , Neurodegenerative Diseases/physiopathology
8.
Brain ; 146(12): 4791-4793, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37873966
10.
Int J Neuropsychopharmacol ; 20(6): 445-454, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28339618

ABSTRACT

Background: Growing evidence supports the existence of neurobiological trait abnormalities in individuals at genetic risk for bipolar disorder. The aim of this study was to examine potential differences in brain-derived neurotrophic factor, cytokines, oxidative stress, and telomere length markers between patients with bipolar disorder, their siblings, and healthy controls. Methods: Thirty-six patients with bipolar disorder type I, 39 siblings, and 44 healthy controls were assessed. Serum levels of brain-derived neurotrophic factor, interleukin-6, interleukin-10, tumor necrosis factor-α, C-C motif chemokine 11, C-C motif chemokine 24, and 3-nitrotyrosine were measured, as were the activities of glutathione peroxidase, glutathione reductase, and glutathione S-transferase. Telomere length (T/S ratio) was measured using quantitative polymerase chain reaction. Results: Telomere length was different between the 3 groups (P = .041) with both patients and siblings showing a shorter T/S ratio compared with healthy controls. Patients showed increased levels of interleukin-6 (P = .005) and interleukin-10 (P = .002) compared with controls as well as increased levels of interleukin-6 (p = 0.014) and CCL24 (P = .016) compared with their siblings. C-C motif chemokine 11 levels were increased in siblings compared with controls (P = .015), and a similar tendency was found in patients compared with controls (P = .045). Glutathione peroxidase activity was decreased in patients compared with controls (P = .006) and siblings (P = .025). No differences were found for the other markers. Conclusions: The present results suggest that unaffected siblings may present accelerated aging features. These neurobiological findings may be considered as endophenotypic traits. Further prospective studies are warranted.


Subject(s)
Bipolar Disorder/metabolism , Cellular Senescence/physiology , Inflammation/blood , Oxidative Stress/physiology , Siblings , Telomere/metabolism , Biomarkers/blood , Bipolar Disorder/drug therapy , Cross-Sectional Studies , Female , Genetic Predisposition to Disease , Humans , Interview, Psychological , Male , Middle Aged
13.
J Neural Transm (Vienna) ; 122(5): 643-51, 2015 May.
Article in English | MEDLINE | ID: mdl-25233798

ABSTRACT

Streptococcus pneumoniae is a common cause of bacterial meningitis, with a high mortality rate and neurological sequelae. In contrast, folic acid plays an important role in neuroplasticity and the preservation of neuronal integrity. In the present study, we evaluated the influence of folic acid on memory, oxidative damage, enzymatic defence, and brain-derived neurotrophic factor (BDNF) expression in experimental pneumococcal meningitis. In animals that received folic acid at a dose of 10 or 50 mg, there was a reduction in both crossing and rearing during an open-field task compared with the training session, demonstrating habituation memory. During a step-down inhibitory avoidance task, there was a difference between the training and the test sessions, demonstrating aversive memory. In the hippocampus, BDNF expression decreased in the meningitis group; however, adjuvant treatment with 10 mg of folic acid increased BDNF expression, decreased lipid peroxidation, protein carbonylation, nitrate/nitrite levels, and myeloperoxidase activity and increased superoxide dismutase activity. In frontal cortex adjuvant treatment with 10 mg of folic acid decreased lipid peroxidation and protein carbonylation. There is substantial interest in the role of folic acid and related pathways in nervous system function and in folic acid's potential therapeutic effects. Here, adjuvant treatment with vitamin B9 prevented memory impairment in experimental pneumococcal meningitis.


Subject(s)
Cognition Disorders/prevention & control , Folic Acid/pharmacology , Frontal Lobe/drug effects , Hippocampus/drug effects , Meningitis, Pneumococcal/drug therapy , Nootropic Agents/pharmacology , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Brain-Derived Neurotrophic Factor/metabolism , Cognition Disorders/etiology , Cognition Disorders/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Frontal Lobe/physiopathology , Hippocampus/physiopathology , Inhibition, Psychological , Male , Memory/drug effects , Meningitis, Pneumococcal/complications , Meningitis, Pneumococcal/physiopathology , Motor Activity/drug effects , Motor Activity/physiology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Random Allocation , Rats, Wistar
14.
Int J Neuropsychopharmacol ; 17(1): 33-40, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24067327

ABSTRACT

Recent reports suggest that brain-derived neurotrophic factor (BDNF) could be a biomarker for relapse, drug craving and withdrawal severity. In particular, elevated BDNF levels among former cocaine users have been associated with higher rates of relapse in 90 d. However, no data are available on BDNF levels at baseline and during crack cocaine withdrawal. This study evaluated BDNF among crack cocaine users during inpatient treatment, before and after withdrawal, vs. healthy controls. Clinical correlates with changes in BDNF levels were also assessed. Serum BDNF was evaluated in 49 male crack users on the first and last days of hospitalization and in 97 healthy controls. Serum BDNF was assayed using a sandwich ELISA kit. BDNF levels were significantly lower upon admission when compared to controls, even after adjustment for age, length of inpatient treatment, number of crack rocks used in the last 30 d, years of crack use and interaction between the latter two variables. At discharge, BDNF levels between patients and controls were similar. Number of crack rocks used in the last 30 d and years of crack use were inversely correlated with the outcome. Our findings show that BDNF levels increase during early crack cocaine withdrawal, at an inverse correlation with number of crack rocks used in the last 30 d and years of crack use.


Subject(s)
Brain-Derived Neurotrophic Factor/blood , Cocaine-Related Disorders/blood , Crack Cocaine/adverse effects , Substance Withdrawal Syndrome/blood , Adult , Biomarkers/blood , Case-Control Studies , Hospitalization , Humans , Male , Young Adult
15.
Int J Neuropsychopharmacol ; 18(1)2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25522387

ABSTRACT

BACKGROUND: Impaired stress resilience and a dysfunctional hypothalamic-pituitary-adrenal (HPA) axis are suggested to play key roles in the pathophysiology of illness progression in bipolar disorder (BD), but the mechanisms leading to this dysfunction have never been elucidated. This study aimed to examine HPA axis activity and underlying molecular mechanisms in patients with BD and unaffected siblings of BD patients. METHODS: Twenty-four euthymic patients with BD, 18 siblings of BD patients, and 26 healthy controls were recruited for this study. All subjects underwent a dexamethasone suppression test followed by analyses associated with the HPA axis and the glucocorticoid receptor (GR). RESULTS: Patients with BD, particularly those at a late stage of illness, presented increased salivary post-dexamethasone cortisol levels when compared to controls (p = 0.015). Accordingly, these patients presented reduced ex vivo GR responsiveness (p = 0.008) and increased basal protein levels of FK506-binding protein 51 (FKBP51, p = 0.012), a co-chaperone known to desensitize GR, in peripheral blood mononuclear cells. Moreover, BD patients presented increased methylation at the FK506-binding protein 5 (FKBP5) gene. BD siblings presented significantly lower FKBP51 protein levels than BD patients, even though no differences were found in FKBP5 basal mRNA levels. CONCLUSIONS: Our data suggest that the epigenetic modulation of the FKBP5 gene, along with increased FKBP51 levels, is associated with the GR hyporesponsiveness seen in BD patients. Our findings are consistent with the notion that unaffected first-degree relatives of BD patients share biological factors that influence the disorder, and that such changes are more pronounced in the late stages of the illness.


Subject(s)
Bipolar Disorder/metabolism , Hydrocortisone/metabolism , Receptors, Glucocorticoid/metabolism , Tacrolimus Binding Proteins/metabolism , Adrenocorticotropic Hormone/blood , Adult , Bipolar Disorder/drug therapy , Bipolar Disorder/genetics , Dexamethasone/pharmacology , Disease Progression , Epigenesis, Genetic , Female , Glucocorticoids/pharmacology , Humans , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Male , Methylation , Middle Aged , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , RNA, Messenger/metabolism , Saliva/metabolism , Siblings , Tacrolimus Binding Proteins/genetics
16.
FEBS J ; 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38426291

ABSTRACT

Huntington's disease (HD) is a currently incurable neurogenerative disorder and is typically characterized by progressive movement disorder (including chorea), cognitive deficits (culminating in dementia), psychiatric abnormalities (the most common of which is depression), and peripheral symptoms (including gastrointestinal dysfunction). There are currently no approved disease-modifying therapies available for HD, with death usually occurring approximately 10-25 years after onset, but some therapies hold promising potential. HD subjects are often burdened by chronic diarrhea, constipation, esophageal and gastric inflammation, and a susceptibility to diabetes. Our understanding of the microbiota-gut-brain axis in HD is in its infancy and growing evidence from preclinical and clinical studies suggests a role of gut microbial population imbalance (gut dysbiosis) in HD pathophysiology. The gut and the brain can communicate through the enteric nervous system, immune system, vagus nerve, and microbiota-derived-metabolites including short-chain fatty acids, bile acids, and branched-chain amino acids. This review summarizes supporting evidence demonstrating the alterations in bacterial and fungal composition that may be associated with HD. We focus on mechanisms through which gut dysbiosis may compromise brain and gut health, thus triggering neuroinflammatory responses, and further highlight outcomes of attempts to modulate the gut microbiota as promising therapeutic strategies for HD. Ultimately, we discuss the dearth of data and the need for more longitudinal and translational studies in this nascent field. We suggest future directions to improve our understanding of the association between gut microbes and the pathogenesis of HD, and other 'brain and body disorders'.

17.
J Affect Disord ; 363: 520-531, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39043310

ABSTRACT

BACKGROUND: Compulsive- and anxiety-like behaviour can be efficiently modelled in SAPAP3 knockout (KO) mice, a preclinical model of relevance to obsessive-compulsive disorder (OCD). Although there is emerging evidence in the clinical literature of gastrointestinal dysfunction in OCD, no previous studies have investigated gut function in preclinical models of relevance to OCD. Similarly, the effects of voluntary exercise (EX) or environmental enrichment (EE) have not yet been explored in this context. METHOD: We comprehensively phenotyped the SAPAP3 KO mouse model, including the assessment of grooming microstructure, anxiety- and depressive-like behaviour, and gastrointestinal function. Mice were exposed to either standard housing (SH), exercise (EX, provided by giving mice access to running wheels), or environmental enrichment (EE) for 4 weeks to investigate the effects of enriched housing conditions in this animal model relevant to OCD. FINDINGS: Our study is the first to assess grooming microstructure, perseverative locomotor activity, and gastrointestinal function in SAPAP3 KO mice. We are also the first to report a sexually dimorphic effect of grooming in young-adult SAPAP3 KO mice; along with changes to grooming patterning and indicators of gut dysfunction, which occurred in the absence of gut dysbiosis in this model. Overall, we found no beneficial effects of voluntary exercise or environmental enrichment interventions in this mouse model; and unexpectedly, we revealed a deleterious effect of wheel-running exercise on grooming behaviour. We suspect that the detrimental effects of experimental housing in our study may be indicative of off-target effects of stress-a conclusion that warrants further investigation into the effects of chronic stress in this preclinical model of compulsive behaviour.


Subject(s)
Compulsive Behavior , Disease Models, Animal , Grooming , Mice, Knockout , Obsessive-Compulsive Disorder , Animals , Grooming/physiology , Mice , Male , Obsessive-Compulsive Disorder/physiopathology , Obsessive-Compulsive Disorder/genetics , Compulsive Behavior/physiopathology , Compulsive Behavior/genetics , Female , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/deficiency , Behavior, Animal/physiology , Anxiety/physiopathology , Depression/physiopathology , Gastrointestinal Diseases/physiopathology , Environment , Physical Conditioning, Animal/physiology
18.
Nat Neurosci ; 27(7): 1274-1284, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38773348

ABSTRACT

Retrotransposons are mobile DNA sequences duplicated via transcription and reverse transcription of an RNA intermediate. Cis-regulatory elements encoded by retrotransposons can also promote the transcription of adjacent genes. Somatic LINE-1 (L1) retrotransposon insertions have been detected in mammalian neurons. It is, however, unclear whether L1 sequences are mobile in only some neuronal lineages or therein promote neurodevelopmental gene expression. Here we report programmed L1 activation by SOX6, a transcription factor critical for parvalbumin (PV) interneuron development. Mouse PV interneurons permit L1 mobilization in vitro and in vivo, harbor unmethylated L1 promoters and express full-length L1 mRNAs and proteins. Using nanopore long-read sequencing, we identify unmethylated L1s proximal to PV interneuron genes, including a novel L1 promoter-driven Caps2 transcript isoform that enhances neuron morphological complexity in vitro. These data highlight the contribution made by L1 cis-regulatory elements to PV interneuron development and transcriptome diversity, uncovered due to L1 mobility in this milieu.


Subject(s)
Interneurons , Long Interspersed Nucleotide Elements , Parvalbumins , Animals , Interneurons/metabolism , Interneurons/physiology , Mice , Long Interspersed Nucleotide Elements/genetics , Parvalbumins/metabolism , Retroelements/genetics , Male , Neurogenesis/physiology , Neurogenesis/genetics , Mice, Inbred C57BL , Gene Expression Regulation, Developmental/genetics
19.
Methods Mol Biol ; 2687: 77-91, 2023.
Article in English | MEDLINE | ID: mdl-37464164

ABSTRACT

Mice and other rodent models have been widely used to understand the role of the gut microbiome in various neurological and psychiatric disorders. Here we describe a protocol to characterize the structural and functional phenotype of the rodent gut and to examine the gut microbiota composition through V4 16S rRNA gene sequencing and microbiome profiling. This protocol will have utility for those investigating the gut, and associated microbiota, in a wide range of different rodent models of human disorders.


Subject(s)
Mental Disorders , Microbiota , Humans , Mice , Animals , Rodentia/genetics , RNA, Ribosomal, 16S/genetics , Microbiota/genetics , Gastrointestinal Tract , Mental Disorders/genetics
20.
Gastroenterol Rep (Oxf) ; 10: goac017, 2022.
Article in English | MEDLINE | ID: mdl-35582476

ABSTRACT

Recent research has been uncovering the role of the gut microbiota for brain health and disease. These studies highlight the role of gut microbiota on regulating brain function and behavior through immune, metabolic, and neuronal pathways. In this review we provide an overview of the gut microbiota axis pathways to lay the groundwork for upcoming sessions on the links between the gut microbiota and neurogenerative disorders. We also discuss how the gut microbiota may act as an intermediate factor between the host and the environment to mediate disease onset and neuropathology. Based on the current literature, we further examine the potential for different microbiota-based therapeutic strategies to prevent, to modify, or to halt the progress of neurodegeneration.

SELECTION OF CITATIONS
SEARCH DETAIL