Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Commun Biol ; 7(1): 80, 2024 01 10.
Article in English | MEDLINE | ID: mdl-38200107

ABSTRACT

Vagus nerve signaling is a key component of the gut-brain axis and regulates diverse physiological processes that decline with age. Gut to brain vagus firing patterns are regulated by myenteric intrinsic primary afferent neuron (IPAN) to vagus neurotransmission. It remains unclear how IPANs or the afferent vagus age functionally. Here we identified a distinct ageing code in gut to brain neurotransmission defined by consistent differences in firing rates, burst durations, interburst and intraburst firing intervals of IPANs and the vagus, when comparing young and aged neurons. The aminosterol squalamine changed aged neurons firing patterns to a young phenotype. In contrast to young neurons, sertraline failed to increase firing rates in the aged vagus whereas squalamine was effective. These results may have implications for improved treatments involving pharmacological and electrical stimulation of the vagus for age-related mood and other disorders. For example, oral squalamine might be substituted for or added to sertraline for the aged.


Subject(s)
Sensory Receptor Cells , Sertraline , Cholestanols , Vagus Nerve
2.
Biol Psychiatry ; 95(4): 310-318, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-37839790

ABSTRACT

Second only to early life, adolescence is a period of dramatic change and growth. For the developing young adult, this occurs against a backdrop of distinct environmental challenges and stressors. A significant body of work has identified an important role for the microbiota-gut-brain (MGB) axis in the development and function of the brain. Given that the MGB axis is both highly plastic during the teenage years and vulnerable to environmental stressors, more attention needs to be drawn to its potential role in the emergence of psychiatric illnesses, many of which first manifest during adolescence. Here, we review the current literature surrounding the developing microbiome, enteric nervous system, vagus nerve, and brain during the adolescent period. We also examine preclinical and clinical research involving the MGB axis during this dynamic developmental window and argue that more research is needed to further understand the role of the MGB in the pathogenesis of brain disorders. Greater understanding of the adolescent MGB axis will open up the exciting potential for new microbial-based therapeutics for the treatment of these often-refractory psychiatric illnesses.


Subject(s)
Gastrointestinal Microbiome , Mental Disorders , Adolescent , Humans , Brain-Gut Axis , Gastrointestinal Microbiome/physiology , Brain
3.
Mol Nutr Food Res ; 66(22): e2200177, 2022 11.
Article in English | MEDLINE | ID: mdl-36068654

ABSTRACT

SCOPE: Milk fat globule membrane (MFGM) is an essential component of milk. Bovine MFGM (bMFGM) has been shown to support cognitive development and increase relative concentrations of serum phospholipids. This study investigates bioavailability of bMFGM components after oral administration in two preclinical models to explore whether dietary bMFGM induces parallel changes to plasma and brain lipidomes. METHODS AND RESULTS: Transgenic APOE*3.Leiden mice (n = 18 per group) and Sprague-Dawley rats (n = 12 per group) are fed bMFGM-enriched (MFGM+) or Control diet, followed by phospholipid profile-determination in plasma, hippocampus, and prefrontal cortex tissue by targeted mass spectrometry. Multivariate analysis of lipidomic profiles demonstrates a separation between MFGM+ and Control plasma across rodents. In plasma, sphingomyelins contributed the most to the separation of lipid patterns among both models, where three sphingomyelins (d18:1/14:0, d18:1/23:0, d18:1/23:1[9Z]) are consistently higher in the circulation of MFGM+ groups. A similar trend is observed in rat prefrontal cortex, although no significant separation of the brain lipidome is demonstrated. CONCLUSION: bMFGM-enriched diet alters plasma phospholipid composition in rodents, predominantly increasing sphingomyelin levels in the systemic circulation with similar, but non-significant, trends in central brain regions. These changes may contribute to the beneficial effects of bMFGM on neurodevelopment during early life.


Subject(s)
Dietary Supplements , Glycolipids , Glycoproteins , Lipid Droplets , Lipidomics , Animals , Mice , Rats , Brain , Lipid Droplets/chemistry , Phospholipids/pharmacology , Rats, Sprague-Dawley , Sphingomyelins/pharmacology , Glycoproteins/administration & dosage , Glycolipids/administration & dosage
4.
Sci Rep ; 11(1): 21130, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34702901

ABSTRACT

The vagus nerve relays mood-altering signals originating in the gut lumen to the brain. In mice, an intact vagus is required to mediate the behavioural effects of both intraluminally applied selective serotonin reuptake inhibitors and a strain of Lactobacillus with antidepressant-like activity. Similarly, the prodepressant effect of lipopolysaccharide is vagus nerve dependent. Single vagal fibres are broadly tuned to respond by excitation to both anti- and prodepressant agents, but it remains unclear how neural responses encode behaviour-specific information. Here we demonstrate using ex vivo experiments that for single vagal fibres within the mesenteric neurovascular bundle supplying the mouse small intestine, a unique neural firing pattern code is common to both chemical and bacterial vagus-dependent antidepressant luminal stimuli. This code is qualitatively and statistically discernible from that evoked by lipopolysaccharide, a non-vagus-dependent antidepressant or control non-antidepressant Lactobacillus strain and are not affected by sex status. We found that all vagus dependent antidepressants evoked a decrease in mean spike interval, increase in spike burst duration, decrease in gap duration between bursts and increase in intra-burst spike intervals. Our results offer a novel neuronal electrical perspective as one explanation for mechanisms of action of gut-derived vagal dependent antidepressants. We expect that our ex vivo individual vagal fibre recording model will improve the design and operation of new, extant electroceutical vagal stimulation devices currently used to treat major depression. Furthermore, use of this vagal antidepressant code should provide a valuable screening tool for novel potential oral antidepressant candidates in preclinical animal models.


Subject(s)
Action Potentials/drug effects , Antidepressive Agents , Lactobacillus/chemistry , Selective Serotonin Reuptake Inhibitors , Vagus Nerve/physiopathology , Animals , Antidepressive Agents/chemistry , Antidepressive Agents/pharmacology , Female , Male , Mice , Mice, Inbred BALB C , Selective Serotonin Reuptake Inhibitors/chemistry , Selective Serotonin Reuptake Inhibitors/pharmacology
5.
Sci Rep ; 11(1): 13756, 2021 07 02.
Article in English | MEDLINE | ID: mdl-34215822

ABSTRACT

Intestinal bacteria have diverse and complex influence on their host. Evidence is accumulating that this may be mediated in part by bacterial extracellular membrane vesicles (MV), nanometer-sized particles important for intercellular communication. Little is known about the composition of MV from gram-positive beneficial bacteria nor how they interact with intestinal epithelial cells (IEC). Here we demonstrate that MV from Lacticaseibacillus rhamnosus JB-1 are endocytosed in a likely clathrin-dependent manner by both mouse and human IEC in vitro and by mouse IEC in vivo. We further show that JB-1 MV contain lipoteichoic acid (LTA) that activates Toll-like receptor 2 (TLR2) and induces immunoregulatory interleukin-10 expression by dendritic cells in an internalization-dependent manner. By contrast, neither LTA nor TLR2 appear to be required for JB-1 MV endocytosis by IEC. These results demonstrate a novel mechanism by which bacterial MV can influence host physiology and suggest one potential route for beneficial influence of certain bacteria and probiotics.


Subject(s)
Extracellular Vesicles/chemistry , Interleukin-10/genetics , Lacticaseibacillus rhamnosus/genetics , Toll-Like Receptor 2/genetics , Animals , Cell Membrane/drug effects , Endocytosis/drug effects , Gene Expression Regulation/drug effects , Gram-Positive Bacteria/chemistry , Humans , Intestinal Mucosa/drug effects , Lacticaseibacillus rhamnosus/chemistry , Mice
6.
Neuropharmacology ; 195: 108682, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34175326

ABSTRACT

The vagus nerve is one of the major signalling components between the gut microbiota and brain. However, the exact relationship between gut-brain signaling along the vagus and the effects of gut microbes on brain function and behaviour is unclear. In particular, the relationship between the vagus nerve and immune signaling, that also appears to play a critical role in microbiota-gut-brain communication, has not been delineated. The aim of the present study was to determine the effect of subdiaphragmatic vagotomy on peripheral and central immune changes associated with the anxiolytic actions of L.rhamnosus. Male mice underwent vagotomy or sham surgery, followed by administration of L.rhamnosus for 14 days. L.rhamnosus administration following sham surgery resulted in reduced anxiety-like behaviour, and an attenuation of the hypothalamic-pituitary-adrenal axis (HPA axis), as indicated by reduced plasma corticosterone after acute restraint stress. These effects were associated with an increase in splenic T regulatory cells and a decrease in activated microglia in the hippocampus. The anxiolytic effects, HPA modulation and increase in T regulatory cells were prevented by vagotomy, whereas vagotomy alone led to a significant increase in activated microglia in the hippocampus that was not altered with L.rhamnosus treatment. Thus, both microbe induced and constitutive vagal signaling influences critical immune components of the microbiota-gut-brain axis. These findings suggest that, rather than acting as a direct neural link to the central nervous system, the role of the vagus nerve in gut-microbe to brain signalling is as an integral component of a bi-directional neuroimmunoendocrine pathway.


Subject(s)
Behavior, Animal/drug effects , Brain-Gut Axis/drug effects , Corticosterone/blood , Hypothalamo-Hypophyseal System/diagnostic imaging , Lacticaseibacillus rhamnosus , Pituitary-Adrenal System/drug effects , Animals , Brain-Gut Axis/immunology , Hypothalamo-Hypophyseal System/immunology , Male , Mice , Pituitary-Adrenal System/immunology , Vagotomy
7.
Neurobiol Pain ; 10: 100064, 2021.
Article in English | MEDLINE | ID: mdl-34151049

ABSTRACT

Visceral pain refers to pain arising from the internal organs and is distinctly different from the expression and mechanisms of somatic pain. Diseases and disorders with increased visceral pain are associated with significantly reduced quality of life and incur large financial costs due to medical visits and lost work productivity. In spite of the notable burden of illness associated with those disorders involving increased visceral pain, and some knowledge regarding etiology, few successful therapeutics have emerged, and thus increased attention to animal models of visceral hypersensitivity is warranted in order to elucidate new treatment opportunities. Altered microbiota-gut-brain (MGB) axis communication is central to the comorbid gastrointestinal/psychiatric diseases of which increased visceral (intestinal) sensitivity is a hallmark. This has led to a particular focus on intestinal microbiome disruption and its potential role in the etiology of heightened visceral pain. Here we provide a review of studies examining models of heightened visceral pain due to altered bidirectional communication of the MGB axis, many of which are conducted on a background of stress exposure. We discuss work in which the intestinal microbiota has either been directly manipulated (as with germ-free, antibiotic, and fecal microbial transplantation studies) or indirectly affected through early life or adult stress, inflammation, and infection. Animal models of visceral pain alterations with accompanying changes to the intestinal microbiome have the highest face and construct validity to the human condition and are the focus of the current review.

8.
Mod Trends Psychiatry ; 32: 58-67, 2021.
Article in English | MEDLINE | ID: mdl-34032645

ABSTRACT

There is currently enormous interest in the impact of the intestinal microbiota on the development and function of the brain via activity of the microbiota-gut-brain axis. It has long been recognised that symbiotic microorganisms influence host behaviour, but in recent years evidence has accumulated that this can, in fact, be beneficial to the host. Indeed, substantial research has now demonstrated an influence of the intestinal microbiota on a wide range of mammalian behaviours. Here, we review what is currently known about the influence of intestinal microbiota on learning and memory, olfaction, social behaviours, and circadian processes. While work in animal models is compelling, further work is required to elucidate mechanisms whereby bacterial influence is occurring, as well as to determine the extent to which gut microbiota can influence similar phenotypes in humans.


Subject(s)
Behavior/physiology , Behavioral Symptoms/microbiology , Brain , Gastrointestinal Microbiome/physiology , Animals , Behavior, Animal/physiology , Brain/microbiology , Brain/physiology , Humans , Mental Processes/physiology , Psychophysiology
9.
Neuronal Signal ; 4(4): NS20200007, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33343931

ABSTRACT

Visceral hypersensitivity is a hallmark of many functional and stress-related gastrointestinal disorders, and there is growing evidence that the gut microbiota may play a role in its pathophysiology. It has previously been shown that early life stress-induced visceral sensitivity is reduced by various probiotic strains of bacteria (including Lactobacillus rhamnosus GG (LGG)) alone or in combination with prebiotic fibres in rat models. However, the exact mechanisms underpinning such effects remain unresolved. Here, we investigated if soluble mediators derived from LGG can mimic the bacteria's effects on visceral hypersensitivity and the microbiota-gut-brain axis. Rats were exposed to maternal separation (MS) from postnatal days 2-12. From weaning onwards both non-separated (NS) and MS offspring were provided drinking water with or without supplementation of standardized preparations of the LGG soluble mediators (LSM). Our results show that MS led to increased visceral sensitivity and exaggerated corticosterone plasma levels following restraint stress in adulthood, and both of these effects were ameliorated through LSM supplementation. Differential regulation of various genes in the spinal cord of MS versus NS rats was observed, 41 of which were reversed by LSM supplementation. At the microbiota composition level MS led to changes in beta diversity and abundance of specific bacteria including parabacteroides, which were ameliorated by LSM. These findings support probiotic soluble mediators as potential interventions in the reduction of symptoms of visceral hypersensitivity.

10.
Sci Rep ; 10(1): 18501, 2020 Oct 23.
Article in English | MEDLINE | ID: mdl-33097815

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

11.
Sci Rep ; 10(1): 18355, 2020 Oct 22.
Article in English | MEDLINE | ID: mdl-33093646

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

12.
Sci Rep ; 10(1): 13485, 2020 08 10.
Article in English | MEDLINE | ID: mdl-32778662

ABSTRACT

Chronic social defeat (CSD) in mice has been suggested as a model for studying post-traumatic stress disorder (PTSD). Our previous work indicated that exposure to Lactobacillus rhamnosus JB-1 (JB-1) during CSD can attenuate subsequent behavioural and immune disruption, suggesting a potential for microbe based therapeutic approaches in PTSD. In the current study, we assessed the ability of JB-1 to mitigate the behavioral consequences of CSD when treatment is instigated in the early post-stress period and compared the probiotic effects with those of the selective serotonin reuptake inhibitor (SSRI), sertraline. JB-1 or sertraline were administered orally 48 h following 10-days of CSD in male C57BL/6 mice. Contrary to our hypothesis of a beneficial effect, 30 days of treatment with either JB-1 or sertraline increased the persistence of both aggressor avoidance and reduced sociability in defeated mice. This was accompanied by lower hippocampal mRNA expression for genes related to fear memory. Defeated mice treated with either JB-1 or sertraline also exhibited systemic immune changes, with a decrease in Th1 cells, activated monocytes, and the monocyte chemoattractant CCL2. This study identifies potentially detrimental effects of both JB-1 and sertraline if administered in the early post-trauma period and suggests caution should be applied when considering psychobiotic or SSRI based approaches for early intervention in trauma related psychiatric disorders.


Subject(s)
Avoidance Learning/drug effects , Oligopeptides/pharmacology , Animals , Attention/drug effects , Behavior, Animal/drug effects , Brain/metabolism , Fear/drug effects , Hippocampus/metabolism , Lacticaseibacillus rhamnosus/metabolism , Male , Memory/drug effects , Mice , Mice, Inbred C57BL , Selective Serotonin Reuptake Inhibitors/pharmacology , Sertraline/pharmacology , Social Defeat , Stress Disorders, Post-Traumatic/psychology , Stress, Psychological/complications
13.
Sci Rep ; 10(1): 10318, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32587382

ABSTRACT

There is increasing awareness of the need to consider potential long-term effects of antibiotics on the health of children. In addition to being associated with immune and metabolic diseases, there is evidence that early-life antibiotic exposure can affect neurodevelopment. Here we investigated the effect of low dose of penicillin V on mice when administered for 1 week immediately prior to weaning. We demonstrated that exposure to the antibiotic during the pre-weaning period led to long-term changes in social behaviour, but not anxiety-like traits, in male mice only. The change in behaviour of males was associated with decreased hippocampal expression of AVPR1A and AVPR1B while expression of both receptors was increased in females. Spleens of male mice also showed an increase in the proportion of activated dendritic cells and a corresponding decrease in regulatory T cells with penicillin exposure. All changes in brain, behaviour and immune cell populations, associated with penicillin exposure, were absent in mice that received L. rhamnosus JB-1 supplementation concurrent with the antibiotic. Our study indicates that post-natal exposure to a clinically relevant dose of antibiotic has long-term, sex dependent effects on the CNS and may have implications for the development of neuropsychiatric disorders. Importantly, we also provide further evidence that probiotic based strategies may be of use in counteracting detrimental effects of early-life antibiotics on neurodevelopment.


Subject(s)
Anti-Bacterial Agents/adverse effects , Brain/drug effects , Gastrointestinal Microbiome/drug effects , Lacticaseibacillus rhamnosus , Probiotics/administration & dosage , Administration, Oral , Animals , Anti-Bacterial Agents/administration & dosage , Anxiety/immunology , Anxiety/microbiology , Anxiety/physiopathology , Anxiety/prevention & control , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/growth & development , Brain/pathology , Brain/physiopathology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Feces/microbiology , Female , Gastrointestinal Microbiome/physiology , Humans , Male , Mice , Penicillins/administration & dosage , Penicillins/adverse effects , Sex Factors , Social Behavior , Spleen/cytology , Spleen/drug effects , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Weaning
14.
Neuropharmacology ; 170: 108067, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32224131

ABSTRACT

There is accumulating evidence that certain gut microbes modulate brain chemistry and have antidepressant-like behavioural effects. However, it is unclear which brain regions respond to bacteria-derived signals or how signals are transmitted to distinct regions. We investigated the role of the vagus in mediating neuronal activation following oral treatment with Lactobacillus rhamnosus (JB-1). Male Balb/c mice were orally administered a single dose of saline or a live or heat-killed preparation of a physiologically active bacterial strain, Lactobacillus rhamnosus (JB-1). 165 min later, c-Fos immunoreactivity in the brain was mapped, and mesenteric vagal afferent fibre firing was recorded. Mice also underwent sub-diaphragmatic vagotomy to investigate whether severing the vagus prevented JB-1-induced c-Fos expression. Finally, we examined the ΔFosB response following acute versus chronic bacterial treatment. While a single exposure to live and heat-killed bacteria altered vagal activity, only live treatment induced rapid neural activation in widespread but distinct brain regions, as assessed by c-Fos expression. Sub-diaphragmatic vagotomy abolished c-Fos immunoreactivity in most, but not all, previously responsive regions. Chronic, but not acute treatment induced a distinct pattern of ΔFosB expression, including in previously unresponsive brain regions. These data identify that specific brain regions respond rapidly to gut microbes via vagal-dependent and independent pathways, and demonstrate that acute versus long-term exposure is associated with differential responses in distinct brain regions.


Subject(s)
Brain/metabolism , Brain/microbiology , Lacticaseibacillus rhamnosus/metabolism , Neurons/metabolism , Neurons/microbiology , Vagus Nerve/metabolism , Vagus Nerve/microbiology , Administration, Oral , Animals , Male , Mice , Mice, Inbred BALB C , Vagotomy/trends , Vagus Nerve/surgery
15.
Brain Behav Immun ; 88: 451-460, 2020 08.
Article in English | MEDLINE | ID: mdl-32276029

ABSTRACT

Over the past decade there has been increasing interest in the involvement of the microbiota-gut-brain axis in mental health. However, there are major gaps in our knowledge regarding the complex signaling systems through which gut microbes modulate the CNS. The immune system is a recognized mediator in the bidirectional communication continuously occurring between gut and brain. We previously demonstrated that Lactobacillus rhamnosus JB-1 (JB-1), a bacterial strain that has anxiolytic- and antidepressant-like effects in mice, modulates the immune system through induction of immunosuppressive T regulatory cells. Here we examined a potential causal relationship between JB-1 induced regulatory T cells and the observed effects on behaviour. We found that depletion of regulatory T cells, via treatment with monoclonal antibody against CD25, inhibited the antidepressant- and anxiolytic-like effects induced by 4-week oral administration of JB-1 in mice. Ly6Chi monocytes were found to be decreased in JB-1 fed mice with intact regulatory T cells, but not in JB-1 fed mice following depletion. Furthermore, adoptive transfer of CD4+CD25+ cells, from JB-1 treated donor mice, but not from controls, induced antidepressant- and anxiolytic-like effects in recipient mice. Ly6Chi monocytes were also significantly decreased in mice receiving CD4+CD25+ cells from JB1 fed donors. This study identifies cells within the CD4+CD25+ population, most likely regulatory T cells, as both necessary and sufficient in JB-1-induced antidepressant- and anxiolytic-like effects in mice, providing novel mechanistic insight into microbiota-gut-brain communication in addition to highlighting the potential for immunotherapy in psychiatric disorders.


Subject(s)
Lacticaseibacillus rhamnosus , Adoptive Transfer , Animals , Male , Mice , Mice, Inbred BALB C , Monocytes , T-Lymphocytes, Regulatory
16.
J Neuroimmunol ; 343: 577213, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32278229

ABSTRACT

Deficiencies in the adaptive immune system have been linked to anxiety-like behaviours and stress reactivity. Mice lacking T lymphocytes through knockout of the T cell receptor (TCR) ß and δ chains were compared to wild type C57Bl/6 mice. Central stress circuitry gene expression was assessed following repeated restraint stress. TCRß-/-δ-/- mice showed an increased baseline plasma corticosterone and exaggerated changes in stress-related gene expression after repeated restraint stress. Sexual dimorphic stress responses were observed in wild-type C57Bl/6 mice but not in TCRß-/-δ-/- mice. These data suggest that T cell-brain interactions influence sex-differences in CNS stress circuitry and stress reactivity.


Subject(s)
Adaptive Immunity/immunology , Neuroimmunomodulation/physiology , Psychological Distress , Sex Characteristics , T-Lymphocytes , Animals , Brain/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell/deficiency , Restraint, Physical , Transcriptome/immunology
17.
Eur J Neurosci ; 51(4): 1042-1058, 2020 02.
Article in English | MEDLINE | ID: mdl-31339598

ABSTRACT

Nutritional interventions targeting the microbiota-gut-brain axis are proposed to modulate stress-induced dysfunction of physiological processes and brain development. Maternal separation (MS) in rats induces long-term alterations to behaviour, pain responses, gut microbiome and brain neurochemistry. In this study, the effects of dietary interventions (milk fat globule membrane [MFGM] and a polydextrose/galacto-oligosaccharide prebiotic blend) were evaluated. Diets were provided from postnatal day 21 to both non-separated and MS offspring. Spatial memory, visceral sensitivity and stress reactivity were assessed in adulthood. Gene transcripts associated with cognition and stress and the caecal microbiota composition were analysed. MS-induced visceral hypersensitivity was ameliorated by MFGM and to greater extent with the combination of MFGM and prebiotic blend. Furthermore, spatial learning and memory were improved by prebiotics and MFGM alone and with the combination. The prebiotic blend and the combination of the prebiotics and MFGM appeared to facilitate return to baseline with regard to HPA axis response to the restraint stress, which can be beneficial in times where coping mechanisms to stressful events are required. Interestingly, the combination of MFGM and prebiotic reduced the long-term impact of MS on a marker of myelination in the prefrontal cortex. MS affected the microbiota at family level only, while MFGM, the prebiotic blend and the combination influenced abundance at family and genus level as well as influencing beta-diversity levels. In conclusion, intervention with MFGM and prebiotic blend significantly impacted the composition of the microbiota as well as ameliorating some of the long-term effects of early-life stress.


Subject(s)
Gastrointestinal Microbiome , Maternal Deprivation , Microbiota , Animals , Brain , Glycolipids , Glycoproteins , Hypothalamo-Hypophyseal System , Lipid Droplets , Pituitary-Adrenal System , Prebiotics , Rats , Stress, Physiological
18.
Brain Behav Immun ; 84: 154-163, 2020 02.
Article in English | MEDLINE | ID: mdl-31785396

ABSTRACT

Growing evidence suggests that environmental disruptors of maternal microbes may have significant detrimental consequences for the developing fetus. Antibiotic exposure during early life can have long-term effects on neurodevelopment in mice and humans. Here we explore whether exposure to low-dose penicillin during only the last week of gestation in mice has long-term effects on offspring behaviour, brain, immune function, and gut microbiota. We found that this treatment had sex-specific effects in the adult mouse offspring. Female, but not male, mice demonstrated decreased anxiety-like behaviours, while male, but not female, mice had abnormal social behaviours which correlated with altered brain expression of AVPR1A, AVPR1B, and OXTR, and decreases in the balance of splenic FOXP3+ regulatory T cells. Prenatal penicillin exposure also led to distinct microbiota compositions that clustered differently by sex. These data suggest that exposure of pregnant mice to even a low dose of penicillin through only the last week before birth is nonetheless sufficient to induce long-term sex-specific developmental changes in both male and female offspring.


Subject(s)
Behavior, Animal/drug effects , Gastrointestinal Microbiome/drug effects , Immunity/drug effects , Penicillins/administration & dosage , Penicillins/pharmacology , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/physiopathology , Sex Characteristics , Social Behavior , Animals , Brain/drug effects , Brain/metabolism , Female , Male , Mice , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/microbiology
19.
Sci Rep ; 9(1): 14290, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31582799

ABSTRACT

The vagus nerve can transmit signals to the brain resulting in a reduction in depressive behavior as evidenced by the long-term beneficial effects of electrical stimulation of the vagus in patients with intractable depression. The vagus is the major neural connection between gut and brain, and we have previously shown that ingestion of beneficial bacteria modulates behaviour and brain neurochemistry via this pathway. Given the high levels of serotonin in the gut, we considered if gut-brain signaling, and specifically the vagal pathway, might contribute to the therapeutic effect of oral selective serotonin reuptake inhibitors (SSRI). Mesenteric nerve recordings were conducted in mice after treatment with SSRI to ascertain if this class of drugs resulted in increased vagal excitability. Patch clamp recordings of enteric neurons were carried out to measure activity of primary afferent neurons in the gut in response to SSRI and to assess the importance of gut epithelium in transducing signal. The tail suspension test (TST) was used following 14d feeding of SSRI in vagotomised and surgical sham mice to measure depressive-like behaviour. Brain mRNA expression was examined via PCR and the intestinal microbiome was assessed. Mesenteric nerve recordings in BALB/c mice demonstrated that oral treatment with SSRI leads to a significant increase in vagal activity. This effect was not observed in mice treated with a representative noradrenaline-dopamine reuptake inhibitor. It is known that signals from the gut can be transmitted to the vagus via the enteric nervous system. Exposure of the gut to SSRI increased the excitability of intrinsic primary afferent neurons in the myenteric plexus, through an intestinal epithelium dependent mechanism, and alpha-diversity of gut microbiota was altered. Critically, blocking vagal signaling from gut to brain, via subdiaphragmatic vagotomy, abolished the antidepressive effects of oral SSRI treatment as determined by the tail suspension test. This work suggests that vagus nerve dependent gut-brain signaling contributes to the effects of oral SSRI and further, highlights the potential for pharmacological approaches to treatment of mood disorders that focus on vagal stimulation and may not even require therapeutic agents to enter the circulation.


Subject(s)
Brain/drug effects , Enteric Nervous System/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Vagus Nerve/drug effects , Administration, Oral , Animals , Brain/physiology , Digestive System/drug effects , Digestive System/innervation , Enteric Nervous System/physiology , Male , Mice , Mice, Inbred BALB C , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Serotonin/metabolism , Selective Serotonin Reuptake Inhibitors/administration & dosage , Vagus Nerve/physiology
20.
Nutr Neurosci ; 22(6): 425-434, 2019 Jun.
Article in English | MEDLINE | ID: mdl-29173065

ABSTRACT

Early life is a period of significant brain development when the brain is at its most plastic and vulnerable. Stressful episodes during this window of development have long-lasting effects on the central nervous system. Rodent maternal separation (MS) is a reliable model of early-life stress and induces alterations in both physiology and behaviour. Intriguingly, the gut microbiota of MS offspring differ from that of non-separated offspring, suggesting a mechanistic role for the microbiota-gut-brain axis. Hence, we tested whether dietary factors known to affect the gut microbiota alter the neurobehavioural effects of MS. The impact of consuming diet containing prebiotics polydextrose (PDX) and galactooligosaccharide (GOS) alone or in combination with live bacteria Lactobacillus rhamnosus GG (LGG) from weaning onwards in rats subjected to early-life MS was assessed. Adult offspring were assessed for anxiety-like behaviour in the open field test, spatial memory using the Morris water maze, and reactivity to restraint stress. Brains were examined via PCR for changes in mRNA gene expression. Here, we demonstrate that diets containing a combination of PDX/GOS and LGG attenuates the effects of early-life MS on anxiety-like behaviour and hippocampal-dependent learning with changes to hippocampal mRNA expression of genes related to stress circuitry, anxiety and learning.


Subject(s)
Behavior, Animal , Glucans/administration & dosage , Lacticaseibacillus rhamnosus , Maternal Deprivation , Oligosaccharides/administration & dosage , Prebiotics/administration & dosage , Stress, Psychological/microbiology , Animals , Anxiety/microbiology , Exploratory Behavior , Female , Hippocampus/metabolism , Hippocampus/microbiology , Male , Probiotics/administration & dosage , Rats, Sprague-Dawley , Spatial Memory
SELECTION OF CITATIONS
SEARCH DETAIL
...