Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
Gut Microbes ; 15(2): 2282796, 2023 Dec.
Article En | MEDLINE | ID: mdl-38010168

Human aging is characterized by gut microbiome alteration and differential loss of gut commensal species associated with the onset of frailty. The administration of cultured commensal strains to replenish lost taxa could potentially promote healthy aging. To investigate the interaction of whole microbiomes and administered strains, we transplanted gut microbiota from a frail or healthy elderly subject into germ-free mice. We supplemented the frail-donor recipient group with a defined consortium of taxa (the "S7") that we identified by analyzing healthy aging subjects in our previous studies and whose abundance correlated with health-promoting dietary intervention. Inoculation with a frail or a healthy donor microbiome resulted in differential microbiota compositions in murine recipients 5 weeks post-transplantation. Fecal acetate levels were significantly higher in healthy donor recipient mice than in frail donor recipient mice after 4 weeks. However, the frailty-related phenotype was not replicated in recipient mice with single-dose microbiota transplantation from a healthy and a frail donor. Five S7 species colonized successfully in germ-free mice, with a relatively high abundance of Barnesiella intestinihominis and Eubacterium rectale. The engraftment of five S7 species in germ-free mice increased fecal acetate levels and reduced colon permeability and plasma TNF-ɑ concentration. Supplementation with the S7 in frail-microbiota recipient mice did not increase alpha-diversity but significantly increased the abundance of Barnesiella intestinihominis. S7 supplementation showed the potential for improving spatial reference memory in frail-microbiota recipient mice. Collectively, these data highlight the challenge of elderly microbiota engraftment in the germ-free mouse model but show promise for modulating the gut microbiome of frail elderly subjects by administering an artificial gut microbe consortium associated with healthy aging.


Frailty , Gastrointestinal Microbiome , Humans , Animals , Mice , Aged , Gastrointestinal Microbiome/genetics , Bacteroidetes , Feces/microbiology , Fecal Microbiota Transplantation , Acetates
2.
Microbiome ; 11(1): 168, 2023 08 02.
Article En | MEDLINE | ID: mdl-37528457

BACKGROUND: Typically, animal models studying gastrointestinal microbiotas compromised in early life have employed either germ-free animals or mice treated with a cocktail of antibiotics. Such studies intend to mimic scenarios of infants born by caesarean section and/or subjected to antibiotic treatment. However, the antibiotics used in these studies are rarely prescribed to infants. Therefore, an early life model was developed in which the murine gastrointestinal microbiota was severely disrupted by clindamycin treatment. RESULTS: In this mouse model, we investigated the extent supplementation with a synbiotic mixture of prebiotics, being scGOS/lcFOS with the human milk oligosaccharide 2'-Fucosyllactose (2'-FL), in combination with or without single strain or mix of "infant type" bifidobacteria, can rescue an antibiotic-compromised microbiota. Shotgun metagenomic sequencing showed that the microbiota was severely disrupted by the clindamycin challenge. No recovery was observed 3 weeks post-challenge in the scGOS/lcFOS/2'FL group, while the group that received the synbiotic treatment of scGOS/lcFOS/2'-FL with Bifidobacterium breve NRBB01 showed partial recovery. Strikingly in the scGOS/lcFOS/2'-FL group receiving the mixture of bifidobacteria resulted in a recovery of the microbiota disruption. Histological analyses showed that the clindamycin-treated animals at the end of the experiment still suffered from mild oedema and villi/colonic crypt irregularities which was ameliorated by the synbiotic intervention. CONCLUSION: Our study demonstrates that supplementation of synbiotic mixture of scGOS/lcFOS/2'-FL in combination with a specific mix of infant-type bifidobacterial strains is able to partially revive an antibiotic-perturbed gastrointestinal microbiota. Video Abstract.


Gastrointestinal Microbiome , Synbiotics , Humans , Infant , Animals , Pregnancy , Mice , Female , Bifidobacterium , Anti-Bacterial Agents/pharmacology , Cesarean Section , Clindamycin , Oligosaccharides
3.
Gut Microbes ; 15(1): 2229948, 2023.
Article En | MEDLINE | ID: mdl-37424323

A high-fat (HF) diet reduces resistance to the foodborne pathogen Listeria monocytogenes. We demonstrate that short-term gavage with A. muciniphila increases resistance to oral and systemic L. monocytogenes infection in mice fed a HF diet. A. muciniphila reduced inflammation in the gut and liver of mice fed a high-fat diet prior to infection and reduced inflammatory cell infiltration in the ileum to levels similar to mice fed a low-fat (LF) diet. Akkermansia administration had minimal impacts upon the microbiota and microbial metabolites and did not affect individual taxa or impact the Bacteroidetes to Firmicutes ratio. In summary, A. muciniphila increased resistance to L. monocytogenes infection in mice fed a HF diet by moderating immune/physiological effects through specific interaction between A. muciniphila and the host gut.


Gastrointestinal Microbiome , Listeria monocytogenes , Listeriosis , Animals , Mice , Diet, High-Fat/adverse effects , Verrucomicrobia/physiology , Mice, Inbred C57BL
4.
Gut Microbes ; 15(1): 2194794, 2023.
Article En | MEDLINE | ID: mdl-36994608

Ruminococcus gnavus is a prevalent gut microbe reported to occur in higher abundance among individuals with inflammatory bowel disease (IBD). This study reports the isolation and characterization of six bacteriophages (phages) isolated from human fecal material and environmental samples that infect this species. Isolated phages have a siphovirus morphology, with genomes ranging between 36.5 and 37.8 kbp. Genome analysis indicates that the phages have a temperate lifestyle, which was confirmed by their ability to form lysogens on their host bacterial species. In contrast to the finding that phages lyse their host in liquid medium, results from a mouse trial indicate these phages can co-exist with the host bacterium in the gut without causing a significant reduction of R. gnavus. The bacterial counts in the feces of phage-treated mice did not significantly differ in the presence of phage. Furthermore, analysis of publicly available gut virome sequence data indicates a high abundance of these phages among individuals suffering from IBD. This work provides the first insight into how phages interact with R. gnavus in the human gut microbiome.


Bacteriophages , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Humans , Mice , Animals , Bacteriophages/genetics , Mucins , Ruminococcus/genetics , Gastrointestinal Microbiome/genetics , Bacteria
5.
Gut Microbes ; 14(1): 2149023, 2022.
Article En | MEDLINE | ID: mdl-36420990

The mechanisms by which early microbial colonizers of the neonate influence gut development are poorly understood. Bacterial bile salt hydrolase (BSH) acts as a putative colonization factor that influences bile acid signatures and microbe-host signaling pathways and we considered whether this activity can influence infant gut development. In silico analysis of the human neonatal gut metagenome confirmed that BSH enzyme sequences are present as early as one day postpartum. Gastrointestinal delivery of cloned BSH to immature gnotobiotic mice accelerated shortening of the colon and regularized gene expression profiles, with monocolonised mice more closely resembling conventionally raised animals. In situ expression of BSH decreased markers of cell proliferation (Ki67, Hes2 and Ascl2) and strongly increased expression of ALPI, a marker of cell differentiation and barrier function. These data suggest an evolutionary paradigm whereby microbial BSH activity potentially influences bacterial colonization and in-turn benefits host gastrointestinal maturation.


Gastrointestinal Microbiome , Transcriptome , Female , Humans , Mice , Animals , Amidohydrolases/genetics , Amidohydrolases/metabolism , Gastrointestinal Tract/microbiology , Bacteria/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism
6.
Nutr Neurosci ; 25(4): 657-669, 2022 Apr.
Article En | MEDLINE | ID: mdl-32723167

Introduction: Adolescence is an important stage of maturation for various brain structures. It is during this time therefore that the brain may be more vulnerable to environmental factors such as diet that may influence mood and memory. Diets high in fat and sugar (termed a cafeteria diet) during adolescence have been shown to negatively impact upon cognitive performance, which may be reversed by switching to a standard diet during adulthood. Consumption of a cafeteria diet increases both peripheral and central levels of interleukin-1ß (IL-1ß), a pro-inflammatory cytokine which is also implicated in cognitive impairment during the ageing process. It is unknown whether adolescent exposure to a cafeteria diet potentiates the negative effects of IL-1ß on cognitive function during adulthood.Methods: Male Sprague-Dawley rats consumed a cafeteria diet during adolescence after which time they received a lentivirus injection in the hippocampus to induce chronic low-grade overexpression of IL-1ß. After viral integration, metabolic parameters, circulating and central pro-inflammatory cytokine levels, and cognitive behaviours were assessed.Results: Our data demonstrate that rats fed the cafeteria diet exhibit metabolic dysregulations in adulthood, which were concomitant with low-grade peripheral and central inflammation. Overexpression of hippocampal IL-1ß in adulthood impaired spatial working memory. However, adolescent exposure to a cafeteria diet, combined with or without hippocampal IL-1ß in adulthood did not induce any lasting cognitive deficits when the diet was replaced with a standard diet in adulthood. Discussion: These data demonstrate that cafeteria diet consumption during adolescence induces metabolic and inflammatory changes, but not behavioural changes in adulthood.


Diet , Spatial Memory , Animals , Brain/metabolism , Diet, High-Fat/adverse effects , Hippocampus/metabolism , Male , Rats , Rats, Sprague-Dawley
7.
Brain Behav Immun ; 99: 327-338, 2022 01.
Article En | MEDLINE | ID: mdl-34732365

Stress during critical periods of neurodevelopment is associated with an increased risk of developing stress-related psychiatric disorders, which are more common in women than men. Hippocampal neurogenesis (the birth of new neurons) is vulnerable to maternal separation (MS) and inflammatory stressors, and emerging evidence suggests that hippocampal neurogenesis is more sensitive to stress in the ventral hippocampus (vHi) than in the dorsal hippocampus (dHi). Although research into the effects of MS stress on hippocampal neurogenesis is well documented in male rodents, the effect in females remains underexplored. Similarly, reports on the impact of inflammatory stressors on hippocampal neurogenesis in females are limited, especially when female bias in the prevalence of stress-related psychiatric disorders begins to emerge. Thus, in this study we investigated the effects of MS followed by an inflammatory stressor (lipopolysaccharide, LPS) in early adolescence on peripheral and hippocampal inflammatory responses and hippocampal neurogenesis in juvenile female rats. We show that MS enhanced an LPS-induced increase in the pro-inflammatory cytokine IL-1ß in the vHi but not in the dHi. However, microglial activation was similar following LPS alone or MS alone in both hippocampal regions, while MS prior to LPS reduced microglial activation in both dHi and vHi. The production of new neurons was unaffected by MS and LPS. MS and LPS independently reduced the dendritic complexity of new neurons, and MS exacerbated LPS-induced reductions in the complexity of distal dendrites of new neurons in the vHi but not dHi. These data highlight that MS differentially primes the physiological response to LPS in the juvenile female rat hippocampus.


Maternal Deprivation , Neuroinflammatory Diseases , Animals , Female , Hippocampus , Lipopolysaccharides/pharmacology , Male , Microglia , Neurogenesis/physiology , Neurons , Rats
8.
Front Microbiol ; 11: 69, 2020.
Article En | MEDLINE | ID: mdl-32082288

Significant evidence supports a relationship between the gut microbiome, inflammation, host response, and health, including the finding that a number of disorders are associated with disruption of the microbiome. In these disorders, a number of dietary interventions (including prebiotics, live probiotics, or heat-killed microbes) have been proposed to be curative or preventative agents. The use of heat-killed microbes has a number of benefits over living organisms, including reduced infection risk in vulnerable individuals, extended shelf life and the potential for use in combination with antimicrobial agents. We previously reported that murine chow supplemented with 5% ADR-159, a heat-treated fermentate generated by two Lactobacillus strains, altered both behavior and the microbiome of male mice. Now we show that ADR-159 fed female mice also display a similar microbiome shift as determined by 16S rDNA analysis. In particular, we observed a reduction of levels of Turicibacter and Clostridium sensu stricto. These subtle changes in the bacterial component of the microbiome were mirrored by changes in the virome. Extended consumption of the ADR-159 diet had no negative effect on general health and lipocalin 2 levels (LCN2; a proxy for inflammation), but we observed increased IL-17f and decreased IL-12α expression in the colon and decreased short chain fatty acid levels in the ADR-159 fed animals. Four weeks into the diet, half of the animals were dosed with Citrobacter to determine the effect of ADR-159 on infection and on pathogen induced colitis. Overall, our results suggest that while the ADR-159 diet does not prevent Citrobacter infection, it had an effect on Citrobacter-induced inflammation. In contrast to animals fed standard chow, ADR-159 fed animals did not show a reduction of small intestine length and increase of colon crypt depth, which occurred in control mice. These microbiological, histological, and immunological results provide evidence to support the impact of heat-treated microorganisms and their metabolites on the murine microbiome and health.

9.
Brain Behav Immun ; 83: 172-179, 2020 01.
Article En | MEDLINE | ID: mdl-31604142

Both neuroinflammation and adult hippocampal neurogenesis (AHN) are implicated in many neurodegenerative disorders as well as in neuropsychiatric disorders, which often become symptomatic during adolescence. A better knowledge of the impact that chronic neuroinflammation has on the hippocampus during the adolescent period could lead to the discovery of new therapeutics for some of these disorders. The hippocampus is particularly vulnerable to altered concentrations of the pro-inflammatory cytokine interleukin-1ß (IL-1ß), with elevated levels implicated in the aetiology of neurodegenerative disorders such as Alzheimer's and Parkinson's, and stress-related disorders such as depression. The effect of acutely and chronically elevated concentrations of hippocampal IL-1ß have been shown to reduce AHN in adult rodents. However, the effect of exposure to chronic overexpression of hippocampal IL-1ß during adolescence, a time of increased vulnerability, hasn't been fully interrogated. Thus, in this study we utilized a lentiviral approach to induce chronic overexpression of IL-1ß in the dorsal hippocampus of adolescent male Sprague Dawley rats for 5 weeks, during which time its impact on cognition and hippocampal neurogenesis were examined. A reduction in hippocampal neurogenesis was observed along with a reduced level of neurite branching on hippocampal neurons. However, there was no effect of IL-1ß overexpression on performance in pattern separation, novel object recognition or spontaneous alternation in the Y maze. Our study has highlighted that chronic IL-1ß overexpression in the hippocampus during the adolescent period exerts a negative impact on neurogenesis independent of cognitive performance, and suggests a degree of resilience of the adolescent hippocampus to inflammatory insult.


Aging/metabolism , Cognition , Hippocampus/cytology , Hippocampus/metabolism , Interleukin-1beta/metabolism , Neurogenesis , Animals , Male , Rats , Rats, Sprague-Dawley
10.
Stress ; 23(4): 368-385, 2020 07.
Article En | MEDLINE | ID: mdl-31591928

In response to stressor exposure, expression of the inflammatory cytokine interleukin-1ß (IL-1) is increased within the paraventricular nucleus of the hypothalamus (PVN). Surgical removal of the adrenal glands (ADX) potentiated stress-induced IL-1 expression, suggesting a role for adrenal-derived hormones in constraining stress-evoked increases in IL-1. While corticosterone (CORT) is a primary factor inhibiting IL-1 expression, progesterone (PROG) is also released by the adrenal glands in male rats in response to stress and also has potent anti-inflammatory properties. This series of studies first established doses of CORT and PROG that adequately recapitulate the normal stress-induced rise, and then tested for individual and combined roles of CORT and PROG in mitigating stress-induced expression of inflammatory genes. We found that CORT injection alone attenuated ADX-induced increases in IL-1 expression and normalized the HPA axis response to stress. In general, PROG replacement had little effect on changes in HPA axis responsivity or stress-induced inflammatory measures. When CORT and PROG were co-administered, a small effect on expression of the decoy receptor, IL-1R2 was observed, suggestive of an anti-inflammatory response. Overall, these results suggest that although CORT is likely to be the primary stress-related hormone responsible for constraining cytokine expression evoked by stress, CORT and PROG may exert certain combined actions that temper stress-induced neuroinflammation.LAY SUMMARYExposure to stress promoted expression of inflammation-related genes in the PVN and BNST. This inflammation was mainly suppressed by the adrenal hormone corticosterone, whereas progesterone had a smaller role in mitigating post-stress inflammation.


Corticosterone , Hypothalamo-Hypophyseal System , Animals , Corticosterone/pharmacology , Male , Paraventricular Hypothalamic Nucleus , Pituitary-Adrenal System , Progesterone/pharmacology , Rats , Stress, Physiological , Stress, Psychological
11.
Microbiome ; 7(1): 7, 2019 01 18.
Article En | MEDLINE | ID: mdl-30658700

BACKGROUND: A westernized diet comprising a high caloric intake from animal fats is known to influence the development of pathological inflammatory conditions. However, there has been relatively little focus upon the implications of such diets for the progression of infectious disease. Here, we investigated the influence of a high-fat (HF) diet upon parameters that influence Listeria monocytogenes infection in mice. RESULTS: We determined that short-term administration of a HF diet increases the number of goblet cells, a known binding site for the pathogen, in the gut and also induces profound changes to the microbiota and promotes a pro-inflammatory gene expression profile in the host. Host physiological changes were concordant with significantly increased susceptibility to oral L. monocytogenes infection in mice fed a HF diet relative to low fat (LF)- or chow-fed animals. Prior to Listeria infection, short-term consumption of HF diet elevated levels of Firmicutes including Coprococcus, Butyricicoccus, Turicibacter and Clostridium XIVa species. During active infection with L. monocytogenes, microbiota changes were further exaggerated but host inflammatory responses were significantly downregulated relative to Listeria-infected LF- or chow-fed groups, suggestive of a profound tempering of the host response influenced by infection in the context of a HF diet. The effects of diet were seen beyond the gut, as a HF diet also increased the sensitivity of mice to systemic infection and altered gene expression profiles in the liver. CONCLUSIONS: We adopted a systems approach to identify the effects of HF diet upon L. monocytogenes infection through analysis of host responses and microbiota changes (both pre- and post-infection). Overall, the results indicate that short-term consumption of a westernized diet has the capacity to significantly alter host susceptibility to L. monocytogenes infection concomitant with changes to the host physiological landscape. The findings suggest that diet should be a consideration when developing models that reflect human infectious disease.


Diet, High-Fat/adverse effects , Listeria monocytogenes/pathogenicity , Listeriosis/etiology , Microbiota/drug effects , Obesity/genetics , Animals , Diet, Western/adverse effects , Disease Models, Animal , Feces/microbiology , Female , Firmicutes/drug effects , Firmicutes/genetics , Firmicutes/isolation & purification , Gene Expression Regulation/drug effects , Goblet Cells/cytology , Goblet Cells/drug effects , Listeriosis/genetics , Listeriosis/immunology , Metagenome/drug effects , Mice , Obesity/complications , Obesity/etiology , Sequence Analysis, DNA
12.
J Neuroimmunol ; 331: 87-96, 2019 06 15.
Article En | MEDLINE | ID: mdl-28844503

The orphan nuclear receptor TLX (Nr2e1) is a key regulator of hippocampal neurogenesis. Impaired adult hippocampal neurogenesis has been reported in neurodegenerative and psychiatric conditions including dementia and stress-related depression. Neuroinflammation is also implicated in the neuropathology of these disorders, and has been shown to negatively affect hippocampal neurogenesis. To investigate a role for TLX in hippocampal neuroinflammation, we assessed microglial activation in the hippocampus of mice with a spontaneous deletion of TLX. Results from our study suggest that a lack of TLX is implicated in deregulation of microglial phenotype and that consequently, the survival and function of newborn cells in the hippocampus is impaired. TLX may be an important target in understanding inflammatory-associated impairments in neurogenesis.


Hippocampus/pathology , Microglia/physiology , Neurogenesis/physiology , Receptors, Cytoplasmic and Nuclear/deficiency , Animals , Cell Survival , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Doublecortin Protein , Hippocampus/metabolism , Inflammation , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Male , Mice , Mice, Knockout , Receptors, Cytoplasmic and Nuclear/physiology , Stem Cell Niche
13.
Brain Behav Immun ; 74: 252-264, 2018 11.
Article En | MEDLINE | ID: mdl-30217534

Understanding the long-term consequences of chronic inflammation in the hippocampus may help to develop therapeutic targets for the treatment of cognitive disorders related to stress, ageing and neurodegeneration. The hippocampus is particularly vulnerable to increases in the pro-inflammatory cytokine interleukin-1ß (IL-1ß), a mediator of neuroinflammation, with elevated levels implicated in the aetiology of neurodegenerative diseases such as Alzheimer's and Parkinson's, and in stress-related disorders such as depression. Acute increases in hippocampal IL-1ß have been shown to impair cognition and reduce adult hippocampal neurogenesis, the birth of new neurons. However, the impact of prolonged increases in IL-1ß, as evident in clinical conditions, on cognition has not been fully explored. Therefore, the present study utilized a lentiviral approach to induce long-term overexpression of IL-1ß in the dorsal hippocampus of adult male Sprague Dawley rats and examine its impact on cognition. Following three weeks of viral integration, pattern separation, a process involving hippocampal neurogenesis, was impaired in IL-1ß-treated rats in both object-location and touchscreen operant paradigms. This was coupled with a decrease in the number and neurite complexity of immature neurons in the hippocampus. Conversely, tasks involving the hippocampus, but not sensitive to disruption of hippocampal neurogenesis, including spontaneous alternation, novel object and location recognition were unaffected. Touchscreen operant visual discrimination, a cognitive task involving the prefrontal cortex, was largely unaffected by IL-1ß overexpression. In conclusion, these findings suggest that chronically elevated IL-1ß in the hippocampus selectively impairs pattern separation. Inflammatory-mediated disruption of adult hippocampal neurogenesis may contribute to the cognitive decline associated with neurodegenerative and stress-related disorders.


Cognitive Dysfunction/metabolism , Hippocampus/metabolism , Interleukin-1beta/metabolism , Animals , Hippocampus/pathology , Inflammation/metabolism , Inflammation/pathology , Male , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurogenesis/physiology , Neurons/metabolism , Neurons/pathology , Rats , Rats, Sprague-Dawley , Temporal Lobe/metabolism , Temporal Lobe/pathology
14.
Brain Behav Immun ; 70: 268-279, 2018 05.
Article En | MEDLINE | ID: mdl-29518529

TLX is an orphan nuclear receptor highly expressed within neural progenitor cells (NPCs) in the hippocampus where is regulates proliferation. Inflammation has been shown to have negative effects on hippocampal function as well as on NPC proliferation. Specifically, the pro-inflammatory cytokine IL-1ß suppresses NPC proliferation as well as TLX expression in the hippocampus. However, it is unknown whether TLX itself is involved in regulating the inflammatory response in the hippocampus. To explore the role of TLX in inflammation, we assessed changes in the transcriptional landscape of the hippocampus of TLX knockout mice (TLX-/-) compared to wildtype (WT) littermate controls with and without intrahippocampal injection of IL-1ß using a whole transcriptome RNA sequencing approach. We demonstrated that there is an increase in the transcription of genes involved in the promotion of inflammation and regulation of cell chemotaxis (Tnf, Il1b, Cxcr1, Cxcr2, Tlr4) and a decrease in the expression of genes relating to synaptic signalling (Lypd1, Syt4, Cplx2) in cannulated TLX-/- mice compared to WT controls. We demonstrate that mice lacking in TLX share a similar increase in 176 genes involved in regulating inflammation (e.g. Cxcl1, Tnf, Il1b) as WT mice injected with IL-1ß into the hippocampus. Moreover, TLX-/- mice injected with IL-1ß displayed a blunted transcriptional profile compared to WT mice injected with IL-1ß. Thus, TLX-/- mice, which already have an exaggerated inflammatory profile after cannulation surgery, are primed to respond differently to an inflammatory stimulus such as IL-1ß. Together, these results demonstrate that TLX regulates hippocampal inflammatory transcriptome response to brain injury (in this case cannulation surgery) and cytokine stimulation.


Orphan Nuclear Receptors/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Cell Proliferation , Cytokines , Hippocampus/metabolism , Inflammation , Interleukin-1beta/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/immunology , Neural Stem Cells/physiology , Neurogenesis , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction , Transcriptome , Tumor Necrosis Factor-alpha
15.
Neuroscience ; 365: 226-236, 2017 Dec 04.
Article En | MEDLINE | ID: mdl-28939260

Hippocampal neurogenesis and associated cognitive behaviors are regulated by a number of factors including stress, inflammation, and exercise. However, the interplay between these factors remains relatively unexplored, especially across the lifespan. In the current study, the effect of social isolation stress during the adolescent period on neurogenesis and hippocampal-dependent cognitive behaviors was examined. This period of the lifespan has been demonstrated to be an important time for hippocampal growth and plasticity, during which changes to hippocampal neurogenesis may have long lasting effects. Additionally, we aimed to determine whether a 'dual-hit' of adolescent stress and adult chronic neuroinflammation would potentiate any negative effects of either insult alone. Lastly, the potential positive effects of exercise during adolescence was examined to determine whether exercise could attenuate any negative impacts of these insults on hippocampal neurogenesis and behavior. The results from the current study demonstrate that social isolation stress during adolescence followed by intra-hippocampal exposure to the pro-inflammatory cytokine IL-1ß in early adulthood produces deficits in both spontaneous alternations and novel object recognition. Exercise attenuated deficits in neurogenesis and novel object recognition in mice that had been exposed to the 'dual-hit' of stress and neuroinflammation. These findings indicate that adolescence represents a key period of the lifespan during which external factors such as stress and exercise can impact on hippocampal development, and may alter the response to challenges such as neuroinflammation in later life.


Hippocampus/physiopathology , Inflammation/etiology , Neurogenesis/physiology , Physical Conditioning, Animal/physiology , Social Isolation , Stress, Psychological/pathology , Animals , Animals, Newborn , Body Weight , Bromodeoxyuridine/metabolism , Cell Survival , Doublecortin Domain Proteins , Interleukin-1beta/metabolism , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Neuropeptides/metabolism , Phosphopyruvate Hydratase/metabolism , Recognition, Psychology/physiology , Stress, Psychological/physiopathology
16.
J Abnorm Psychol ; 125(4): 482-94, 2016 05.
Article En | MEDLINE | ID: mdl-27054346

Extensive evidence highlights the role of inflammatory processes in major depressive disorder (MDD). However, most studies have examined a consistent set of inflammatory cytokines and there is evidence that other immune-derived products may play a role in MDD. In this article, we present data from 3 complimentary studies that support the role of a novel cytokine, interleukin-33 (IL-33), in depression risk. First, we show that a 2-SNP haplotype in the IL-33 gene (rs11792633 and rs7044343) moderated the link between women's history of childhood abuse and their history of recurrent MDD (rMDD), such that the link between childhood abuse and rMDD was stronger among women with fewer copies of the protective IL-33 CT haplotype. Second, linking these findings to differences in circulating cytokine levels, we show in a separate sample that those with a history of rMDD had higher peripheral levels of IL-33 and IL-1ß compared with women with a single MDD episode or no history of MDD. Third, providing initial evidence of brain regions underlying these effects using archival rat brain tissue, we show that an acute stressor increased IL-33 expression in the paraventricular nucleus of the hypothalamus and, to a lesser extent, the prefrontal cortex, key brain regions underlying stress response and emotion regulation. These findings provide converging support for the potential role of IL-33 in risk for recurrent MDD. (PsycINFO Database Record


Depressive Disorder, Major/blood , Depressive Disorder, Major/genetics , Interleukin-33/blood , Interleukin-33/genetics , Adult , Animals , Brain/metabolism , Female , Gene-Environment Interaction , Haplotypes , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Rats , Rats, Sprague-Dawley , Risk Factors , Stress, Psychological/metabolism
17.
Behav Brain Res ; 306: 36-47, 2016 06 01.
Article En | MEDLINE | ID: mdl-26970576

The nuclear receptor Tlx is a key regulator of embryonic and adult hippocampal neurogenesis and has been genetically linked to bipolar disorder. Mice lacking Tlx (Nr2e1(-/-)) display deficits in adult hippocampal neurogenesis and behavioural abnormalities. However, whether Tlx regulates behaviour during adolescence or in a sex-dependent manner remains unexplored. Therefore, we investigated the role of Tlx in a series of behavioural tasks in adolescent male and female mice with a spontaneous deletion of Tlx (Nr2e1(-/-) mice). Testing commenced at adolescence (postnatal day 28) and continued until adulthood (postnatal day 67). Adolescent male and female Nr2e1(-/-) mice were hyperactive in an open field, an effect that persisted in adulthood. Male but not female Nr2e1(-/-) mice exhibited reduced thigmotaxis during adolescence and adulthood. Impairments in rotarod motor performance developed in male and female Nr2e1(-/-) mice at the onset of adulthood. Spontaneous alternation in the Y-maze, a hippocampus-dependent task, was impaired in adolescent but not adult male and female Nr2e1(-/-) mice. Contextual fear conditioning was impaired in adolescent male Nr2e1(-/-) mice only, but both male and female adolescent Nr2e1(-/-) mice showed impaired cued fear conditioning, a hippocampal-amygdala dependent cognitive process. These deficits persisted into adulthood in males but not females. In conclusion, deletion of Tlx impairs motor, cognitive and anxiety-related behaviours during adolescence and adulthood in male and female mice with most effects occurring during adolescence rather than adulthood, independent of housing conditions. This suggests that Tlx has functions beyond regulation of adult hippocampal neurogenesis, and may be an important target in understanding neurobiological disorders.


Aging , Anxiety/genetics , Cognition Disorders/genetics , Hyperkinesis/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Sex Characteristics , Aging/genetics , Animals , Animals, Newborn , Body Weight/genetics , Conditioning, Classical/physiology , Cues , Exploratory Behavior/physiology , Fear/physiology , Female , Locomotion/genetics , Male , Maze Learning/physiology , Mice , Mice, Transgenic , Receptors, Cytoplasmic and Nuclear/genetics , Reflex/genetics
18.
Neuroendocrinology ; 100(2-3): 162-77, 2014.
Article En | MEDLINE | ID: mdl-25300872

Exposure to stressors such as foot shock (FS) leads to increased expression of multiple inflammatory factors, including the proinflammatory cytokine interleukin-1 (IL-1) in the brain. Studies have indicated that there are sex differences in stress reactivity, suggesting that the fluctuations in gonadal steroid levels across the estrous cycle may play a regulatory role in the stress-induced cytokine expression. The present studies were designed to investigate the role of 17-ß-estradiol (E2) and progesterone (Pg) in regulating the cytokine response within the paraventricular nucleus (PVN) of the hypothalamus through analysis of gene expression with real-time RT-PCR. Regularly cycling female rats showed a stress-induced increase in PVN IL-1 levels during the diestrous, proestrous, and estrous stages. During the metestrous stage, no change in IL-1 levels was seen following FS; however, estrogen receptor (ER)-ß levels did increase. Ovariectomy resulted in an increase in PVN IL-1 levels, which was attenuated by treatment with estradiol benzoate (10 or 50 µg), indicating an E2-mediated anti-inflammatory effect. Ovariectomized rats treated with Pg (500 or 1,250 µg) showed no alteration in IL-1 levels, but Pg did up-regulate ER-ß gene expression. The results from the current study implicate a potential mechanism through which high availability of endogenous Pg during the metestrous stage increases ER-ß sensitivity, which in turn attenuates the PVN IL-1 response to stress. Thus, the interaction between gonadal steroid hormones and their central receptors may exert a powerful inhibitory effect on neuroimmune consequences of stress throughout the estrous cycle.


Estradiol/metabolism , Estrous Cycle/metabolism , Interleukin-1/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Progesterone/metabolism , Stress, Psychological/metabolism , Animals , Disease Models, Animal , Electroshock , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor beta/metabolism , Female , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/physiology , Ovariectomy , Paraventricular Hypothalamic Nucleus/drug effects , Progesterone/administration & dosage , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Reproductive Control Agents/pharmacology
19.
Behav Pharmacol ; 25(5-6): 582-98, 2014 Sep.
Article En | MEDLINE | ID: mdl-25083574

Stress leads to neuroinflammatory and behavioral consequences through upregulation of inflammatory-related cytokines within the central nervous system such as interleukin-1ß (IL-1ß), which may be indicative of microglial priming/activation. Evidence suggests that the P2X7 receptor (P2X7R) may play an important role in the synthesis and conversion of IL-1ß. In a series of six experiments, adult male rats were intubated with a highly selective P2X7R antagonist (A-804598) before footshock exposure. As expected, footshock increased IL-1ß and CD14 mRNA in the paraventricular nucleus, and A-804598 (25 mg/kg) partially attenuated these effects. Footshock also increased hypothalamic IL-1 protein in whole hypothalamic blocks, but no effect was observed on the formation of pro-IL-1ß or IL-1ß in the paraventricular nucleus as assessed using western blotting. A-804598 also did not reverse the suppression in exploration produced by stress exposure. The present findings support the use of the footshock paradigm as a method for inducing stress-related neuroimmune and behavioral changes, but the evidence to support the role of A-804598 as a potential tool to reverse such changes remains modest. This study is the first to examine the role of P2X7R in vivo following footshock exposure. Further characterization of P2X7R may have implications for understanding the relationship between stress and inflammation.


Exploratory Behavior/drug effects , Guanidines/pharmacology , Neuroimmunomodulation/drug effects , Psychotropic Drugs/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , Quinolines/pharmacology , Stress, Psychological/drug therapy , Animals , Blotting, Western , Electroshock , Enzyme-Linked Immunosorbent Assay , Exploratory Behavior/physiology , Foot , Hypothalamus/drug effects , Hypothalamus/physiopathology , Interleukin-1beta/metabolism , Lipopolysaccharide Receptors/metabolism , Male , Neuroimmunomodulation/physiology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2X7/metabolism , Stress, Psychological/physiopathology
20.
Endocrinology ; 155(9): 3527-37, 2014 Sep.
Article En | MEDLINE | ID: mdl-24926824

Although progesterone is most commonly regarded in terms of its role in the female estrous cycle, reproductive behavior, and pregnancy, progesterone is also a precursor to corticosterone (CORT) and is released from the adrenal glands of both sexes in response to stress. However, the relationship between plasma CORT and progesterone during times of stress has not been well established. To better characterize dynamic changes in progesterone release as a result of stressor exposure, plasma progesterone levels were measured using enzyme immunoassay under multiple conditions, including after stress exposure (footshock, restraint, and forced swim), manipulations of the hypothalamic-pituitary-adrenal axis (injection of metyrapone or dexamethasone, or adrenalectomy), and in response to CRH and ACTH injections. When plasma levels of ACTH, CORT, and progesterone were analyzed across studies, CORT and progesterone were found to be highly correlated and rarely dissociated. Additionally, it appears that in male rats, the adrenal glands are the principle source of circulating progesterone in response to stress. Interestingly, a detailed time course indicated lack of a circadian rhythm of progesterone secretion, despite a robust rhythm of circulating CORT. The current studies provide critical new information on the coincident release of progesterone and CORT in response to stress and their somewhat paradoxical dissociation across the circadian rhythm. These findings provide an important foundation for future studies that will examine the role of stress-induced progesterone in behavioral, neuroimmune, and neuroendocrine responses to stress.


Progesterone/blood , Stress, Physiological , Adrenal Glands/metabolism , Adrenocorticotropic Hormone/blood , Animals , Corticosterone/blood , Female , Male , Rats , Rats, Sprague-Dawley
...