Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Brain Behav Immun ; 104: 39-53, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35569797

ABSTRACT

Sepsis associated encephalopathy, occurs in 70% of severe septic cases, following which survivors exhibit long-term cognitive impairment or global loss of cognitive function. Currently there is no clearly defined neurochemical basis of septic encephalopathy. Moreover, the lingering neurological complications associated with the severe acute respiratory syndrome CoV 2 (SARS-CoV-2) and the significant worsening in outcomes for those individuals with SARS-Cov-2 following sepsis underscore the need to define factors underlying the susceptibility to acute toxic encephalitis. In this study, differential neurochemical sequelae in response to sepsis (lipopolysaccharide (LPS)-induced endotoxemia and caecal ligation and puncture (CLP)), were evaluated in two inbred mouse strains, known to differ in behaviour, immune profile, and neurotransmitter levels, namely BALB/c and C57BL/6J. It was hypothesized that these strains would differ in sepsis severity, cytokine profile, peripheral tryptophan metabolism and central monoamine turnover. BALB/c mice exhibited more pronounced sickness behavioural scores, hypothermia, and significant upregulation of cytokines in the LPS model relative to C57BL/6J mice. Increased plasma kynurenine/tryptophan ratio, hippocampal serotonin and brainstem dopamine turnover were evident in both strains, but the magnitude was greater in BALB/c mice. In addition, CLP significantly enhanced kynurenine levels and hippocampal serotonergic and dopaminergic neurotransmission in C57BL/6J mice. Overall, these studies depict consistent changes in kynurenine, serotonin, and dopamine post sepsis. Further evaluation of these monoamines in the context of septic encephalopathy and cognitive decline is warranted. Moreover, these data suggest the continued evaluation of altered peripheral kynurenine metabolism as a potential blood-based biomarker of sepsis.

2.
J Neuroinflammation ; 12: 67, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25890218

ABSTRACT

BACKGROUND: Lipopolysaccharide (LPS) and interferon-γ (IFNγ) increase expression of tumour necrosis factor-α (TNFα) that characterizes the M1 activation state of macrophages. Whereas it is accepted that the immune system undergoes changes with age, there is inconsistency in the literature with respect to the impact of age on the response of macrophages to inflammatory stimuli. Here, we investigate the effect of age on the responsiveness of bone marrow-derived macrophages (BMDMs) to LPS and IFNγ. The context for addressing this question is that macrophages, which infiltrate the brain of aged animals, will encounter the neuroinflammatory environment that has been described with age. METHODS: Brain tissue, prepared from young and aged rats, was assessed for expression of inflammatory markers by PCR and for evidence of infiltration of macrophages by flow cytometry. BMDMs were prepared from the long bones of young and aged rats, maintained in culture for 8 days and incubated in the presence or absence of LPS (100 ng/ml) or IFNγ (50 ng/ml). Cells were harvested and assessed for mRNA expression of markers of M1 activation including TNFα and NOS2, or for expression of IFNγR1 and TLR4 by western immunoblotting. To assess whether BMDMs induced glial activation, mixed glial cultures were incubated in the presence of conditioned media obtained from unstimulated BMDMs of young and aged rats and evaluated for expression of inflammatory markers. RESULTS: Markers associated with M1 activation were expressed to a greater extent in BMDMs from aged rats in response to LPS and IFNγ, compared with cells from young rats. The increased responsiveness was associated with increases in IFNγ receptor (IFNγR) and Toll-like receptor 4 (TLR4). The data show that conditioned media from BMDMs of aged rats increased the expression of pro-inflammatory mediators in glial cells. Significantly, there was an age-related increase in macrophage infiltration into the brain, and this was combined with increased expression of IFNγ and the Toll-like receptor 4 agonist, high-mobility group protein B1 (HMGB1). CONCLUSION: Exposure of infiltrating macrophages to the inflammatory microenvironment that develops in the brain with age is likely to contribute to a damaging cascade that negatively impacts neuronal function.


Subject(s)
Aging , Cytokines/metabolism , Gene Expression Regulation/drug effects , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Cells, Cultured , Cytokines/genetics , Enzyme-Linked Immunosorbent Assay , Male , Nitric Oxide Synthase Type II/metabolism , RNA, Messenger , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/metabolism
3.
Neurobiol Aging ; 33(1): 162-75, 2012 Jan.
Article in English | MEDLINE | ID: mdl-20382448

ABSTRACT

Neuroinflammation is a significant and consistent feature of many neurodegenerative disorders, including Alzheimer's disease (AD) and Parkinson's disease (PD). The greatest risk factor for neurodegenerative disorders is age and a proinflammatory phenotype in the aged brain is believed to contribute to these neurodegenerative conditions. In animal models, neuroinflammatory changes, characterized by increased microglial activation, have been associated with a loss of synaptic plasticity and here we show that treatment of aged rats with the PPARγ agonist, rosiglitazone, modulates the inflammatory changes and restores synaptic function. The evidence presented highlights an important role for astrocytes in inducing inflammatory changes and suggests that the age-related astrogliosis and astrocytosis is responsible for the increase in the proinflammatory cytokine, tumor necrosis factor alpha (TNF-α). Magnetic resonance (MR) imaging revealed an age-related increase in T1 relaxation time and, importantly, treatment of aged rats with rosiglitazone reversed the age-related increases in astrogliosis and astrocytosis, TNF-α concentration and T1 relaxation time. The evidence indicates that the site of action for rosiglitazone is endothelial cells, and suggests that its effect on astrocytes is secondary to its effect on endothelial cells.


Subject(s)
Aging/pathology , Aging/physiology , Astrocytes/pathology , Astrocytes/physiology , Endothelial Cells/drug effects , Hippocampus/cytology , Hypoglycemic Agents/pharmacology , Long-Term Potentiation/drug effects , Thiazolidinediones/pharmacology , Animals , Cells, Cultured , Hippocampus/pathology , Hippocampus/physiopathology , Inflammation Mediators/metabolism , Magnetic Resonance Imaging , Male , PPAR gamma/agonists , Rats , Rats, Wistar , Rosiglitazone , Tumor Necrosis Factor-alpha/metabolism
4.
Neurobiol Aging ; 33(5): 1005.e23-35, 2012 May.
Article in English | MEDLINE | ID: mdl-22071124

ABSTRACT

In view of the increase in the aging population and the unavoidable parallel increase in the incidence of age-related neurodegenerative diseases, a key challenge in neuroscience is the identification of clinical signatures which change with age and impact on neuronal and cognitive function. Early diagnosis offers the possibility of early therapeutic intervention, thus magnetic resonance imaging (MRI) is potentially a powerful diagnostic tool. We evaluated age-related changes in relaxometry, blood flow, and blood-brain barrier (BBB) permeability in the rat by magnetic resonance imaging and assessed these changes in the context of the age-related decrease in synaptic plasticity. We report that T2 relaxation time was decreased with age; this was coupled with a decrease in gray matter perfusion, suggesting that the observed microglial activation, as identified by increased expression of CD11b, MHCII, and CD68 by immunohistochemistry, flow cytometry, or polymerase chain reaction (PCR), might be a downstream consequence of these changes. Increased permeability of the blood-brain barrier was observed in the perivascular area and the hippocampus of aged, compared with young, rats. Similarly there was an age-related increase in CD45-positive cells by flow cytometry, which are most likely infiltrating macrophages, with a parallel increase in the messenger mRNA expression of chemokines IP-10 and MCP-1. These combined changes may contribute to the deficit in long-term potentiation (LTP) in perforant path-granule cell synapses of aged animals.


Subject(s)
Aging/physiology , Blood-Brain Barrier/physiology , Cerebrovascular Circulation/physiology , Long-Term Potentiation/physiology , Neurodegenerative Diseases/physiopathology , Aging/pathology , Animals , Capillary Permeability/physiology , Cerebral Cortex/blood supply , Cerebral Cortex/pathology , Cerebral Cortex/physiology , Hippocampus/blood supply , Hippocampus/pathology , Hippocampus/physiology , Magnetic Resonance Imaging/methods , Male , Neurodegenerative Diseases/pathology , Perfusion Imaging/methods , Rats , Rats, Wistar
5.
J Neuroimmunol ; 220(1-2): 34-42, 2010 Mar 30.
Article in English | MEDLINE | ID: mdl-20061033

ABSTRACT

Evidence suggests that noradrenaline has a tonic anti-inflammatory action in the central nervous system (CNS) via its ability to inhibit expression of inflammatory mediators from glial cells. Consequently it is suggested that noradrenaline may play an endogenous neuroprotective role in CNS disorders where inflammatory events contribute to pathology. Infiltration of peripheral immune cells into the brain is driven by increased chemokine and cell adhesion molecule (CAM) expression, and is known to exacerbate neuroinflammation and thereby contribute to the disease process in a number of neurodegenerative disease states. Here we demonstrate that treatment of rats with the noradrenaline reuptake inhibitors (NRIs) desipramine and atomoxetine, agents that increase extracellular noradrenaline in the CNS, suppressed chemokine and cell adhesion molecule (CAM) expression in rat brain following a systemic challenge with bacterial lipopolysaccharide (LPS). Specifically, these agents reduced expression of the chemokines, interferon-inducible protein-10 (IP-10, CXCL-10) and regulated upon activation normal T-cell expressed and secreted (RANTES, CCL-5), and the CAMs, vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule (ICAM-1) in cortex and hippocampus. The inhibitory action of NRIs on chemokines and CAM expression was mimicked by in vitro exposure of cultured glial cells to noradrenaline, but not to the NRIs themselves. These data indicate that the suppressive action of NRIs on chemokine and CAM expression that occurs in vivo is due to increased noradrenaline availability at glial cells, as opposed to a direct action of the drugs on glial cells per se. These results support the theory that noradrenaline has anti-inflammatory properties, and agents that increase noradrenaline availability in vivo can play a role in combating brain inflammation by reducing expression of chemokines and CAMs; molecules that facilitate leucocyte influx into the CNS.


Subject(s)
Adrenergic Uptake Inhibitors/pharmacology , Brain/drug effects , Cell Adhesion Molecules/drug effects , Chemokines/drug effects , Encephalitis/drug therapy , Adrenergic Uptake Inhibitors/therapeutic use , Animals , Animals, Newborn , Biomarkers/blood , Brain/immunology , Brain/metabolism , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cell Adhesion Molecules/metabolism , Cells, Cultured , Chemokine CCL5/metabolism , Chemokine CXCL1/metabolism , Chemokine CXCL10/metabolism , Chemokines/metabolism , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Encephalitis/immunology , Encephalitis/physiopathology , Inflammation Mediators/pharmacology , Intercellular Adhesion Molecule-1/metabolism , Lipopolysaccharides , Male , Neuroglia/drug effects , Neuroglia/immunology , Neuroglia/metabolism , Norepinephrine/metabolism , Rats , Rats, Sprague-Dawley , Vascular Cell Adhesion Molecule-1/metabolism
6.
J Neurochem ; 110(5): 1547-56, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19627440

ABSTRACT

Several neurodegenerative disorders are associated with evidence of inflammation, one feature of which is increased activation of microglia, the most likely cellular source of inflammatory cytokines like interleukin-1beta. It is now recognized that interaction of microglia with other cells contributes to maintenance of microglia in a quiescent state and the complementary distribution of the chemokine, fractalkine (CX(3)CL1) on neurons and its receptor (CX(3)CR1) on microglia, suggests that this interaction may play a role in modulating microglial activation. Here we demonstrate that both soluble and membrane-bound fractalkine attenuate lipopolysaccharide-induced microglial activation in vitro. We also show that fractalkine expression is reduced in the brain of aged rats and this is accompanied by an age-related increase in microglial activation. Treatment of aged rats with fractalkine attenuates the age-related increase in microglial activation and the evidence indicates that fractalkine-induced activation of the phosphatidylinositol-3 kinase pathway is required to maintain microglia in a quiescent state both in vivo and in vitro.


Subject(s)
Chemokine CX3CL1/physiology , Microglia/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/physiology , Age Factors , Aging/metabolism , Animals , Animals, Newborn , Cells, Cultured , Enzyme Activation/physiology , Lipopolysaccharides/physiology , Male , Membrane Proteins/physiology , Microglia/metabolism , Phosphatidylinositol 3-Kinases/physiology , Rats , Rats, Wistar , Solubility
7.
Int J Neuropsychopharmacol ; 12(5): 687-99, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19046481

ABSTRACT

Evidence suggests that noradrenaline has a tonic anti-inflammatory action in the central nervous system (CNS) via its ability to suppress microglial and astrocytic activation, and inhibit production of inflammatory mediators. Consequently it is suggested that noradrenaline may play an endogenous neuroprotective role in CNS disorders where inflammatory events contribute to pathology. Here we demonstrate that acute treatment of rats with the noradrenaline reuptake inhibitors (NRIs) desipramine and atomoxetine elicited anti-inflammatory actions in rat cortex following a systemic challenge with bacterial lipopolysaccharide (LPS). This was characterized by a reduction in cortical gene expression of the pro-inflammatory cytokines interleukin-1beta (IL-1beta) and tumour necrosis factor-alpha (TNF-alpha), the enzyme inducible nitric oxide synthase (iNOS), and the microglial activation markers CD11b and CD40. These anti-inflammatory actions of NRIs were associated with reduced activation of nuclear factor-kappa B (NF-kappaB); a transcription factor that is considered the major regulator of inflammation in the CNS. To determine whether NRI administration directly altered glial expression of these inflammatory markers, primary cortical glial cells were exposed in vitro to the NRIs desipramine or atomoxetine. In vitro treatment with NRIs largely failed to alter mRNA expression of IL-1beta, TNF-alpha, iNOS, CD11b and CD40, following stimulation with LPS. Similarly, LPS-induced TNF-alpha and IL-1beta protein production from glial cells was unaffected by NRI treatment. In contrast, in vitro exposure of cultured glial cells to noradrenaline suppressed IL-1beta, TNF-alpha, iNOS and CD40 expression. These results suggest that in vivo administration of NRIs limit inflammatory events in the brain, probably by increasing noradrenaline availability. Overall, this study has yielded significant insights into the ability of noradrenaline-augmentation strategies to limit neuroinflammation.


Subject(s)
Adrenergic Uptake Inhibitors/therapeutic use , Cerebral Cortex/metabolism , Depression/metabolism , Inflammation Mediators/antagonists & inhibitors , Nerve Degeneration/metabolism , Norepinephrine/metabolism , Adrenergic Uptake Inhibitors/pharmacology , Animals , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Depression/drug therapy , Depression/pathology , Inflammation/drug therapy , Inflammation/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/toxicity , Male , Nerve Degeneration/drug therapy , Norepinephrine/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
8.
Neurosci Lett ; 441(1): 29-34, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18584961

ABSTRACT

Inflammation-mediated dysregulation of the kynurenine pathway has been implicated as a contributor to a number of major brain disorders. Consequently, we examined the impact of a systemic inflammatory challenge on kynurenine pathway enzyme expression in rat brain. Indoleamine 2,3-dioxygenase (IDO) expression was induced in cortex and hippocampus following systemic lipopolysaccharide (LPS) administration. Whilst IDO expression was paralleled by increased circulating interferon (IFN)-gamma concentrations, IFN-gamma expression in the brain was only modestly altered following LPS administration. In contrast, induction of IDO was associated with increased central tumour necrosis factor (TNF)-alpha and interleukin (IL)-6 expression. Similarly, in cultured glial cells LPS-induced IDO expression was accompanied by increased TNF-alpha and IL-6 expression, whereas IFN-gamma was not detectable. These findings indicate that IFN-gamma is not required for LPS-induced IDO expression in brain. A robust increase in kynurenine-3-monooxygenase (KMO) expression was observed in rat brain 24h post LPS, without any change in kynurenine aminotransferase II (KAT II) expression. In addition, we report that constitutive expression of KAT II is approximately 8-fold higher than KMO in cortex and 20-fold higher in hippocampus. Similarly, in glial cells constitutive expression of KAT II was approximately 16-fold higher than KMO, and expression of KMO but not KAT II was induced by LPS. These data are the first to demonstrate that a systemic inflammatory challenge stimulates KMO expression in brain; a situation that is likely to favour kynurenine metabolism in a neurotoxic direction. However, our observation that expression of KAT II is much higher than KMO in rat brain is likely to counteract potential neurotoxicity that could arise from KMO induction following an acute inflammation.


Subject(s)
Brain/enzymology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Inflammation/blood , Inflammation/pathology , Interferon-gamma/blood , Kynurenine 3-Monooxygenase/metabolism , Animals , Brain/drug effects , Brain/pathology , Inflammation/chemically induced , Interferon-alpha/genetics , Interferon-alpha/metabolism , Interferon-gamma/genetics , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides , Male , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
10.
Neuroimmunomodulation ; 10(6): 367-78, 2002.
Article in English | MEDLINE | ID: mdl-12907844

ABSTRACT

Administration of bacterial lipopolysaccharide (LPS) to rodents induces hypophagia, body weight loss and hypolocomotion, a constellation of symptoms collectively referred to as 'sickness behaviour'. We examined the role of the gaseous transmitter nitric oxide (NO) in mediating LPS-induced sickness behaviour in rats. Treatment with the non-selective NO synthase (NOS) inhibitor N(G)-nitro-L-arginine (L-NA) (20 mg/kg; i.p.) increased the severity of LPS-induced sickness behaviour in rats, suggesting that endogenous NO does not act as a mediator of LPS-induced sickness behaviour, but may rather have a protective role, acting in an inhibitory feedback manner to limit LPS-induced sickness. To evaluate the role of the different NOS isoforms in this response, we examined the effect of the neuronal NOS inhibitor, 7-nitroindazole (7-NI; 25 and 50 mg/kg; i.p.), and the inducible NOS inhibitor, aminoguanidine (AGN; 50 and 100 mg/kg; i.p.). Neither 7-NI nor AGN significantly altered LPS-induced sickness behaviour. Therefore, it is likely that the endothelial isoform of NOS mediates the effect of L-NA on LPS-induced sickness behaviour. As pro-inflammatory cytokines are mediators of LPS-induced sickness behaviour, we examined the effect of L-NA (20 mg/kg; i.p.) on LPS-induced interleukin (IL)-1beta, IL-6 and tumour necrosis factor (TNF)-alpha production. L-NA increased LPS-induced TNF-alpha without significantly altering IL-1beta or IL-6 production. Moreover, pre-treatment with the TNF-alpha inhibitor pentoxyfilline (25 mg/kg; i.p.) largely reversed the augmenting effect of L-NA on LPS-induced sickness behaviour, suggesting that the ability of L-NA to increase TNF-alpha production underpinned its ability to increase the severity of sickness. In conclusion, L-NA increases the severity of LPS-induced sickness behaviour, most likely by blocking the tonic inhibitory action of constitutively produced NO on TNF-alpha production.


Subject(s)
Behavior, Animal/physiology , Lipopolysaccharides/adverse effects , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide/metabolism , Sick Role , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Feedback, Physiological/immunology , Interleukin-1/metabolism , Interleukin-6/metabolism , Male , Nitroarginine/pharmacology , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...