Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Neuropathol Appl Neurobiol ; 45(3): 262-277, 2019 04.
Article in English | MEDLINE | ID: mdl-29797340

ABSTRACT

AIM: The insular cortex consists of a heterogenous cytoarchitecture and diverse connections and is thought to integrate autonomic, cognitive, emotional and interoceptive functions to guide behaviour. In Parkinson's disease (PD) and dementia with Lewy bodies (DLB), it reveals α-synuclein pathology in advanced stages. The aim of this study is to assess the insular cortex cellular and subregional vulnerability to α-synuclein pathology in well-characterized PD and DLB subjects. METHODS: We analysed postmortem insular tissue from 24 donors with incidental Lewy body disease, PD, PD with dementia (PDD), DLB and age-matched controls. The load and distribution of α-synuclein pathology and tyrosine hydroxylase (TH) cells were studied throughout the insular subregions. The selective involvement of von Economo neurons (VENs) in the anterior insula and astroglia was assessed in all groups. RESULTS: A decreasing gradient of α-synuclein pathology load from the anterior periallocortical agranular towards the intermediate dysgranular and posterior isocortical granular insular subregions was found. Few VENs revealed α-synuclein inclusions while astroglial synucleinopathy was a predominant feature in PDD and DLB. TH neurons were predominant in the agranular and dysgranular subregions but did not reveal α-synuclein inclusions or significant reduction in density in patient groups. CONCLUSIONS: Our study highlights the vulnerability of the anterior agranular insula to α-synuclein pathology in PD, PDD and DLB. Whereas VENs and astrocytes were affected in advanced disease stages, insular TH neurons were spared. Owing to the anterior insula's affective, cognitive and autonomic functions, its greater vulnerability to pathology indicates a potential contribution to nonmotor deficits in PD and DLB.


Subject(s)
Cerebral Cortex/pathology , Lewy Body Disease/pathology , Parkinson Disease/pathology , Tissue Banks , alpha-Synuclein/metabolism , Aged , Aged, 80 and over , Cerebral Cortex/metabolism , Female , Humans , Lewy Body Disease/metabolism , Male , Parkinson Disease/metabolism
2.
Gene Ther ; 17(5): 662-71, 2010 May.
Article in English | MEDLINE | ID: mdl-20182518

ABSTRACT

Neuroinflammation, as defined by activation of local glial cells and production of various inflammatory mediators, is an important feature of many neurological disorders. Expression of pro-inflammatory mediators produced by glial cells in the central nervous system (CNS) is considered to contribute to the neuropathology observed in those diseases. To diminish the production or action of pro-inflammatory mediators, we have used lentiviral (LV) vector-mediated encoding rat interleukin-10 (rIL-10) or rat interleukin-1 receptor antagonist (rIL-1ra) to direct the local, long-term expression of these anti-inflammatory cytokines in the CNS. We have shown that cultured macrophages or astroglia transduced with LV-rIL-10 or LV-rIL-1ra produced far less tumor necrosis factor (TNF)alpha or IL-6, respectively in response to pro-inflammatory stimuli. Moreover, intracerebroventricular (i.c.v.) administration of LV-rIL-10 or LV-rIL-1ra resulted in transduction of glial cells and macrophages and, subsequently reduced TNFalpha, IL-6 and inducible nitric oxide synthase (iNOS) expression in various brain regions induced by inflammatory stimuli, whereas peripheral expression of these mediators remained unaffected. In addition, expression levels of the anti-inflammatory cytokines IL-4 and transforming growth factor-beta were not altered in either brain or pituitary gland. Furthermore, i.c.v. administration of LV-rIL-10 or LV-rIL-1ra given during the remission phase of chronic-relapsing experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis, improved the clinical outcome of the relapse phase. Thus, local application of LV vectors expressing anti-inflammatory cytokines could be of therapeutic interest to counteract pro-inflammatory processes in the brain without interfering with the peripheral production of inflammatory mediators.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Genetic Therapy/methods , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin-10/genetics , Animals , Brain/metabolism , Brain/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Genetic Vectors , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Interleukin-4/analysis , Interleukin-4/metabolism , Interleukin-6/analysis , Interleukin-6/metabolism , Lentivirus , Macrophages/metabolism , Male , Neuroglia/metabolism , Nitric Oxide Synthase Type II/analysis , Nitric Oxide Synthase Type II/metabolism , Rats , Rats, Wistar , Transduction, Genetic , Transforming Growth Factor beta/analysis , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/metabolism
3.
Eur Neuropsychopharmacol ; 20(1): 1-17, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19748235

ABSTRACT

Adult hippocampal neurogenesis, a once unorthodox concept, has changed into one of the most rapidly growing fields in neuroscience. The present report results from the ECNP targeted expert meeting in 2007 during which cellular plasticity changes were addressed in the adult brain, focusing on neurogenesis and apoptosis in hippocampus and frontal cortex. We discuss recent studies investigating factors that regulate neurogenesis with special emphasis on effects of stress, sleep disruption, exercise and inflammation, a group of seemingly unrelated factors that share at least two unifying properties, namely that they all regulate adult hippocampal neurogenesis and have all been implicated in the pathophysiology of mood disorders. We conclude that although neurogenesis has been implicated in cognitive function and is stimulated by antidepressant drugs, its functional impact and contribution to the etiology of depression remains unclear. A lasting reduction in neurogenesis following severe or chronic stress exposure, either in adult or early life, may represent impaired hippocampal plasticity and can contribute to the cognitive symptoms of depression, but is, by itself, unlikely to produce the full mood disorder. Normalization of reductions in neurogenesis appears at least partly, implicated in antidepressant action.


Subject(s)
Exercise/physiology , Inflammation/physiopathology , Neurogenesis/physiology , Sleep Wake Disorders/physiopathology , Stress, Psychological/pathology , Adult Stem Cells/physiology , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Depressive Disorder/drug therapy , Depressive Disorder/physiopathology , Hippocampus/pathology , Humans , Inflammation/pathology
4.
Brain Behav Immun ; 23(1): 92-100, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18835435

ABSTRACT

Multiple Sclerosis (MS) is an autoimmune inflammatory disease that presents clinically with a range of symptoms including motor, sensory, and cognitive dysfunction as well as demyelination and lesion formation in brain and spinal cord. A variety of animal models of MS have been developed that share many of the pathological hallmarks of MS including motor deficits (ascending paralysis), demyelination and axonal damage of central nervous system (CNS) tissue. In recent years, neuropathic pain has been recognized as a prevalent symptom of MS in a majority of patients. To date, there have been very few investigations into sensory disturbances in animal models of MS. The current work contains the first assessment of hind paw mechanical allodynia (von Frey test) over the course of a relapsing-remitting myelin oligodendrocyte glycoprotein induced experimental autoimmune encephalomyelitis (MOG-EAE) rat model of MS and establishes the utility of this model in examining autoimmune induced sensory dysfunction. We demonstrate periods of both decreased responsiveness to touch that precedes the onset of hind limb paralysis, and increased responsiveness (allodynia) that occurs during the period of motor deficit amelioration traditionally referred to as symptom remission. Furthermore, we tested the ability of our recently characterized anti-inflammatory IL-10 gene therapy to treat the autoimmune inflammation induced behavioral symptoms and tissue histopathological changes. This therapy is shown here to reverse inflammation induced paralysis, to reduce disease associated reduction in sensitivity to touch, to prevent the onset of allodynia, to reverse disease associated loss of body weight, and to suppress CNS glial activation associated with disease progression in this model.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Genetic Therapy/methods , Interleukin-10/physiology , Multiple Sclerosis/therapy , Analysis of Variance , Animals , Body Weight/physiology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Glial Fibrillary Acidic Protein/metabolism , Hindlimb , Immunohistochemistry , Inflammation/immunology , Inflammation/therapy , Interleukin-10/genetics , Male , Motor Activity/physiology , Multiple Sclerosis/chemically induced , Multiple Sclerosis/physiopathology , Myelin Proteins , Myelin-Associated Glycoprotein , Myelin-Oligodendrocyte Glycoprotein , Neuralgia/physiopathology , Neuralgia/therapy , Paralysis/physiopathology , Paralysis/therapy , Rats , Rats, Inbred Strains , Spinal Cord/metabolism , Time Factors
5.
Genes Brain Behav ; 7 Suppl 1: 28-42, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18184368

ABSTRACT

This review addresses the role of adult hippocampal neurogenesis and stem cells in some of the most common neurodegenerative disorders and their related animal models. We discuss recent literature in relation to Alzheimer's disease and dementia, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, alcoholism, ischemia, epilepsy and major depression.


Subject(s)
Hippocampus/pathology , Neurodegenerative Diseases/pathology , Neurons/pathology , Adult , Animals , Brain/pathology , Cell Division , Disease Models, Animal , Humans , Mice
6.
Brain ; 130(Pt 12): 3321-35, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18055496

ABSTRACT

Doublecortin (DCX) is a microtubule-associated protein expressed by migrating neuroblasts and is considered to be a reliable marker of neurogenesis. DCX has been used to study the relation between neurogenesis in adult human brain and neurological and neurodegenerative disease processes in the search for putative therapeutic strategies. Using autopsy and surgically resected tissue from a total of 60 patients, we present evidence that DCX is present in several cellular compartments of differentiated astrocytes in the adult human neocortex. One of these compartments consisted of peripheral processes forming punctate envelopes around mature neuronal cell bodies. Markers of glial activation, such as GFAP and HLA, were not associated with DCX immunoreactivity, however, the presence of cytoarchitectural alterations tended to correlate with reduced DCX staining of astrocytic somata. Interestingly, local Alzheimer pathology that showed no relation with cytoarchitectural abnormalities appeared to correlate negatively with the expression of DCX in the astrocytic somata. In combination with the literature our data support the view that DCX in the adult human neocortex may have a function in glia-to-neuron communication. Furthermore, our results indicate that in the adult human neocortex DCX is neither a reliable nor a selective marker of neurogenesis.


Subject(s)
Astrocytes/metabolism , Microtubule-Associated Proteins/metabolism , Neocortex/metabolism , Neurodegenerative Diseases/metabolism , Neuropeptides/metabolism , Adolescent , Adult , Age Distribution , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Biomarkers/metabolism , Cell Differentiation , Child , Child, Preschool , Doublecortin Domain Proteins , Doublecortin Protein , Female , Humans , Male , Middle Aged , Neurodegenerative Diseases/pathology
7.
J Neuroimmunol ; 169(1-2): 59-67, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16198427

ABSTRACT

Inflammation in the central nervous system is an early hallmark of many neurodegenerative diseases including Alzheimer's disease (AD). Recently, increasing evidence suggests that hypercholesterolemia during midlife and abnormalities in the cholesterol metabolism could have an important role in the pathogenesis of AD. In the present study, we have evaluated the effect of high cholesterol (HC) diet on the expression of interleukin-6 (IL-6), a cytokine involved in neurodegeneration, and caspase-1, that is responsible for the cleavage of the precursors of interleukin-1 beta (IL-1 beta) and interleukin-18 (IL-18) in the brain of apolipoprotein E (Apo E) knock-out (KO) and wild type (WT) mice. The density of IL-6-positive cells was increased in the hippocampus (p<0.0001) and the dorsal part of the cortex (p<0.001) of KO and WT mice on HC diet (KOHC and WTHC mice, respectively) compared to KO and WT mice on ND (KOND and WTND mice, respectively). KOHC mice had increased caspase-1 positive cells and staining intensity in the hippocampus in comparison with WTHC mice (p<0.01). In the hippocampus, the density of caspase-1 positive cells was also higher in KOHC compared to KOND mice (p<0.05) and KOHC compared with WTHC mice (p<0.01). There was a major increase in caspase-1 immunoreactivity and cell density in both the dosal part of the cortex (p<0.001) and the lateral part of the cortex (p<0.005) in KO and WT mice on HC diet compared to ND. The findings of the present study indicate that chronic exposure to HC diet increases the expression of the two important inflammatory mediators IL-6 and caspase-1 in the brain of KO and WT mice. In the case of caspase-1, we report a major difference in the effect of HC diet on the KO mice compared to WT mice in the hippocampus. Increased expression of inflammatory mediators involved in neurodegeneration could be a potential mechanism by which hypercholesterolemia and HC diet increase the risk of AD.


Subject(s)
Apolipoproteins E/deficiency , Brain/drug effects , Caspase 1/metabolism , Cholesterol, Dietary/adverse effects , Interleukin-6/metabolism , Animals , Apolipoprotein E4 , Brain/cytology , Brain/metabolism , Cell Count/methods , Cytokines/metabolism , Immunohistochemistry/methods , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/metabolism
8.
J Neuroimmunol ; 118(2): 203-11, 2001 Aug 30.
Article in English | MEDLINE | ID: mdl-11498255

ABSTRACT

The expression of interleukin-1beta was examined in dorsal root ganglion (DRG) neurons from adult rats using non-radioactive in situ hybridization and immunocytochemistry. At all spinal levels, approximately 70% of the DRG neurons appeared to express IL-1beta mRNA; about 80% of these DRG neurons actually appeared to produce the IL-1beta protein at markedly varying levels. The expression of IL-1beta was found in large as well as in intermediate diameter sensory neurons but only sporadically in the population of small sensory neurons. The population of IL-1beta immunopositive sensory neurons included most of the large calretinin-positive Ia afferents, but only a few of the small substance P/CGRP positive sensory neurons. In situ hybridization staining for the detection of type 1 IL-1 receptor showed expression of this receptor by most of the sensory neurons as well as by supportive glial-like cells, presumably satellite cells. The functional significance of IL-1beta in the DRG neurons needs to be elucidated, but we speculate that IL-1beta produced by DRG neurons may be an auto/paracrine signalling molecule in sensory transmission.


Subject(s)
Ganglia, Spinal/metabolism , Interleukin-1/biosynthesis , Neurons, Afferent/metabolism , Animals , Calbindin 2 , Calcitonin Gene-Related Peptide/biosynthesis , Cell Size , Cytoplasm/metabolism , Cytoplasm/ultrastructure , Female , Ganglia, Spinal/cytology , Immunohistochemistry , In Situ Hybridization , Interleukin-1/genetics , Male , Microscopy, Immunoelectron , Neuroglia/cytology , Neuroglia/metabolism , Neurons, Afferent/cytology , Polyribosomes/metabolism , Polyribosomes/ultrastructure , RNA, Messenger/biosynthesis , Rats , Receptors, Interleukin-1/biosynthesis , Receptors, Interleukin-1 Type I , S100 Calcium Binding Protein G/biosynthesis , Substance P/biosynthesis
9.
J Biol Chem ; 276(12): 8820-4, 2001 Mar 23.
Article in English | MEDLINE | ID: mdl-11136731

ABSTRACT

Septic shock is the most common cause of death in intensive care units and no effective treatment is available at present. Lipopolysaccharide (LPS) is the primary mediator of Gram-negative sepsis by inducing the production of macrophage-derived cytokines. Previously, we showed that apolipoprotein E (apoE), an established modulator of lipid metabolism, can bind LPS, thereby redirecting LPS from macrophages to hepatocytes in vivo. We now report that intravenously administered LPS strongly increases the serum levels of apoE. In addition, apoE can prevent the LPS-induced production of cytokines and subsequent death in rodents. Finally, apoE-deficient mice show a significantly higher sensitivity toward LPS than control wild-type mice. These findings indicate that apoE may have a physiological role in the protection against sepsis, and recombinant apoE may be used therapeutically to protect against LPS-induced endotoxemia.


Subject(s)
Apolipoproteins E/physiology , Lipopolysaccharides/antagonists & inhibitors , Salmonella/pathogenicity , Sepsis/therapy , Animals , Apolipoproteins E/metabolism , Enzyme-Linked Immunosorbent Assay , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar , Sepsis/microbiology
10.
Neurosci Lett ; 285(3): 169-72, 2000 May 19.
Article in English | MEDLINE | ID: mdl-10806313

ABSTRACT

In the present study, we examined whether the vagus nerve is involved in mediating lipopolysaccharide (LPS)-induced appearance of IL-1beta immunoreactive cells in the brain and pituitary gland. Rats were either sham-operated or subjected to subdiaphragmatic vagotomy. Four weeks later, pyrogen free saline or 400 microg/kg LPS was administered to the rats intraperitoneally. Four and 8 h later, the animals were intracardially perfused with 4% paraformaldehyde and tissues were prepared for IL-1beta immunocytochemistry. IL-1beta positive cells were observed at both time-intervals after LPS administration in the choroid plexus, meninges, circumventricular organs and pituitary gland of both sham-operated and vagotomized rats. We conclude that under the conditions studied, the vagus nerve does not mediate LPS-induced appearance of IL-1beta in the rat brain and pituitary gland.


Subject(s)
Brain/metabolism , Interleukin-1/metabolism , Pituitary Gland/metabolism , Vagus Nerve/physiology , Animals , Brain/drug effects , Lipopolysaccharides/pharmacology , Male , Pituitary Gland/drug effects , Rats , Rats, Sprague-Dawley , Vagotomy , Vagus Nerve/drug effects
11.
Glia ; 30(2): 134-42, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10719355

ABSTRACT

The pro-inflammatory cytokines interleukin-1beta (IL-1beta), IL-6, tumor necrosis factor-alpha (TNF-alpha), and nitric oxide (NO) can be produced by activated glial cells and play a critical role in various neurological diseases. Using primary co-cultures of rat microglial and astroglial cells, we investigated the effects of the anti-inflammatory cytokines transforming growth factor-beta1 (TGF-beta1)/beta2, IL-4, and IL-10 on the production of (pro-) inflammatory mediators after stimulation of the cells with lipopolysaccharide (LPS; 0.1 micrograms/ml, 24 h). IL-10 (10 and 100 ng/ml) and IL-4 (5 and 50 U/ml) suppressed the LPS-induced production of NO, IL-6, and TNF-alpha in a dose-dependent manner, whereas TGF-beta1/beta2 (2 and 20 ng/ml) only suppressed NO production. LPS-induced levels of IL-1beta were suppressed by IL-10, but not by IL-4 and TGF-beta1/beta2. Conversely, co-incubation of the glial cells with LPS and antibodies to TGF-beta1/beta2 selectively enhanced LPS-induced NO production, whereas co-incubation with antibody to IL-10 enhanced LPS-induced production of all pro-inflammatory cytokines and NO. This finding strongly suggests that effective concentrations of TGF-beta1/beta2 and IL-10 are produced by LPS-stimulated glial cell co-cultures. Production of IL-10 in these co-cultures was confirmed by measurement of rat IL-10 by radioimmunoassay. We conclude that anti-inflammatory cytokines affect the production of inflammatory mediators in LPS-activated co-cultures of microglial and astroglial cells differentially.


Subject(s)
Astrocytes/metabolism , Cytokines/biosynthesis , Interleukin-10/pharmacology , Interleukin-4/pharmacology , Microglia/metabolism , Nitric Oxide/biosynthesis , Transforming Growth Factor beta/pharmacology , Animals , Antibodies/pharmacology , Astrocytes/cytology , Astrocytes/drug effects , Cells, Cultured , Interleukin-1/biosynthesis , Interleukin-1/immunology , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-4/immunology , Interleukin-4/metabolism , Interleukin-6/biosynthesis , Interleukin-6/immunology , Lipopolysaccharides , Microglia/cytology , Microglia/drug effects , Neutralization Tests , Nitrites/metabolism , Rats , Rats, Wistar , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
12.
Auton Neurosci ; 85(1-3): 133-40, 2000 Dec 20.
Article in English | MEDLINE | ID: mdl-11189020

ABSTRACT

Vagotomy suppresses fever and hyperalgesia caused by intraperitoneal lipopolysaccharide (LPS) but has little effect on the febrile response to intravenous or intramuscular LPS. This suggests that some vagus-mediated mechanisms are recruited only when LPS is administered via the intraperitoneal route. We hypothesized that such mechanisms are associated with LPS transport from the peritoneal cavity to the circulation. Adult Wistar rats underwent total subdiaphragmatic, bilateral selective celiac, or sham vagotomy. On day 28-32 after surgery, they were injected IP with Escherichia coli LPS (5, 20, or 100 microg/kg) or saline and decapitated 90 min thereafter. Their plasma levels of LPS and their plasma interleukin-6, adrenocorticotropin, and corticosterone responses to LPS were measured. Success of intraperitoneal administration of LPS was verified by increased interleukin-1beta and interleukin-6 concentrations in the peritoneal lavage fluid. Effectiveness of vagotomies was confirmed by increased stomach mass (food retention) and pancreas mass (hypertrophy). In the shams, LPS caused a dose-dependent endotoxemia and increased plasma levels of interleukin-6, adrenocorticotropin, and corticosterone. Neither celiac nor total vagotomy affected any of these responses. LPS escapes from the peritoneal cavity by two primary routes, viz., the hematogenous (via the portal vein) and lymphogenous (via the lymphatic system). The design of the present study did not allow for evaluating the rapid, hematogenous transport. The results obtained suggest that the abdominal vagus does not control the slow. lymphogenous escape of LPS from the peritoneal cavity.


Subject(s)
Lipopolysaccharides/pharmacokinetics , Peritoneal Cavity , Peritoneum/metabolism , Vagus Nerve/physiology , Adrenocorticotropic Hormone/blood , Animals , Ascitic Fluid/chemistry , Ascitic Fluid/metabolism , Corticosterone/blood , Diaphragm , Dose-Response Relationship, Drug , Eating/physiology , Fever/chemically induced , Fever/immunology , Fever/metabolism , Hypertrophy , Interleukin-1/analysis , Interleukin-6/analysis , Interleukin-6/blood , Lipopolysaccharides/blood , Male , Pancreas/pathology , Peritoneum/blood supply , Peritoneum/innervation , Rats , Rats, Wistar , Stomach/physiology , Vagotomy/methods , Vagus Nerve/surgery
13.
Neuroscience ; 93(3): 915-30, 1999.
Article in English | MEDLINE | ID: mdl-10473257

ABSTRACT

The temporal and anatomical distribution of members of the interleukin-1 system in the rat brain following intraperitoneal kainic acid administration was studied in relation to neurodegeneration as detected with in situ end labelling. Kainic acid administration (10 mg/kg, i.p.) resulted in the induced expression of interleukin-1beta, interleukin- receptor antagonist and caspase-1p10 immunoreactivity in areas known to display neuronal and tissue damage upon excitotoxic lesions. The induction of these proteins was transient. Interleukin-1 immunoreactivity appeared at 5 h, and the interleukin-1 receptor antagonist-immunoreactive cells were first detected at 12 h, whereas the induction of caspase- 1p10 expression was first detected 24 h after kainic acid injection. Double labelling with the microglial marker Ox42 confirmed that both interleukin-1beta and interleukin-1 receptor antagonist were mainly localized in microglial cells. The regional distribution of in situ end-labelled neurons was similar to the distribution of cells expressing interleukin-1beta and interleukin-1 receptor antagonist, whereas the distribution of caspase-1 was more limited. The in situ end-labelled neurons, were, similarly to the interleukin-1beta-positive cells, first detected at 5 h, which is earlier than the induction of caspase-1. Our results show that the induction of IL-1beta and IL-1 receptor antagonist proteins after kainic acid are closely associated with the temporal as well as the anatomical distribution of in situ end-labelled neurons, whereas the induction of caspase-1 protein exhibited a delayed temporal profile and limited distribution. Since cytokine production occurs in activated microglial cells, the inflammatory component seems to be a strong mediator of this type of excitotoxic damage. The late onset of the caspase-1 expression would seem to indicate that this enzyme has no fundamental role in directly causing neuronal cell death induced by systemic kainic acid.


Subject(s)
Brain Chemistry/drug effects , Caspase 1/analysis , Excitatory Amino Acid Agonists/pharmacology , Interleukin-1/analysis , Kainic Acid/pharmacology , Nerve Degeneration/metabolism , Nerve Tissue Proteins/analysis , Sialoglycoproteins/analysis , Animals , Apoptosis/drug effects , Caspase 1/biosynthesis , Caspase 1/genetics , Enzyme Induction , Gene Expression Regulation/drug effects , Interleukin 1 Receptor Antagonist Protein , Interleukin-1/biosynthesis , Interleukin-1/genetics , Male , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurons/chemistry , Neurons/pathology , Rats , Rats, Sprague-Dawley , Sialoglycoproteins/biosynthesis , Sialoglycoproteins/genetics
14.
AIDS ; 13(3): 317-26, 1999 Feb 25.
Article in English | MEDLINE | ID: mdl-10199221

ABSTRACT

OBJECTIVES: To determine the occurrence and cellular localization of inducible nitric oxide synthase (iNOS), NOS activity and its association with cell death in brains of AIDS and AIDS dementia complex (ADC) patients. DESIGN AND METHODS: Post-mortem cerebral cortex tissue of eight AIDS patients, eight ADC patients and eight control subjects was processed for iNOS immunocytochemistry, NADPH-diaphorase activity staining as an index of NOS activity, and in situ end-labelling to detect cell death. RESULTS: iNOS-positive cells were present in the white matter of 14 out of 16 AIDS and ADC patients, whereas two out of eight control subjects showed iNOS-positive cells. iNOS immunoreactivity was exclusively localized in activated macrophages and microglial cells that both showed NADPH-diaphorase activity. In addition, NADPH-diaphorase activity, not related to iNOS immunoreactivity, was observed in astrocytes in both white and grey matter of AIDS and ADC patients. All AIDS and ADC patients, and only one control subject showed characteristic features of apoptotic cell death. CONCLUSIONS: Different forms of NOS are present in microglial cells and astrocytes of AIDS and ADC patients but are largely absent in control subjects. Although more NOS-expressing cells occur in ADC than in AIDS patients, apoptotic cell death was found in both patient groups to the same extent. We postulate that NO production in brains of AIDS patients results in cumulative cortical cell loss, which becomes neurologically evident at later stages of disease and is expressed as ADC.


Subject(s)
AIDS Dementia Complex/physiopathology , Acquired Immunodeficiency Syndrome/physiopathology , Apoptosis , Cerebral Cortex/pathology , Nitric Oxide Synthase/metabolism , AIDS Dementia Complex/pathology , Acquired Immunodeficiency Syndrome/pathology , Adult , Aged , Antibodies, Monoclonal , Autopsy , Cell Death , Cerebral Cortex/virology , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Middle Aged , NADPH Dehydrogenase
15.
Neuroscience ; 89(1): 137-47, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10051223

ABSTRACT

Interleukin-1 receptor type I and interleukin-1 receptor antagonist were found in magnocellular neurons of the paraventricular and supraoptic nuclei of the rat hypothalamus by immunohistochemical detection. Double-labelling experiments revealed that both proteins occurred in vasopressin-containing neurons. A similar distribution pattern was observed in a group of vasopressin-positive accessory magnocellular neurons. Axons emanating from the interleukin-1 receptor type I- and interleukin-1 receptor antagonist-immunoreactive neuronal cell bodies could be seen within the hypothalamic nuclei, and varicosities expressing interleukin-1 receptor antagonist immunoreactivity were observed in the internal zone of the median eminence, as well as in the hypothalamo-pituitary projection. The co-localization of interleukin-1 receptor type I with vasopressin is in agreement with findings that interleukin-1 has a stimulatory effect on vasopressin synthesis and release. The hypothalamic neurons may serve as a source of interleukin-1 receptor antagonist to balance the effects of interleukin-1.


Subject(s)
Neurons/physiology , Paraventricular Hypothalamic Nucleus/cytology , Receptors, Interleukin-1/physiology , Supraoptic Nucleus/cytology , Vasopressins/physiology , Animals , Antibodies, Monoclonal , Blotting, Western , Corticotropin-Releasing Hormone/physiology , Fluorescent Antibody Technique , Immunoenzyme Techniques , Male , Neurons/chemistry , Paraventricular Hypothalamic Nucleus/chemistry , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-1/analysis , Receptors, Interleukin-1/immunology , Receptors, Interleukin-1 Type I , Supraoptic Nucleus/chemistry
16.
Am J Physiol ; 276(3): R652-8, 1999 03.
Article in English | MEDLINE | ID: mdl-10070124

ABSTRACT

Interleukin (IL)-6 has been proposed to mediate several sickness responses, including brain-mediated neuroendocrine, temperature, and behavioral changes. However, the exact mechanisms and sites of action of IL-6 are still poorly understood. In the present study, we describe the effects of central administration of species-homologous recombinant rat IL-6 (rrIL-6) on the induction of hypothalamic-pituitary-adrenal (HPA) activity, fever, social investigatory behavior, and immobility. After intracerebroventricular administration of rrIL-6 (50 or 100 ng/rat), rats demonstrated HPA and febrile responses. In contrast, rrIL-6 alone did not induce changes in social investigatory and locomotor behavior at doses of up to 400 ng/rat. Coadministration of rrIL-6 (100 ng/rat) and rrIL-1beta (40 ng/rat), which alone did not affect the behavioral responses, reduced social investigatory behavior and increased the duration of immobility. Compared with rhIL-6, intracerebroventricular administration of rrIL-6 (100 ng/rat) induced higher HPA responses and early-phase febrile responses. This is consistent with a higher potency of rrIL-6, compared with rhIL-6, in the murine B9 bioassay. We conclude that species-homologous rrIL-6 alone can act in the brain to induce HPA and febrile responses, whereas it only reduces social investigatory behavior and locomotor activity in the presence of IL-1beta.


Subject(s)
Behavior, Animal/drug effects , Fever/chemically induced , Hypothalamo-Hypophyseal System/physiology , Interleukin-6/pharmacology , Pituitary-Adrenal System/physiology , Adrenocorticotropic Hormone/blood , Animals , Body Temperature/drug effects , Corticosterone/blood , Humans , Hypothalamo-Hypophyseal System/drug effects , Injections, Intraventricular , Male , Motor Activity/drug effects , Pituitary-Adrenal System/drug effects , Rats , Rats, Wistar , Recombinant Proteins , Social Behavior
17.
J Neurosci Res ; 54(6): 820-7, 1998 Dec 15.
Article in English | MEDLINE | ID: mdl-9856866

ABSTRACT

Injection of Fluoro-Gold (FG) into the whisker pad of rats yields stable retrograde labeling of facial motoneurons. Subsequent removal of 10 mm from all facial nerve branches permanently deprives the FG-labeled motoneurons from their targets and the motoneurons gradually die. Neuronal debris is phagocytized by two types of neuronophages: parenchymal microglia (monoclonal antibody [MAb] OX42-positive, MAb ED2-negative) and perivascular phagocytes (OX42-negative, ED2-positive). Because both types of neuronophages express major histocompatibility complex (MHC) class II glycoproteins (MAb OX6-positive), they are considered to be the potential antigen-presenting cells of the brain. To check this hypothesis, we tested whether both types of neuronophages also synthetize the co-stimulatory cytokine interleukin-1beta (IL-1beta) immunocytochemically visualized by MAbs SILK-5/6. Employing combined fluorescent visualization of antigens (OX6, ED2, and SILK-5/6) in sections containing fluorescent (FG-prelabeled) neuronophages, we found that, during slowly occurring neuronal loss, the vast majority of IL-1beta immunoreactive neuronophages were of perivascular (ED2-positive) origin. We concluded that, during delayed neuronal death "behind" an intact blood-brain barrier, the perivascular phagocytes were more likely to function as antigen-presenting cells than the parenchymal microglia.


Subject(s)
Facial Nerve/immunology , Histocompatibility Antigens Class II/metabolism , Interleukin-1/biosynthesis , Oligodendroglia/immunology , Phagocytosis , Animals , Antigen-Presenting Cells , Blood-Brain Barrier , Facial Nerve Injuries , Female , Oligodendroglia/metabolism , Oligodendroglia/ultrastructure , Rats , Rats, Wistar
18.
Eur Cytokine Netw ; 9(3): 279-88, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9831177

ABSTRACT

IL-1alpha and IL-1beta have potent effects on the central nervous system resulting in fever, activation of the hypothalamic-pituitary-adrenal axis and behavioural depression. These effects have mainly been studied in rats, using recombinant human and mouse IL-1. Because IL-1alpha and IL-1beta show some species specificity in the potency of their biological activities, the objective of the present work was to directly compare the effects of recombinant rat IL-1alpha and IL-1beta in the rat system as a first step to dissect out the mechanisms that are involved in these effects. In vitro, recombinant rat IL-1alpha and IL-1beta bound with the same affinity as human IL-1 to the rat insulinoma Rin m5F cell line that mainly expresses type I IL-1 receptors. This binding activated IL-1 receptors, as shown by induction of the synthesis of TNF-alpha mRNA. In vivo, recombinant rat IL-1alpha and IL-1beta enhanced body temperature, increased plasma levels of corticosterone and ACTH, and depressed social behaviour. All these effects were obtained at doses 100-1,000 fold lower when IL-1 was injected centrally than when it was administered peripherally, indicating that they are centrally mediated. The relative potencies of recombinant rat IL-1alpha and IL-1beta were not the same depending on the endpoint and the route of injection, indicating that different mechanisms are likely to be involved in the various effects of IL-1 on the brain.


Subject(s)
Body Temperature/drug effects , Brain/drug effects , Cerebral Ventricles/physiology , Interleukin-1/pharmacology , Animals , Brain/physiology , Cerebral Ventricles/drug effects , Cloning, Molecular , Escherichia coli , Exploratory Behavior/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiology , Injections, Intraventricular , Insulinoma , Interleukin-1/administration & dosage , Interleukin-1/metabolism , Male , Mice , Pancreatic Neoplasms , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/physiology , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-1/physiology , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Social Behavior , Transcription, Genetic/drug effects , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...